1
|
Luo W, Zhang C, Dong L. Rhodium(III)-Catalyzed Annulation Synthesis of Difluorinated Quinazolinone Derivatives Using an Amide Carbonyl as the Directing Group. J Org Chem 2024; 89:9627-9640. [PMID: 38888955 DOI: 10.1021/acs.joc.3c02596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/20/2024]
Abstract
The use of amide carbonyl groups of substrates as weakly coordinating directing groups has received a significant amount of attention. Recently, difluoromethylene alkynes have been successfully used in fluorination reactions, resulting in the preparation of various fluorine-containing compounds. This work describes a [4+2] annulation method for creating a range of fluorinated quinolino[2,1-b]quinazolinone derivatives. The derivatives are formed through Rh(III)-catalyzed cascade cyclization of 3-phenylquinazolinones and gem-difluoromethylene alkynes.
Collapse
Affiliation(s)
- Wen Luo
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Chao Zhang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Lin Dong
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| |
Collapse
|
2
|
Raji Reddy C, Neeliveettil A, Ajaykumar U, Punna N, Neuville L, Masson G. Access to N-Fused Quinazolinones by Radical-Promoted Cascade Annulations of Alkenyl N-Cyanamides with Aromatic Aldehydes. J Org Chem 2024; 89:7115-7124. [PMID: 38691342 DOI: 10.1021/acs.joc.4c00494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2024]
Abstract
A cascade radical cyclization of alkenyl N-cyanamides with aromatic aldehydes has been achieved for an expeditious synthesis of keto-methylated dihydropyrrolo-quinazolinones. Benzoyl radicals, generated from aryl aldehydes in the presence of di-tert-butyl peroxide (DTBP), promoted the domino annulations leading to distinctive functionalized quinazolinones in good yields. In addition, the robustness of the present protocol is validated by employing heterocyclic and natural product-based aldehydes.
Collapse
Affiliation(s)
- Chada Raji Reddy
- Department of Organic Synthesis & Process Chemistry CSIR-Indian Institute of Chemical Technology (CSIR-IICT), Hyderabad 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Anootha Neeliveettil
- Department of Organic Synthesis & Process Chemistry CSIR-Indian Institute of Chemical Technology (CSIR-IICT), Hyderabad 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Uprety Ajaykumar
- Department of Organic Synthesis & Process Chemistry CSIR-Indian Institute of Chemical Technology (CSIR-IICT), Hyderabad 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Nagender Punna
- Department of Organic Synthesis & Process Chemistry CSIR-Indian Institute of Chemical Technology (CSIR-IICT), Hyderabad 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Luc Neuville
- Institut de Chimie des Substances Naturelles (ICSN), CNRS UPR 2301, Université Paris-Saclay, 1 avenue de la Terrasse, 91198 Gif-sur-Yvette Cedex, France
| | - Geraldine Masson
- Institut de Chimie des Substances Naturelles (ICSN), CNRS UPR 2301, Université Paris-Saclay, 1 avenue de la Terrasse, 91198 Gif-sur-Yvette Cedex, France
| |
Collapse
|
3
|
Huang FP, Qin WJ, Pan XY, Yang K, Wang K, Teng QH. Visible-Light-Induced Chemodivergent Synthesis of Tetracyclic Quinazolinones and 3-Iminoisoindoliones via the Substrate Control Strategy. J Org Chem 2024; 89:4395-4405. [PMID: 38501298 DOI: 10.1021/acs.joc.3c02501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2024]
Abstract
A visible-light-induced chemodivergent synthesis of tetracyclic quinazolinones and 3-iminoisoindoliones has been developed. This chemodivergent reaction afforded two kinds of different products by substrate control. A detailed investigation of the reaction mechanism revealed that this consecutive photoinduced electron transfer (ConPET) cascade cyclization involved a radical process, and the aryl radical was the crucial intermediate. This method employed 4-DPAIPN as a photocatalyst and i-Pr2NEt as a sacrificial electron donor leading to metal-free conditions.
Collapse
Affiliation(s)
- Fang-Ping Huang
- Guangxi Key Laboratory of Electrochemical and Magnetochemical Functional Materials, College of Chemistry and Bioengineering, Guilin University of Technology, Guilin 541004, China
| | - Wen-Jian Qin
- Guangxi Key Laboratory of Electrochemical and Magnetochemical Functional Materials, College of Chemistry and Bioengineering, Guilin University of Technology, Guilin 541004, China
| | - Xin-Yao Pan
- Guangxi Key Laboratory of Electrochemical and Magnetochemical Functional Materials, College of Chemistry and Bioengineering, Guilin University of Technology, Guilin 541004, China
| | - Kun Yang
- Guangxi Key Laboratory of Electrochemical and Magnetochemical Functional Materials, College of Chemistry and Bioengineering, Guilin University of Technology, Guilin 541004, China
| | - Kai Wang
- Guangxi Key Laboratory of Electrochemical and Magnetochemical Functional Materials, College of Chemistry and Bioengineering, Guilin University of Technology, Guilin 541004, China
| | - Qing-Hu Teng
- Guangxi Key Laboratory of Electrochemical and Magnetochemical Functional Materials, College of Chemistry and Bioengineering, Guilin University of Technology, Guilin 541004, China
| |
Collapse
|
4
|
Kumar V, Jangid K, Kumar N, Kumar V, Kumar V. 3D-QSAR-based pharmacophore modelling of quinazoline derivatives for the identification of acetylcholinesterase inhibitors through virtual screening, molecular docking, molecular dynamics and DFT studies. J Biomol Struct Dyn 2024:1-15. [PMID: 38329085 DOI: 10.1080/07391102.2024.2313157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 08/12/2023] [Indexed: 02/09/2024]
Abstract
Alzheimer's disease (AD) is a progressive neurological disorder responsible for the cognitive dysfunction and cognitive impairment in the patients. Acetylcholinesterase inhibitors (AChEIs) are used to treat AD however, these only provided symptomatic relief and more efficient drug molecules are desired for the effective treatment of the disease. In this article, ligand-based drug-designing strategy was used to develop and validate a field-based 3D-QSAR pharmacophore model on quinazoline-based AChEIs reported in the literature. The validated pharmacophore model (AAAHR_1) was used as a prefilter to screen an ASINEX database via virtual screening workflow (VSW). The hits generated were subjected to MM-GBSA to identify potential AChEIs and top three scoring molecules (BAS 05264565, LEG 12727144 and SYN 22339886) were evaluated for thermodynamic stability at the target site using molecular dynamic simulations. Additionally, DFT study was performed to predict the reactivity of lead molecules towards acetylcholinesterase (AChE). Thus, by utilising various computational tools, three molecules were identified as potent AChEIs that can be developed as potential drug candidates for the treatment of AD.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Vijay Kumar
- Department of Chemistry, Laboratory of Organic and Medicinal Chemistry, Central University of Punjab, Bathinda, India
| | - Kailash Jangid
- Department of Chemistry, Laboratory of Organic and Medicinal Chemistry, Central University of Punjab, Bathinda, India
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, Bathinda, India
| | - Naveen Kumar
- Department of Chemistry, Laboratory of Organic and Medicinal Chemistry, Central University of Punjab, Bathinda, India
| | - Vinay Kumar
- Department of Chemistry, Laboratory of Organic and Medicinal Chemistry, Central University of Punjab, Bathinda, India
| | - Vinod Kumar
- Department of Chemistry, Laboratory of Organic and Medicinal Chemistry, Central University of Punjab, Bathinda, India
| |
Collapse
|
5
|
Moftah HK, Mousa MHA, Elrazaz EZ, Kamel AS, Lasheen DS, Georgey HH. Novel quinazolinone Derivatives: Design, synthesis and in vivo evaluation as potential agents targeting Alzheimer disease. Bioorg Chem 2024; 143:107065. [PMID: 38150939 DOI: 10.1016/j.bioorg.2023.107065] [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: 10/19/2023] [Revised: 12/02/2023] [Accepted: 12/23/2023] [Indexed: 12/29/2023]
Abstract
Since Alzheimer disease is one of the most prevalent types of dementia with a high mortality and disability rate, so development of multi-target drugs becomes the major strategy for battling AD. This study shows the development of a series of quinazolinone based derivatives as novel, multifunctional anti-AD drugs that exhibit both cholinesterase inhibitoryand anti-inflammatory properties. The preliminary results of the in vitro AChE inhibition activity showed that compounds 4b, 5a, 6f, 6h and 7b were better represented for further evaluation. Furthermore, in-vivo AChE inhibition activity and behavior Morris water maze test against donepezil as reference drug were evaluated. Additionally, hippocampal inflammatory markers; TNF-α, NFĸB, IL-1β and IL-6 and antioxidant markers; SOD and MDA were assessed to evaluate the efficacy of quinazolinone derivatives against AD hallmarks. The results showed that 6f, 6h and 7b have promising anti-acetylcholinesterase, anti-inflammatory and antioxidant activities thus, have a significant effect in treatment of AD. Moreover, Histopathological examination revealed that 6f, 6h and 7b derivatives have neuroprotective effect against neuronal damage caused by induced scopolamine model in mice. Finally, the binding ability of the synthesized derivatives to the target, AChE was investigated through molecular docking which reflected significant interactions to the target based on their docking binding scores. Hence, the newly designed quinazolinone derivatives possess promising anti-acetylcholinesterase activity and challenging for the management of AD in the future.
Collapse
Affiliation(s)
- Hadeer K Moftah
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Drug Technology, Egyptian Chinese University, Gesr El Suez st, PO 11786, Cairo, Egypt
| | - Mai H A Mousa
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Drug Technology, Egyptian Chinese University, Gesr El Suez st, PO 11786, Cairo, Egypt
| | - Eman Z Elrazaz
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Ain Shams University, African Union Organization Street, Cairo 11566, Egypt
| | - Ahmed S Kamel
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, Cairo P.O. Box 11562, Egypt; Department of Pharmacology and Toxicology, Faculty of Pharmacy and Drug Technology, Egyptian Chinese University, Gesr El Suez st, PO 11786 Cairo, Egypt
| | - Deena S Lasheen
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Ain Shams University, African Union Organization Street, Cairo 11566, Egypt
| | - Hanan H Georgey
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, Cairo P.O. Box 11562, Egypt.
| |
Collapse
|
6
|
Ko N, Min S, Moon K, Shin H, Kwon NY, Mishra NK, Rakshit A, Singh P, Kim IS. Catalyst-Controlled C-H Allylation and Annulation of 2-Aryl Quinazolinones with 2-Methylidene Cyclic Carbonate. J Org Chem 2023; 88:13315-13326. [PMID: 37668242 DOI: 10.1021/acs.joc.3c01652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/06/2023]
Abstract
The site-selective modification of quinazolinone as a privileged bicyclic N-heterocycle is an attractive topic in medicinal chemistry and material science. We herein report the ruthenium(II)-catalyzed C-H allylation of 2-aryl quinazolinones with 2-methylidene cyclic carbonate. In addition, tandem C-H allylation and annulation are achieved under rhodium(III) catalysis, resulting in the formation of tetracyclic quinazolinones including a tertiary carbon center. Post-transformations of the synthesized products demonstrate the potential of the developed methodology. A series of mechanistic investigations were also performed.
