1
|
Jiao Y, Zhang X, Duan L, Cheng R, Yang N, Peng Z, Li B, Xu L, Chen W, Chen J, Liu Y, Yan H. Association of plasma zinc and copper levels with mild cognitive impairment in patients with type 2 diabetes. Front Nutr 2025; 12:1532080. [PMID: 40144573 PMCID: PMC11936807 DOI: 10.3389/fnut.2025.1532080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 02/27/2025] [Indexed: 03/28/2025] Open
Abstract
Background Type 2 diabetes mellitus (T2DM) is a significant risk factor for cognitive impairment. Zinc deficiency contributes to T2DM development, while copper may exacerbate diabetes through prooxidant mechanisms. Higher zinc levels may protect against copper toxicity. This study investigates the association of plasma zinc and copper levels with mild cognitive impairment (MCI) in T2DM patients. Methods T2DM patients admitted to Tongji Hospital from 2012 to 2018 were classified into MCI (n = 136) and control (n = 136) groups, matched by age (± 3 years) and gender. Conditional logistic regression was used to assess the associations between plasma zinc, copper levels and MCI. A generalized additive model (GAM) evaluated the dose-response relationship between plasma zinc, copper levels and Mini-Mental State Examination (MMSE) scores. Results The median of plasma metal levels in MCI and control groups were 831.31 μg/L and 936.29 μg/L for zinc, 932.07 μg/L and 860.47 μg/L for copper, and 0.91 and 1.11 for the zinc-to-copper (Zn/Cu) ratio. Compared to participants in the lowest tertile, the multivariable-adjusted odds ratios with 95% confidence intervals (CI) for MCI in the highest tertile were 0.33 (0.13, 0.79) for zinc, 3.56 (1.42, 8.94) for copper, and 0.37 (0.15, 0.93) for the Zn/Cu ratio. Plasma Aβ40 levels were significantly lower (p = 0.009) and plasma Aβ42/40 levels were significantly higher (p = 0.008) in MCI group compared with those in control group. Zinc concentration was positively associated with Aβ42. For per SD (327.71 μg/L) increase in plasma zinc levels, the percent change (95% CI) of Aβ42 were 2.90 (0.85, 4.99). Conclusion Higher plasma zinc levels and higher Zn/Cu ratio were associated with lower odds of MCI in T2DM patients, while higher copper levels increased the risk of MCI. This study provides insights on plasma zinc, copper, and Zn/Cu ratio and Aβ of MCI, further studies are needed to clarify the underlying mechanisms for novel therapies that could prevent or cure multiple T2DM-related cognitive impairments.
Collapse
Affiliation(s)
- Yang Jiao
- Department of Health Toxicology, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xing Zhang
- Department of Health Toxicology, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lian Duan
- Department of Health Toxicology, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ruijie Cheng
- Department of Health Toxicology, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ning Yang
- Department of Health Toxicology, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhao Peng
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ben Li
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Changsha Institute for Food and Drug Control, Changsha, China
| | - Lu Xu
- Xiangyang Public Inspection and Testing Center, Xiangyang, China
| | - Wenwen Chen
- Department of Health Toxicology, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jingrong Chen
- Department of Health Toxicology, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yanchao Liu
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hong Yan
- Department of Health Toxicology, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
2
|
Talukder MEK, Akhter S, Ahammad F, Aktar A, Islam MS, Laboni AA, Afroze M, Khan M, Uddin MJ, Rahman MM. Multi-modal neuroprotection of Argemone mexicana L. against Alzheimer's disease: In vitro and in silico study. Heliyon 2024; 10:e37178. [PMID: 39286063 PMCID: PMC11402773 DOI: 10.1016/j.heliyon.2024.e37178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 08/28/2024] [Indexed: 09/19/2024] Open
Abstract
Argemone mexicana L. is a medicinal plant, but its impact on Alzheimer's disease (AD) is right now undetermined. We intended to investigate the in-vitro anti-AD potential of leaves and flowers of A. mexicana methanol, ethanol, and ethyl extracts and to identify multi-modal anti-AD phytochemicals by computational approaches. Molecular docking of 196 phytochemicals identified three hit phytochemicals (protoberberine, protopine, and codeine) with higher binding affinity and multi-targeting ability toward AChE, BChE, BACE-1, and GSK-3β. Further MM-GBSA assays confirmed the integrity of these phytochemicals as the hit phytochemicals. However, these phytochemicals demonstrated favorable pharmacokinetics (PK) and drugable properties having no toxicity. Molecular dynamics simulations confirmed the binding strength of the hit phytoconstituents in the active pockets of AChE, BChE, BACE-1, and GSK-3β with multi-targeting inhibitory activities. All the extracts exhibited dose-dependent antioxidant and anti-cholinesterase activities supporting the in silico results in the context of oxidative stress and cholinergic pathways. Our results offer scientific validation of the anti-AD properties of Argemone mexicana L. and identified protoberberine, protopine, and codeine that could be used for the development of multi-modal inhibitors of AChE, BChE, BACE-1, and GSK-3β to combat AD. Additional in vivo validation is recommended to ensure a thorough assessment in the present research.
Collapse
Affiliation(s)
- Md Enamul Kabir Talukder
- Molecular and Cellular Biology Laboratory, Department of Genetic Engineering and Biotechnology, Jashore University of Science and Technology, Jashore, 7408, Bangladesh
| | - Shahina Akhter
- Department of Biochemistry and Biotechnology, University of Science and Technology Chittagong (USTC), Foy's Lake, Chittagong, 4202, Bangladesh
| | - Foysal Ahammad
- Laboratory of Computational Biology, Biological Solution Centre (BioSol Centre), Jashore, 7408, Bangladesh
| | - Asmim Aktar
- Molecular and Cellular Biology Laboratory, Department of Genetic Engineering and Biotechnology, Jashore University of Science and Technology, Jashore, 7408, Bangladesh
| | - Md Saidul Islam
- Korea Institute of Radiological & Medical Sciences, 75, Nowon-ro, Nowon-gu, Seoul, South Korea
| | - Aysha Akter Laboni
- Bangladesh Reference Institute for Chemical Measurements (BRICM), Bangladesh Council of Scientific and Industrial Research, Dr Qudrat-i-Khuda Road, Dhanmondi, Dhaka, 1205, Bangladesh
| | - Mirola Afroze
- Bangladesh Reference Institute for Chemical Measurements (BRICM), Bangladesh Council of Scientific and Industrial Research, Dr Qudrat-i-Khuda Road, Dhanmondi, Dhaka, 1205, Bangladesh
| | - Mala Khan
- Bangladesh Reference Institute for Chemical Measurements (BRICM), Bangladesh Council of Scientific and Industrial Research, Dr Qudrat-i-Khuda Road, Dhanmondi, Dhaka, 1205, Bangladesh
| | - Mohammad Jashim Uddin
- Department of Pharmacy, Jashore University of Science and Technology, Jashore, 7408, Bangladesh
| | - Md Mashiar Rahman
- Molecular and Cellular Biology Laboratory, Department of Genetic Engineering and Biotechnology, Jashore University of Science and Technology, Jashore, 7408, Bangladesh
| |
Collapse
|
3
|
Hahn DF, Gapsys V, de Groot BL, Mobley DL, Tresadern G. Current State of Open Source Force Fields in Protein-Ligand Binding Affinity Predictions. J Chem Inf Model 2024; 64:5063-5076. [PMID: 38895959 PMCID: PMC11234369 DOI: 10.1021/acs.jcim.4c00417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 04/23/2024] [Accepted: 04/25/2024] [Indexed: 06/21/2024]
Abstract
In drug discovery, the in silico prediction of binding affinity is one of the major means to prioritize compounds for synthesis. Alchemical relative binding free energy (RBFE) calculations based on molecular dynamics (MD) simulations are nowadays a popular approach for the accurate affinity ranking of compounds. MD simulations rely on empirical force field parameters, which strongly influence the accuracy of the predicted affinities. Here, we evaluate the ability of six different small-molecule force fields to predict experimental protein-ligand binding affinities in RBFE calculations on a set of 598 ligands and 22 protein targets. The public force fields OpenFF Parsley and Sage, GAFF, and CGenFF show comparable accuracy, while OPLS3e is significantly more accurate. However, a consensus approach using Sage, GAFF, and CGenFF leads to accuracy comparable to OPLS3e. While Parsley and Sage are performing comparably based on aggregated statistics across the whole dataset, there are differences in terms of outliers. Analysis of the force field reveals that improved parameters lead to significant improvement in the accuracy of affinity predictions on subsets of the dataset involving those parameters. Lower accuracy can not only be attributed to the force field parameters but is also dependent on input preparation and sampling convergence of the calculations. Especially large perturbations and nonconverged simulations lead to less accurate predictions. The input structures, Gromacs force field files, as well as the analysis Python notebooks are available on GitHub.
Collapse
Affiliation(s)
- David F. Hahn
- Computational
Chemistry, Janssen Research & Development, Turnhoutseweg 30, Beerse 2340, Belgium
| | - Vytautas Gapsys
- Computational
Chemistry, Janssen Research & Development, Turnhoutseweg 30, Beerse 2340, Belgium
- Computational
Biomolecular Dynamics Group, Max Planck
Institute for Multidisciplinary Sciences, Am Fassberg 11, Göttingen 37077, Germany
| | - Bert L. de Groot
- Computational
Biomolecular Dynamics Group, Max Planck
Institute for Multidisciplinary Sciences, Am Fassberg 11, Göttingen 37077, Germany
| | - David L. Mobley
- Department
of Chemistry, University of California, Irvine, California 92697, United States
- Department
of Pharmaceutical Sciences, University of
California, Irvine, California 92697, United States
| | - Gary Tresadern
- Computational
Chemistry, Janssen Research & Development, Turnhoutseweg 30, Beerse 2340, Belgium
| |
Collapse
|
4
|
Narayanan AP, Jayan J, Sudevan ST, Dhyani A, Zachariah SM, Mathew B. Flavonoid and Chalcone Scaffolds as Inhibitors of BACE1: Recent Updates. Comb Chem High Throughput Screen 2024; 27:1243-1256. [PMID: 37519205 DOI: 10.2174/1386207326666230731092409] [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: 06/26/2022] [Revised: 02/27/2023] [Accepted: 03/08/2023] [Indexed: 08/01/2023]
Abstract
Flavonoids and chalcones are two major classes of chemical moieties that have a vast background of pharmacological activities. Chalcone is a subclass of flavonoids whose therapeutic potential has been implicated due to an array of bioactivities. A lot of research works have shown interest in investigating the neuroprotective effect of these molecules, and have revealed them to be much more potent molecules that can be used to treat neurodegenerative disorders. Beta-site APP cleaving enzyme (BACE1), which is majorly found in the brain, is one of the reasons behind the development of Alzheimer's disease (AD). Flavonoids and chalcones have proven clinical data that they inhibit the production of Aβ plaques that are involved in the progression of AD. In this article, we have provided a detailed chronological review of the research work on the BACE1 inhibiting potency of both flavonoids and chalcones. Almost all the flavonoids and chalcones mentioned in this article have shown very good in vitro and in vivo BACE1 inhibiting activity. The docking studies and the structural importance of some BACE1-inhibiting flavonoids, as well as chalcones, are also mentioned here.
