1
|
Pitman M, Hahn DF, Tresadern G, Mobley DL. To Design Scalable Free Energy Perturbation Networks, Optimal Is Not Enough. J Chem Inf Model 2023; 63:1776-1793. [PMID: 36878475 PMCID: PMC10547263 DOI: 10.1021/acs.jcim.2c01579] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2023]
Abstract
Drug discovery is accelerated with computational methods such as alchemical simulations to estimate ligand affinities. In particular, relative binding free energy (RBFE) simulations are beneficial for lead optimization. To use RBFE simulations to compare prospective ligands in silico, researchers first plan the simulation experiment, using graphs where nodes represent ligands and graph edges represent alchemical transformations between ligands. Recent work demonstrated that optimizing the statistical architecture of these perturbation graphs improves the accuracy of the predicted changes in the free energy of ligand binding. Therefore, to improve the success rate of computational drug discovery, we present the open-source software package High Information Mapper (HiMap)─a new take on its predecessor, Lead Optimization Mapper (LOMAP). HiMap removes heuristics decisions from design selection and instead finds statistically optimal graphs over ligands clustered with machine learning. Beyond optimal design generation, we present theoretical insights for designing alchemical perturbation maps. Some of these results include that for n number of nodes, the precision of perturbation maps is stable at n·ln(n) edges. This result indicates that even an "optimal" graph can result in unexpectedly high errors if a plan includes too few alchemical transformations for the given number of ligands and edges. And, as a study compares more ligands, the performance of even optimal graphs will deteriorate with linear scaling of the edge count. In this sense, ensuring an A- or D-optimal topology is not enough to produce robust errors. We additionally find that optimal designs will converge more rapidly than radial and LOMAP designs. Moreover, we derive bounds for how clustering reduces cost for designs with a constant expected relative error per cluster, invariant of the size of the design. These results inform how to best design perturbation maps for computational drug discovery and have broader implications for experimental design.
Collapse
Affiliation(s)
- Mary Pitman
- Department of Pharmacy & Pharmaceutical Sciences, University of California, Irvine, CA 92697, USA
| | - David F. Hahn
- Computational Chemistry, Janssen Research & Development, Turnhoutseweg 30, Beerse B-2340, Belgium
| | - Gary Tresadern
- Computational Chemistry, Janssen Research & Development, Turnhoutseweg 30, Beerse B-2340, Belgium
| | - David L. Mobley
- Department of Pharmacy & Pharmaceutical Sciences, University of California, Irvine, CA 92697, USA
- Department of Chemistry, University of California, Irvine, CA 92697, USA
| |
Collapse
|
2
|
Mukerjee N, Das A, Jawarkar RD, Maitra S, Das P, Castrosanto MA, Paul S, Samad A, Zaki MEA, Al-Hussain SA, Masand VH, Hasan MM, Bukhari SNA, Perveen A, Alghamdi BS, Alexiou A, Kamal MA, Dey A, Malik S, Bakal RL, Abuzenadah AM, Ghosh A, Md Ashraf G. Repurposing food molecules as a potential BACE1 inhibitor for Alzheimer's disease. Front Aging Neurosci 2022; 14:878276. [PMID: 36072483 PMCID: PMC9443073 DOI: 10.3389/fnagi.2022.878276] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 07/07/2022] [Indexed: 11/13/2022] Open
Abstract
Alzheimer's disease (AD) is a severe neurodegenerative disorder of the brain that manifests as dementia, disorientation, difficulty in speech, and progressive cognitive and behavioral impairment. The emerging therapeutic approach to AD management is the inhibition of β-site APP cleaving enzyme-1 (BACE1), known to be one of the two aspartyl proteases that cleave β-amyloid precursor protein (APP). Studies confirmed the association of high BACE1 activity with the proficiency in the formation of β-amyloid-containing neurotic plaques, the characteristics of AD. Only a few FDA-approved BACE1 inhibitors are available in the market, but their adverse off-target effects limit their usage. In this paper, we have used both ligand-based and target-based approaches for drug design. The QSAR study entails creating a multivariate GA-MLR (Genetic Algorithm-Multilinear Regression) model using 552 molecules with acceptable statistical performance (R 2 = 0.82, Q 2 loo = 0.81). According to the QSAR study, the activity has a strong link with various atoms such as aromatic carbons and ring Sulfur, acceptor atoms, sp2-hybridized oxygen, etc. Following that, a database of 26,467 food compounds was primarily used for QSAR-based virtual screening accompanied by the application of the Lipinski rule of five; the elimination of duplicates, salts, and metal derivatives resulted in a truncated dataset of 8,453 molecules. The molecular descriptor was calculated and a well-validated 6-parametric version of the QSAR model was used to predict the bioactivity of the 8,453 food compounds. Following this, the food compounds whose predicted activity (pKi) was observed above 7.0 M were further docked into the BACE1 receptor which gave rise to the Identification of 4-(3,4-Dihydroxyphenyl)-2-hydroxy-1H-phenalen-1-one (PubChem I.D: 4468; Food I.D: FDB017657) as a hit molecule (Binding Affinity = -8.9 kcal/mol, pKi = 7.97 nM, Ki = 10.715 M). Furthermore, molecular dynamics simulation for 150 ns and molecular mechanics generalized born and surface area (MMGBSA) study aided in identifying structural motifs involved in interactions with the BACE1 enzyme. Molecular docking and QSAR yielded complementary and congruent results. The validated analyses can be used to improve a drug/lead candidate's inhibitory efficacy against the BACE1. Thus, our approach is expected to widen the field of study of repurposing nutraceuticals into neuroprotective as well as anti-cancer and anti-viral therapeutic interventions.