Collapse
Affiliation(s)
- Nayoung Ko
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Sujin Min
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Kyeongwon Moon
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Hyungjin Shin
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Na Yeon Kwon
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | | | - Amitava Rakshit
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Pargat Singh
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - In Su Kim
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| |
Collapse
|
7
|
Hatami M, Basri Z, Sakhvidi BK, Mortazavi M. Thiadiazole – A promising structure in design and development of anti-Alzheimer agents. Int Immunopharmacol 2023; 118:110027. [PMID: 37011500 DOI: 10.1016/j.intimp.2023.110027] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 02/28/2023] [Accepted: 03/09/2023] [Indexed: 04/03/2023]
Abstract
The design and development of effective multitargeted agents in treating Alzheimer disease (AD) has always been a hot topic in the field of drug discovery. Since AD is a multifactorial disorder, various key hidden players such as deficit of acetylcholine (ACh), tau-protein aggregation, and oxidative stress have been associated with the incidence and progress of AD. In pursuit of improving efficacy and expanding the range of pharmacological activities of current AD drugs, the molecular hybridization method is also used intensively. Five-membered heterocyclic systems such as thiadiazole scaffolds have previously been shown to have therapeutic activity. Thiadiazole analogs as an anti-oxidant compound have been known to include a wide range of biological activity from anti-cancer to anti-Alzheimer properties. The suitable pharmacokinetic and physicochemical properties of the thiadiazole scaffold have introduced it as a therapeutic target in medicinal chemistry. The current review portrays the critical role of the thiadiazole scaffold in the design of various compounds with potential effects in the treatment of Alzheimer's disease. Furthermore, the rationale used behind hybrid-based design strategies and the outcomes achieved through the hybridization of Thiadiazole analogs with various core structures have been discussed. In addition, the data in the present review may help researchers in the design of new multidrug combinations that may provide new options for the treatment of AD.
Collapse
Affiliation(s)
- Maryam Hatami
- Department of Biotechnology, Institute of Science and High Technology and Environmental Sciences, Graduate University of Advanced Technology, Kerman, Iran
| | - Zahra Basri
- Department of Biotechnology, Institute of Science and High Technology and Environmental Sciences, Graduate University of Advanced Technology, Kerman, Iran
| | - Batool Khani Sakhvidi
- Department of Biotechnology, Institute of Science and High Technology and Environmental Sciences, Graduate University of Advanced Technology, Kerman, Iran
| | - Mojtaba Mortazavi
- Department of Biotechnology, Institute of Science and High Technology and Environmental Sciences, Graduate University of Advanced Technology, Kerman, Iran.
| |
Collapse
|
8
|
Zhang Z, Wang J, Guo S, Fan J, Fan X. t-BuOK-Catalyzed Regio- and Stereoselective Intramolecular Hydroamination Reaction Leading to Phthalazinoquinazolinone Derivatives. J Org Chem 2023; 88:1282-1291. [PMID: 36594406 DOI: 10.1021/acs.joc.2c02638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
We report herein an efficient and practical strategy for the preparation of 5H-phthalazino[1,2-b]quinazolin-8(6H)-one derivatives through a t-BuOK-catalyzed intramolecular hydroamination reaction of functionalized quinazolinones under extremely mild reaction conditions. A variety of quinazolinone substrates are well tolerated to furnish the corresponding products in good to high yields via an exclusive 6-exo-dig cyclization process. The present protocol has the advantages of readily obtainable starting materials, broad substrate scope, and high regio- and stereoselectivity.
Collapse
Affiliation(s)
- Ziyi Zhang
- NMPA Key Laboratory for Research and Evaluation of Innovative Drug, Key Laboratory of Green Chemical Media and Reactions, Ministry of Education, Collaborative Innovation Center of Henan Province for Green Manufacturing of Fine Chemicals, School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang 453007, Henan, China
| | - Jin Wang
- School of Environment, Key Laboratory for Yellow River and Huai River Water Environment and Pollution Control, Ministry of Education, Henan Normal University, Xinxiang 453007, Henan, China
| | - Shenghai Guo
- NMPA Key Laboratory for Research and Evaluation of Innovative Drug, Key Laboratory of Green Chemical Media and Reactions, Ministry of Education, Collaborative Innovation Center of Henan Province for Green Manufacturing of Fine Chemicals, School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang 453007, Henan, China
| | - Jing Fan
- School of Environment, Key Laboratory for Yellow River and Huai River Water Environment and Pollution Control, Ministry of Education, Henan Normal University, Xinxiang 453007, Henan, China
| | - Xuesen Fan
- NMPA Key Laboratory for Research and Evaluation of Innovative Drug, Key Laboratory of Green Chemical Media and Reactions, Ministry of Education, Collaborative Innovation Center of Henan Province for Green Manufacturing of Fine Chemicals, School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang 453007, Henan, China
| |
Collapse
|
9
|
Pang B, Wang Y, Hao L, Wu G, Ma Z, Ji Y. Tandem C-C/C-N Bond Formation via Rh(III)-Catalyzed α-Fluoroalkenylation and Sequential Annulation of 2-Arylquinazolinones and gem-Difluorostyrenes. J Org Chem 2023; 88:143-153. [PMID: 36563294 DOI: 10.1021/acs.joc.2c02006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
An efficient method of Rh(III)-catalyzed coupling reaction between 2-arylquinazolinones and gem-difluorostyrenes has been developed. In this work, two diverse structures of monofluoroalkenes and isoindolo[1,2-b]quinazolin-10(12H)-one derivatives were respectively synthesized by controlling the amount of additives (Ca(OH)2 and AgNTf2) to achieve controlled stepwise breaking of the C-F bonds of gem-difluorostyrenes. This reaction has the characteristics of a wide range of substrates and good functional group tolerance. Meanwhile, several control experiments were conducted and a plausible mechanism was proposed.
Collapse
Affiliation(s)
- Binghan Pang
- Engineering Research Centre of Pharmaceutical Process Chemistry, Ministry of Education, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, P. R. China
| | - Yangyang Wang
- Engineering Research Centre of Pharmaceutical Process Chemistry, Ministry of Education, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, P. R. China
| | - Liqiang Hao
- Engineering Research Centre of Pharmaceutical Process Chemistry, Ministry of Education, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, P. R. China
| | - Gaorong Wu
- Engineering Research Centre of Pharmaceutical Process Chemistry, Ministry of Education, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, P. R. China
| | - Zhihong Ma
- Biotalk Co., LTD, Shanghai 200092, China
| | - Yafei Ji
- Engineering Research Centre of Pharmaceutical Process Chemistry, Ministry of Education, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, P. R. China
| |
Collapse
|
10
|
Access to new phosphonate- and imidazolidine-benzopyrimidinone derivatives as antityrosinase and anti-acetylcholinesterase agents: Design, synthesis and molecular docking. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.133693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
11
|
Stern N, Gacs A, Tátrai E, Flachner B, Hajdú I, Dobi K, Bágyi I, Dormán G, Lőrincz Z, Cseh S, Kígyós A, Tóvári J, Goldblum A. Dual Inhibitors of AChE and BACE-1 for Reducing Aβ in Alzheimer's Disease: From In Silico to In Vivo. Int J Mol Sci 2022; 23:13098. [PMID: 36361906 PMCID: PMC9655245 DOI: 10.3390/ijms232113098] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 10/03/2022] [Accepted: 10/05/2022] [Indexed: 07/30/2023] Open
Abstract
Alzheimer's disease (AD) is a complex and widespread condition, still not fully understood and with no cure yet. Amyloid beta (Aβ) peptide is suspected to be a major cause of AD, and therefore, simultaneously blocking its formation and aggregation by inhibition of the enzymes BACE-1 (β-secretase) and AChE (acetylcholinesterase) by a single inhibitor may be an effective therapeutic approach, as compared to blocking one of these targets or by combining two drugs, one for each of these targets. We used our ISE algorithm to model each of the AChE peripheral site inhibitors and BACE-1 inhibitors, on the basis of published data, and constructed classification models for each. Subsequently, we screened large molecular databases with both models. Top scored molecules were docked into AChE and BACE-1 crystal structures, and 36 Molecules with the best weighted scores (based on ISE indexes and docking results) were sent for inhibition studies on the two enzymes. Two of them inhibited both AChE (IC50 between 4-7 μM) and BACE-1 (IC50 between 50-65 μM). Two additional molecules inhibited only AChE, and another two molecules inhibited only BACE-1. Preliminary testing of inhibition by F681-0222 (molecule 2) on APPswe/PS1dE9 transgenic mice shows a reduction in brain tissue of soluble Aβ42.