Collapse
Affiliation(s)
- Anishma Payyappilliparambil Narayanan
- Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, Kochi, Kerala-682041, India
| | - Jayalakshmi Jayan
- Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, Kochi, Kerala-682041, India
| | - Sachithra Thazhathuveedu Sudevan
- Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, Kochi, Kerala-682041, India
| | - Archana Dhyani
- School of Pharmacy, Graphic Era Hill University, Dehradun, 248007, Uttarakhand, India
| | - Subin Mary Zachariah
- Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, Kochi, Kerala-682041, India
| | - Bijo Mathew
- Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, Kochi, Kerala-682041, India
| |
Collapse
|
5
|
Baumann H, Dybeck E, McClendon CL, Pickard FC, Gapsys V, Pérez-Benito L, Hahn DF, Tresadern G, Mathiowetz AM, Mobley DL. Broadening the Scope of Binding Free Energy Calculations Using a Separated Topologies Approach. J Chem Theory Comput 2023; 19:5058-5076. [PMID: 37487138 PMCID: PMC10413862 DOI: 10.1021/acs.jctc.3c00282] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Indexed: 07/26/2023]
Abstract
Binding free energy calculations predict the potency of compounds to protein binding sites in a physically rigorous manner and see broad application in prioritizing the synthesis of novel drug candidates. Relative binding free energy (RBFE) calculations have emerged as an industry-standard approach to achieve highly accurate rank-order predictions of the potency of related compounds; however, this approach requires that the ligands share a common scaffold and a common binding mode, restricting the methods' domain of applicability. This is a critical limitation since complex modifications to the ligands, especially core hopping, are very common in drug design. Absolute binding free energy (ABFE) calculations are an alternate method that can be used for ligands that are not congeneric. However, ABFE suffers from a known problem of long convergence times due to the need to sample additional degrees of freedom within each system, such as sampling rearrangements necessary to open and close the binding site. Here, we report on an alternative method for RBFE, called Separated Topologies (SepTop), which overcomes the issues in both of the aforementioned methods by enabling large scaffold changes between ligands with a convergence time comparable to traditional RBFE. Instead of only mutating atoms that vary between two ligands, this approach performs two absolute free energy calculations at the same time in opposite directions, one for each ligand. Defining the two ligands independently allows the comparison of the binding of diverse ligands without the artificial constraints of identical poses or a suitable atom-atom mapping. This approach also avoids the need to sample the unbound state of the protein, making it more efficient than absolute binding free energy calculations. Here, we introduce an implementation of SepTop. We developed a general and efficient protocol for running SepTop, and we demonstrated the method on four diverse, pharmaceutically relevant systems. We report the performance of the method, as well as our practical insights into the strengths, weaknesses, and challenges of applying this method in an industrial drug design setting. We find that the accuracy of the approach is sufficiently high to rank order ligands with an accuracy comparable to traditional RBFE calculations while maintaining the additional flexibility of SepTop.
Collapse
Affiliation(s)
- Hannah
M. Baumann
- Department
of Pharmaceutical Sciences, University of
California, Irvine, Irvine, California 92697, United States
| | - Eric Dybeck
- Pfizer
Worldwide Research, Development, and Medical, 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - Christopher L. McClendon
- Pfizer
Worldwide Research, Development, and Medical, 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - Frank C. Pickard
- Pfizer
Worldwide Research, Development, and Medical, 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - Vytautas Gapsys
- Computational
Chemistry, Janssen Research & Development, Janssen Pharmaceutica N. V., Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Laura Pérez-Benito
- Computational
Chemistry, Janssen Research & Development, Janssen Pharmaceutica N. V., Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - David F. Hahn
- Computational
Chemistry, Janssen Research & Development, Janssen Pharmaceutica N. V., Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Gary Tresadern
- Computational
Chemistry, Janssen Research & Development, Janssen Pharmaceutica N. V., Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Alan M. Mathiowetz
- Pfizer
Worldwide Research, Development, and Medical, 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - David L. Mobley
- Department
of Pharmaceutical Sciences, University of
California, Irvine, Irvine, California 92697, United States
- Department
of Chemistry, University of California,
Irvine, Irvine, California 92697, United States
| |
Collapse
|
6
|
Guanidine-based β amyloid precursor protein cleavage enzyme 1 (BACE-1) inhibitors for the Alzheimer's disease (AD): A review. Bioorg Med Chem 2022; 74:117047. [DOI: 10.1016/j.bmc.2022.117047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 09/16/2022] [Accepted: 10/04/2022] [Indexed: 11/02/2022]
|
7
|
Bieniek MK, Cree B, Pirie R, Horton JT, Tatum NJ, Cole DJ. An open-source molecular builder and free energy preparation workflow. Commun Chem 2022; 5:136. [PMID: 36320862 PMCID: PMC9607723 DOI: 10.1038/s42004-022-00754-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 10/11/2022] [Indexed: 01/27/2023] Open
Abstract
Automated free energy calculations for the prediction of binding free energies of congeneric series of ligands to a protein target are growing in popularity, but building reliable initial binding poses for the ligands is challenging. Here, we introduce the open-source FEgrow workflow for building user-defined congeneric series of ligands in protein binding pockets for input to free energy calculations. For a given ligand core and receptor structure, FEgrow enumerates and optimises the bioactive conformations of the grown functional group(s), making use of hybrid machine learning/molecular mechanics potential energy functions where possible. Low energy structures are optionally scored using the gnina convolutional neural network scoring function, and output for more rigorous protein-ligand binding free energy predictions. We illustrate use of the workflow by building and scoring binding poses for ten congeneric series of ligands bound to targets from a standard, high quality dataset of protein-ligand complexes. Furthermore, we build a set of 13 inhibitors of the SARS-CoV-2 main protease from the literature, and use free energy calculations to retrospectively compute their relative binding free energies. FEgrow is freely available at https://github.com/cole-group/FEgrow, along with a tutorial.
Collapse
Affiliation(s)
- Mateusz K. Bieniek
- School of Natural and Environmental Sciences, Newcastle University, Newcastle upon Tyne, NE1 7RU UK
| | - Ben Cree
- School of Natural and Environmental Sciences, Newcastle University, Newcastle upon Tyne, NE1 7RU UK
| | - Rachael Pirie
- School of Natural and Environmental Sciences, Newcastle University, Newcastle upon Tyne, NE1 7RU UK
| | - Joshua T. Horton
- School of Natural and Environmental Sciences, Newcastle University, Newcastle upon Tyne, NE1 7RU UK
| | - Natalie J. Tatum
- Newcastle University Centre for Cancer, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH UK
| | - Daniel J. Cole
- School of Natural and Environmental Sciences, Newcastle University, Newcastle upon Tyne, NE1 7RU UK
| |
Collapse
|
8
|
Hahn DF, Bayly CI, Boby ML, Macdonald HEB, Chodera JD, Gapsys V, Mey ASJS, Mobley DL, Benito LP, Schindler CEM, Tresadern G, Warren GL. Best practices for constructing, preparing, and evaluating protein-ligand binding affinity benchmarks [Article v0.1]. LIVING JOURNAL OF COMPUTATIONAL MOLECULAR SCIENCE 2022; 4:1497. [PMID: 36382113 PMCID: PMC9662604 DOI: 10.33011/livecoms.4.1.1497] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/01/2023]
Abstract
Free energy calculations are rapidly becoming indispensable in structure-enabled drug discovery programs. As new methods, force fields, and implementations are developed, assessing their expected accuracy on real-world systems (benchmarking) becomes critical to provide users with an assessment of the accuracy expected when these methods are applied within their domain of applicability, and developers with a way to assess the expected impact of new methodologies. These assessments require construction of a benchmark-a set of well-prepared, high quality systems with corresponding experimental measurements designed to ensure the resulting calculations provide a realistic assessment of expected performance when these methods are deployed within their domains of applicability. To date, the community has not yet adopted a common standardized benchmark, and existing benchmark reports suffer from a myriad of issues, including poor data quality, limited statistical power, and statistically deficient analyses, all of which can conspire to produce benchmarks that are poorly predictive of real-world performance. Here, we address these issues by presenting guidelines for (1) curating experimental data to develop meaningful benchmark sets, (2) preparing benchmark inputs according to best practices to facilitate widespread adoption, and (3) analysis of the resulting predictions to enable statistically meaningful comparisons among methods and force fields. We highlight challenges and open questions that remain to be solved in these areas, as well as recommendations for the collection of new datasets that might optimally serve to measure progress as methods become systematically more reliable. Finally, we provide a curated, versioned, open, standardized benchmark set adherent to these standards (PLBenchmarks) and an open source toolkit for implementing standardized best practices assessments (arsenic) for the community to use as a standardized assessment tool. While our main focus is free energy methods based on molecular simulations, these guidelines should prove useful for assessment of the rapidly growing field of machine learning methods for affinity prediction as well.
Collapse
Affiliation(s)
- David F. Hahn
- Computational Chemistry,Janssen Research & Development, Turnhoutseweg 30, Beerse B-2340, Belgium
| | | | - Melissa L. Boby
- Computational and Systems Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065 USA
| | - Hannah E. Bruce Macdonald
- Computational and Systems Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065 USA
- MSD R&D Innovation Centre, 120 Moorgate, London EC2M 6UR, United Kingdom
| | - John D. Chodera
- Computational and Systems Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065 USA
| | - Vytautas Gapsys
- Computational Biomolecular Dynamics Group, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | - Antonia S. J. S. Mey
- EaStCHEM School of Chemistry, David Brewster Road, Joseph Black Building, The King’s Buildings, Edinburgh, EH9 3FJ, UK
| | - David L. Mobley
- Departments of Pharmaceutical Sciences and Chemistry, University of California, Irvine, CA USA
| | - Laura Perez Benito
- Computational Chemistry,Janssen Research & Development, Turnhoutseweg 30, Beerse B-2340, Belgium
| | | | - Gary Tresadern
- Computational Chemistry,Janssen Research & Development, Turnhoutseweg 30, Beerse B-2340, Belgium
| | | |
Collapse
|
9
|
Bhati A, Coveney PV. Large Scale Study of Ligand-Protein Relative Binding Free Energy Calculations: Actionable Predictions from Statistically Robust Protocols. J Chem Theory Comput 2022; 18:2687-2702. [PMID: 35293737 PMCID: PMC9009079 DOI: 10.1021/acs.jctc.1c01288] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Indexed: 12/28/2022]
Abstract
The accurate and reliable prediction of protein-ligand binding affinities can play a central role in the drug discovery process as well as in personalized medicine. Of considerable importance during lead optimization are the alchemical free energy methods that furnish an estimation of relative binding free energies (RBFE) of similar molecules. Recent advances in these methods have increased their speed, accuracy, and precision. This is evident from the increasing number of retrospective as well as prospective studies employing them. However, such methods still have limited applicability in real-world scenarios due to a number of important yet unresolved issues. Here, we report the findings from a large data set comprising over 500 ligand transformations spanning over 300 ligands binding to a diverse set of 14 different protein targets which furnish statistically robust results on the accuracy, precision, and reproducibility of RBFE calculations. We use ensemble-based methods which are the only way to provide reliable uncertainty quantification given that the underlying molecular dynamics is chaotic. These are implemented using TIES (Thermodynamic Integration with Enhanced Sampling). Results achieve chemical accuracy in all cases. Ensemble simulations also furnish information on the statistical distributions of the free energy calculations which exhibit non-normal behavior. We find that the "enhanced sampling" method known as replica exchange with solute tempering degrades RBFE predictions. We also report definitively on numerous associated alchemical factors including the choice of ligand charge method, flexibility in ligand structure, and the size of the alchemical region including the number of atoms involved in transforming one ligand into another. Our findings provide a key set of recommendations that should be adopted for the reliable application of RBFE methods.