Collapse
Affiliation(s)
- Nobendu Mukerjee
- Department of Microbiology, Ramakrishna Mission Vivekananda Centenary College, Khardaha, India
- Department of Health Sciences, Novel Global Community Educational Foundation, Hebersham, NSW, Australia
| | - Anubhab Das
- Institute of Health Sciences, Presidency University, Kolkata, India
| | - Rahul D. Jawarkar
- Department of Medicinal Chemistry, Dr. Rajendra Gode Institute of Pharmacy, Amravati, India
| | - Swastika Maitra
- Department of Microbiology, Adamas University, Kolkata, India
| | | | - Melvin A. Castrosanto
- Institute of Chemistry, University of the Philippines Los Baños, Los Baños, Philippines
| | - Soumyadip Paul
- Department of Microbiology, Ramakrishna Mission Vivekananda Centenary College, Khardaha, India
| | - Abdul Samad
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tishk International University, Erbil, Iraq
| | - Magdi E. A. Zaki
- Department of Chemistry, Faculty of Science, Imam Mohammad Ibn Saud Islamic University, Riyadh, Saudi Arabia
| | - Sami A. Al-Hussain
- Department of Chemistry, Faculty of Science, Imam Mohammad Ibn Saud Islamic University, Riyadh, Saudi Arabia
| | - Vijay H. Masand
- Department of Chemistry, Vidya Bharati Mahavidyalaya, Amravati, India
| | - Mohammad Mehedi Hasan
- Department of Biochemistry and Molecular Biology, Faculty of Life Sciences, Mawlana Bhashani Science and Technology University, Tangail, Bangladesh
| | - Syed Nasir Abbas Bukhari
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jouf University, Sakaka, Saudi Arabia
| | - Asma Perveen
- Glocal School of Life Sciences, Glocal University, Saharanpur, India
| | - Badrah S. Alghamdi
- Department of Physiology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
- Pre-Clinical Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- The Neuroscience Research Unit, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Athanasios Alexiou
- Department of Science and Engineering, Novel Global Community Educational Foundation, Hebersham, NSW, Australia
- AFNP Med, Vienna, Austria
| | - Mohammad Amjad Kamal
- Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
- Enzymoics, Novel Global Community Educational Foundation, Hebersham, NSW, Australia
| | - Abhijit Dey
- Department of Life Sciences, Presidency University, Kolkata, India
| | - Sumira Malik
- Amity Institute of Biotechnology, Amity University, Jharkhand, Ranchi, India
| | - Ravindra L. Bakal
- Department of Medicinal Chemistry, Dr. Rajendra Gode Institute of Pharmacy, Amravati, India
| | - Adel Mohammad Abuzenadah
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Arabinda Ghosh
- Microbiology Division, Department of Botany, Gauhati University, Guwahati, India
| | - Ghulam Md Ashraf
- Pre-Clinical Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
3
|
Patel S, Bansoad AV, Singh R, Khatik GL. BACE1: A Key Regulator in Alzheimer's Disease Progression and Current Development of its Inhibitors. Curr Neuropharmacol 2022; 20:1174-1193. [PMID: 34852746 PMCID: PMC9886827 DOI: 10.2174/1570159x19666211201094031] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 11/26/2021] [Accepted: 11/28/2021] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is a chronic neurodegenerative disease with no specific disease-modifying treatment. β-secretase (BACE1) is considered the potential and rationale target because it is involved in the rate-limiting step, which produces toxic Aβ42 peptides that leads to deposits in the form of amyloid plaques extracellularly, resulting in AD. OBJECTIVE This study aims to discuss the role and implications of BACE1 and its inhibitors in the management of AD. METHODS We have searched and collected the relevant quality work from PubMed using the following keywords "BACE1", BACE2", "inhibitors", and "Alzheimer's disease". In addition, we included the work which discusses the role of BACE1 in AD and the recent work on its inhibitors. RESULTS In this review, we have discussed the importance of BACE1 in regulating AD progression and the current development of BACE1 inhibitors. However, the development of a BACE1 inhibitor is very challenging due to the large active site of BACE1. Nevertheless, some of the BACE1 inhibitors have managed to enter advanced phases of clinical trials, such as MK-8931 (Verubecestat), E2609 (Elenbecestat), AZD3293 (Lanabecestat), and JNJ-54861911 (Atabecestat). This review also sheds light on the prospect of BACE1 inhibitors as the most effective therapeutic approach in delaying or preventing AD progression. CONCLUSION BACE1 is involved in the progression of AD. The current ongoing or failed clinical trials may help understand the role of BACE1 inhibition in regulating the Aβ load and cognitive status of AD patients.
Collapse
Affiliation(s)
| | - Ankush Vardhaman Bansoad
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research-Raebareli, New Transit Campus, Bijnor-Sisendi Road, Sarojini Nagar, Near CRPF Base Camp, Lucknow (Uttar Pradesh), 226002, India
| | - Rakesh Singh
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research-Raebareli, New Transit Campus, Bijnor-Sisendi Road, Sarojini Nagar, Near CRPF Base Camp, Lucknow (Uttar Pradesh), 226002, India
| | - Gopal L. Khatik
- Department of Medicinal Chemistry, ,Address correspondence to this author at the Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research- Raebareli, New Transit Campus, Bijnor-Sisendi Road, Sarojini Nagar, Near CRPF Base Camp, Lucknow, Uttar Pradesh, India, 226002; E-mail: ,
| |
Collapse
|
4
|
Peng C, Wang J, Shi Y, Xu Z, Zhu W. Increasing the Sampling Efficiency of Protein Conformational Change by Combining a Modified Replica Exchange Molecular Dynamics and Normal Mode Analysis. J Chem Theory Comput 2020; 17:13-28. [PMID: 33351613 DOI: 10.1021/acs.jctc.0c00592] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Understanding conformational change at an atomic level is significant when determining a protein functional mechanism. Replica exchange molecular dynamics (REMD) is a widely used enhanced sampling method to explore protein conformational space. However, REMD with an explicit solvent model requires huge computational resources, immensely limiting its application. In this study, a variation of parallel tempering metadynamics (PTMetaD) with the omission of solvent-solvent interactions in exchange attempts and the use of low-frequency modes calculated by normal-mode analysis (NMA) as collective variables (CVs), namely ossPTMetaD, is proposed with the aim to accelerate MD simulations simultaneously in temperature and geometrical spaces. For testing the performance of ossPTMetaD, five protein systems with diverse biological functions and motion patterns were selected, including large-scale domain motion (AdK), flap movement (HIV-1 protease and BACE1), and DFG-motif flip in kinases (p38α and c-Abl). The simulation results showed that ossPTMetaD requires much fewer numbers of replicas than temperature REMD (T-REMD) with a reduction of ∼70% to achieve a similar exchange ratio. Although it does not obey the detailed balance condition, ossPTMetaD provides consistent results with T-REMD and experimental data. The high accessibility of the large conformational change of protein systems by ossPTMetaD, especially in simulating the very challenging DFG-motif flip of protein kinases, demonstrated its high efficiency and robustness in the characterization of the large-scale protein conformational change pathway and associated free energy profile.
Collapse
Affiliation(s)
- Cheng Peng
- CAS Key Laboratory of Receptor Research; Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, China.,University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing, 100049, China
| | - Jinan Wang
- CAS Key Laboratory of Receptor Research; Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, China
| | - Yulong Shi
- CAS Key Laboratory of Receptor Research; Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, China.,University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing, 100049, China
| | - Zhijian Xu
- CAS Key Laboratory of Receptor Research; Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, China.,University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing, 100049, China
| | - Weiliang Zhu
- CAS Key Laboratory of Receptor Research; Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, China.,Open Studio for Druggability Research of Marine Lead Compounds, Qingdao National Laboratory for Marine Science and Technology, 1 Wenhai Road, Aoshanwei, Jimo, Qingdao 266237, China.,University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing, 100049, China
| |
Collapse
|
5
|
Rombouts F, Kusakabe KI, Hsiao CC, Gijsen HJM. Small-molecule BACE1 inhibitors: a patent literature review (2011 to 2020). Expert Opin Ther Pat 2020; 31:25-52. [PMID: 33006491 DOI: 10.1080/13543776.2021.1832463] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
INTRODUCTION Inhibition of β-site amyloid precursor protein cleaving enzyme 1 (BACE1) has been extensively pursued as potential disease-modifying treatment for Alzheimer's disease (AD). Clinical failures with BACE inhibitors have progressively raised the bar forever cleaner candidates with reduced cardiovascular liability, toxicity risk, and increased selectivity over cathepsin D (CatD) and BACE2. AREAS COVERED This review provides an overview of patented BACE1 inhibitors between 2011 and 2020 per pharmaceutical company or research group and highlights the progress that was made in dialing out toxicity liabilities. EXPERT OPINION Despite an increasingly crowded IP situation, significant progress was made using highly complex chemistry in avoiding toxicity liabilities, with BACE1/BACE2 selectivity being the most remarkable achievement. However, clinical trial data suggest on-target toxicity is likely a contributing factor, which implies the only potential future of BACE1 inhibitors lies in careful titration of highly selective compounds in early populations where the amyloid burden is still minimal as prophylactic therapy, or as an affordable oral maintenance therapy following amyloid-clearing therapies.