Collapse
Affiliation(s)
- Noa Stern
- Molecular Modeling and Drug Design Lab, Institute for Drug Research, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel
| | - Alexandra Gacs
- Department of Experimental Pharmacology, National Institute of Oncology, H-1122 Budapest, Hungary
| | - Enikő Tátrai
- Department of Experimental Pharmacology, National Institute of Oncology, H-1122 Budapest, Hungary
- KINETO Lab Ltd., H-1032 Budapest, Hungary
| | | | - István Hajdú
- TargetEx Ltd., H-2120 Dunakeszi, Hungary
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, H-1117 Budapest, Hungary
| | | | | | | | | | | | | | - József Tóvári
- KINETO Lab Ltd., H-1032 Budapest, Hungary
- Department of Tumor Biology, National Korányi Institute of TB and Pulmonology, H-1121 Budapest, Hungary
| | - Amiram Goldblum
- Molecular Modeling and Drug Design Lab, Institute for Drug Research, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel
| |
Collapse
|
12
|
Patel S, Bansoad AV, Singh R, Khatik GL. BACE1: A Key Regulator in Alzheimer's Disease Progression and Current Development of its Inhibitors. Curr Neuropharmacol 2022; 20:1174-1193. [PMID: 34852746 PMCID: PMC9886827 DOI: 10.2174/1570159x19666211201094031] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 11/26/2021] [Accepted: 11/28/2021] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is a chronic neurodegenerative disease with no specific disease-modifying treatment. β-secretase (BACE1) is considered the potential and rationale target because it is involved in the rate-limiting step, which produces toxic Aβ42 peptides that leads to deposits in the form of amyloid plaques extracellularly, resulting in AD. OBJECTIVE This study aims to discuss the role and implications of BACE1 and its inhibitors in the management of AD. METHODS We have searched and collected the relevant quality work from PubMed using the following keywords "BACE1", BACE2", "inhibitors", and "Alzheimer's disease". In addition, we included the work which discusses the role of BACE1 in AD and the recent work on its inhibitors. RESULTS In this review, we have discussed the importance of BACE1 in regulating AD progression and the current development of BACE1 inhibitors. However, the development of a BACE1 inhibitor is very challenging due to the large active site of BACE1. Nevertheless, some of the BACE1 inhibitors have managed to enter advanced phases of clinical trials, such as MK-8931 (Verubecestat), E2609 (Elenbecestat), AZD3293 (Lanabecestat), and JNJ-54861911 (Atabecestat). This review also sheds light on the prospect of BACE1 inhibitors as the most effective therapeutic approach in delaying or preventing AD progression. CONCLUSION BACE1 is involved in the progression of AD. The current ongoing or failed clinical trials may help understand the role of BACE1 inhibition in regulating the Aβ load and cognitive status of AD patients.
Collapse
Affiliation(s)
| | - Ankush Vardhaman Bansoad
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research-Raebareli, New Transit Campus, Bijnor-Sisendi Road, Sarojini Nagar, Near CRPF Base Camp, Lucknow (Uttar Pradesh), 226002, India
| | - Rakesh Singh
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research-Raebareli, New Transit Campus, Bijnor-Sisendi Road, Sarojini Nagar, Near CRPF Base Camp, Lucknow (Uttar Pradesh), 226002, India
| | - Gopal L. Khatik
- Department of Medicinal Chemistry, ,Address correspondence to this author at the Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research- Raebareli, New Transit Campus, Bijnor-Sisendi Road, Sarojini Nagar, Near CRPF Base Camp, Lucknow, Uttar Pradesh, India, 226002; E-mail: ,
| |
Collapse
|
13
|
Huang X, An Z, Yu Y, Feng X, Wang Y. Synthesis and Evaluation of Novel Ferulic Amide Derivatives and the Treatment of Alzheimer's Disease. ChemistrySelect 2022. [DOI: 10.1002/slct.202200650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Xian‐Feng Huang
- School of Pharmacy & School of Medicine Changzhou University Changzhou Jiangsu, 213164 PR China
| | - Zhe An
- School of Pharmacy & School of Medicine Changzhou University Changzhou Jiangsu, 213164 PR China
| | - Ying‐Cong Yu
- School of Pharmacy & School of Medicine Changzhou University Changzhou Jiangsu, 213164 PR China
| | - Xiao‐Qing Feng
- School of Pharmacy & School of Medicine Changzhou University Changzhou Jiangsu, 213164 PR China
- School of Pharmacy & School of Medicine Changzhou University Changzhou Jiangsu, 213164 PR China
| | - Ya‐Jing Wang
- School of Pharmacy & School of Medicine Changzhou University Changzhou Jiangsu, 213164 PR China
| |
Collapse
|
14
|
Zeng Y, Nie L, Liu L, Niu C, Li Y, Bozorov K, Zhao J, Shen J, Aisa HA. Design, Synthesis, in vitro Evaluation of a New Pyrrolo[1,2‐
a
]thiazolo[5,4‐
d
]pyrimidinone Derivatives as Cholinesterase Inhibitors Against Alzheimer's Disease. J Heterocycl Chem 2022. [DOI: 10.1002/jhet.4452] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Yan Zeng
- State Key Laboratory Basis of Xinjiang Indigenous Medicinal Plants Resource Utilization Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences Urumqi China
- University of Chinese Academy of Sciences Beijing China
| | - Lifei Nie
- State Key Laboratory Basis of Xinjiang Indigenous Medicinal Plants Resource Utilization Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences Urumqi China
| | - Liu Liu
- State Key Laboratory Basis of Xinjiang Indigenous Medicinal Plants Resource Utilization Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences Urumqi China
| | - Chao Niu
- State Key Laboratory Basis of Xinjiang Indigenous Medicinal Plants Resource Utilization Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences Urumqi China
- University of Chinese Academy of Sciences Beijing China
| | - Yi Li
- State Key Laboratory Basis of Xinjiang Indigenous Medicinal Plants Resource Utilization Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences Urumqi China
- University of Chinese Academy of Sciences Beijing China
| | - Khurshed Bozorov
- State Key Laboratory Basis of Xinjiang Indigenous Medicinal Plants Resource Utilization Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences Urumqi China
- Faculty of Chemistry Samarkand State University Samarkand Uzbekistan
| | - Jiangyu Zhao
- State Key Laboratory Basis of Xinjiang Indigenous Medicinal Plants Resource Utilization Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences Urumqi China
- University of Chinese Academy of Sciences Beijing China
| | - Jingshan Shen
- State Key Laboratory Basis of Xinjiang Indigenous Medicinal Plants Resource Utilization Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences Urumqi China
- Shanghai Institute of Materia Medica Chinese Academy of Sciences Shanghai China
| | - Haji Akber Aisa
- State Key Laboratory Basis of Xinjiang Indigenous Medicinal Plants Resource Utilization Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences Urumqi China
- University of Chinese Academy of Sciences Beijing China
| |
Collapse
|
15
|
Haghighijoo Z, Zamani L, Moosavi F, Emami S. Therapeutic potential of quinazoline derivatives for Alzheimer's disease: A comprehensive review. Eur J Med Chem 2022; 227:113949. [PMID: 34742016 DOI: 10.1016/j.ejmech.2021.113949] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 10/02/2021] [Accepted: 10/22/2021] [Indexed: 12/24/2022]
Abstract
Quinazolines are considered as a promising class of bioactive heterocyclic compounds with broad properties. Particularly, the quinazoline scaffold has an impressive role in the design and synthesis of new CNS-active drugs. The drug-like properties and pharmacological characteristics of quinazoline could lead to different drugs with various targets. Among CNS disorders, Alzheimer's disease (AD) is a progressive neurodegenerative disorder with memory loss, cognitive decline and language dysfunction. AD is a complex and multifactorial disease therefore, the need for finding multi-target drugs against this devastative disease is urgent. A literature survey revealed that quinazoline derivatives have diverse therapeutic potential for AD as modulators/inhibitors of β-amyloid, tau protein, cholinesterases, monoamine oxidases, and phosphodiesterases as well as other protective effects. Thus, we describe here the most relevant and recent studies about anti-AD agents with quinazoline structure which can further aid the development and discovery of new anti-AD agents.
Collapse
Affiliation(s)
- Zahra Haghighijoo
- Department of Chemistry, University of Louisiana at Lafayette, Lafayette, LA, 70504, USA
| | - Leila Zamani
- Worcester Polytechnic Institute, 100 Institute Road, Worcester, MA, 01609, USA
| | - Fatemeh Moosavi
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Saeed Emami
- Department of Medicinal Chemistry and Pharmaceutical Sciences Research Center, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran.
| |
Collapse
|
16
|
Ly D, Nguyen TT, Tran CTH, Nguyen VPT, Nguyen KX, Pham PH, Le NTH, Nguyen TT, Phan NTS. Metal-Free Annulation of 2-Nitrobenzyl Alcohols and Tetrahydroisoquinolines toward the Divergent Synthesis of Quinazolinones and Quinazolinethiones. J Org Chem 2021; 87:103-113. [PMID: 34918926 DOI: 10.1021/acs.joc.1c02017] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
A simple metal-free method for the synthesis of quinazolinones from commercially available 2-nitrobenzyl alcohols and tetrahydroisoquinolines is developed. The reaction conditions were tolerant of an array of functionalities such as halogen, tertiary amine, protected alcohol, and ester groups. Under nearly identical conditions, quinazolinethiones were obtained in the presence of elemental sulfur and suitable mediators.