Collapse
Affiliation(s)
- Agastya
P. Bhati
- Centre
for Computational Science, Department of Chemistry, University College London, London WC1H 0AJ, United Kingdom
| | - Peter V. Coveney
- Centre
for Computational Science, Department of Chemistry, University College London, London WC1H 0AJ, United Kingdom
- Informatics
Institute, University of Amsterdam, P.O. Box 94323, 1090 GH Amsterdam, Netherlands
| |
Collapse
|
10
|
Pirotte B, Florence X, Goffin E, Leleux F, Lebrun P. Research Advancements on Fluorinated and Non-Fluorinated 4-Phenyl(thio)ureido-Substituted 2,2-Dimethylchromans Acting as Inhibitors of Insulin Release and Smooth Muscle Relaxants. Med Chem 2022; 18:884-894. [PMID: 35189799 DOI: 10.2174/1573406418666220221145500] [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: 10/22/2021] [Revised: 11/29/2021] [Accepted: 12/21/2021] [Indexed: 11/22/2022]
Abstract
AIMS The present study aimed at characterizing the impact of the presence or absence of fluorine atoms on the phenyl and benzopyran rings of 4-phenyl(thio)ureido-substituted 2,2-dimethylchromans on their ability to inhibit insulin release from pancreatic -cells or to relax vascular smooth muscle cells. METHODS Most compounds were found to inhibit insulin secretion and to provoke a marked myorelaxant activity. RESULTS The lack of a fluorine or a chlorine atom at the 6-position of the 2,2-dimethylchroman core structure reduced the inhibitory activity on the pancreatic endocrine tissue. One of the most active compounds on both tissues, compound 11h (BPDZ 678), was selected for further pharmacological investigations. CONCLUSION The biological data suggested that 11h mainly expressed the profile of a KATP channel opener on pancreatic -cells, although a calcium entry blockade effect was also observed. On vascular smooth muscle cells, 11h behaved as a calcium entry blocker.
Collapse
Affiliation(s)
- Bernard Pirotte
- Laboratoire de Chimie Pharmaceutique, Center for Interdisciplinary Research on Medicines (CIRM), Université de Liège, Quartier Hôpital, Avenue Hippocrate 15, B-4000 Liège, Belgium
| | - Xavier Florence
- Laboratoire de Chimie Pharmaceutique, Center for Interdisciplinary Research on Medicines (CIRM), Université de Liège, Quartier Hôpital, Avenue Hippocrate 15, B-4000 Liège, Belgium
- Laboratoire de Physiologie et Pharmacologie, Université Libre de Bruxelles, Faculté de Médecine, 808 Route de Lennik, B-1070 Bruxelles, Belgium
| | - Eric Goffin
- Laboratoire de Chimie Pharmaceutique, Center for Interdisciplinary Research on Medicines (CIRM), Université de Liège, Quartier Hôpital, Avenue Hippocrate 15, B-4000 Liège, Belgium
| | - Fabienne Leleux
- Laboratoire de Physiologie et Pharmacologie, Université Libre de Bruxelles, Faculté de Médecine, 808 Route de Lennik, B-1070 Bruxelles, Belgium
| | - Philippe Lebrun
- Laboratoire de Physiologie et Pharmacologie, Université Libre de Bruxelles, Faculté de Médecine, 808 Route de Lennik, B-1070 Bruxelles, Belgium
| |
Collapse
|
11
|
Xi JM, Liao WW. Radical addition to the CC bond meets (1, n)-HAT: recent advances in the remote C(sp 3)–H or C(sp 2)–H functionalization of alkenes. Org Chem Front 2022. [DOI: 10.1039/d2qo00793b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
This review summarizes the recent development of remote C(sp3)–H bond or aldehydic C(sp2)–H functionalizations enabled by intermolecular radical addition to CC bond/(1,n)-HAT tandem sequences.
Collapse
Affiliation(s)
- Ji-Ming Xi
- Department of Organic Chemistry, College of Chemistry, Jilin University, Changchun 130012, P. R. China
| | - Wei-Wei Liao
- Department of Organic Chemistry, College of Chemistry, Jilin University, Changchun 130012, P. R. China
- State Key Laboratory of Organometallic Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 200032, P. R. China
| |
Collapse
|
12
|
Computational screening of promising beta-secretase 1 inhibitors through multi-step molecular docking and molecular dynamics simulations - Pharmacoinformatics approach. J Mol Struct 2020. [DOI: 10.1016/j.molstruc.2019.127660] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
13
|
Iraji A, Khoshneviszadeh M, Firuzi O, Khoshneviszadeh M, Edraki N. Novel small molecule therapeutic agents for Alzheimer disease: Focusing on BACE1 and multi-target directed ligands. Bioorg Chem 2020; 97:103649. [PMID: 32101780 DOI: 10.1016/j.bioorg.2020.103649] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Revised: 01/05/2020] [Accepted: 02/03/2020] [Indexed: 12/17/2022]
Abstract
Alzheimer's Disease (AD) is a progressive neurodegenerative disorder that effects 50 million people worldwide. In this review, AD pathology and the development of novel therapeutic agents targeting AD were fully discussed. In particular, common approaches to prevent Aβ production and/or accumulation in the brain including α-secretase activators, specific γ-secretase modulators and small molecules BACE1 inhibitors were reviewed. Additionally, natural-origin bioactive compounds that provide AD therapeutic advances have been introduced. Considering AD is a multifactorial disease, the therapeutic potential of diverse multi target-directed ligands (MTDLs) that combine the efficacy of cholinesterase (ChE) inhibitors, MAO (monoamine oxidase) inhibitors, BACE1 inhibitors, phosphodiesterase 4D (PDE4D) inhibitors, for the treatment of AD are also reviewed. This article also highlights descriptions on the regulator of serotonin receptor (5-HT), metal chelators, anti-aggregants, antioxidants and neuroprotective agents targeting AD. Finally, current computational methods for evaluating the structure-activity relationships (SAR) and virtual screening (VS) of AD drugs are discussed and evaluated.
Collapse
Affiliation(s)
- Aida Iraji
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahsima Khoshneviszadeh
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Omidreza Firuzi
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mehdi Khoshneviszadeh
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Medicinal Chemistry, Faculty of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Najmeh Edraki
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
14
|
Gapsys V, Pérez-Benito L, Aldeghi M, Seeliger D, van Vlijmen H, Tresadern G, de Groot BL. Large scale relative protein ligand binding affinities using non-equilibrium alchemy. Chem Sci 2019; 11:1140-1152. [PMID: 34084371 PMCID: PMC8145179 DOI: 10.1039/c9sc03754c] [Citation(s) in RCA: 154] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 12/01/2019] [Indexed: 12/14/2022] Open
Abstract
Ligand binding affinity calculations based on molecular dynamics (MD) simulations and non-physical (alchemical) thermodynamic cycles have shown great promise for structure-based drug design. However, their broad uptake and impact is held back by the notoriously complex setup of the calculations. Only a few tools other than the free energy perturbation approach by Schrödinger Inc. (referred to as FEP+) currently enable end-to-end application. Here, we present for the first time an approach based on the open-source software pmx that allows to easily set up and run alchemical calculations for diverse sets of small molecules using the GROMACS MD engine. The method relies on theoretically rigorous non-equilibrium thermodynamic integration (TI) foundations, and its flexibility allows calculations with multiple force fields. In this study, results from the Amber and Charmm force fields were combined to yield a consensus outcome performing on par with the commercial FEP+ approach. A large dataset of 482 perturbations from 13 different protein-ligand datasets led to an average unsigned error (AUE) of 3.64 ± 0.14 kJ mol-1, equivalent to Schrödinger's FEP+ AUE of 3.66 ± 0.14 kJ mol-1. For the first time, a setup is presented for overall high precision and high accuracy relative protein-ligand alchemical free energy calculations based on open-source software.
Collapse
Affiliation(s)
- Vytautas Gapsys
- Computational Biomolecular Dynamics Group, Department of Theoretical and Computational Biophysics, Max Planck Institute for Biophysical Chemistry D-37077 Göttingen Germany
| | - Laura Pérez-Benito
- Computational Chemistry, Janssen Research & Development, Janssen Pharmaceutica N. V. Turnhoutseweg 30 B-2340 Beerse Belgium
| | - Matteo Aldeghi
- Computational Biomolecular Dynamics Group, Department of Theoretical and Computational Biophysics, Max Planck Institute for Biophysical Chemistry D-37077 Göttingen Germany
| | - Daniel Seeliger
- Medicinal Chemistry, Boehringer Ingelheim Pharma GmbH & Co. KG Birkendorfer Strasse 65 D-88397 Biberach a.d. Riss Germany
| | - Herman van Vlijmen
- Computational Chemistry, Janssen Research & Development, Janssen Pharmaceutica N. V. Turnhoutseweg 30 B-2340 Beerse Belgium
| | - Gary Tresadern
- Computational Chemistry, Janssen Research & Development, Janssen Pharmaceutica N. V. Turnhoutseweg 30 B-2340 Beerse Belgium
| | - Bert L de Groot
- Computational Biomolecular Dynamics Group, Department of Theoretical and Computational Biophysics, Max Planck Institute for Biophysical Chemistry D-37077 Göttingen Germany
| |
Collapse
|
15
|
Chen J, Wang J, Yin B, Pang L, Wang W, Zhu W. Molecular Mechanism of Binding Selectivity of Inhibitors toward BACE1 and BACE2 Revealed by Multiple Short Molecular Dynamics Simulations and Free-Energy Predictions. ACS Chem Neurosci 2019; 10:4303-4318. [PMID: 31545898 DOI: 10.1021/acschemneuro.9b00348] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The β-amyloid cleaving enzymes 1 and 2 (BACE1 and BACE2) have been regarded as the prospective targets for clinically treating Alzheimer's disease (AD) in the last two decades. Thus, insight into the binding differences of inhibitors to BACE1 and BACE2 is of significance for designing highly selective inhibitors toward the two proteins. In this work, multiple short molecular dynamics (MSMD) simulations are coupled with the molecular mechanics generalized Born surface area (MM-GBSA) method to probe the binding selectivity of three inhibitors DBO, CS9, and SC7 on BACE1 over BACE2. The results show that the entropy effect plays a key role in selectivity identification of inhibitors toward BACE1 and BACE2, which determines that DBO has better selectivity toward BACE2 over BACE1, while CS9 and CS7 can more favorably bind to BACE1 than BACE2. The hierarchical clustering analysis based on energetic contributions of residues suggests that BACE1 and BACE2 share the common hot interaction spots. The residue-based free-energy decomposition method was applied to compute the inhibitor-residue interaction spectrum, and the results recognize four common binding subpockets corresponding to the different groups of inhibitors, which can be used as efficient targets for designing highly selective inhibitors toward BACE1 and BACE2. Therefore, these results provide a useful molecular basis and dynamics information for development of highly selective inhibitors targeting BACE1 and BACE2.