Collapse
Affiliation(s)
- Frederik Rombouts
- Medicinal Chemistry, Janssen Research & Development , Beerse, Belgium
| | - Ken-Ichi Kusakabe
- Laboratory for Medicinal Chemistry Research, Shionogi & Co., Ltd ., Toyonaka, Osaka, Japan
| | - Chien-Chi Hsiao
- Medicinal Chemistry, Janssen Research & Development , Beerse, Belgium
| | - Harrie J M Gijsen
- Medicinal Chemistry, Janssen Research & Development , Beerse, Belgium
| |
Collapse
|
6
|
Oehlrich D, Peschiulli A, Tresadern G, Van Gool M, Vega JA, De Lucas AI, Alonso de Diego SA, Prokopcova H, Austin N, Van Brandt S, Surkyn M, De Cleyn M, Vos A, Rombouts FJR, Macdonald G, Moechars D, Gijsen HJM, Trabanco AA. Evaluation of a Series of β-Secretase 1 Inhibitors Containing Novel Heteroaryl-Fused-Piperazine Amidine Warheads. ACS Med Chem Lett 2019; 10:1159-1165. [PMID: 31413800 DOI: 10.1021/acsmedchemlett.9b00181] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Accepted: 07/02/2019] [Indexed: 02/08/2023] Open
Abstract
Despite several years of research, only a handful of β-secretase (BACE) 1 inhibitors have entered clinical trials as potential therapeutics against Alzheimer's disease. The intrinsic basic nature of low molecular weight, amidine-containing BACE 1 inhibitors makes them far from optimal as central nervous system drugs. Herein we present a set of novel heteroaryl-fused piperazine amidine inhibitors designed to lower the basicity of the key, enzyme binding, amidine functionality. This study resulted in the identification of highly potent (IC50 ≤ 10 nM), permeable lead compounds with a reduced propensity to suffer from P-glycoprotein-mediated efflux.
Collapse
Affiliation(s)
| | | | | | - Michiel Van Gool
- Discovery Sciences Medicinal Chemistry, Janssen Research & Development, Janssen−Cilag S.A., C/Jarama 75A, 45007 Toledo, Spain
| | - Juan Antonio Vega
- Discovery Sciences Medicinal Chemistry, Janssen Research & Development, Janssen−Cilag S.A., C/Jarama 75A, 45007 Toledo, Spain
| | - Ana Isabel De Lucas
- Discovery Sciences Medicinal Chemistry, Janssen Research & Development, Janssen−Cilag S.A., C/Jarama 75A, 45007 Toledo, Spain
| | - Sergio A. Alonso de Diego
- Discovery Sciences Medicinal Chemistry, Janssen Research & Development, Janssen−Cilag S.A., C/Jarama 75A, 45007 Toledo, Spain
| | | | | | | | | | | | | | | | | | | | | | - Andrés A. Trabanco
- Discovery Sciences Medicinal Chemistry, Janssen Research & Development, Janssen−Cilag S.A., C/Jarama 75A, 45007 Toledo, Spain
| |
Collapse
|
7
|
Diastereoselective synthesis of fused cyclopropyl-3-amino-2,4-oxazine β-amyloid cleaving enzyme (BACE) inhibitors and their biological evaluation. Bioorg Med Chem Lett 2018; 28:1111-1115. [DOI: 10.1016/j.bmcl.2018.01.056] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 01/24/2018] [Accepted: 01/26/2018] [Indexed: 11/19/2022]
|
8
|
Islam MA, Pillay TS. β-secretase inhibitors for Alzheimer’s disease: identification using pharmacoinformatics. J Biomol Struct Dyn 2018; 37:503-522. [DOI: 10.1080/07391102.2018.1430619] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Affiliation(s)
- Md Ataul Islam
- Faculty of Health Sciences, Department of Chemical Pathology, University of Pretoria and National Health Laboratory Service Tshwane Academic Division, Pretoria, South Africa
- School of Health Sciences, University of Kwazulu-Natal, Westville Campus, Durban, South Africa
| | - Tahir S. Pillay
- Faculty of Health Sciences, Department of Chemical Pathology, University of Pretoria and National Health Laboratory Service Tshwane Academic Division, Pretoria, South Africa
- Division of Chemical Pathology, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
9
|
Gutiérrez LJ, Parravicini O, Sánchez E, Rodríguez R, Cobo J, Enriz RD. New substituted aminopyrimidine derivatives as BACE1 inhibitors: in silico design, synthesis and biological assays. J Biomol Struct Dyn 2018; 37:229-246. [DOI: 10.1080/07391102.2018.1424036] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Lucas J. Gutiérrez
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Chacabuco 915, 5700 San Luis, Argentina
- IMIBIO-CONICET, UNSL, Chacabuco 915, 5700 San Luis, Argentina
| | - Oscar Parravicini
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Chacabuco 915, 5700 San Luis, Argentina
- IMIBIO-CONICET, UNSL, Chacabuco 915, 5700 San Luis, Argentina
| | - Emilse Sánchez
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Chacabuco 915, 5700 San Luis, Argentina
- IMIBIO-CONICET, UNSL, Chacabuco 915, 5700 San Luis, Argentina
| | - Ricaurte Rodríguez
- Departamento de Química, Universidad Nacional de Colombia, Ciudad Universitaria, Carrera 30, No. 45-03, Bogotá, Colombia
| | - Justo Cobo
- Departamento de Química Inorgánica y Orgánica, Universidad de Jaén, Campus Las Lagunillas s/n, 23071 Jaén, Spain
| | - Ricardo D. Enriz
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Chacabuco 915, 5700 San Luis, Argentina
- IMIBIO-CONICET, UNSL, Chacabuco 915, 5700 San Luis, Argentina
| |
Collapse
|
10
|
Tresadern G, Rombouts FJR, Oehlrich D, Macdonald G, Trabanco AA. Industrial medicinal chemistry insights: neuroscience hit generation at Janssen. Drug Discov Today 2017; 22:1478-1488. [PMID: 28669605 DOI: 10.1016/j.drudis.2017.05.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 05/18/2017] [Accepted: 05/25/2017] [Indexed: 12/16/2022]
Abstract
The role of medicinal chemistry has changed over the past 10 years. Chemistry had become one step in a process; funneling the output of high-throughput screening (HTS) on to the next stage. The goal to identify the ideal clinical compound remains, but the means to achieve this have changed. Modern medicinal chemistry is responsible for integrating innovation throughout early drug discovery, including new screening paradigms, computational approaches, novel synthetic chemistry, gene-family screening, investigating routes of delivery, and so on. In this Foundation Review, we show how a successful medicinal chemistry team has a broad impact and requires multidisciplinary expertise in these areas.
Collapse
Affiliation(s)
- Gary Tresadern
- Discovery Sciences, Janssen Research & Development, C/ Jarama 75A, 45007 Toledo, Spain.
| | - Frederik J R Rombouts
- Neuroscience Medicinal Chemistry, Janssen Research & Development, Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Daniel Oehlrich
- Neuroscience Medicinal Chemistry, Janssen Research & Development, Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Gregor Macdonald
- Neuroscience Medicinal Chemistry, Janssen Research & Development, Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Andres A Trabanco
- Neuroscience Medicinal Chemistry, Janssen Research & Development, C/ Jarama 75A, 45007 Toledo, Spain.
| |
Collapse
|
11
|
Palakurti R, Vadrevu R. Pharmacophore based 3D-QSAR modeling, virtual screening and docking for identification of potential inhibitors of β-secretase. Comput Biol Chem 2017; 68:107-117. [DOI: 10.1016/j.compbiolchem.2017.03.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Revised: 02/07/2017] [Accepted: 03/01/2017] [Indexed: 12/19/2022]
|
12
|
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: 1.9] [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
|
13
|
Keränen H, Pérez-Benito L, Ciordia M, Delgado F, Steinbrecher TB, Oehlrich D, van Vlijmen HWT, Trabanco AA, Tresadern G. Acylguanidine Beta Secretase 1 Inhibitors: A Combined Experimental and Free Energy Perturbation Study. J Chem Theory Comput 2017; 13:1439-1453. [PMID: 28103438 DOI: 10.1021/acs.jctc.6b01141] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
A series of acylguanidine beta secretase 1 (BACE1) inhibitors with modified scaffold and P3 pocket substituent was synthesized and studied with free energy perturbation (FEP) calculations. The resulting molecules showed potencies in enzymatic BACE1 inhibition assays up to 1 nM. The correlation between the predicted activity from the FEP calculations and the experimental activity was good for the P3 pocket substituents. The average mean unsigned error (MUE) between prediction and experiment was 0.68 ± 0.17 kcal/mol for the default 5 ns lambda window simulation time improving to 0.35 ± 0.13 kcal/mol for 40 ns. FEP calculations for the P2' pocket substituents on the same acylguanidine scaffold also showed good agreement with experiment and the results remained stable with repeated simulations and increased simulation time. It proved more difficult to use FEP calculations to study the scaffold modification from increasing 5 to 6 and 7 membered-rings. Although prediction and experiment were in agreement for short 2 ns simulations, as the simulation time increased the results diverged. This was improved by the use of a newly developed "Core Hopping FEP+" approach, which also showed improved stability in repeat calculations. The origins of these differences along with the value of repeat and longer simulation times are discussed. This work provides a further example of the use of FEP as a computational tool for molecular design.