Collapse
Affiliation(s)
- Duc Ly
- Faculty of Chemical Engineering, Ho Chi Minh City University of Technology (HCMUT), 268 Ly Thuong Kiet Street, District 10, Ho Chi Minh City 700000, Vietnam.,Vietnam National University Ho Chi Minh City, Linh Trung Ward, Thu Duc District, Ho Chi Minh City 700000, Vietnam
| | - Thao T Nguyen
- Faculty of Chemical Engineering, Ho Chi Minh City University of Technology (HCMUT), 268 Ly Thuong Kiet Street, District 10, Ho Chi Minh City 700000, Vietnam.,Vietnam National University Ho Chi Minh City, Linh Trung Ward, Thu Duc District, Ho Chi Minh City 700000, Vietnam
| | - Cam T H Tran
- Faculty of Chemical Engineering, Ho Chi Minh City University of Technology (HCMUT), 268 Ly Thuong Kiet Street, District 10, Ho Chi Minh City 700000, Vietnam.,Vietnam National University Ho Chi Minh City, Linh Trung Ward, Thu Duc District, Ho Chi Minh City 700000, Vietnam
| | - Vy P T Nguyen
- Faculty of Chemical Engineering, Ho Chi Minh City University of Technology (HCMUT), 268 Ly Thuong Kiet Street, District 10, Ho Chi Minh City 700000, Vietnam.,Vietnam National University Ho Chi Minh City, Linh Trung Ward, Thu Duc District, Ho Chi Minh City 700000, Vietnam
| | - Khang X Nguyen
- Faculty of Chemical Engineering, Ho Chi Minh City University of Technology (HCMUT), 268 Ly Thuong Kiet Street, District 10, Ho Chi Minh City 700000, Vietnam.,Vietnam National University Ho Chi Minh City, Linh Trung Ward, Thu Duc District, Ho Chi Minh City 700000, Vietnam
| | - Phuc H Pham
- Faculty of Chemical Engineering, Ho Chi Minh City University of Technology (HCMUT), 268 Ly Thuong Kiet Street, District 10, Ho Chi Minh City 700000, Vietnam.,Vietnam National University Ho Chi Minh City, Linh Trung Ward, Thu Duc District, Ho Chi Minh City 700000, Vietnam
| | - Nhan T H Le
- Faculty of Chemical Engineering, Ho Chi Minh City University of Technology (HCMUT), 268 Ly Thuong Kiet Street, District 10, Ho Chi Minh City 700000, Vietnam.,Vietnam National University Ho Chi Minh City, Linh Trung Ward, Thu Duc District, Ho Chi Minh City 700000, Vietnam
| | - Tung T Nguyen
- Faculty of Chemical Engineering, Ho Chi Minh City University of Technology (HCMUT), 268 Ly Thuong Kiet Street, District 10, Ho Chi Minh City 700000, Vietnam.,Vietnam National University Ho Chi Minh City, Linh Trung Ward, Thu Duc District, Ho Chi Minh City 700000, Vietnam
| | - Nam T S Phan
- Faculty of Chemical Engineering, Ho Chi Minh City University of Technology (HCMUT), 268 Ly Thuong Kiet Street, District 10, Ho Chi Minh City 700000, Vietnam.,Vietnam National University Ho Chi Minh City, Linh Trung Ward, Thu Duc District, Ho Chi Minh City 700000, Vietnam
| |
Collapse
|
17
|
Nerella A, Jeripothula M. Design, synthesis and biological evaluation of novel deoxyvasicinone-indole as multi-target agents for Alzheimer's disease. Bioorg Med Chem Lett 2021; 49:128212. [PMID: 34153471 DOI: 10.1016/j.bmcl.2021.128212] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 05/31/2021] [Accepted: 06/16/2021] [Indexed: 11/17/2022]
Abstract
In this study, a series of multifunctional hybrids (6a-6l) against Alzheimer's disease were designed and obtained by conjugating the pharmacophores of deoxyvasicinone and indole. These analogs of deoxyvasicinone-indole were evaluated as inhibitors of acetylcholinesterase (AChE) and butyrylcholinesterase (BuChE), and as inhibitors of amyloid aggregation (Aβ1-42) for treatment of Alzheimer's disease (AD). Subsequently, AChE induced Aβ aggregation inhibition test was also performed for selected compounds. Biological activity results demonstrated that compound 6b was the most potent and balanced dual ChEs inhibitor with IC50 values 0.12 µM and 0.15 µM for eeAChE and eqBuChE, respectively. Kinetic analysis and docking study indicated that compound 6b was a mixed-type inhibitor for both AChE and BuChE. Compound 6b also found to be the best inhibitors of self-induced Aβ1-42 aggregation with IC50 values of 1.21 µM. Compound 6b also afforded excellent inhibition of AChE-induced Aβ1-42 aggregation by 81.1%. Overall, these results indicate that 6b may be considered as lead compound for the development of highly effective anti-AD drugs.
Collapse
Affiliation(s)
- Ashok Nerella
- Department of Chemistry, Kakatiya University, Warangal, Telangana, India; Government Polytechnic, Warangal, Telangana, India
| | | |
Collapse
|
18
|
Manzoor S, Gabr MT, Rasool B, Pal K, Hoda N. Dual targeting of acetylcholinesterase and tau aggregation: Design, synthesis and evaluation of multifunctional deoxyvasicinone analogues for Alzheimer's disease. Bioorg Chem 2021; 116:105354. [PMID: 34562674 DOI: 10.1016/j.bioorg.2021.105354] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 08/18/2021] [Accepted: 09/08/2021] [Indexed: 12/13/2022]
Abstract
Development of multitargeted ligands have demonstrated remarkable efficiency as potential therapeutics for Alzheimer's disease (AD). Herein, we reported a new series of deoxyvasicinone analogues as dual inhibitor of acetylcholinesterase (AChE) and tau aggregation that function as multitargeted ligands for AD. All the multitargeted ligands 11(a-j) and 15(a-g) were designed, synthesized, and validated by 1HNMR, 13CNMR and mass spectrometry. All the synthesized compounds 11(a-j) and 15(a-g) were screened for their ability to inhibit AChE, BACE1, amyloid fibrillation, α-syn aggregation, and tau aggregation. All the screened compounds possessed weak inhibition of BACE-1, Aβ42 and α-syn aggregation. However, several compounds were identified as potential hits in the AChE inhibitory screening assay and cellular tau aggregation screening. Among all compounds, 11f remarkably inhibited AChE activity and cellular tau oligomerization at single-dose screening (10 µM). Moreover, 11f displayed a half-maximal inhibitory concentration (IC50) value of 0.91 ± 0.05 µM and half-maximal effective concentration (EC50) value of 3.83 ± 0.51 µM for the inhibition of AChE and cellular tau oligomerization, respectively. In addition, the neuroprotective effect of 11f was determined in tau-expressing SH-SY5Y cells incubated with Aβ oligomers. These findings highlighted the potential of 11f to function as a multifunctional ligand for the development of promising anti-AD drugs.
Collapse
Affiliation(s)
- Shoaib Manzoor
- Drug Design and Synthesis Laboratory, Department of Chemistry, Jamia Millia Islamia, New Delhi 110025, India.
| | - Moustafa T Gabr
- Department of Radiology, Stanford University, Stanford, CA 94305, United States.
| | - Bisma Rasool
- Drug Design and Synthesis Laboratory, Department of Chemistry, Jamia Millia Islamia, New Delhi 110025, India
| | - Kavita Pal
- Drug Design and Synthesis Laboratory, Department of Chemistry, Jamia Millia Islamia, New Delhi 110025, India
| | - Nasimul Hoda
- Drug Design and Synthesis Laboratory, Department of Chemistry, Jamia Millia Islamia, New Delhi 110025, India.
| |
Collapse
|
19
|
Bowroju SK, Penthala NR, Lakkaniga NR, Balasubramaniam M, Ayyadevara S, Shmookler Reis RJ, Crooks PA. Novel hydroxybenzylamine-deoxyvasicinone hybrids as anticholinesterase therapeutics for Alzheimer's disease. Bioorg Med Chem 2021; 45:116311. [PMID: 34304133 DOI: 10.1016/j.bmc.2021.116311] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 07/09/2021] [Accepted: 07/10/2021] [Indexed: 12/31/2022]
Abstract
A series of novel 2-hydroxybenzylamine-deoxyvasicinone hybrid analogs (8a-8n) have been synthesized and evaluated as inhibitors of acetylcholinesterase (AChE) and butyrylcholinesterase (BuChE), and as inhibitors of amyloid peptide (Aβ1-42) aggregation, for treatment of Alzheimer's disease (AD). These dual acting compounds exhibited good AChE inhibitory activities ranging from 0.34 to 6.35 µM. Analogs8g and 8n were found to be the most potent AChE inhibitors in the series with IC50values of 0.38 µM and 0.34 µM, respectively. All the analogs (8a-8n) exhibited weak BuChE inhibitory activities ranging from 14.60 to 21.65 µM. Analogs8g and 8n exhibited BuChE with IC50values of 15.38 µM and 14.60 µM, respectively, demonstrating that these analogs were greater than 40-fold more selective for inhibition of AChE over BuChE. Additionally, compounds8g and 8n were also found to be the best inhibitors of self-induced Aβ1-42 peptide aggregation with IC50values of 3.91 µM and 3.22 µM, respectively; 8g and 8n also inhibited AChE-induced Aβ1-42 peptide aggregation by 68.7% and 72.6%, respectively. Kinetic analysis and molecular docking studies indicate that analogs 8g and 8n bind to a new allosteric pocket (site B) on AChE. In addition, the observed inhibition of AChE-induced Aβ1-42 peptide aggregation by 8n is likely due to allosteric inhibition of the binding of this peptide at the CAS site on AChE. Overall, these results indicate that 8g and 8n are examples of dual-acting lead compounds for the development of highly effective anti-AD drugs.
Collapse
Affiliation(s)
- Suresh K Bowroju
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States
| | - Narsimha R Penthala
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States
| | - Naga Rajiv Lakkaniga
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, United States
| | | | - Srinivas Ayyadevara
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States
| | - Robert J Shmookler Reis
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States; Central Arkansas Veterans Healthcare Service, Little Rock, AR 72205, United States; BioInformatics Program, University of Arkansas for Medical Sciences and University of Arkansas at Little Rock, Little Rock, AR 72205, United States
| | - Peter A Crooks
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States.
| |
Collapse
|
20
|
De Boer D, Nguyen N, Mao J, Moore J, Sorin EJ. A Comprehensive Review of Cholinesterase Modeling and Simulation. Biomolecules 2021; 11:580. [PMID: 33920972 PMCID: PMC8071298 DOI: 10.3390/biom11040580] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 04/08/2021] [Accepted: 04/11/2021] [Indexed: 01/18/2023] Open
Abstract
The present article reviews published efforts to study acetylcholinesterase and butyrylcholinesterase structure and function using computer-based modeling and simulation techniques. Structures and models of both enzymes from various organisms, including rays, mice, and humans, are discussed to highlight key structural similarities in the active site gorges of the two enzymes, such as flexibility, binding site location, and function, as well as differences, such as gorge volume and binding site residue composition. Catalytic studies are also described, with an emphasis on the mechanism of acetylcholine hydrolysis by each enzyme and novel mutants that increase catalytic efficiency. The inhibitory activities of myriad compounds have been computationally assessed, primarily through Monte Carlo-based docking calculations and molecular dynamics simulations. Pharmaceutical compounds examined herein include FDA-approved therapeutics and their derivatives, as well as several other prescription drug derivatives. Cholinesterase interactions with both narcotics and organophosphate compounds are discussed, with the latter focusing primarily on molecular recognition studies of potential therapeutic value and on improving our understanding of the reactivation of cholinesterases that are bound to toxins. This review also explores the inhibitory properties of several other organic and biological moieties, as well as advancements in virtual screening methodologies with respect to these enzymes.