Collapse
Affiliation(s)
- Jianzhong Chen
- School of Science, Shandong Jiaotong University, Jinan 250357 China
| | - Jinan Wang
- Drug Discovery and Design Center, CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China
| | - Baohua Yin
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Laixue Pang
- School of Science, Shandong Jiaotong University, Jinan 250357 China
| | - Wei Wang
- School of Science, Shandong Jiaotong University, Jinan 250357 China
| | - Weiliang Zhu
- Drug Discovery and Design Center, CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China
| |
Collapse
|
16
|
Ganeshpurkar A, Swetha R, Kumar D, Gangaram GP, Singh R, Gutti G, Jana S, Kumar D, Kumar A, Singh SK. Protein-Protein Interactions and Aggregation Inhibitors in Alzheimer's Disease. Curr Top Med Chem 2019; 19:501-533. [PMID: 30836921 DOI: 10.2174/1568026619666190304153353] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Revised: 10/31/2018] [Accepted: 11/20/2018] [Indexed: 12/13/2022]
Abstract
BACKGROUND Alzheimer's Disease (AD), a multifaceted disorder, involves complex pathophysiology and plethora of protein-protein interactions. Thus such interactions can be exploited to develop anti-AD drugs. OBJECTIVE The interaction of dynamin-related protein 1, cellular prion protein, phosphoprotein phosphatase 2A and Mint 2 with amyloid β, etc., studied recently, may have critical role in progression of the disease. Our objective has been to review such studies and their implications in design and development of drugs against the Alzheimer's disease. METHODS Such studies have been reviewed and critically assessed. RESULTS Review has led to show how such studies are useful to develop anti-AD drugs. CONCLUSION There are several PPIs which are current topics of research including Drp1, Aβ interactions with various targets including PrPC, Fyn kinase, NMDAR and mGluR5 and interaction of Mint2 with PDZ domain, etc., and thus have potential role in neurodegeneration and AD. Finally, the multi-targeted approach in AD may be fruitful and opens a new vista for identification and targeting of PPIs in various cellular pathways to find a cure for the disease.
Collapse
Affiliation(s)
- Ankit Ganeshpurkar
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi 221005, India
| | - Rayala Swetha
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi 221005, India
| | - Devendra Kumar
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi 221005, India
| | - Gore P Gangaram
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi 221005, India
| | - Ravi Singh
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi 221005, India
| | - Gopichand Gutti
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi 221005, India
| | - Srabanti Jana
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi 221005, India
| | - Dileep Kumar
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi 221005, India
| | - Ashok Kumar
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi 221005, India
| | - Sushil K Singh
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi 221005, India
| |
Collapse
|
17
|
Yuan Q, Zhang H, Zhang X, Huang P, Liu R, Song YQ, Wu W, Lin ZX. Association Between Axonopathy and Amyloid Plaques in the Spinal Cord of the Transgenic Mice of Alzheimer's Disease. Neuroscience 2019; 409:152-161. [PMID: 31034974 DOI: 10.1016/j.neuroscience.2019.04.037] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 04/12/2019] [Accepted: 04/16/2019] [Indexed: 11/29/2022]
Abstract
Axonopathy manifested by axon swellings might constitute one of the earliest pathological features of Alzheimer's disease. It has been proposed that axonopathy might be associated with the origin of Aβ plaques. However, how axonopathy leads to Aβ plaque pathogenesis remains elusive. Our previous studies have shown that Aβ neuropathology (mainly diffuse plaques) selectively occurred in the regions of corticospinal tract (CST) pathway and its innervated region in the spinal cord of TgCRND8 mice. In this study, we investigated the occurrence and progression of axonopathy and the possible implication in Aβ plaque pathogenesis in the spinal cord of TgCRND8 mice. By anterograde labeling of CST system with a neuroanatomical tracer, we found that dilated corticospinal axons started to appear at 7 months, then exhibited an age-dependent increase. These abnormal structures appear before any plaque deposits are visible in the spinal cord of the mice. Importantly, they colocalized with Aβ plaques in either the white matter or gray matter of the spinal cord at later stages, suggesting that these axonal swellings might represent the initial stages of Aβ plaque formation, and could play a role in Aβ plaque pathogenesis. Furthermore, using ultrastructural analysis we demonstrated that intracellular contents in the axonal dystrophies such as various dense vesicles leaked out into the extracellular matrix under a condition of axon swelling rupture in CST pathways of spinal cord. This provided precise structural evidence that how the Aβ leaks out from the axonal dystrophies into extracellular matrix and how an axonal swelling might serve as a nidus of amyloid plaque formation.
Collapse
Affiliation(s)
- Qiuju Yuan
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong; Brain Research Centre, Faculty of Medicine, The Chinese University of Hong Kong.
| | - Hongwei Zhang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong
| | - Xie Zhang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong
| | - Pengyun Huang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong
| | - Rong Liu
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong
| | - You-Qiang Song
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong
| | - Wutian Wu
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong; GHM Institute of CNS regeneration, Jinan University, Guangzhou, China
| | - Zhi-Xiu Lin
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong; Brain Research Centre, Faculty of Medicine, The Chinese University of Hong Kong.
| |
Collapse
|
18
|
Fujimoto K, Matsuoka E, Asada N, Tadano G, Yamamoto T, Nakahara K, Fuchino K, Ito H, Kanegawa N, Moechars D, Gijsen HJM, Kusakabe KI. Structure-Based Design of Selective β-Site Amyloid Precursor Protein Cleaving Enzyme 1 (BACE1) Inhibitors: Targeting the Flap to Gain Selectivity over BACE2. J Med Chem 2019; 62:5080-5095. [DOI: 10.1021/acs.jmedchem.9b00309] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
19
|
Yu Y, Yin T, Peng Q, Kong L, Li C, Tang D, Yin X. Simultaneous Monitoring of Amyloid-β (Aβ) Oligomers and Fibrils for Effectively Evaluating the Dynamic Process of Aβ Aggregation. ACS Sens 2019; 4:471-478. [PMID: 30693761 DOI: 10.1021/acssensors.8b01493] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Herein, we provide a proof of concept for a novel strategy that targets the assessment of the aggregation of amyloid-β (Aβ) by simultaneously determining its oligomers (Aβo) and fibrils (Aβf) in one analytical system. By fabricating and combining two immunosensors for Aβo and Aβf, respectively, we constructed a two-channel electrochemical system. The ratio of Aβf to Aβo was calculated and taken as a possible criterion for evaluating the extent of aggregation. Thereby, the presence of and transformation between oligomers and fibrils were accurately probed by incubating the Aβ monomer for different times and then calculating the ratios of Aβf to Aβo. The applicability of this method was further validated by tracking the dynamic progress of Aβ aggregation in the cerebrospinal fluid and tissues of Alzheimer's disease (AD) rats, which revealed that the ratio of Aβf to Aβo in rat brain gradually increased with the progression of AD, which was indicative of the severity of peptide aggregation during this process. Overall, this study represents the first example of a quantitative strategy for precisely evaluating the aggregation process that is related to pathological events in AD brain.
Collapse
Affiliation(s)
- Yanyan Yu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou 221004, Jiangsu, P. R. China
- Department of Pharmaceutical Analysis, School of Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou 221004, Jiangsu, P. R. China
| | - Tianxiao Yin
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou 221004, Jiangsu, P. R. China
| | - Qiwen Peng
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou 221004, Jiangsu, P. R. China
| | - Lingna Kong
- Department of Chemistry, East China Normal University, 500 Dongchuan Road, Shanghai 200241, P. R. China
| | - Chenglin Li
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou 221004, Jiangsu, P. R. China
| | - Daoquan Tang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou 221004, Jiangsu, P. R. China
- Department of Pharmaceutical Analysis, School of Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou 221004, Jiangsu, P. R. China
| | - Xiaoxing Yin
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou 221004, Jiangsu, P. R. China
| |
Collapse
|
20
|
Wang YB, Xie JQ, Liu W, Zhang RZ, Huang SH, Xing YH. BACE1 gene silencing alleviates isoflurane anesthesia‑induced postoperative cognitive dysfunction in immature rats by activating the PI3K/Akt signaling pathway. Mol Med Rep 2018; 18:4259-4270. [PMID: 30221701 PMCID: PMC6172366 DOI: 10.3892/mmr.2018.9453] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 08/01/2018] [Indexed: 12/29/2022] Open
Abstract
Postoperative cognitive dysfunction (POCD) is a severe complication characterized by cognitive dysfunction following anesthesia and surgery. The aim of the present study was to investigate the effects of β-site amyloid precursor protein cleavage enzyme 1 (BACE1) gene silencing on isoflurane anesthesia-induced POCD in immature rats via the phosphatidylinositol-3-kinase (PI3K)/protein kinase B (Akt) signaling pathway. Rat models were established and then transfected with BACE1 small interfering RNA and wortmannin (an inhibitor of PI3K). Blood gas analysis was performed, and a series of behavioral experiments were conducted to evaluate the cognitive function, learning ability and locomotor activity of rats. Reverse transcription quantitative polymerase chain reaction and western blot analysis were employed to determine the mRNA and protein expression of the associated genes. An ELISA was used to detect the inflammatory indicators and the content of amyloid precursor protein (APP) and amyloid-β (Aβ). Apoptosis of the hippocampal CA1 region was observed by terminal deoxynucleotidyl transferase dUTP nick-end labeling staining. Initially, it was revealed that the percentage of stagnation time in rats was increased by BACE1 gene silencing; the escape latency and swimming distance were markedly reduced from the 4th to the 6th day, the time the rats spent in first passing the target area was shortened, and the times of passing the target area were increased by BACE1 gene silencing, demonstrating that BACE1 gene silencing enhanced the spatial memory ability of rats. Additionally, it was determined that silencing BACE1 improved the pathological state induced by isoflurane anesthesia in immature rats, and attenuated the inflammatory response and the levels of APP and Aβ in hippocampal tissues. Furthermore, it was suggested that silencing BACE1 may have promoted the activation of the PI3K/Akt signaling pathway, thereby inhibiting the apoptosis of the hippocampal CA1 region. Taken together, these results indicated that BACE1 gene silencing may improve isoflurane anesthesia-induced POCD in immature rats by activating the PI3K/Akt signaling pathway and inhibiting the Aβ generated by APP.