Collapse
Affiliation(s)
- Henrik Keränen
- Computational Chemistry, Janssen Research & Development, Janssen Pharmaceutica N. V. , Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Laura Pérez-Benito
- Laboratori de Medicina Computacional Unitat de Bioestadistica, Facultat de Medicina, Universitat Autonoma de Barcelona , 08193, Bellaterra, Spain.,Computational Chemistry, Janssen Research and Development, Janssen-Cilag , c/ Jarama 75A, 45007, Toledo, Spain
| | - Myriam Ciordia
- Neuroscience Medicinal Chemistry, Janssen Research and Development, Janssen-Cilag , c/ Jarama 75A, 45007, Toledo, Spain
| | - Francisca Delgado
- Neuroscience Medicinal Chemistry, Janssen Research and Development, Janssen-Cilag , c/ Jarama 75A, 45007, Toledo, Spain
| | | | - Daniel Oehlrich
- Neuroscience Medicinal Chemistry, Janssen Research & Development, Janssen Pharmaceutica N. V. , Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Herman W T van Vlijmen
- Computational Chemistry, Janssen Research & Development, Janssen Pharmaceutica N. V. , Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Andrés A Trabanco
- Neuroscience Medicinal Chemistry, Janssen Research and Development, Janssen-Cilag , c/ Jarama 75A, 45007, Toledo, Spain
| | - Gary Tresadern
- Computational Chemistry, Janssen Research and Development, Janssen-Cilag , c/ Jarama 75A, 45007, Toledo, Spain
| |
Collapse
|
14
|
Bochevarov AD, Watson MA, Greenwood JR, Philipp DM. Multiconformation, Density Functional Theory-Based pKa Prediction in Application to Large, Flexible Organic Molecules with Diverse Functional Groups. J Chem Theory Comput 2016; 12:6001-6019. [PMID: 27951674 DOI: 10.1021/acs.jctc.6b00805] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Art D. Bochevarov
- Schrödinger, Inc., 120 West 45th Street, New York, New York 10036, United States
| | - Mark A. Watson
- Schrödinger, Inc., 120 West 45th Street, New York, New York 10036, United States
| | - Jeremy R. Greenwood
- Schrödinger, Inc., 120 West 45th Street, New York, New York 10036, United States
| | - Dean M. Philipp
- Schrödinger, Inc., 101 SW Main Street, Suite 1300, Portland, Oregon 97204, United States
| |
Collapse
|
15
|
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: 10.9] [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
|
16
|
|
17
|
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: 1.3] [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
|
18
|
Boy KM, Guernon JM, Wu YJ, Zhang Y, Shi J, Zhai W, Zhu S, Gerritz SW, Toyn JH, Meredith JE, Barten DM, Burton CR, Albright CF, Good AC, Grace JE, Lentz KA, Olson RE, Macor JE, Thompson LA. Macrocyclic prolinyl acyl guanidines as inhibitors of β-secretase (BACE). Bioorg Med Chem Lett 2015; 25:5040-7. [DOI: 10.1016/j.bmcl.2015.10.031] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 10/08/2015] [Accepted: 10/12/2015] [Indexed: 02/02/2023]
|
19
|
Rombouts FJR, Tresadern G, Delgado O, Martínez-Lamenca C, Van Gool M, García-Molina A, Alonso de Diego SA, Oehlrich D, Prokopcova H, Alonso JM, Austin N, Borghys H, Van Brandt S, Surkyn M, De Cleyn M, Vos A, Alexander R, Macdonald G, Moechars D, Gijsen H, Trabanco AA. 1,4-Oxazine β-Secretase 1 (BACE1) Inhibitors: From Hit Generation to Orally Bioavailable Brain Penetrant Leads. J Med Chem 2015; 58:8216-35. [DOI: 10.1021/acs.jmedchem.5b01101] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Frederik J. R. Rombouts
- Neuroscience Medicinal Chemistry, Janssen Research & Development, Janssen Pharmaceutica NV, Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Gary Tresadern
- Discovery Sciences, Janssen Research & Development, Janssen−Cilag SA, C/Jarama 75A, 45007 Toledo, Spain
| | - Oscar Delgado
- Neuroscience Medicinal Chemistry, Janssen Research & Development, Janssen−Cilag SA, C/Jarama 75A, 45007 Toledo, Spain
| | - Carolina Martínez-Lamenca
- Neuroscience Medicinal Chemistry, Janssen Research & Development, Janssen Pharmaceutica NV, Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Michiel Van Gool
- Neuroscience Medicinal Chemistry, Janssen Research & Development, Janssen−Cilag SA, C/Jarama 75A, 45007 Toledo, Spain
| | - Aránzazu García-Molina
- Neuroscience Medicinal Chemistry, Janssen Research & Development, Janssen−Cilag SA, C/Jarama 75A, 45007 Toledo, Spain
| | - Sergio A. Alonso de Diego
- Neuroscience Medicinal Chemistry, Janssen Research & Development, Janssen−Cilag SA, C/Jarama 75A, 45007 Toledo, Spain
| | - Daniel Oehlrich
- Neuroscience Medicinal Chemistry, Janssen Research & Development, Janssen Pharmaceutica NV, Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Hana Prokopcova
- Neuroscience Medicinal Chemistry, Janssen Research & Development, Janssen Pharmaceutica NV, Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - José Manuel Alonso
- Discovery Sciences, Janssen Research & Development, Janssen−Cilag SA, C/Jarama 75A, 45007 Toledo, Spain
| | - Nigel Austin
- Discovery Sciences, Janssen Research & Development, Janssen Pharmaceutica NV, Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Herman Borghys
- Discovery Sciences, Janssen Research & Development, Janssen Pharmaceutica NV, Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Sven Van Brandt
- Neuroscience Medicinal Chemistry, Janssen Research & Development, Janssen Pharmaceutica NV, Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Michel Surkyn
- Neuroscience Medicinal Chemistry, Janssen Research & Development, Janssen Pharmaceutica NV, Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Michel De Cleyn
- Neuroscience Medicinal Chemistry, Janssen Research & Development, Janssen Pharmaceutica NV, Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Ann Vos
- Discovery Sciences, Janssen Research & Development, Janssen Pharmaceutica NV, Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Richard Alexander
- Biologics Research, Janssen Research & Development, 1400 McKean Road, Spring House, Pennsylvania 19477, United States
| | - Gregor Macdonald
- Neuroscience Medicinal Chemistry, Janssen Research & Development, Janssen Pharmaceutica NV, Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Dieder Moechars
- Neuroscience Biology, Janssen Research & Development, Janssen Pharmaceutica NV, Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Harrie Gijsen
- Neuroscience Medicinal Chemistry, Janssen Research & Development, Janssen Pharmaceutica NV, Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Andrés A. Trabanco
- Neuroscience Medicinal Chemistry, Janssen Research & Development, Janssen−Cilag SA, C/Jarama 75A, 45007 Toledo, Spain
| |
Collapse
|
20
|
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: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Indexed: 12/19/2022]
|
21
|
Ahmad A, Ramasamy K, Majeed ABA, Mani V. Enhancement of β-secretase inhibition and antioxidant activities of tempeh, a fermented soybean cake through enrichment of bioactive aglycones. PHARMACEUTICAL BIOLOGY 2015; 53:758-66. [PMID: 25756802 DOI: 10.3109/13880209.2014.942791] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
CONTEXT Soybean and its fermented products are the most common source of isoflavones in human food. OBJECTIVE The present study quantifies the major glycosides and aglycones in soybean and its fermented product tempeh isoflavone extracts. The comparision of antioxidant effects and BACE1 inhibitory activity between the isoflavones of soybean and tempeh were also established. MATERIALS AND METHODS The major isoflavones such as daidzein and genistein (aglycones), and their sugar conjugates (glycosides) daidzin and genistin in soybean and tempeh isoflavones were quantified using HPLC analysis. Comparative studies on BACE 1 (β-site amyloid precursor protein cleaving enzyme 1 or β-secretase 1) inhibition and free-radical scavenging activities (diphenyl-1-picrylhydrazyl (DPPH) and ferrous ion chelating ability) were conducted. RESULTS The amount of actives (mg/100 g) in soybean isoflavone compared with tempeh isoflavone is as follows: daidzein 16.72 mg/100 g versus 38.91 mg/100 g, genistein 11.10 mg/100 g versus 24.03 mg/100 g, daidzin 6.16 mg/100 g versus 0.69 mg/100 g, and genistin 24.61 mg/100 g versus 6.57 mg/100 g. The IC50 values of soybean and tempeh isoflavones against BACE1 were 10.87 and 5.47 mg/ml, respectively. The tempeh isoflavone had a more potent DPPH free-radical scavenging activity (IC50 = 2.67 mg/ml) than the soybean isoflavone (IC50 = 10 mg/ml). The ferrous ion chelating ability of the isoflavones was practically similar (IC50 = 10.40 mg/ml, soybean and 11.13 mg/ml, tempeh). DISCUSSION AND CONCLUSION The present study indicates that tempeh is a healthy supplement to alleviate oxidative stress through the enrichment of aglycones.