Collapse
Affiliation(s)
- Danna De Boer
- Department of Chemistry & Biochemistry, California State University, Long Beach, CA 90840, USA;
| | - Nguyet Nguyen
- Department of Chemical Engineering, California State University, Long Beach, CA 90840, USA; (N.N.); (J.M.)
| | - Jia Mao
- Department of Chemical Engineering, California State University, Long Beach, CA 90840, USA; (N.N.); (J.M.)
| | - Jessica Moore
- Department of Biomedical Engineering, California State University, Long Beach, CA 90840, USA;
| | - Eric J. Sorin
- Department of Chemistry & Biochemistry, California State University, Long Beach, CA 90840, USA;
| |
Collapse
|
21
|
Yuan X, Cui Y, Zhang X, Qin L, Sun Q, Duan X, Chen L, Li G, Qiu J, Guo K. Electrochemical Tri‐ and Difluoromethylation‐Triggered Cyclization Accompanied by the Oxidative Cleavage of Indole Derivatives. Chemistry 2021; 27:6522-6528. [DOI: 10.1002/chem.202005368] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/25/2021] [Indexed: 01/02/2023]
Affiliation(s)
- Xin Yuan
- College of Biotechnology and Pharmaceutical Engineering Nanjing Tech University 30 Puzhu Rd S Nanjing 211816 P. R. China
| | - Yu‐Sheng Cui
- College of Biotechnology and Pharmaceutical Engineering Nanjing Tech University 30 Puzhu Rd S Nanjing 211816 P. R. China
| | - Xin‐Peng Zhang
- College of Biotechnology and Pharmaceutical Engineering Nanjing Tech University 30 Puzhu Rd S Nanjing 211816 P. R. China
| | - Long‐Zhou Qin
- College of Biotechnology and Pharmaceutical Engineering Nanjing Tech University 30 Puzhu Rd S Nanjing 211816 P. R. China
| | - Qi Sun
- College of Biotechnology and Pharmaceutical Engineering Nanjing Tech University 30 Puzhu Rd S Nanjing 211816 P. R. China
| | - Xiu Duan
- College of Biotechnology and Pharmaceutical Engineering Nanjing Tech University 30 Puzhu Rd S Nanjing 211816 P. R. China
| | - Lin Chen
- College of Biotechnology and Pharmaceutical Engineering Nanjing Tech University 30 Puzhu Rd S Nanjing 211816 P. R. China
| | - Guigen Li
- Institute of Chemistry & Biomedical Science Nanjing University No.163, Xianlin Avenue, Qixia District Nanjing 210093 P. R. China
- Department of Chemistry and Biochemistry Texas Tech University Lubbock TX 79409-1061 USA
| | - Jiang‐Kai Qiu
- College of Biotechnology and Pharmaceutical Engineering Nanjing Tech University 30 Puzhu Rd S Nanjing 211816 P. R. China
| | - Kai Guo
- College of Biotechnology and Pharmaceutical Engineering Nanjing Tech University 30 Puzhu Rd S Nanjing 211816 P. R. China
| |
Collapse
|
22
|
Choi JH, Do Kim H, Kang JY, Jeong T, Ghosh P, Kim IS. Ruthenium(
II
)‐Catalyzed CH/NH Carbonylative Cyclization of
2‐Aryl
Quinazolinones with Isocyanates as
CO
Surrogates. B KOREAN CHEM SOC 2021. [DOI: 10.1002/bkcs.12228] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Jin Ho Choi
- School of Pharmacy Sungkyunkwan University Suwon 16419 Republic of Korea
| | - Hak Do Kim
- School of Pharmacy Sungkyunkwan University Suwon 16419 Republic of Korea
| | - Ju Young Kang
- School of Pharmacy Sungkyunkwan University Suwon 16419 Republic of Korea
| | - Taejoo Jeong
- School of Pharmacy Sungkyunkwan University Suwon 16419 Republic of Korea
| | - Prithwish Ghosh
- School of Pharmacy Sungkyunkwan University Suwon 16419 Republic of Korea
| | - In Su Kim
- School of Pharmacy Sungkyunkwan University Suwon 16419 Republic of Korea
| |
Collapse
|
23
|
Huang J, Chen W, Liang J, Yang Q, Deng Z, Song Z, Peng Y. Rhodium( iii)-catalyzed annulation of 3-arylquinazolinones with alkynes via double C–H activation: an efficient route for quinolino[2,1- b]quinazolinones. Org Chem Front 2021. [DOI: 10.1039/d1qo01186c] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
An effective method for the synthesis of quinolino[2,1-b]quinazolinones has been described.
Collapse
Affiliation(s)
- Jian Huang
- Key Laboratory for Green Chemistry of Jiangxi Province, Key Laboratory of Functional Small Molecules for Ministry of Education, Jiangxi Normal University, 99 Ziyang Avenue, Nanchang 330022, China
| | - Wei Chen
- Key Laboratory for Green Chemistry of Jiangxi Province, Key Laboratory of Functional Small Molecules for Ministry of Education, Jiangxi Normal University, 99 Ziyang Avenue, Nanchang 330022, China
| | - Jiazhi Liang
- Key Laboratory for Green Chemistry of Jiangxi Province, Key Laboratory of Functional Small Molecules for Ministry of Education, Jiangxi Normal University, 99 Ziyang Avenue, Nanchang 330022, China
| | - Qin Yang
- Key Laboratory for Green Chemistry of Jiangxi Province, Key Laboratory of Functional Small Molecules for Ministry of Education, Jiangxi Normal University, 99 Ziyang Avenue, Nanchang 330022, China
| | - Zhihong Deng
- Key Laboratory for Green Chemistry of Jiangxi Province, Key Laboratory of Functional Small Molecules for Ministry of Education, Jiangxi Normal University, 99 Ziyang Avenue, Nanchang 330022, China
| | - Zhibin Song
- Key Laboratory for Green Chemistry of Jiangxi Province, Key Laboratory of Functional Small Molecules for Ministry of Education, Jiangxi Normal University, 99 Ziyang Avenue, Nanchang 330022, China
| | - Yiyuan Peng
- Key Laboratory for Green Chemistry of Jiangxi Province, Key Laboratory of Functional Small Molecules for Ministry of Education, Jiangxi Normal University, 99 Ziyang Avenue, Nanchang 330022, China
| |
Collapse
|
24
|
Choi JH, Kim K, Oh H, Han S, Mishra NK, Kim IS. Ru(ii)-Catalyzed C-H addition and oxidative cyclization of 2-aryl quinazolinones with activated aldehydes. Org Biomol Chem 2020; 18:9611-9622. [PMID: 33020797 DOI: 10.1039/d0ob01663b] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The ruthenium(ii)-catalyzed cross-coupling reaction between 2-aryl quinazolinones and activated aldehydes is described. This method enables the site-selective hydroxyalkylation under redox-neutral conditions. Moreover, this protocol provides a facile access to various tetracyclic isoindoloquinazolinones by using Cu(OAc)2 as an external oxidant via C-H addition and subsequent intramolecular cyclization. A wide substrate scope and a high level of chemoselectivity as well as broad functional group tolerance are observed.
Collapse
Affiliation(s)
- Jin Ho Choi
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| | | | | | | | | | | |
Collapse
|
25
|
Pedrood K, Sherafati M, Mohammadi-Khanaposhtani M, Asgari MS, Hosseini S, Rastegar H, Larijani B, Mahdavi M, Taslimi P, Erden Y, Günay S, Gulçin İ. Design, synthesis, characterization, enzymatic inhibition evaluations, and docking study of novel quinazolinone derivatives. Int J Biol Macromol 2020; 170:1-12. [PMID: 33352155 DOI: 10.1016/j.ijbiomac.2020.12.121] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 12/12/2020] [Accepted: 12/15/2020] [Indexed: 12/14/2022]
Abstract
In this study, novel quinazolinone derivatives 7a-n were synthesized and evaluated against metabolic enzymes including α-glycosidase, acetylcholinesterase, butyrylcholinesterase, human carbonic anhydrase I, and II. These compounds exhibited high inhibitory activities in comparison to used standard inhibitors with Ki values in the range of 19.28-135.88 nM for α-glycosidase (Ki value for standard inhibitor = 187.71 nM), 0.68-23.01 nM for acetylcholinesterase (Ki value for standard inhibitor = 53.31 nM), 1.01-29.56 nM for butyrylcholinesterase (Ki value for standard inhibitor = 58.16 nM), 10.25-126.05 nM for human carbonic anhydrase I (Ki value for standard inhibitor = 248.18 nM), and 13.46-178.35 nM for human carbonic anhydrase II (Ki value for standard inhibitor = 323.72). Furthermore, the most potent compounds against each enzyme were selected in order to evaluate interaction modes of these compounds in the active site of the target enzyme. Cytotoxicity assay of the title compounds 7a-n against cancer cell lines MCF-7 and LNCaP demonstrated that these compounds do not show significant cytotoxic effects.
Collapse
Affiliation(s)
- Keyvan Pedrood
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Maedeh Sherafati
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Mohammadi-Khanaposhtani
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | | | - Samanesadat Hosseini
- Department of Pharmaceutical Chemistry, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hossein Rastegar
- Cosmetic Products Research Center, Iranian Food and Drug Administration, MOHE, Tehran, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Mahdavi
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.
| | - Parham Taslimi
- Department of Biotechnology, Faculty of Science, Bartin University, 74100 Bartin, Turkey.
| | - Yavuz Erden
- Department of Molecular Biology and Genetics, Faculty of Science, Bartin University, 74100 Bartin, Turkey
| | - Sevilay Günay
- Department of Molecular Biology and Genetics, Faculty of Science, Bartin University, 74100 Bartin, Turkey
| | - İlhami Gulçin
- Department of Chemistry, Faculty of Sciences, Ataturk University, 25240, Erzurum, Turkey
| |
Collapse
|
26
|
Du H, Jiang X, Ma M, Xu H, Liu S, Ma F. Novel deoxyvasicinone and tetrahydro-beta-carboline hybrids as inhibitors of acetylcholinesterase and amyloid beta aggregation. Bioorg Med Chem Lett 2020; 30:127659. [PMID: 33137375 DOI: 10.1016/j.bmcl.2020.127659] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 09/30/2020] [Accepted: 10/27/2020] [Indexed: 12/17/2022]
Abstract
A novel series of deoxyvasicinone-tetrahydro-beta-carboline hybrids were synthesized and evaluated as acetylcholinesterase (AChE) and β-amyloid peptide (Aβ) aggregation inhibitors for the treatment of Alzheimer's disease. The results revealed that the derivatives had multifunctional profiles, including AChE inhibition, Aβ1-42 aggregation inhibition, and neuroprotective properties. Inspiringly, hybrids 8b and 8d displayed excellent inhibitory activities against hAChE (IC50 = 0.93 and 1.08 nM, respectively) and Aβ1-42 self-aggregation (IC50 = 19.71 and 2.05 μM, respectively). In addition, 8b and 8d showed low cytotoxicity and good neuroprotective activity against Aβ1-42-induced damage in SH-SY5Y cells.