Collapse
Affiliation(s)
- Ying-Bin Wang
- Department of Anesthesiology, Lanzhou University Second Hospital, Lanzhou, Gansu 730030, P.R. China
| | - Jian-Qin Xie
- Department of Anesthesiology, Lanzhou University Second Hospital, Lanzhou, Gansu 730030, P.R. China
| | - Wei Liu
- Department of Anesthesiology, Lanzhou University Second Hospital, Lanzhou, Gansu 730030, P.R. China
| | - Rong-Zhi Zhang
- Department of Anesthesiology, Lanzhou University Second Hospital, Lanzhou, Gansu 730030, P.R. China
| | - Sheng-Hui Huang
- Department of Anesthesiology, Lanzhou University Second Hospital, Lanzhou, Gansu 730030, P.R. China
| | - Yan-Hong Xing
- Department of Anesthesiology, Lanzhou University Second Hospital, Lanzhou, Gansu 730030, P.R. China
| |
Collapse
|
21
|
Coimbra JRM, Marques DFF, Baptista SJ, Pereira CMF, Moreira PI, Dinis TCP, Santos AE, Salvador JAR. Highlights in BACE1 Inhibitors for Alzheimer's Disease Treatment. Front Chem 2018; 6:178. [PMID: 29881722 PMCID: PMC5977085 DOI: 10.3389/fchem.2018.00178] [Citation(s) in RCA: 122] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 05/04/2018] [Indexed: 12/22/2022] Open
Abstract
Alzheimer's disease (AD) is a severe neurodegenerative disorder and the most common type of dementia in the elderly. The clinical symptoms of AD include a progressive loss of memory and impairment of cognitive functions interfering with daily life activities. The main neuropathological features consist in extracellular amyloid-β (Aβ) plaque deposition and intracellular Neurofibrillary tangles (NFTs) of hyperphosphorylated Tau. Understanding the pathophysiological mechanisms that underlie neurodegeneration in AD is essential for rational design of neuroprotective agents able to prevent disease progression. According to the "Amyloid Cascade Hypothesis" the critical molecular event in the pathogenesis of AD is the accumulation of Aβ neurotoxic oligomers. Since the proteolytic processing of Amyloid Precursor Protein (APP) by β-secretase (beta-site APP cleaving enzyme 1, BACE1) is the rate-limiting step in the production of Aβ, this enzyme is considered a major therapeutic target and BACE1 inhibitors have the potential to be disease-modifying drugs for AD treatment. Therefore, intensive efforts to discover and develop inhibitors that can reach the brain and effectively inhibit BACE1 have been pursued by several groups worldwide. The aim of this review is to highlight the progress in the discovery of potent and selective small molecule BACE1 inhibitors over the past decade.
Collapse
Affiliation(s)
- Judite R. M. Coimbra
- Laboratory of Pharmaceutical Chemistry, Faculty of Pharmacy, University of CoimbraCoimbra, Portugal
- Center for Neuroscience and Cell Biology, University of CoimbraCoimbra, Portugal
| | - Daniela F. F. Marques
- Laboratory of Pharmaceutical Chemistry, Faculty of Pharmacy, University of CoimbraCoimbra, Portugal
- Center for Neuroscience and Cell Biology, University of CoimbraCoimbra, Portugal
| | - Salete J. Baptista
- Laboratory of Pharmaceutical Chemistry, Faculty of Pharmacy, University of CoimbraCoimbra, Portugal
- Center for Neuroscience and Cell Biology, University of CoimbraCoimbra, Portugal
- Chem4Pharma, Edifício IPN IncubadoraCoimbra, Portugal
| | - Cláudia M. F. Pereira
- Center for Neuroscience and Cell Biology, University of CoimbraCoimbra, Portugal
- Faculty of Medicine, University of CoimbraCoimbra, Portugal
| | - Paula I. Moreira
- Center for Neuroscience and Cell Biology, University of CoimbraCoimbra, Portugal
- Laboratory of Physiology, Faculty of Medicine, University of CoimbraCoimbra, Portugal
| | - Teresa C. P. Dinis
- Center for Neuroscience and Cell Biology, University of CoimbraCoimbra, Portugal
- Laboratory of Biochemistry, Faculty of Pharmacy, University of CoimbraCoimbra, Portugal
| | - Armanda E. Santos
- Center for Neuroscience and Cell Biology, University of CoimbraCoimbra, Portugal
- Laboratory of Biochemistry, Faculty of Pharmacy, University of CoimbraCoimbra, Portugal
| | - Jorge A. R. Salvador
- Laboratory of Pharmaceutical Chemistry, Faculty of Pharmacy, University of CoimbraCoimbra, Portugal
- Center for Neuroscience and Cell Biology, University of CoimbraCoimbra, Portugal
| |
Collapse
|
22
|
Nakahara K, Fuchino K, Komano K, Asada N, Tadano G, Hasegawa T, Yamamoto T, Sako Y, Ogawa M, Unemura C, Hosono M, Ito H, Sakaguchi G, Ando S, Ohnishi S, Kido Y, Fukushima T, Dhuyvetter D, Borghys H, Gijsen HJM, Yamano Y, Iso Y, Kusakabe KI. Discovery of Potent and Centrally Active 6-Substituted 5-Fluoro-1,3-dihydro-oxazine β-Secretase (BACE1) Inhibitors via Active Conformation Stabilization. J Med Chem 2018; 61:5525-5546. [DOI: 10.1021/acs.jmedchem.8b00011] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
23
|
Fuchino K, Mitsuoka Y, Masui M, Kurose N, Yoshida S, Komano K, Yamamoto T, Ogawa M, Unemura C, Hosono M, Ito H, Sakaguchi G, Ando S, Ohnishi S, Kido Y, Fukushima T, Miyajima H, Hiroyama S, Koyabu K, Dhuyvetter D, Borghys H, Gijsen HJM, Yamano Y, Iso Y, Kusakabe KI. Rational Design of Novel 1,3-Oxazine Based β-Secretase (BACE1) Inhibitors: Incorporation of a Double Bond To Reduce P-gp Efflux Leading to Robust Aβ Reduction in the Brain. J Med Chem 2018; 61:5122-5137. [DOI: 10.1021/acs.jmedchem.8b00002] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
24
|
Johansson P, Kaspersson K, Gurrell IK, Bäck E, Eketjäll S, Scott CW, Cebers G, Thorne P, McKenzie MJ, Beaton H, Davey P, Kolmodin K, Holenz J, Duggan ME, Budd Haeberlein S, Bürli RW. Toward β-Secretase-1 Inhibitors with Improved Isoform Selectivity. J Med Chem 2018; 61:3491-3502. [DOI: 10.1021/acs.jmedchem.7b01716] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Patrik Johansson
- Discovery Sciences, IMED Biotech Unit, AstraZeneca, S-43183 Mölndal, Sweden
| | - Karin Kaspersson
- Discovery Sciences, IMED Biotech Unit, AstraZeneca, S-43183 Mölndal, Sweden
| | - Ian K. Gurrell
- Neuroscience, IMED Biotech Unit, AstraZeneca, Cambridge CB21 6GH, U.K
| | - Elisabeth Bäck
- Discovery Sciences, IMED Biotech Unit, AstraZeneca, S-43183 Mölndal, Sweden
| | - Susanna Eketjäll
- Cardiovascular and Metabolic Diseases, IMED Biotech Unit, AstraZeneca, 141 57 Huddinge, Sweden
| | - Clay W. Scott
- Discovery Safety, Drug Safety and Metabolism, IMED Biotech Unit, AstraZeneca, Waltham, Massachusetts 02451, United States
| | - Gvido Cebers
- Neuroscience, IMED Biotech Unit, AstraZeneca, Cambridge CB21 6GH, U.K
| | | | | | | | - Paul Davey
- Oncology Chemistry, IMED Biotech Unit, AstraZeneca, Cambridge CB4 0WG, U.K
| | | | - Jörg Holenz
- Neuroscience, IMED Biotech Unit, AstraZeneca, Cambridge CB21 6GH, U.K
| | - Mark E. Duggan
- Neuroscience, IMED Biotech Unit, AstraZeneca, Cambridge CB21 6GH, U.K
| | | | - Roland W. Bürli
- Neuroscience, IMED Biotech Unit, AstraZeneca, Cambridge CB21 6GH, U.K
| |
Collapse
|
25
|
Pirotte B, Florence X, Goffin E, Lebrun P. Deciphering Structure-Activity Relationships in a Series of 2,2-Dimethylchromans Acting as Inhibitors of Insulin Release and Smooth Muscle Relaxants. ChemMedChem 2017; 12:1810-1817. [DOI: 10.1002/cmdc.201700409] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 09/20/2017] [Indexed: 11/09/2022]
Affiliation(s)
- Bernard Pirotte
- Laboratoire de Chimie Pharmaceutique; Center for Interdisciplinary Research on Medicines (CIRM); Université de Liège; Quartier Hôpital, Avenue Hippocrate 15 4000 Liège Belgium
| | - Xavier Florence
- Laboratoire de Chimie Pharmaceutique; Center for Interdisciplinary Research on Medicines (CIRM); Université de Liège; Quartier Hôpital, Avenue Hippocrate 15 4000 Liège Belgium
- Laboratoire de Physiologie et Pharmacologie; Université Libre de Bruxelles; Faculté de Médecine; 808 Route de Lennik 1070 Bruxelles Belgium
| | - Eric Goffin
- Laboratoire de Chimie Pharmaceutique; Center for Interdisciplinary Research on Medicines (CIRM); Université de Liège; Quartier Hôpital, Avenue Hippocrate 15 4000 Liège Belgium
| | - Philippe Lebrun
- Laboratoire de Physiologie et Pharmacologie; Université Libre de Bruxelles; Faculté de Médecine; 808 Route de Lennik 1070 Bruxelles Belgium
| |
Collapse
|
26
|
Prati F, Bottegoni G, Bolognesi ML, Cavalli A. BACE-1 Inhibitors: From Recent Single-Target Molecules to Multitarget Compounds for Alzheimer’s Disease. J Med Chem 2017; 61:619-637. [DOI: 10.1021/acs.jmedchem.7b00393] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Federica Prati
- Drug Discovery Unit,
Division of Biological Chemistry and Drug Discovery, College of Life
Sciences, University of Dundee, Dow Street, Dundee, DD1 5EH, Scotland, U.K
| | - Giovanni Bottegoni
- CompuNet, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy
- Heptares Therapeutics Ltd., BioPark, Broadwater Road, Welwyn Garden City, Hertfordshire AL7 3AX, U.K
| | - Maria Laura Bolognesi
- Department
of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Via Belmeloro 6, 40126 Bologna, Italy
| | - Andrea Cavalli
- CompuNet, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy
- Department
of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Via Belmeloro 6, 40126 Bologna, Italy
| |
Collapse
|
27
|
Low JD, Bartberger MD, Chen K, Cheng Y, Fielden MR, Gore V, Hickman D, Liu Q, Allen Sickmier E, Vargas HM, Werner J, White RD, Whittington DA, Wood S, Minatti AE. Development of 2-aminooxazoline 3-azaxanthene β-amyloid cleaving enzyme (BACE) inhibitors with improved selectivity against Cathepsin D. MEDCHEMCOMM 2017; 8:1196-1206. [PMID: 30108829 PMCID: PMC6072065 DOI: 10.1039/c7md00106a] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 04/20/2017] [Indexed: 12/20/2022]
Abstract
As part of an ongoing effort at Amgen to develop a disease-modifying therapy for Alzheimer's disease, we have previously used the aminooxazoline xanthene (AOX) scaffold to generate potent and orally efficacious BACE1 inhibitors. While AOX-BACE1 inhibitors demonstrated acceptable cardiovascular safety margins, a retinal pathological finding in rat toxicological studies demanded further investigation. It has been widely postulated that such retinal toxicity might be related to off-target inhibition of Cathepsin D (CatD), a closely related aspartyl protease. We report the development of AOX-BACE1 inhibitors with improved selectivity against CatD by following a structure- and property-based approach. Our efforts culminated in the discovery of a picolinamide-substituted 3-aza-AOX-BACE1 inhibitor absent of retinal effects in an early screening rat toxicology study.