Collapse
Affiliation(s)
- Aliya Ahmad
- Faculty of Pharmacy, Puncak Alam Campus, Universiti Teknologi MARA (UiTM) , Selangor Darul Ehsan , Malaysia and
| | | | | | | |
Collapse
|
22
|
Hossain T, Mukherjee A, Saha A. Chemometric design to explore pharmacophore features of BACE inhibitors for controlling Alzheimer's disease. MOLECULAR BIOSYSTEMS 2015; 11:549-57. [DOI: 10.1039/c4mb00540f] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Pharmacophoric features of potent BACE inhibitors derived from multi-chemometric studies.
Collapse
Affiliation(s)
- Tabassum Hossain
- Department of Chemical Technology
- University of Calcutta
- Kolkata-700009
- India
| | - Arup Mukherjee
- Department of Chemical Technology
- University of Calcutta
- Kolkata-700009
- India
| | - Achintya Saha
- Department of Chemical Technology
- University of Calcutta
- Kolkata-700009
- India
| |
Collapse
|
23
|
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: 12.6] [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]
|
24
|
Viklund J, Kolmodin K, Nordvall G, Swahn BM, Svensson M, Gravenfors Y, Rahm F. Creation of Novel Cores for β-Secretase (BACE-1) Inhibitors: A Multiparameter Lead Generation Strategy. ACS Med Chem Lett 2014; 5:440-5. [PMID: 24900855 DOI: 10.1021/ml5000433] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Accepted: 02/03/2014] [Indexed: 12/18/2022] Open
Abstract
In order to find optimal core structures as starting points for lead optimization, a multiparameter lead generation workflow was designed with the goal of finding BACE-1 inhibitors as a treatment for Alzheimer's disease. De novo design of core fragments was connected with three predictive in silico models addressing target affinity, permeability, and hERG activity, in order to guide synthesis. Taking advantage of an additive SAR, the prioritized cores were decorated with a few, well-characterized substituents from known BACE-1 inhibitors in order to allow for core-to-core comparisons. Prediction methods and analyses of how physicochemical properties of the core structures correlate to in vitro data are described. The syntheses and in vitro data of the test compounds are reported in a separate paper by Ginman et al. [J. Med. Chem. 2013, 56, 4181-4205]. The affinity predictions are described in detail by Roos et al. [J. Chem. Inf. 2014, DOI: 10.1021/ci400374z].
Collapse
Affiliation(s)
- Jenny Viklund
- Department of Medicinal Chemistry AstraZeneca R&D Södertälje, SE-151 85 Södertälje, Sweden
| | - Karin Kolmodin
- Department of Medicinal Chemistry AstraZeneca R&D Södertälje, SE-151 85 Södertälje, Sweden
| | - Gunnar Nordvall
- Department of Medicinal Chemistry AstraZeneca R&D Södertälje, SE-151 85 Södertälje, Sweden
| | - Britt-Marie Swahn
- Department of Medicinal Chemistry AstraZeneca R&D Södertälje, SE-151 85 Södertälje, Sweden
| | - Mats Svensson
- Department of Medicinal Chemistry AstraZeneca R&D Södertälje, SE-151 85 Södertälje, Sweden
| | - Ylva Gravenfors
- Department of Medicinal Chemistry AstraZeneca R&D Södertälje, SE-151 85 Södertälje, Sweden
| | - Fredrik Rahm
- Department of Medicinal Chemistry AstraZeneca R&D Södertälje, SE-151 85 Södertälje, Sweden
| |
Collapse
|
25
|
Roos K, Viklund J, Meuller J, Kaspersson K, Svensson M. Potency prediction of β-secretase (BACE-1) inhibitors using density functional methods. J Chem Inf Model 2014; 54:818-25. [PMID: 24456077 DOI: 10.1021/ci400374z] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Scoring potency is a main challenge for structure based drug design. Inductive effects of subtle variations in the ligand are not possible to accurately predict by classical computational chemistry methods. In this study, the problem of predicting potency of ligands with electronic variations participating in key interactions with the protein was addressed. The potency was predicted for a large set of cyclic amidine and guanidine cores extracted from β-secretase (BACE-1) inhibitors. All cores were of similar size and had equal interaction motifs but were diverse with respect to electronic substitutions. A density functional theory approach, in combination with a representation of the active site of a protein using only key residues, was shown to be predictive. This computational approach was used to guide and support drug design, within the time frame of a normal drug discovery design cycle.
Collapse
Affiliation(s)
- Katarina Roos
- Department of Medicinal Chemistry and §Discovery Sciences, AstraZeneca R&D Mölndal , SE-431 83 Mölndal, Sweden
| | | | | | | | | |
Collapse
|
26
|
Dessolin J. N-(3-(2-amino-6,6-difluoro-4,4a,5,6,7,7a-hexahydro-cyclopenta[e][1,3]oxazin-4-yl)-phenyl)-amides as BACE1 inhibitors: a patent evaluation of WO2013041499. Expert Opin Ther Pat 2013; 24:239-42. [PMID: 24219148 DOI: 10.1517/13543776.2014.859246] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
A new series of oxazin amides have been synthesized from isoxazoles using a reaction to increase the heterocyclic ring size and were evaluated as BACE1 inhibitors. The innovative compounds were able to diminish amyloid-β peptide concentration in cell and proved to be selective toward peptidases from the same family. Further studies on the toxicity of this series showed that these new molecules were not recognized by P-glycoprotein and that they were unsusceptible to rapid metabolization by cytochrome P450 or glutathione conjugation. These results indicate that such compounds could be useful in developing drugs to fight Alzheimer's disease and that this novel oxazin scaffold should be considered as a starting point to tackle this pathology.