Collapse
Affiliation(s)
- Hongtao Du
- College of Life Science, Xinyang Normal University, Xinyang 464000, China; College of Science, Northwest A&F University, Yangling 712100, Shaanxi Province, China.
| | - Xinyu Jiang
- College of Life Science, Xinyang Normal University, Xinyang 464000, China
| | - Meng Ma
- College of Life Science, Xinyang Normal University, Xinyang 464000, China
| | - Huili Xu
- College of Life Science, Xinyang Normal University, Xinyang 464000, China
| | - Shuang Liu
- College of Life Science, Xinyang Normal University, Xinyang 464000, China
| | - Fang Ma
- School of Geographic Sciences, Xinyang Normal University, Xinyang 464000, China.
| |
Collapse
|
27
|
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: 6.4] [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
|
28
|
Huang WY, Zhang XR, Lyu L, Wang SQ, Zhang XT. Pyridazino[1,6-b]quinazolinones as new anticancer scaffold: Synthesis, DNA intercalation, topoisomerase I inhibition and antitumor evaluation in vitro and in vivo. Bioorg Chem 2020; 99:103814. [PMID: 32278208 DOI: 10.1016/j.bioorg.2020.103814] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 02/26/2020] [Accepted: 03/31/2020] [Indexed: 12/21/2022]
Abstract
A new anticancer N-containing heterocyclic scaffold was designed and 30 pyridazino[1,6-b]quinazolinone derivatives were synthesized and characterized. Antiproliferation evaluation in vitro against four human cancer cell lines including SK-OV-3(ovarian cell), CNE-2(nasopharyngeal cell), MGC-803(gastric cell) and NCI-H460(lung cell) indicated that most of them exhibited potent anticancer activity and the IC50 value of the most potent compound lowered to sub-μM. DNA interaction assay indicated that compounds 4e, 4g, 6o, 6p, 8o can intercalate into DNA. Compounds 6 and 8 also demonstrated potent topoisomerase I (topo I) activity. Annexin V- FITC/propidium iodide dual staining assay and cell cycle analysis indicated that 2-(4-bromophenyl)-4-((3-(diethylamino)propyl)amino) -10H-pyridazino [1,6-b]quinazolin- 10-one (8p) could induce arrest cell cycle at G2 phase and apoptosis in MGC-803 cells in a dose-dependent manner. The in vivo antitumor efficiency of compound 8p was also evaluated on MGC-803 xenograft nude mice, and the relative tumor growth inhibition was up to 55.9% at a dose of 20 mg/kg per two days. The results suggested that pyridazino[1,6-b]-quinazolinones might serve as a promising novel scaffold for the development of new antitumor agents.
Collapse
Affiliation(s)
- Wan-Yun Huang
- Department of Pharmaceutical Chemistry, College of Pharmacy, Guilin Medical University, Guilin 541004, China.
| | - Xiao-Rong Zhang
- Department of Pharmaceutical Chemistry, College of Pharmacy, Guilin Medical University, Guilin 541004, China
| | - Liang Lyu
- Department of Pharmacology, College of Pharmacy, Guilin Medical University, Guilin 541004, China.
| | - Shu-Qin Wang
- Department of Pharmaceutical Chemistry, College of Pharmacy, Guilin Medical University, Guilin 541004, China
| | - Xiao-Ting Zhang
- Department of Pharmaceutical Chemistry, College of Pharmacy, Guilin Medical University, Guilin 541004, China
| |
Collapse
|
29
|
Du H, Liu X, Xie J, Ma F. Novel Deoxyvasicinone-Donepezil Hybrids as Potential Multitarget Drug Candidates for Alzheimer's Disease. ACS Chem Neurosci 2019; 10:2397-2407. [PMID: 30720268 DOI: 10.1021/acschemneuro.8b00699] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
In this study, we designed and synthesized a series of deoxyvasicinone-donepezil hybrids and determined whether they could be used as novel multitarget inhibitors for Alzheimer's disease. In vitro studies showed that most of the hybrids demonstrated moderate to potent inhibition of hAChE, BACE1, and Aβ1-42 aggregation. In particular, the hybrids 10a, 10d, 11a, and 11j exhibited excellent inhibitory activities against hAChE (IC50 = 56.14, 5.91, 3.29, and 8.65 nM, respectively), BACE1 (IC50 = 0.834, 0.167, 0.129, and 0.085 μM, respectively), and Aβ1-42 aggregation (IC50 = 13.26, 19.43, 9.26, and 5.41 μM, respectively). In addition, 10a and 11a exhibited very low cytotoxicity and showed remarkable neuroprotective activity against Aβ1-42-induced damage in SH-SY5Y cells.
Collapse
Affiliation(s)
- Hongtao Du
- College of Life Science, Xinyang Normal University, Xinyang 464000, China
- College of Plant Protection, Northwest A&F University, Yangling 712100, Shaanxi Province, China
| | - Xinlian Liu
- College of Life Science, Xinyang Normal University, Xinyang 464000, China
- Tea Plant Biology Key Laboratory of Henan Province, Xinyang 464000, China
| | - Jusen Xie
- College of Life Science, Xinyang Normal University, Xinyang 464000, China
- Tea Plant Biology Key Laboratory of Henan Province, Xinyang 464000, China
| | - Fang Ma
- School of Geographic Sciences, Xinyang Normal University, Xinyang 464000, China
| |
Collapse
|
30
|
Zhang Y, Shao Y, Gong J, Zhu J, Cheng T, Chen J. Selenium-Catalyzed Oxidative C–H Amination of (E)-3-(Arylamino)-2-styrylquinazolin-4(3H)-ones: A Metal-Free Synthesis of 1,2-Diarylpyrazolo[5,1-b]quinazolin-9(1H)-ones. J Org Chem 2019; 84:2798-2807. [DOI: 10.1021/acs.joc.8b03179] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Yetong Zhang
- College of Chemistry & Materials Engineering, Wenzhou University, Wenzhou 325035, China
| | - Yinlin Shao
- College of Chemistry & Materials Engineering, Wenzhou University, Wenzhou 325035, China
| | - Julin Gong
- College of Chemistry & Materials Engineering, Wenzhou University, Wenzhou 325035, China
| | - Jianghe Zhu
- College of Chemistry & Materials Engineering, Wenzhou University, Wenzhou 325035, China
| | - Tianxing Cheng
- College of Chemistry & Materials Engineering, Wenzhou University, Wenzhou 325035, China
| | - Jiuxi Chen
- College of Chemistry & Materials Engineering, Wenzhou University, Wenzhou 325035, China
| |
Collapse
|
31
|
Sukumaran SD, Faraj FL, Lee VS, Othman R, Buckle MJC. 2-Aryl-3-(arylideneamino)-1,2-dihydroquinazoline-4(3 H)-ones as inhibitors of cholinesterases and self-induced β-amyloid (Aβ) aggregation: biological evaluations and mechanistic insights from molecular dynamics simulations. RSC Adv 2018; 8:7818-7831. [PMID: 35539141 PMCID: PMC9078462 DOI: 10.1039/c7ra11872d] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Accepted: 01/19/2018] [Indexed: 11/21/2022] Open
Abstract
A series of 2-aryl-3-(arylideneamino)-1,2-dihydroquinazoline-4(3H)-ones were evaluated as inhibitors of acetylcholinesterase (AChE), butyrylcholinesterase (BuChE) and self-induced β-amyloid (Aβ) aggregation. All the compounds were found to inhibit both forms of cholinesterase (IC50 in the range 4-32 μM) with some selectivity for BuChE. Most of the compounds also showed self-induced Aβ aggregation inhibitory activities, which were comparable or higher than those obtained for reference compounds, curcumin and myricetin. Docking and molecular dynamics (MD) simulation experiments suggested that the compounds are able to disrupt the dimer form of Aβ.
Collapse
Affiliation(s)
- Sri Devi Sukumaran
- Department of Pharmacy, Faculty of Medicine, University of Malaya 50603 Kuala Lumpur Malaysia +60-3-7967-4959
- Drug Design and Development Research Group (DDDRG), University of Malaya 50603 Kuala Lumpur Malaysia
| | - Fadhil Lafta Faraj
- Department of Chemistry, Faculty of Science, University of Diyala Diyala Governorate Iraq
| | - Vannajan Sanghiran Lee
- Drug Design and Development Research Group (DDDRG), University of Malaya 50603 Kuala Lumpur Malaysia
- Department of Chemistry, Faculty of Science, University of Malaya 50603 Kuala Lumpur Malaysia +60 163208906
| | - Rozana Othman
- Department of Pharmacy, Faculty of Medicine, University of Malaya 50603 Kuala Lumpur Malaysia +60-3-7967-4959
- Drug Design and Development Research Group (DDDRG), University of Malaya 50603 Kuala Lumpur Malaysia
| | - Michael J C Buckle
- Department of Pharmacy, Faculty of Medicine, University of Malaya 50603 Kuala Lumpur Malaysia +60-3-7967-4959
- Drug Design and Development Research Group (DDDRG), University of Malaya 50603 Kuala Lumpur Malaysia
| |
Collapse
|
32
|
Dolles D, Hoffmann M, Gunesch S, Marinelli O, Möller J, Santoni G, Chatonnet A, Lohse MJ, Wittmann HJ, Strasser A, Nabissi M, Maurice T, Decker M. Structure-Activity Relationships and Computational Investigations into the Development of Potent and Balanced Dual-Acting Butyrylcholinesterase Inhibitors and Human Cannabinoid Receptor 2 Ligands with Pro-Cognitive in Vivo Profiles. J Med Chem 2018; 61:1646-1663. [PMID: 29400965 DOI: 10.1021/acs.jmedchem.7b01760] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The enzyme butyrylcholinesterase (BChE) and the human cannabinoid receptor 2 (hCB2R) represent promising targets for pharmacotherapy in the later stages of Alzheimer's disease. We merged pharmacophores for both targets into small benzimidazole-based molecules, investigated SARs, and identified several dual-acting ligands with a balanced affinity/inhibitory activity and an excellent selectivity over both hCB1R and hAChE. A homology model for the hCB2R was developed based on the hCB1R crystal structure and used for molecular dynamics studies to investigate binding modes. In vitro studies proved hCB2R agonism. Unwanted μ-opioid receptor affinity could be designed out. One well-balanced dual-acting and selective hBChE inhibitor/hCB2R agonist showed superior in vivo activity over the lead CB2 agonist with regards to cognition improvement. The data shows the possibility to combine a small molecule with selective and balanced GPCR-activity/enzyme inhibition and in vivo activity for the therapy of AD and may help to rationalize the development of other dual-acting ligands.