Collapse
Affiliation(s)
- Jonathan D Low
- Department of Medicinal Chemistry , Amgen Inc. , One Amgen Center Drive , Thousand Oaks , CA 91320 , USA . ; Tel: +1 805 447 4721
| | - Michael D Bartberger
- Department of Molecular Engineering , Amgen Inc. , One Amgen Center Drive , Thousand Oaks , CA 91320 , USA
| | - Kui Chen
- Department Discovery Technologies , Amgen Inc. , One Amgen Center Drive , Thousand Oaks , CA 91320 , USA
| | - Yuan Cheng
- Department of Medicinal Chemistry , Amgen Inc. , One Amgen Center Drive , Thousand Oaks , CA 91320 , USA . ; Tel: +1 805 447 4721
| | - Mark R Fielden
- Comparative Biology and Safety Sciences , Amgen Inc. , One Amgen Center Drive , Thousand Oaks , CA 91320 , USA
| | - Vijay Gore
- Department of Medicinal Chemistry , Amgen Inc. , One Amgen Center Drive , Thousand Oaks , CA 91320 , USA . ; Tel: +1 805 447 4721
| | - Dean Hickman
- Department of Pharmacokinetics and Drug Metabolism , Amgen Inc. , One Amgen Center Drive , Thousand Oaks , CA 91320 , USA
| | - Qingyian Liu
- Department of Medicinal Chemistry , Amgen Inc. , One Amgen Center Drive , Thousand Oaks , CA 91320 , USA . ; Tel: +1 805 447 4721
| | - E Allen Sickmier
- Department of Molecular Engineering , Amgen Inc. , 360 Binney Street , Cambridge , MA 02142 , USA
| | - Hugo M Vargas
- Comparative Biology and Safety Sciences , Amgen Inc. , One Amgen Center Drive , Thousand Oaks , CA 91320 , USA
| | - Jonathan Werner
- Comparative Biology and Safety Sciences , Amgen Inc. , One Amgen Center Drive , Thousand Oaks , CA 91320 , USA
| | - Ryan D White
- Department of Medicinal Chemistry , Amgen Inc. , 360 Binney Street , Cambridge , MA 02142 , USA
| | - Douglas A Whittington
- Department of Molecular Engineering , Amgen Inc. , 360 Binney Street , Cambridge , MA 02142 , USA
| | - Stephen Wood
- Department of Neuroscience , Amgen Inc. , One Amgen Center Drive , Thousand Oaks , CA 91320 , USA
| | - Ana E Minatti
- Department of Medicinal Chemistry , Amgen Inc. , One Amgen Center Drive , Thousand Oaks , CA 91320 , USA . ; Tel: +1 805 447 4721
| |
Collapse
|
28
|
Nie X, Cheng C, Zhu G. Palladium-Catalyzed Remote Aryldifluoroalkylation of Alkenyl Aldehydes. Angew Chem Int Ed Engl 2017. [DOI: 10.1002/ange.201611697] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Xingliang Nie
- Department of Chemistry; Zhejiang Normal University; 688 Yingbin Road Jinhua 321004 P.R. China
| | - Cungui Cheng
- Department of Chemistry; Zhejiang Normal University; 688 Yingbin Road Jinhua 321004 P.R. China
| | - Gangguo Zhu
- Department of Chemistry; Zhejiang Normal University; 688 Yingbin Road Jinhua 321004 P.R. China
| |
Collapse
|
29
|
Nie X, Cheng C, Zhu G. Palladium-Catalyzed Remote Aryldifluoroalkylation of Alkenyl Aldehydes. Angew Chem Int Ed Engl 2017; 56:1898-1902. [DOI: 10.1002/anie.201611697] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Indexed: 11/11/2022]
Affiliation(s)
- Xingliang Nie
- Department of Chemistry; Zhejiang Normal University; 688 Yingbin Road Jinhua 321004 P.R. China
| | - Cungui Cheng
- Department of Chemistry; Zhejiang Normal University; 688 Yingbin Road Jinhua 321004 P.R. China
| | - Gangguo Zhu
- Department of Chemistry; Zhejiang Normal University; 688 Yingbin Road Jinhua 321004 P.R. China
| |
Collapse
|
30
|
Qu F, Yang M, Rasooly A. Dual Signal Amplification Electrochemical Biosensor for Monitoring the Activity and Inhibition of the Alzheimer’s Related Protease β-Secretase. Anal Chem 2016; 88:10559-10565. [DOI: 10.1021/acs.analchem.6b02659] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Affiliation(s)
- Fengli Qu
- College
of Chemistry and Chemical Engineering, Central South University, Changsha, 410083, China
- College
of Chemistry and Chemical Engineering, Qufu Normal University, Qufu, Shandong 273165, China
| | - Minghui Yang
- College
of Chemistry and Chemical Engineering, Central South University, Changsha, 410083, China
| | - Avraham Rasooly
- National
Cancer Institute, National Institutes of Health, Rockville, Maryland 20850, United States
| |
Collapse
|
31
|
Ciordia M, Pérez-Benito L, Delgado F, Trabanco AA, Tresadern G. Application of Free Energy Perturbation for the Design of BACE1 Inhibitors. J Chem Inf Model 2016; 56:1856-71. [PMID: 27500414 DOI: 10.1021/acs.jcim.6b00220] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Novel spiroaminodihydropyrroles probing for optimized interactions at the P3 pocket of β-secretase 1 (BACE1) were designed with the use of free energy perturbation (FEP) calculations. The resulting molecules showed pIC50 potencies in enzymatic BACE1 inhibition assays ranging from approximately 5 to 7. Good correlation was observed between the predicted activity from the FEP calculations and experimental activity. Simulations run with a default 5 ns approach delivered a mean unsigned error (MUE) between prediction and experiment of 0.58 and 0.91 kcal/mol for retrospective and prospective applications, respectively. With longer simulations of 10 and 20 ns, the MUE was in both cases 0.57 kcal/mol for the retrospective application, and 0.69 and 0.59 kcal/mol for the prospective application. Other considerations that impact the quality of the calculations are discussed. This work provides an example of the value of FEP as a computational tool for drug discovery.
Collapse
Affiliation(s)
- Myriam Ciordia
- Janssen Research and Development , c/Jarama 75A, 45007 Toledo, Spain.,Departamento de Química y Bioquímica, Facultad de Farmacia, Universidad CEU San Pablo , Urbanización Montepríncipe Ctra., Boadilla del Monte Km 5.3, 28668 Madrid, Spain
| | - Laura Pérez-Benito
- Janssen Research and Development , c/Jarama 75A, 45007 Toledo, Spain.,Laboratori de Medicina Computacional Unitat de Bioestadistica, Facultat de Medicina, Universitat Autonoma de Barcelona , 08193 Bellaterra, Spain
| | - Francisca Delgado
- Janssen Research and Development , c/Jarama 75A, 45007 Toledo, Spain
| | - Andrés A Trabanco
- Janssen Research and Development , c/Jarama 75A, 45007 Toledo, Spain
| | - Gary Tresadern
- Janssen Research and Development , c/Jarama 75A, 45007 Toledo, Spain
| |
Collapse
|
32
|
Mandal M, Wu Y, Misiaszek J, Li G, Buevich A, Caldwell JP, Liu X, Mazzola RD, Orth P, Strickland C, Voigt J, Wang H, Zhu Z, Chen X, Grzelak M, Hyde LA, Kuvelkar R, Leach PT, Terracina G, Zhang L, Zhang Q, Michener MS, Smith B, Cox K, Grotz D, Favreau L, Mitra K, Kazakevich I, McKittrick BA, Greenlee W, Kennedy ME, Parker EM, Cumming JN, Stamford AW. Structure-Based Design of an Iminoheterocyclic β-Site Amyloid Precursor Protein Cleaving Enzyme (BACE) Inhibitor that Lowers Central Aβ in Nonhuman Primates. J Med Chem 2016; 59:3231-48. [DOI: 10.1021/acs.jmedchem.5b01995] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- Mihirbaran Mandal
- Department of Global Chemistry, ‡Department of Neuroscience, §Department of Safety Assessment and
Laboratory Animal Research, ∥Department of Discovery Pharmaceutical Sciences, and ⊥Department of
Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Yusheng Wu
- Department of Global Chemistry, ‡Department of Neuroscience, §Department of Safety Assessment and
Laboratory Animal Research, ∥Department of Discovery Pharmaceutical Sciences, and ⊥Department of
Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Jeffrey Misiaszek
- Department of Global Chemistry, ‡Department of Neuroscience, §Department of Safety Assessment and
Laboratory Animal Research, ∥Department of Discovery Pharmaceutical Sciences, and ⊥Department of
Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Guoqing Li
- Department of Global Chemistry, ‡Department of Neuroscience, §Department of Safety Assessment and
Laboratory Animal Research, ∥Department of Discovery Pharmaceutical Sciences, and ⊥Department of
Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Alexei Buevich
- Department of Global Chemistry, ‡Department of Neuroscience, §Department of Safety Assessment and
Laboratory Animal Research, ∥Department of Discovery Pharmaceutical Sciences, and ⊥Department of
Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - John P. Caldwell
- Department of Global Chemistry, ‡Department of Neuroscience, §Department of Safety Assessment and
Laboratory Animal Research, ∥Department of Discovery Pharmaceutical Sciences, and ⊥Department of
Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Xiaoxiang Liu
- Department of Global Chemistry, ‡Department of Neuroscience, §Department of Safety Assessment and
Laboratory Animal Research, ∥Department of Discovery Pharmaceutical Sciences, and ⊥Department of
Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Robert D. Mazzola
- Department of Global Chemistry, ‡Department of Neuroscience, §Department of Safety Assessment and
Laboratory Animal Research, ∥Department of Discovery Pharmaceutical Sciences, and ⊥Department of
Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Peter Orth
- Department of Global Chemistry, ‡Department of Neuroscience, §Department of Safety Assessment and
Laboratory Animal Research, ∥Department of Discovery Pharmaceutical Sciences, and ⊥Department of
Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Corey Strickland
- Department of Global Chemistry, ‡Department of Neuroscience, §Department of Safety Assessment and
Laboratory Animal Research, ∥Department of Discovery Pharmaceutical Sciences, and ⊥Department of
Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Johannes Voigt
- Department of Global Chemistry, ‡Department of Neuroscience, §Department of Safety Assessment and
Laboratory Animal Research, ∥Department of Discovery Pharmaceutical Sciences, and ⊥Department of
Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Hongwu Wang
- Department of Global Chemistry, ‡Department of Neuroscience, §Department of Safety Assessment and
Laboratory Animal Research, ∥Department of Discovery Pharmaceutical Sciences, and ⊥Department of
Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Zhaoning Zhu
- Department of Global Chemistry, ‡Department of Neuroscience, §Department of Safety Assessment and
Laboratory Animal Research, ∥Department of Discovery Pharmaceutical Sciences, and ⊥Department of
Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Xia Chen
- Department of Global Chemistry, ‡Department of Neuroscience, §Department of Safety Assessment and
Laboratory Animal Research, ∥Department of Discovery Pharmaceutical Sciences, and ⊥Department of
Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Michael Grzelak
- Department of Global Chemistry, ‡Department of Neuroscience, §Department of Safety Assessment and
Laboratory Animal Research, ∥Department of Discovery Pharmaceutical Sciences, and ⊥Department of
Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Lynn A. Hyde
- Department of Global Chemistry, ‡Department of Neuroscience, §Department of Safety Assessment and
Laboratory Animal Research, ∥Department of Discovery Pharmaceutical Sciences, and ⊥Department of
Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Reshma Kuvelkar
- Department of Global Chemistry, ‡Department of Neuroscience, §Department of Safety Assessment and
Laboratory Animal Research, ∥Department of Discovery Pharmaceutical Sciences, and ⊥Department of
Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Prescott T. Leach
- Department of Global Chemistry, ‡Department of Neuroscience, §Department of Safety Assessment and
Laboratory Animal Research, ∥Department of Discovery Pharmaceutical Sciences, and ⊥Department of
Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Giuseppe Terracina
- Department of Global Chemistry, ‡Department of Neuroscience, §Department of Safety Assessment and
Laboratory Animal Research, ∥Department of Discovery Pharmaceutical Sciences, and ⊥Department of
Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Lili Zhang
- Department of Global Chemistry, ‡Department of Neuroscience, §Department of Safety Assessment and
Laboratory Animal Research, ∥Department of Discovery Pharmaceutical Sciences, and ⊥Department of
Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Qi Zhang
- Department of Global Chemistry, ‡Department of Neuroscience, §Department of Safety Assessment and
Laboratory Animal Research, ∥Department of Discovery Pharmaceutical Sciences, and ⊥Department of
Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Maria S. Michener
- Department of Global Chemistry, ‡Department of Neuroscience, §Department of Safety Assessment and
Laboratory Animal Research, ∥Department of Discovery Pharmaceutical Sciences, and ⊥Department of
Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Brad Smith
- Department of Global Chemistry, ‡Department of Neuroscience, §Department of Safety Assessment and
Laboratory Animal Research, ∥Department of Discovery Pharmaceutical Sciences, and ⊥Department of
Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Kathleen Cox
- Department of Global Chemistry, ‡Department of Neuroscience, §Department of Safety Assessment and
Laboratory Animal Research, ∥Department of Discovery Pharmaceutical Sciences, and ⊥Department of
Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Diane Grotz
- Department of Global Chemistry, ‡Department of Neuroscience, §Department of Safety Assessment and
Laboratory Animal Research, ∥Department of Discovery Pharmaceutical Sciences, and ⊥Department of
Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Leonard Favreau
- Department of Global Chemistry, ‡Department of Neuroscience, §Department of Safety Assessment and
Laboratory Animal Research, ∥Department of Discovery Pharmaceutical Sciences, and ⊥Department of
Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Kaushik Mitra
- Department of Global Chemistry, ‡Department of Neuroscience, §Department of Safety Assessment and
Laboratory Animal Research, ∥Department of Discovery Pharmaceutical Sciences, and ⊥Department of
Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Irina Kazakevich
- Department of Global Chemistry, ‡Department of Neuroscience, §Department of Safety Assessment and
Laboratory Animal Research, ∥Department of Discovery Pharmaceutical Sciences, and ⊥Department of
Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Brian A. McKittrick
- Department of Global Chemistry, ‡Department of Neuroscience, §Department of Safety Assessment and
Laboratory Animal Research, ∥Department of Discovery Pharmaceutical Sciences, and ⊥Department of
Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - William Greenlee
- Department of Global Chemistry, ‡Department of Neuroscience, §Department of Safety Assessment and
Laboratory Animal Research, ∥Department of Discovery Pharmaceutical Sciences, and ⊥Department of
Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Matthew E. Kennedy
- Department of Global Chemistry, ‡Department of Neuroscience, §Department of Safety Assessment and
Laboratory Animal Research, ∥Department of Discovery Pharmaceutical Sciences, and ⊥Department of
Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Eric M. Parker
- Department of Global Chemistry, ‡Department of Neuroscience, §Department of Safety Assessment and
Laboratory Animal Research, ∥Department of Discovery Pharmaceutical Sciences, and ⊥Department of
Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Jared N. Cumming
- Department of Global Chemistry, ‡Department of Neuroscience, §Department of Safety Assessment and
Laboratory Animal Research, ∥Department of Discovery Pharmaceutical Sciences, and ⊥Department of
Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Andrew W. Stamford
- Department of Global Chemistry, ‡Department of Neuroscience, §Department of Safety Assessment and
Laboratory Animal Research, ∥Department of Discovery Pharmaceutical Sciences, and ⊥Department of
Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| |
Collapse
|
33
|
Chambers RK, Khan TA, Olsen DB, Sleebs BE. Synthesis of amino heterocycle aspartyl protease inhibitors. Org Biomol Chem 2016; 14:4970-85. [DOI: 10.1039/c5ob01842k] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Synthetic strategies to access 2-amino heterocycle head groups that inhibit aspartyl proteases, are reviewed.
Collapse
Affiliation(s)
- Rachel K. Chambers
- The Walter and Eliza Hall Institute for Medical Research
- Parkville
- Australia
| | | | | | - Brad E. Sleebs
- The Walter and Eliza Hall Institute for Medical Research
- Parkville
- Australia
- The University of Melbourne
- Parkville
| |
Collapse
|
34
|
Mateu N, Ciordia M, Delgado O, Sánchez-Roselló M, Trabanco AA, Van Gool M, Tresadern G, Pérez-Benito L, Fustero S. A Versatile Approach to CF3-Containing 2-Pyrrolidones by Tandem Michael Addition-Cyclization: Exemplification in the Synthesis of Amidine Class BACE1 Inhibitors. Chemistry 2015; 21:11719-26. [DOI: 10.1002/chem.201501662] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Indexed: 12/19/2022]
|
35
|
Intriguing possibilities and beneficial aspects of transporter-conscious drug design. Bioorg Med Chem 2015; 23:4119-4131. [PMID: 26138194 DOI: 10.1016/j.bmc.2015.06.022] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Revised: 06/02/2015] [Accepted: 06/10/2015] [Indexed: 01/11/2023]
Abstract
It has been revealed that many types of drugs interact with transporter proteins within an organism. Transporter proteins absorb or excrete materials, including drugs and nutrients, across the cell membrane. Some hydrophobic drugs are excreted from the cell as xenobiotics by ATP-binding cassette (ABC) transporters. However, solute carrier (SLC) transporters are tissue-specifically expressed and have substrate specificities. Thus, transporter-conscious drug design is an excellent method of delivering drugs to pharmaceutical target organs and provides advantages in absorption, distribution, excretion, and toxicity of drugs (ADMET) due to transport systems. In fact, based on this strategy, the bioavailability of prodrugs designed as peptide transporter 1 (PEPT1) substrates was better than that of the corresponding parent compounds due to the transport system in the small intestine. Furthermore, in central nervous system (CNS) drug developing, drug delivery into brain across the blood-brain barrier (BBB) is a serious problem. However, this problem can be also solved by the use of the transport systems at the BBB. Therefore, transporter-consciously designed drugs not only may effectively elicit activity but also may control adverse side effects caused by off-targets and drug-drug interactions and, consequently, may show good performance in clinical trials. In this review, I introduce possibilities and advantages of transporter-conscious drug designs.
Collapse
|
36
|
Chen JJ, Liu Q, Yuan C, Gore V, Lopez P, Ma V, Amegadzie A, Qian W, Judd TC, Minatti AE, Brown J, Cheng Y, Xue M, Zhong W, Dineen TA, Epstein O, Human J, Kreiman C, Marx I, Weiss MM, Hitchcock SA, Powers TS, Chen K, Wen PH, Whittington DA, Cheng AC, Bartberger MD, Hickman D, Werner JA, Vargas HM, Everds NE, Vonderfecht SL, Dunn RT, Wood S, Fremeau RT, White RD, Patel VF. Development of 2-aminooxazoline 3-azaxanthenes as orally efficacious β-secretase inhibitors for the potential treatment of Alzheimer’s disease. Bioorg Med Chem Lett 2015; 25:767-74. [DOI: 10.1016/j.bmcl.2014.12.092] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Revised: 12/23/2014] [Accepted: 12/29/2014] [Indexed: 01/25/2023]
|
37
|
Nenajdenko VG, Muzalevskiy VM, Shastin AV. Polyfluorinated ethanes as versatile fluorinated C2-building blocks for organic synthesis. Chem Rev 2015; 115:973-1050. [PMID: 25594605 DOI: 10.1021/cr500465n] [Citation(s) in RCA: 113] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Valentine G Nenajdenko
- Department of Chemistry, Moscow State University , Leninskie Gory, Moscow 119992, Russia
| | | | | |
Collapse
|
38
|
Hu YJ, Wang XB, Li SY, Xie SS, Wang KD, Kong LY. Facile synthesis of spiro chromanone-tetrahydrothiophenes with three contiguous stereocenters via sulfa-Michael/aldol cascade reactions. Tetrahedron Lett 2015. [DOI: 10.1016/j.tetlet.2014.11.026] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
39
|
Rajasekhar K, Chakrabarti M, Govindaraju T. Function and toxicity of amyloid beta and recent therapeutic interventions targeting amyloid beta in Alzheimer's disease. Chem Commun (Camb) 2015; 51:13434-50. [DOI: 10.1039/c5cc05264e] [Citation(s) in RCA: 149] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Our Feature Article details the physiological role of amyloid beta (Aβ), elaborates its toxic effects and outlines therapeutic molecules designed in the last two years targeting different aspects of Aβ for preventing AD.
Collapse
Affiliation(s)
- K. Rajasekhar
- Bioorganic Chemistry Laboratory
- New Chemistry Unit
- Jawaharlal Nehru Centre for Advanced Scientific Research
- Bengaluru 560064
- India
| | - Malabika Chakrabarti
- Bioorganic Chemistry Laboratory
- New Chemistry Unit
- Jawaharlal Nehru Centre for Advanced Scientific Research
- Bengaluru 560064
- India
| | - T. Govindaraju
- Bioorganic Chemistry Laboratory
- New Chemistry Unit
- Jawaharlal Nehru Centre for Advanced Scientific Research
- Bengaluru 560064
- India
| |
Collapse
|
40
|
Thomas AA, Hunt KW, Newhouse B, Watts RJ, Liu X, Vigers G, Smith D, Rhodes SP, Brown KD, Otten JN, Burkard M, Cox AA, Geck Do MK, Dutcher D, Rana S, DeLisle RK, Regal K, Wright AD, Groneberg R, Liao J, Scearce-Levie K, Siu M, Purkey HE, Lyssikatos JP. 8-Tetrahydropyran-2-yl chromans: highly selective beta-site amyloid precursor protein cleaving enzyme 1 (BACE1) inhibitors. J Med Chem 2014; 57:10112-29. [PMID: 25411915 DOI: 10.1021/jm5015132] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
A series of 2,3,4,4a,10,10a-hexahydropyrano[3,2-b]chromene analogs was developed that demonstrated high selectivity (>2000-fold) for BACE1 vs Cathepsin D (CatD). Three different Asp-binding moieties were examined: spirocyclic acyl guanidines, aminooxazolines, and aminothiazolines in order to modulate potency, selectivity, efflux, and permeability. Guided by structure based design, changes to P2' and P3 moieties were explored. A conformationally restricted P2' methyl group provided inhibitors with excellent cell potency (37-137 nM) and selectivity (435 to >2000-fold) for BACE1 vs CatD. These efforts lead to compound 59, which demonstrated a 69% reduction in rat CSF Aβ1-40 at 60 mg/kg (PO).