Collapse
Affiliation(s)
- Jean Dessolin
- Institut Européen de Chimie et Biologie (IECB), CNRS UMR 5248 Chimie et Biologie des Membranes et des Nano-objets (CBMN) CBMN-IECB , 2 rue Robert Escarpit, 33607 Pessac Cedex , France
| |
Collapse
|
27
|
BACE1 inhibitors: A head group scan on a series of amides. Bioorg Med Chem Lett 2013; 23:4239-43. [DOI: 10.1016/j.bmcl.2013.05.003] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2013] [Revised: 04/29/2013] [Accepted: 05/01/2013] [Indexed: 01/09/2023]
|
28
|
Ginman T, Viklund J, Malmström J, Blid J, Emond R, Forsblom R, Johansson A, Kers A, Lake F, Sehgelmeble F, Sterky KJ, Bergh M, Lindgren A, Johansson P, Jeppsson F, Fälting J, Gravenfors Y, Rahm F. Core refinement toward permeable β-secretase (BACE-1) inhibitors with low hERG activity. J Med Chem 2013; 56:4181-205. [PMID: 23126626 DOI: 10.1021/jm3011349] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
By use of iterative design aided by predictive models for target affinity, brain permeability, and hERG activity, novel and diverse compounds based on cyclic amidine and guanidine cores were synthesized with the goal of finding BACE-1 inhibitors as a treatment for Alzheimer's disease. Since synthesis feasibility had low priority in the design of the cores, an extensive synthesis effort was needed to make the relevant compounds. Syntheses of these compounds are reported, together with physicochemical properties and structure-activity relationships based on in vitro data. Four crystal structures of diverse amidines binding in the active site are deposited and discussed. Inhibitors of BACE-1 with 3 μM to 32 nM potencies in cells are shown, together with data on in vivo brain exposure levels for four compounds. The results presented show the importance of the core structure for the profile of the final compounds.
Collapse
Affiliation(s)
- Tobias Ginman
- Department of Medicinal Chemistry, AstraZeneca R&D Södertälje, SE-151 85, Södertälje, Sweden
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Yuan J, Venkatraman S, Zheng Y, McKeever BM, Dillard LW, Singh SB. Structure-based design of β-site APP cleaving enzyme 1 (BACE1) inhibitors for the treatment of Alzheimer's disease. J Med Chem 2013; 56:4156-80. [PMID: 23509904 DOI: 10.1021/jm301659n] [Citation(s) in RCA: 113] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The amyloid hypothesis asserts that excess production or reduced clearance of the amyloid-β (Aβ) peptides in the brain initiates a sequence of events that ultimately lead to Alzheimer's disease and dementia. The Aβ hypothesis has identified BACE1 as a therapeutic target to treat Alzheimer's and led to medicinal chemistry efforts to design its inhibitors both in the pharmaceutical industry and in academia. This review summarizes two distinct categories of inhibitors designed based on conformational states of "closed" and "open" forms of the enzyme. In each category the inhibitors are classified based on the core catalytic interaction group or the aspartyl binding motif (ABM). This review covers the description of inhibitors in each ABM class with X-ray crystal structures of key compounds, their binding modes, related structure-activity data highlighting potency advances, and additional properties such as selectivity profile, P-gp efflux, pharmacokinetic, and pharmacodynamic data.
Collapse
Affiliation(s)
- Jing Yuan
- Vitae Pharmaceuticals, 502 W. Office Center Drive, Fort Washington, Pennsylvania 19034, USA
| | | | | | | | | | | |
Collapse
|
30
|
Edraki N, Firuzi O, Foroumadi A, Miri R, Madadkar-Sobhani A, Khoshneviszadeh M, Shafiee A. Phenylimino-2 H -chromen-3-carboxamide derivatives as novel small molecule inhibitors of β-secretase (BACE1). Bioorg Med Chem 2013; 21:2396-2412. [DOI: 10.1016/j.bmc.2013.01.064] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2012] [Revised: 01/19/2013] [Accepted: 01/22/2013] [Indexed: 11/29/2022]
|
31
|
Hossain T, Islam MA, Pal R, Saha A. Exploring structural requirement and binding interactions of β-amyloid cleavage enzyme inhibitors using molecular modeling techniques. Med Chem Res 2013. [DOI: 10.1007/s00044-013-0481-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
32
|
Butini S, Brogi S, Novellino E, Campiani G, Ghosh AK, Brindisi M, Gemma S. The structural evolution of β-secretase inhibitors: a focus on the development of small-molecule inhibitors. Curr Top Med Chem 2013; 13:1787-807. [PMID: 23931442 PMCID: PMC6034716 DOI: 10.2174/15680266113139990137] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2013] [Accepted: 05/11/2013] [Indexed: 12/12/2022]
Abstract
Effective treatment of Alzheimer's disease (AD) remains a critical unmet need in medicine. The lack of useful treatment for AD led to an intense search for novel therapies based on the amyloid hypothesis, which states that amyloid β-42 (Aβ42) plays an early and crucial role in all cases of AD. β-Secretase (also known as BACE-1 β-site APP-cleaving enzyme, Asp-2 or memapsin-2) is an aspartyl protease representing the rate limiting step in the generation of Aβ peptide fragments, therefore it could represent an important target in the steady hunt for a disease-modifying treatment. Generally, β-secretase inhibitors are grouped into two families: peptidomimetic and nonpeptidomimetic inhibitors. However, irrespective of the class, serious challenges with respect to blood-brain barrier (BBB) penetration and selectivity still remain. Discovering a small molecule inhibitor of β-secretase represents an unnerving challenge but, due to its significant potential as a therapeutic target, growing efforts in this task are evident from both academic and industrial laboratories. In this frame, the rising availability of crystal structures of β-secretase-inhibitor complexes represents an invaluable opportunity for optimization. Nevertheless, beyond the inhibitory activity, the major issue of the current research approaches is about problems associated with BBB penetration and pharmacokinetic properties. This review follows the structural evolution of the early β-secretase inhibitors and gives a snap-shot of the hottest chemical templates in the literature of the last five years, showing research progress in this field.
Collapse
Affiliation(s)
- Stefania Butini
- European Research Centre for Drug Discovery and Development (NatSynDrugs), University of Siena, Italy
| | - Simone Brogi
- European Research Centre for Drug Discovery and Development (NatSynDrugs), University of Siena, Italy
| | - Ettore Novellino
- European Research Centre for Drug Discovery and Development (NatSynDrugs), University of Siena, Italy
- Dipartimento di Farmacia, University of Naples Federico II, Italy
| | - Giuseppe Campiani
- European Research Centre for Drug Discovery and Development (NatSynDrugs), University of Siena, Italy
| | - Arun K. Ghosh
- Department of Chemistry and Department of Medicinal Chemistry, Purdue University, West Lafayette, Indiana 47907, United States
| | - Margherita Brindisi
- European Research Centre for Drug Discovery and Development (NatSynDrugs), University of Siena, Italy
| | - Sandra Gemma
- European Research Centre for Drug Discovery and Development (NatSynDrugs), University of Siena, Italy
| |
Collapse
|
33
|
Gravenfors Y, Viklund J, Blid J, Ginman T, Karlström S, Kihlström J, Kolmodin K, Lindström J, von Berg S, von Kieseritzky F, Bogar K, Slivo C, Swahn BM, Olsson LL, Johansson P, Eketjäll S, Fälting J, Jeppsson F, Strömberg K, Janson J, Rahm F. New aminoimidazoles as β-secretase (BACE-1) inhibitors showing amyloid-β (Aβ) lowering in brain. J Med Chem 2012; 55:9297-311. [PMID: 23017051 DOI: 10.1021/jm300991n] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Amino-2H-imidazoles are described as a new class of BACE-1 inhibitors for the treatment of Alzheimer's disease. Synthetic methods, crystal structures, and structure-activity relationships for target activity, permeability, and hERG activity are reported and discussed. Compound (S)-1m was one of the most promising compounds in this report, with high potency in the cellular assay and a good overall profile. When guinea pigs were treated with compound (S)-1m, a concentration and time dependent decrease in Aβ40 and Aβ42 levels in plasma, brain, and CSF was observed. The maximum reduction of brain Aβ was 40-50%, 1.5 h after oral dosing (100 μmol/kg). The results presented highlight the potential of this new class of BACE-1 inhibitors with good target potency and with low effect on hERG, in combination with a fair CNS exposure in vivo.