Collapse
Affiliation(s)
- Dominik Dolles
- Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy and Food Chemistry, Julius Maximilian University of Würzburg , Am Hubland, D-97074 Würzburg, Germany
| | - Matthias Hoffmann
- Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy and Food Chemistry, Julius Maximilian University of Würzburg , Am Hubland, D-97074 Würzburg, Germany
| | - Sandra Gunesch
- Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy and Food Chemistry, Julius Maximilian University of Würzburg , Am Hubland, D-97074 Würzburg, Germany
| | - Oliviero Marinelli
- School of Pharmacy, Department of Experimental Medicine, University of Camerino , I-62032 Camerino, Italy
| | - Jan Möller
- Institute of Pharmacology and Toxicology, Julius Maximilian University of Würzburg , Versbacher Strabe 9, D-97078 Würzburg, Germany
| | - Giorgio Santoni
- School of Pharmacy, Department of Experimental Medicine, University of Camerino , I-62032 Camerino, Italy
| | - Arnaud Chatonnet
- INRA UMR866, University of Montpellier , F-34060 Montpellier, France
| | - Martin J Lohse
- Institute of Pharmacology and Toxicology, Julius Maximilian University of Würzburg , Versbacher Strabe 9, D-97078 Würzburg, Germany
| | - Hans-Joachim Wittmann
- Pharmaceutical and Medicinal Chemistry II, Institute of Pharmacy, University of Regensburg , D-95053 Regensburg, Germany
| | - Andrea Strasser
- Pharmaceutical and Medicinal Chemistry II, Institute of Pharmacy, University of Regensburg , D-95053 Regensburg, Germany
| | - Massimo Nabissi
- School of Pharmacy, Department of Experimental Medicine, University of Camerino , I-62032 Camerino, Italy
| | - Tangui Maurice
- INSERM UMR-S1198, University of Montpellier, EPHE , F-34095 Montpellier, France
| | - Michael Decker
- Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy and Food Chemistry, Julius Maximilian University of Würzburg , Am Hubland, D-97074 Würzburg, Germany
| |
Collapse
|
33
|
Xu G, Tong C, Cui S, Dai L. A silver catalyzed domino reaction of N-cyanamide alkenes and 1,3-dicarbonyls for the synthesis of quinazolinones. Org Biomol Chem 2018; 16:5899-5906. [DOI: 10.1039/c8ob01252k] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
A silver catalyzed domino reaction of N-cyanamide alkenes and 1,3-dicarbonyls has been developed for the facile synthesis of quinazolinones.
Collapse
Affiliation(s)
- Gang Xu
- College of Chemical and Biological Engineering
- Zhejiang University
- Hangzhou 310027
- P. R. China
| | - Chaodi Tong
- College of Chemical and Biological Engineering
- Zhejiang University
- Hangzhou 310027
- P. R. China
| | - Sunliang Cui
- Institute of Drug Discovery and Design
- College of Pharmaceutical Sciences
- Zhejiang University
- Hangzhou 310058
- P. R. China
| | - Liyan Dai
- College of Chemical and Biological Engineering
- Zhejiang University
- Hangzhou 310027
- P. R. China
| |
Collapse
|
34
|
Ma F, Du H. Novel deoxyvasicinone derivatives as potent multitarget-directed ligands for the treatment of Alzheimer's disease: Design, synthesis, and biological evaluation. Eur J Med Chem 2017; 140:118-127. [DOI: 10.1016/j.ejmech.2017.09.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 09/03/2017] [Accepted: 09/05/2017] [Indexed: 12/31/2022]
|
35
|
Zhang C, Li S, Bureš F, Lee R, Ye X, Jiang Z. Visible Light Photocatalytic Aerobic Oxygenation of Indoles and pH as a Chemoselective Switch. ACS Catal 2016. [DOI: 10.1021/acscatal.6b01969] [Citation(s) in RCA: 119] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Chenhao Zhang
- Key
Laboratory of Natural Medicine and Immuno-Engineering of Henan Province, Henan University, Kaifeng, Henan, China
| | - Sanliang Li
- Key
Laboratory of Natural Medicine and Immuno-Engineering of Henan Province, Henan University, Kaifeng, Henan, China
| | - Filip Bureš
- Institute
of Organic Chemistry and Technology, Faculty of Chemical Technology, University of Pardubice, Studentská 573, Pardubice 53210, Czech Republic
| | - Richmond Lee
- ARC
Centre of Excellence for Electromaterials Science and Research School
of Chemistry, Australian National University, Canberra, ACT 2601, Australia
| | - Xinyi Ye
- Division
of Chemistry and Biological Chemistry, Nanyang Technological University, 21 Nanyang Link, 637371 Singapore
| | - Zhiyong Jiang
- Key
Laboratory of Natural Medicine and Immuno-Engineering of Henan Province, Henan University, Kaifeng, Henan, China
| |
Collapse
|
36
|
Abbas SY, El-Bayouki KAM, Basyouni WM. Utilization of isatoic anhydride in the syntheses of various types of quinazoline and quinazolinone derivatives. SYNTHETIC COMMUN 2016. [DOI: 10.1080/00397911.2016.1177087] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Samir Y. Abbas
- Organometallic and Organometalloid Chemistry Department, National Research Centre, Dokki, Cairo, Egypt
| | - Khairy A. M. El-Bayouki
- Organometallic and Organometalloid Chemistry Department, National Research Centre, Dokki, Cairo, Egypt
| | - Wahid M. Basyouni
- Organometallic and Organometalloid Chemistry Department, National Research Centre, Dokki, Cairo, Egypt
| |
Collapse
|
37
|
Synthesis and Biological Evaluation of Benzochromenopyrimidinones as Cholinesterase Inhibitors and Potent Antioxidant, Non-Hepatotoxic Agents for Alzheimer's Disease. Molecules 2016; 21:molecules21050634. [PMID: 27187348 PMCID: PMC6273488 DOI: 10.3390/molecules21050634] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 04/19/2016] [Accepted: 05/04/2016] [Indexed: 11/16/2022] Open
Abstract
We report herein the straightforward two-step synthesis and biological assessment of novel racemic benzochromenopyrimidinones as non-hepatotoxic, acetylcholinesterase inhibitors with antioxidative properties. Among them, compound 3Bb displayed a mixed-type inhibition of human acetylcholinesterase (IC50 = 1.28 ± 0.03 μM), good antioxidant activity, and also proved to be non-hepatotoxic on human HepG2 cell line.
Collapse
|
38
|
Sawatzky E, Wehle S, Kling B, Wendrich J, Bringmann G, Sotriffer CA, Heilmann J, Decker M. Discovery of Highly Selective and Nanomolar Carbamate-Based Butyrylcholinesterase Inhibitors by Rational Investigation into Their Inhibition Mode. J Med Chem 2016; 59:2067-82. [DOI: 10.1021/acs.jmedchem.5b01674] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Affiliation(s)
- Edgar Sawatzky
- Pharmazeutische
und Medizinische Chemie, Institut für Pharmazie und Lebensmittelchemie, Universität Würzburg, Am Hubland, D-97074 Würzburg, Germany
| | - Sarah Wehle
- Pharmazeutische
und Medizinische Chemie, Institut für Pharmazie und Lebensmittelchemie, Universität Würzburg, Am Hubland, D-97074 Würzburg, Germany
| | - Beata Kling
- Lehrstuhl
für Pharmazeutische Biologie, Institut für Pharmazie, Universität Regensburg, Universitätsstraße 31, D-93053 Regensburg, Germany
| | - Jan Wendrich
- Lehrstuhl
für Organische Chemie I, Institut für Organische Chemie, Universität Würzburg, Am Hubland, D-97074 Würzburg, Germany
| | - Gerhard Bringmann
- Lehrstuhl
für Organische Chemie I, Institut für Organische Chemie, Universität Würzburg, Am Hubland, D-97074 Würzburg, Germany
| | - Christoph A. Sotriffer
- Pharmazeutische
und Medizinische Chemie, Institut für Pharmazie und Lebensmittelchemie, Universität Würzburg, Am Hubland, D-97074 Würzburg, Germany
| | - Jörg Heilmann
- Lehrstuhl
für Pharmazeutische Biologie, Institut für Pharmazie, Universität Regensburg, Universitätsstraße 31, D-93053 Regensburg, Germany
| | - Michael Decker
- Pharmazeutische
und Medizinische Chemie, Institut für Pharmazie und Lebensmittelchemie, Universität Würzburg, Am Hubland, D-97074 Würzburg, Germany
| |
Collapse
|
39
|
Zheng J, Deng Z, Zhang Y, Cui S. Copper-Catalyzed Divergent Trifluoromethylation/Cyclization of Unactivated Alkenes. Adv Synth Catal 2016. [DOI: 10.1002/adsc.201500965] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
40
|
Dgachi Y, Ismaili L, Knez D, Benchekroun M, Martin H, Szałaj N, Wehle S, Bautista-Aguilera OM, Luzet V, Bonnet A, Malawska B, Gobec S, Chioua M, Decker M, Chabchoub F, Marco-Contelles J. Synthesis and Biological Assessment of Racemic Benzochromenopyrimidinimines as Antioxidant, Cholinesterase, and Aβ1−42Aggregation Inhibitors for Alzheimer's Disease Therapy. ChemMedChem 2016; 11:1318-27. [DOI: 10.1002/cmdc.201500539] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Indexed: 11/08/2022]
Affiliation(s)
- Youssef Dgachi
- Laboratory of Applied Chemistry: Heterocycles, Lipids and Polymers; Faculty of Sciences of Sfax; University of Sfax; BP 802 3000 Sfax Tunisia
| | - Lhassane Ismaili
- Neurosciences Intégratives et Cliniques EA 481; Laboratoire de Chimie Organique et Thérapeutique, UFR SMP; Université de Franche-Comté; Université Bourgogne Franche-Comté; 19 rue Ambroise Paré 25000 Besançon France
| | - Damijan Knez
- Faculty of Pharmacy; University of Ljubljana; Aškerčeva 7 1000 Ljubljana Slovenia
| | - Mohamed Benchekroun
- Neurosciences Intégratives et Cliniques EA 481; Laboratoire de Chimie Organique et Thérapeutique, UFR SMP; Université de Franche-Comté; Université Bourgogne Franche-Comté; 19 rue Ambroise Paré 25000 Besançon France
| | - Hélène Martin
- Laboratory of Cell Toxicology, EA 4267; University of Franche-Comté; 19 rue Ambroise Paré 25030 Besançon France
| | - Natalia Szałaj
- Department of Physicochemical Drug Analysis; Jagiellonian University, Medical College; Medyczna 9 Street 30-688 Krakow Poland
| | - Sarah Wehle
- Pharmazeutische und Medizinische Chemie; Institut für Pharmazie und Lebensmittelchemie; Julius-Maximilians-Universität Würzburg; Am Hubland 97074 Würzburg Germany