Collapse
Affiliation(s)
- Allen A Thomas
- Array BioPharma, 3200 Walnut Street, Boulder, Colorado 80301, United States
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Stathakis CI, Neokosmidis E, Koftis TV. A Scalable Synthesis of the Antidepressant Agomelatine by a Tandem Allylic Chlorination-Isomerization Process. European J Org Chem 2014. [DOI: 10.1002/ejoc.201402886] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
42
|
Liu X, Wright M, Hop CECA. Rational use of plasma protein and tissue binding data in drug design. J Med Chem 2014; 57:8238-48. [PMID: 25099658 DOI: 10.1021/jm5007935] [Citation(s) in RCA: 121] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
It is a commonly accepted assumption that only unbound drug molecules are available to interact with their targets. Therefore, one of the objectives in drug design is to optimize the compound structure to increase in vivo unbound drug concentration. In this review, theoretical analyses and experimental observations are presented to illustrate that low plasma protein binding does not necessarily lead to high in vivo unbound plasma concentration. Similarly, low brain tissue binding does not lead to high in vivo unbound brain tissue concentration. Instead, low intrinsic clearance leads to high in vivo unbound plasma concentration, and low efflux transport activity at the blood-brain barrier leads to high unbound brain concentration. Plasma protein and brain tissue binding are very important parameters in understanding pharmacokinetics, pharmacodynamics, and toxicities of drugs, but these parameters should not be targeted for optimization in drug design.
Collapse
Affiliation(s)
- Xingrong Liu
- Genentech, Inc. , South San Francisco, California 94080, United States
| | | | | |
Collapse
|
43
|
The use of spirocyclic scaffolds in drug discovery. Bioorg Med Chem Lett 2014; 24:3673-82. [DOI: 10.1016/j.bmcl.2014.06.081] [Citation(s) in RCA: 564] [Impact Index Per Article: 51.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Revised: 06/17/2014] [Accepted: 06/27/2014] [Indexed: 12/12/2022]
|
44
|
Volgraf M, Chan L, Huestis MP, Purkey HE, Burkard M, Geck Do M, Harris J, Hunt KW, Liu X, Lyssikatos JP, Rana S, Thomas AA, Vigers GP, Siu M. Synthesis, characterization, and PK/PD studies of a series of spirocyclic pyranochromene BACE1 inhibitors. Bioorg Med Chem Lett 2014; 24:2477-80. [DOI: 10.1016/j.bmcl.2014.04.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Revised: 04/01/2014] [Accepted: 04/04/2014] [Indexed: 10/25/2022]
|
45
|
Huang HJ, Lee CC, Chen CYC. In silico design of BACE1 inhibitor for Alzheimer's disease by traditional Chinese medicine. BIOMED RESEARCH INTERNATIONAL 2014; 2014:741703. [PMID: 24900984 PMCID: PMC4034430 DOI: 10.1155/2014/741703] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Accepted: 02/16/2014] [Indexed: 12/22/2022]
Abstract
The β-site APP cleaving enzyme 1 (BACE1) is an important target for causing Alzheimer's disease (AD), due to the brain deposition peptide amyloid beta (Aβ) require cleavages of amyloid precursor protein (APP) by BACE1 and γ-secretase, but treatments of AD still have side effect in recent therapy. This study utilizes the world largest traditional Chinese medicine (TCM) database and database screening to provide potential BACE1 inhibited compound. Molecular dynamics (MD) simulation was carried out to observe the dynamics structure after ligand binding. We found that Triptofordin B1 has less toxicity than pyrimidine analogue, which has more potent binding affinity with BACE1. For trajectory analysis, all conformations are tending to be stable during 5000 ps simulation time. In dynamic protein validation, the residues of binding region are still stable after MD simulation. For snapshot comparison, we found that Triptofordin B1 could reduce the binding cavity; the results reveal that Triptofordin B1 could bind to BACE1 and better than control, which could be used as potential lead drug to design novel BACE1 inhibitor for AD therapy.
Collapse
Affiliation(s)
- Hung-Jin Huang
- Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, College of Pharmacy, China Medical University, Taichung 40402, Taiwan
| | - Cheng-Chun Lee
- School of Medicine, College of Medicine, China Medical University, Taichung 40402, Taiwan
| | - Calvin Yu-Chian Chen
- School of Medicine, College of Medicine, China Medical University, Taichung 40402, Taiwan
- Department of Biomedical Informatics, Asia University, Taichung 41354, Taiwan
| |
Collapse
|
46
|
Oehlrich D, Prokopcova H, Gijsen HJ. The evolution of amidine-based brain penetrant BACE1 inhibitors. Bioorg Med Chem Lett 2014; 24:2033-45. [DOI: 10.1016/j.bmcl.2014.03.025] [Citation(s) in RCA: 126] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Revised: 03/06/2014] [Accepted: 03/07/2014] [Indexed: 01/18/2023]
|
47
|
Takahashi R, Ma S, Deese A, Yue Q, Kim-Kang H, Yi Y, Siu M, Hunt KW, Kallan NC, Hop CECA, Liu X, Khojasteh SC. Elucidating the mechanism of cytochrome P450-mediated pyrimidine ring conversion to pyrazole metabolites with the BACE1 inhibitor GNE-892 in rats. Drug Metab Dispos 2014; 42:890-8. [PMID: 24595682 DOI: 10.1124/dmd.114.057141] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2025] Open
Abstract
We investigated an uncommon biotransformation of pyrimidine during the metabolism of GNE-892 ((R)-2-amino-1,3',3'-trimethyl-7'-(pyrimidin-5-yl)-3',4'-dihydro-2'H-spiro[imidazole-4,1'-naphthalen]-5(1H)-one), a β-secretase 1 inhibitor. Three novel metabolites, formed by conversion of pyrimidine to pyrazole, were observed in the (14)C-radiolabeled mass balance study in rats. Their structures were characterized by high-resolution mass spectrometry and nuclear magnetic resonance. Although these metabolites accounted for <5% of the administered dose, their unique nature prompted us to conduct further investigations. The pyrazole-containing metabolites were formed in vitro with rat hepatocytes and liver microsomes, which supported that they were formed during hepatic metabolism. Further, their generation was inhibited by 1-aminobenzotriazole, indicating involvement of cytochrome P450s. Studies with rat recombinant enzymes identified that CYP2D2 generated the N-hydroxypyrazole metabolite from GNE-892. This biotransformation proceeded through multiple steps from the likely precursor, pyrimidine N-oxide. On the basis of these data, we propose a mechanism in which the pyrimidine is activated via N-oxidation, followed by a second oxidative process that opens the pyrimidine ring to form a formamide intermediate. After hydrolysis of the formamide, a carbon is lost as formic acid, together with ring closure to form the pyrazole ring. This article highlights a mechanistic approach for determining the biotransformation of the pyrimidine to a pyrazole for GNE-892.
Collapse
Affiliation(s)
- Ryan Takahashi
- Departments of Drug Metabolism and Pharmacokinetics (R.T., S.M., Q.Y., C.E.H., X.L., S.C.K), Small Molecule Pharmaceutical Sciences (A.D.), and Discovery Chemistry (M.S.), Genentech, Inc., South San Francisco, California; XenoBiotic Laboratories, Inc., Plainsboro, New Jersey (H.K., Y.Y.); and Array BioPharma, Boulder, Colorado (K.W.H., N.C.K.)
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Takahashi R, Ma S, Yue Q, Kim-Kang H, Yi Y, Lyssikatos JP, Regal K, Hunt KW, Kallan NC, Siu M, Hop CECA, Liu X, Khojasteh SC. Dose-dependent exposure and metabolism of GNE-892, a β-secretase inhibitor, in monkeys: contributions by P450, AO, and P-gp. Eur J Drug Metab Pharmacokinet 2014; 40:171-85. [DOI: 10.1007/s13318-014-0198-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Accepted: 03/26/2014] [Indexed: 02/04/2023]
|
49
|
Thomas AA, Hunt KW, Volgraf M, Watts RJ, Liu X, Vigers G, Smith D, Sammond D, Tang TP, Rhodes SP, Metcalf AT, Brown KD, Otten JN, Burkard M, Cox AA, Do MKG, Dutcher D, Rana S, DeLisle RK, Regal K, Wright AD, Groneberg R, Scearce-Levie K, Siu M, Purkey HE, Lyssikatos JP, Gunawardana IW. Discovery of 7-tetrahydropyran-2-yl chromans: β-site amyloid precursor protein cleaving enzyme 1 (BACE1) inhibitors that reduce amyloid β-protein (Aβ) in the central nervous system. J Med Chem 2014; 57:878-902. [PMID: 24397738 DOI: 10.1021/jm401635n] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
In an attempt to increase selectivity vs Cathepsin D (CatD) in our BACE1 program, a series of 1,3,4,4a,10,10a-hexahydropyrano[4,3-b]chromene analogues was developed. Three different Asp-binding moieties were examined: spirocyclic acyl guanidines, aminooxazolines, and aminothiazolines in order to modulate potency, selectivity, efflux, and permeability. Using structure-based design, substitutions to improve binding to both the S3 and S2' sites of BACE1 were explored. An acyl guanidine moiety provided the most potent analogues. These compounds demonstrated 10-420 fold selectivity for BACE1 vs CatD, and were highly potent in a cell assay measuring Aβ1-40 production (5-99 nM). They also suffered from high efflux. Despite this undesirable property, two of the acyl guanidines achieved free brain concentrations (Cfree,brain) in a guinea pig PD model sufficient to cover their cell IC50s. Moreover, a significant reduction of Aβ1-40 in guinea pig, rat, and cyno CSF (58%, 53%, and 63%, respectively) was observed for compound 62.
Collapse
Affiliation(s)
- Allen A Thomas
- Array BioPharma , 3200 Walnut Street, Boulder, Colorado 80301, United States
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Cacabelos R, Cacabelos P, Torrellas C, Tellado I, Carril JC. Pharmacogenomics of Alzheimer's disease: novel therapeutic strategies for drug development. Methods Mol Biol 2014; 1175:323-556. [PMID: 25150875 DOI: 10.1007/978-1-4939-0956-8_13] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Alzheimer's disease (AD) is a major problem of health and disability, with a relevant economic impact on our society. Despite important advances in pathogenesis, diagnosis, and treatment, its primary causes still remain elusive, accurate biomarkers are not well characterized, and the available pharmacological treatments are not cost-effective. As a complex disorder, AD is a polygenic and multifactorial clinical entity in which hundreds of defective genes distributed across the human genome may contribute to its pathogenesis. Diverse environmental factors, cerebrovascular dysfunction, and epigenetic phenomena, together with structural and functional genomic dysfunctions, lead to amyloid deposition, neurofibrillary tangle formation, and premature neuronal death, the major neuropathological hallmarks of AD. Future perspectives for the global management of AD predict that genomics and proteomics may help in the search for reliable biomarkers. In practical terms, the therapeutic response to conventional drugs (cholinesterase inhibitors, multifactorial strategies) is genotype-specific. Genomic factors potentially involved in AD pharmacogenomics include at least five categories of gene clusters: (1) genes associated with disease pathogenesis; (2) genes associated with the mechanism of action of drugs; (3) genes associated with drug metabolism (phase I and II reactions); (4) genes associated with drug transporters; and (5) pleiotropic genes involved in multifaceted cascades and metabolic reactions. The implementation of pharmacogenomic strategies will contribute to optimize drug development and therapeutics in AD and related disorders.
Collapse
Affiliation(s)
- Ramón Cacabelos
- Chair of Genomic Medicine, Camilo José Cela University, 28692, Villanueva de la Cañada, Madrid, Spain,
| | | | | | | | | |
Collapse
|