Collapse
Affiliation(s)
- Ylva Gravenfors
- Department of Medicinal Chemistry, AstraZeneca R&D Södertälje, SE-151 85 Södertälje, Sweden.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Probst G, Xu YZ. Small-molecule BACE1 inhibitors: a patent literature review (2006 - 2011). Expert Opin Ther Pat 2012; 22:511-40. [PMID: 22512789 DOI: 10.1517/13543776.2012.681302] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Alzheimer's disease is a devastating neurodegenerative disorder for which no disease-modifying therapy exists. The amyloid hypothesis, which implicates Aβ as the toxin initiating a biological cascade leading to neurodegeneration, is the most prominent theory concerning the underlying cause of the disease. BACE1 is one of two aspartyl proteinases that generate Aβ, thus inhibition of BACE1 has the potential to ameliorate the progression of Alzheimer's disease by abating the production of Aβ. AREAS COVERED This review chronicles small-molecule BACE1 inhibitors as described in the patent literature between 2006 and 2011 and their potential use as disease-modifying treatments for Alzheimer's disease. Over the past half a dozen years, numerous BACE1 inhibitors have been published in the patent applications, but often these contain a paltry amount of pertinent biological data (e.g. potency, selectivity, and efficacy). Fortunately, numerous relevant publications containing important data have appeared in the journal literature during this period. The goal in this effort was to create an amalgam of the two records to add value to this review. EXPERT OPINION The pharmaceutical industry has made tremendous progress in the development of small-molecule BACE1 inhibitors that lower Aβ in the central nervous system. Assuming the amyloid hypothesis is veracious, we anticipate a disease-modifying therapy to combat Alzheimer's disease is near.
Collapse
Affiliation(s)
- Gary Probst
- Elan Pharmaceuticals, Molecular Design, 180 Oyster Point Boulevard, South San Francisco, CA 94080, USA.
| | | |
Collapse
|
35
|
Weiss MM, Williamson T, Babu-Khan S, Bartberger MD, Brown J, Chen K, Cheng Y, Citron M, Croghan MD, Dineen TA, Esmay J, Graceffa RF, Harried SS, Hickman D, Hitchcock SA, Horne DB, Huang H, Imbeah-Ampiah R, Judd T, Kaller MR, Kreiman CR, La DS, Li V, Lopez P, Louie S, Monenschein H, Nguyen TT, Pennington LD, Rattan C, San Miguel T, Sickmier E, Wahl RC, Wen PH, Wood S, Xue Q, Yang BH, Patel VF, Zhong W. Design and Preparation of a Potent Series of Hydroxyethylamine Containing β-Secretase Inhibitors That Demonstrate Robust Reduction of Central β-Amyloid. J Med Chem 2012; 55:9009-24. [DOI: 10.1021/jm300119p] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Matthew M. Weiss
- Departments
of Chemistry Research and Discovery, ‡Neuroscience, §HTS Molecular Pharmacology, ∥Molecular Structure, ⊥Pharmacokinetics
and Drug Metabolism, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States,
and One Amgen Center Drive, Thousand Oaks, California 91320, United
States
| | - Toni Williamson
- Departments
of Chemistry Research and Discovery, ‡Neuroscience, §HTS Molecular Pharmacology, ∥Molecular Structure, ⊥Pharmacokinetics
and Drug Metabolism, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States,
and One Amgen Center Drive, Thousand Oaks, California 91320, United
States
| | - Safura Babu-Khan
- Departments
of Chemistry Research and Discovery, ‡Neuroscience, §HTS Molecular Pharmacology, ∥Molecular Structure, ⊥Pharmacokinetics
and Drug Metabolism, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States,
and One Amgen Center Drive, Thousand Oaks, California 91320, United
States
| | - Michael D. Bartberger
- Departments
of Chemistry Research and Discovery, ‡Neuroscience, §HTS Molecular Pharmacology, ∥Molecular Structure, ⊥Pharmacokinetics
and Drug Metabolism, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States,
and One Amgen Center Drive, Thousand Oaks, California 91320, United
States
| | - James Brown
- Departments
of Chemistry Research and Discovery, ‡Neuroscience, §HTS Molecular Pharmacology, ∥Molecular Structure, ⊥Pharmacokinetics
and Drug Metabolism, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States,
and One Amgen Center Drive, Thousand Oaks, California 91320, United
States
| | - Kui Chen
- Departments
of Chemistry Research and Discovery, ‡Neuroscience, §HTS Molecular Pharmacology, ∥Molecular Structure, ⊥Pharmacokinetics
and Drug Metabolism, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States,
and One Amgen Center Drive, Thousand Oaks, California 91320, United
States
| | - Yuan Cheng
- Departments
of Chemistry Research and Discovery, ‡Neuroscience, §HTS Molecular Pharmacology, ∥Molecular Structure, ⊥Pharmacokinetics
and Drug Metabolism, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States,
and One Amgen Center Drive, Thousand Oaks, California 91320, United
States
| | - Martin Citron
- Departments
of Chemistry Research and Discovery, ‡Neuroscience, §HTS Molecular Pharmacology, ∥Molecular Structure, ⊥Pharmacokinetics
and Drug Metabolism, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States,
and One Amgen Center Drive, Thousand Oaks, California 91320, United
States
| | - Michael D. Croghan
- Departments
of Chemistry Research and Discovery, ‡Neuroscience, §HTS Molecular Pharmacology, ∥Molecular Structure, ⊥Pharmacokinetics
and Drug Metabolism, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States,
and One Amgen Center Drive, Thousand Oaks, California 91320, United
States
| | - Thomas A. Dineen
- Departments
of Chemistry Research and Discovery, ‡Neuroscience, §HTS Molecular Pharmacology, ∥Molecular Structure, ⊥Pharmacokinetics
and Drug Metabolism, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States,
and One Amgen Center Drive, Thousand Oaks, California 91320, United
States
| | - Joel Esmay
- Departments
of Chemistry Research and Discovery, ‡Neuroscience, §HTS Molecular Pharmacology, ∥Molecular Structure, ⊥Pharmacokinetics
and Drug Metabolism, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States,
and One Amgen Center Drive, Thousand Oaks, California 91320, United
States
| | - Russell F. Graceffa
- Departments
of Chemistry Research and Discovery, ‡Neuroscience, §HTS Molecular Pharmacology, ∥Molecular Structure, ⊥Pharmacokinetics
and Drug Metabolism, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States,
and One Amgen Center Drive, Thousand Oaks, California 91320, United
States
| | - Scott S. Harried
- Departments
of Chemistry Research and Discovery, ‡Neuroscience, §HTS Molecular Pharmacology, ∥Molecular Structure, ⊥Pharmacokinetics
and Drug Metabolism, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States,
and One Amgen Center Drive, Thousand Oaks, California 91320, United
States
| | - Dean Hickman
- Departments
of Chemistry Research and Discovery, ‡Neuroscience, §HTS Molecular Pharmacology, ∥Molecular Structure, ⊥Pharmacokinetics
and Drug Metabolism, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States,
and One Amgen Center Drive, Thousand Oaks, California 91320, United
States
| | - Stephen A. Hitchcock
- Departments
of Chemistry Research and Discovery, ‡Neuroscience, §HTS Molecular Pharmacology, ∥Molecular Structure, ⊥Pharmacokinetics
and Drug Metabolism, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States,