| | - Oscar M. Bautista-Aguilera
- Neurosciences Intégratives et Cliniques EA 481; Laboratoire de Chimie Organique et Thérapeutique, UFR SMP; Université de Franche-Comté; Université Bourgogne Franche-Comté; 19 rue Ambroise Paré 25000 Besançon France
| | - Vincent Luzet
- Neurosciences Intégratives et Cliniques EA 481; Laboratoire de Chimie Organique et Thérapeutique, UFR SMP; Université de Franche-Comté; Université Bourgogne Franche-Comté; 19 rue Ambroise Paré 25000 Besançon France
| | - Alexandre Bonnet
- Laboratory of Cell Toxicology, EA 4267; University of Franche-Comté; 19 rue Ambroise Paré 25030 Besançon France
| | - Barbara Malawska
- Department of Physicochemical Drug Analysis; Jagiellonian University, Medical College; Medyczna 9 Street 30-688 Krakow Poland
| | - Stanislav Gobec
- Faculty of Pharmacy; University of Ljubljana; Aškerčeva 7 1000 Ljubljana Slovenia
| | - Mourad Chioua
- Laboratory of Medicinal Chemistry (IQOG, CSIC); C/Juan de la Cierva 3 28006 Madrid Spain
| | - Michael Decker
- Pharmazeutische und Medizinische Chemie; Institut für Pharmazie und Lebensmittelchemie; Julius-Maximilians-Universität Würzburg; Am Hubland 97074 Würzburg Germany
| | - Fakher Chabchoub
- Laboratory of Applied Chemistry: Heterocycles, Lipids and Polymers; Faculty of Sciences of Sfax; University of Sfax; BP 802 3000 Sfax Tunisia
| | - José Marco-Contelles
- Laboratory of Medicinal Chemistry (IQOG, CSIC); C/Juan de la Cierva 3 28006 Madrid Spain
| |
Collapse
|
41
|
Dolles D, Nimczick M, Scheiner M, Ramler J, Stadtmüller P, Sawatzky E, Drakopoulos A, Sotriffer C, Wittmann HJ, Strasser A, Decker M. Aminobenzimidazoles and Structural Isomers as Templates for Dual-Acting Butyrylcholinesterase Inhibitors andhCB2R Ligands To Combat Neurodegenerative Disorders. ChemMedChem 2015; 11:1270-83. [DOI: 10.1002/cmdc.201500418] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2015] [Indexed: 11/12/2022]
Affiliation(s)
- Dominik Dolles
- Pharmaceutical and Medicinal Chemistry; Institute of Pharmacy and Food Chemistry; Julius-Maximilians-Universität Würzburg; Am Hubland 97074 Würzburg Germany
| | - Martin Nimczick
- Pharmaceutical and Medicinal Chemistry; Institute of Pharmacy and Food Chemistry; Julius-Maximilians-Universität Würzburg; Am Hubland 97074 Würzburg Germany
| | - Matthias Scheiner
- Pharmaceutical and Medicinal Chemistry; Institute of Pharmacy and Food Chemistry; Julius-Maximilians-Universität Würzburg; Am Hubland 97074 Würzburg Germany
| | - Jacqueline Ramler
- Pharmaceutical and Medicinal Chemistry; Institute of Pharmacy and Food Chemistry; Julius-Maximilians-Universität Würzburg; Am Hubland 97074 Würzburg Germany
| | - Patricia Stadtmüller
- Pharmaceutical and Medicinal Chemistry; Institute of Pharmacy and Food Chemistry; Julius-Maximilians-Universität Würzburg; Am Hubland 97074 Würzburg Germany
| | - Edgar Sawatzky
- Pharmaceutical and Medicinal Chemistry; Institute of Pharmacy and Food Chemistry; Julius-Maximilians-Universität Würzburg; Am Hubland 97074 Würzburg Germany
| | - Antonios Drakopoulos
- Pharmaceutical and Medicinal Chemistry; Institute of Pharmacy and Food Chemistry; Julius-Maximilians-Universität Würzburg; Am Hubland 97074 Würzburg Germany
| | - Christoph Sotriffer
- Pharmaceutical and Medicinal Chemistry; Institute of Pharmacy and Food Chemistry; Julius-Maximilians-Universität Würzburg; Am Hubland 97074 Würzburg Germany
| | - Hans-Joachim Wittmann
- Pharmaceutical and Medicinal Chemistry II; Institute of Pharmacy; University of Regensburg; 95053 Regensburg Germany
| | - Andrea Strasser
- Pharmaceutical and Medicinal Chemistry II; Institute of Pharmacy; University of Regensburg; 95053 Regensburg Germany
| | - Michael Decker
- Pharmaceutical and Medicinal Chemistry; Institute of Pharmacy and Food Chemistry; Julius-Maximilians-Universität Würzburg; Am Hubland 97074 Würzburg Germany
| |
Collapse
|
42
|
Liu H, Fan H, Gao X, Huang X, Liu X, Liu L, Zhou C, Tang J, Wang Q, Liu W. Design, synthesis and preliminary structure-activity relationship investigation of nitrogen-containing chalcone derivatives as acetylcholinesterase and butyrylcholinesterase inhibitors: a further study based on Flavokawain B Mannich base derivatives. J Enzyme Inhib Med Chem 2015; 31:580-9. [PMID: 26186269 DOI: 10.3109/14756366.2015.1050009] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
In order to study the structure-activity relationship of Flavokawain B Mannich-based derivatives as acetylcholinesterase (AChE) inhibitors in our recent investigation, 20 new nitrogen-containing chalcone derivatives (4 a-8d) were designed, synthesized, and evaluated for AChE inhibitory activity in vitro. The results suggested that amino alkyl side chain of chalcone dramatically influenced the inhibitory activity against AChE. Among them, compound 6c revealed the strongest AChE inhibitory activity (IC50 value: 0.85 μmol/L) and the highest selectivity against AChE over BuChE (ratio: 35.79). Enzyme kinetic study showed that the inhibition mechanism of compound 6c against AChE was a mixed-type inhibition. The molecular docking assay showed that this compound can both bind with the catalytic site and the peripheral site of AChE.
Collapse
Affiliation(s)
- Haoran Liu
- a College of Chemistry and Chemical Engineering, Hu'nan University , Changsha , China and
| | - Haoqun Fan
- a College of Chemistry and Chemical Engineering, Hu'nan University , Changsha , China and
| | - Xiaohui Gao
- b College of Pharmacy, Hu'nan University of Chinese Medicine , Changsha , China
| | - Xueqing Huang
- a College of Chemistry and Chemical Engineering, Hu'nan University , Changsha , China and
| | - Xianjun Liu
- a College of Chemistry and Chemical Engineering, Hu'nan University , Changsha , China and
| | - Linbo Liu
- a College of Chemistry and Chemical Engineering, Hu'nan University , Changsha , China and
| | - Chao Zhou
- a College of Chemistry and Chemical Engineering, Hu'nan University , Changsha , China and
| | - Jingjing Tang
- a College of Chemistry and Chemical Engineering, Hu'nan University , Changsha , China and
| | - Qiuan Wang
- a College of Chemistry and Chemical Engineering, Hu'nan University , Changsha , China and
| | - Wukun Liu
- a College of Chemistry and Chemical Engineering, Hu'nan University , Changsha , China and
| |
Collapse
|
43
|
2,3-Dihydroquinazolin-4(1H)-ones: Visible light mediated synthesis, solvatochromism and biological activity. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2015; 143:139-47. [DOI: 10.1016/j.jphotobiol.2014.12.028] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2014] [Revised: 12/12/2014] [Accepted: 12/18/2014] [Indexed: 01/11/2023]
|
44
|
Benchekroun M, Bartolini M, Egea J, Romero A, Soriano E, Pudlo M, Luzet V, Andrisano V, Jimeno ML, López MG, Wehle S, Gharbi T, Refouvelet B, de Andrés L, Herrera-Arozamena C, Monti B, Bolognesi ML, Rodríguez-Franco MI, Decker M, Marco-Contelles J, Ismaili L. Novel Tacrine-Grafted Ugi Adducts as Multipotent Anti-Alzheimer Drugs: A Synthetic Renewal in Tacrine-Ferulic Acid Hybrids. ChemMedChem 2014; 10:523-39. [DOI: 10.1002/cmdc.201402409] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Indexed: 01/09/2023]
|
45
|
Khan I, Ibrar A, Ahmed W, Saeed A. Synthetic approaches, functionalization and therapeutic potential of quinazoline and quinazolinone skeletons: the advances continue. Eur J Med Chem 2014; 90:124-69. [PMID: 25461317 DOI: 10.1016/j.ejmech.2014.10.084] [Citation(s) in RCA: 270] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Revised: 10/10/2014] [Accepted: 10/31/2014] [Indexed: 12/18/2022]
Abstract
The presence of N-heterocycles as an essential structural motif in a variety of biologically active substances has stimulated the development of new strategies and technologies for their synthesis. Among the various N-heterocyclic scaffolds, quinazolines and quinazolinones form a privileged class of compounds with their diverse spectrum of therapeutic potential. The easy generation of complex molecular diversity through broadly applicable, cost-effective, practical and sustainable synthetic methods in a straightforward fashion along with the importance of these motifs in medicinal chemistry, received significant attention from researchers engaged in drug design and heterocyclic methodology development. In this perspective, the current review article is an effort to recapitulate recent developments in the eco-friendly and green procedures for the construction of highly challenging and potentially bioactive quinazoline and quinazolinone compounds in order to help medicinal chemists in designing and synthesizing novel and potent compounds for the treatment of different disorders. The key mechanistic insights for the synthesis of these heterocycles along with potential applications and manipulations of the products have also been conferred. This article also aims to highlight the promising future directions for the easy access to these frameworks in addition to the identification of more potent and specific products for numerous biological targets.
Collapse
Affiliation(s)
- Imtiaz Khan
- School of Chemistry, University of Nottingham, University Park, Nottingham NG7 2RD, United Kingdom
| | - Aliya Ibrar
- Department of Chemistry, Quaid-i-Azam University, Islamabad 45320, Pakistan
| | - Waqas Ahmed
- Office of Research, Innovation and Commercialization, University of Gujrat, Gujrat 50700, Pakistan
| | - Aamer Saeed
- Department of Chemistry, Quaid-i-Azam University, Islamabad 45320, Pakistan.
| |
Collapse
|