and One Amgen Center Drive, Thousand Oaks, California 91320, United
States
| | - Daniel B. Horne
- Departments
of Chemistry Research and Discovery, ‡Neuroscience, §HTS Molecular Pharmacology, ∥Molecular Structure, ⊥Pharmacokinetics
and Drug Metabolism, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States,
and One Amgen Center Drive, Thousand Oaks, California 91320, United
States
| | - Hongbing Huang
- Departments
of Chemistry Research and Discovery, ‡Neuroscience, §HTS Molecular Pharmacology, ∥Molecular Structure, ⊥Pharmacokinetics
and Drug Metabolism, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States,
and One Amgen Center Drive, Thousand Oaks, California 91320, United
States
| | - Ronke Imbeah-Ampiah
- Departments
of Chemistry Research and Discovery, ‡Neuroscience, §HTS Molecular Pharmacology, ∥Molecular Structure, ⊥Pharmacokinetics
and Drug Metabolism, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States,
and One Amgen Center Drive, Thousand Oaks, California 91320, United
States
| | - Ted Judd
- Departments
of Chemistry Research and Discovery, ‡Neuroscience, §HTS Molecular Pharmacology, ∥Molecular Structure, ⊥Pharmacokinetics
and Drug Metabolism, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States,
and One Amgen Center Drive, Thousand Oaks, California 91320, United
States
| | - Matthew R. Kaller
- Departments
of Chemistry Research and Discovery, ‡Neuroscience, §HTS Molecular Pharmacology, ∥Molecular Structure, ⊥Pharmacokinetics
and Drug Metabolism, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States,
and One Amgen Center Drive, Thousand Oaks, California 91320, United
States
| | - Charles R. Kreiman
- Departments
of Chemistry Research and Discovery, ‡Neuroscience, §HTS Molecular Pharmacology, ∥Molecular Structure, ⊥Pharmacokinetics
and Drug Metabolism, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States,
and One Amgen Center Drive, Thousand Oaks, California 91320, United
States
| | - Daniel S. La
- Departments
of Chemistry Research and Discovery, ‡Neuroscience, §HTS Molecular Pharmacology, ∥Molecular Structure, ⊥Pharmacokinetics
and Drug Metabolism, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States,
and One Amgen Center Drive, Thousand Oaks, California 91320, United
States
| | - Vivian Li
- Departments
of Chemistry Research and Discovery, ‡Neuroscience, §HTS Molecular Pharmacology, ∥Molecular Structure, ⊥Pharmacokinetics
and Drug Metabolism, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States,
and One Amgen Center Drive, Thousand Oaks, California 91320, United
States
| | - Patricia Lopez
- Departments
of Chemistry Research and Discovery, ‡Neuroscience, §HTS Molecular Pharmacology, ∥Molecular Structure, ⊥Pharmacokinetics
and Drug Metabolism, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States,
and One Amgen Center Drive, Thousand Oaks, California 91320, United
States
| | - Steven Louie
- Departments
of Chemistry Research and Discovery, ‡Neuroscience, §HTS Molecular Pharmacology, ∥Molecular Structure, ⊥Pharmacokinetics
and Drug Metabolism, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States,
and One Amgen Center Drive, Thousand Oaks, California 91320, United
States
| | - Holger Monenschein
- Departments
of Chemistry Research and Discovery, ‡Neuroscience, §HTS Molecular Pharmacology, ∥Molecular Structure, ⊥Pharmacokinetics
and Drug Metabolism, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States,
and One Amgen Center Drive, Thousand Oaks, California 91320, United
States
| | - Thomas T. Nguyen
- Departments
of Chemistry Research and Discovery, ‡Neuroscience, §HTS Molecular Pharmacology, ∥Molecular Structure, ⊥Pharmacokinetics
and Drug Metabolism, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States,
and One Amgen Center Drive, Thousand Oaks, California 91320, United
States
| | - Lewis D. Pennington
- Departments
of Chemistry Research and Discovery, ‡Neuroscience, §HTS Molecular Pharmacology, ∥Molecular Structure, ⊥Pharmacokinetics
and Drug Metabolism, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States,
and One Amgen Center Drive, Thousand Oaks, California 91320, United
States
| | - Claire Rattan
- Departments
of Chemistry Research and Discovery, ‡Neuroscience, §HTS Molecular Pharmacology, ∥Molecular Structure, ⊥Pharmacokinetics
and Drug Metabolism, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States,
and One Amgen Center Drive, Thousand Oaks, California 91320, United
States
| | - Tisha San Miguel
- Departments
of Chemistry Research and Discovery, ‡Neuroscience, §HTS Molecular Pharmacology, ∥Molecular Structure, ⊥Pharmacokinetics
and Drug Metabolism, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States,
and One Amgen Center Drive, Thousand Oaks, California 91320, United
States
| | - E.Allen Sickmier
- Departments
of Chemistry Research and Discovery, ‡Neuroscience, §HTS Molecular Pharmacology, ∥Molecular Structure, ⊥Pharmacokinetics
and Drug Metabolism, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States,
and One Amgen Center Drive, Thousand Oaks, California 91320, United
States
| | - Robert C. Wahl
- Departments
of Chemistry Research and Discovery, ‡Neuroscience, §HTS Molecular Pharmacology, ∥Molecular Structure, ⊥Pharmacokinetics
and Drug Metabolism, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States,
and One Amgen Center Drive, Thousand Oaks, California 91320, United
States
| | - Paul H. Wen
- Departments
of Chemistry Research and Discovery, ‡Neuroscience, §HTS Molecular Pharmacology, ∥Molecular Structure, ⊥Pharmacokinetics
and Drug Metabolism, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States,
and One Amgen Center Drive, Thousand Oaks, California 91320, United
States
| | - Stephen Wood
- Departments
of Chemistry Research and Discovery, ‡Neuroscience, §HTS Molecular Pharmacology, ∥Molecular Structure, ⊥Pharmacokinetics
and Drug Metabolism, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States,
and One Amgen Center Drive, Thousand Oaks, California 91320, United
States
| | - Qiufen Xue
- Departments
of Chemistry Research and Discovery, ‡Neuroscience, §HTS Molecular Pharmacology, ∥Molecular Structure, ⊥Pharmacokinetics
and Drug Metabolism, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States,
and One Amgen Center Drive, Thousand Oaks, California 91320, United
States
| | - Bryant H. Yang
- Departments
of Chemistry Research and Discovery, ‡Neuroscience, §HTS Molecular Pharmacology, ∥Molecular Structure, ⊥Pharmacokinetics
and Drug Metabolism, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States,
and One Amgen Center Drive, Thousand Oaks, California 91320, United
States
| | - Vinod F. Patel
- Departments
of Chemistry Research and Discovery, ‡Neuroscience, §HTS Molecular Pharmacology, ∥Molecular Structure, ⊥Pharmacokinetics
and Drug Metabolism, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States,
and One Amgen Center Drive, Thousand Oaks, California 91320, United
States
| | - Wenge Zhong
- Departments
of Chemistry Research and Discovery, ‡Neuroscience, §HTS Molecular Pharmacology, ∥Molecular Structure, ⊥Pharmacokinetics
and Drug Metabolism, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States,
and One Amgen Center Drive, Thousand Oaks, California 91320, United
States
| |
Collapse
|
36
|
Secretase Inhibitors and Modulators as a Disease-Modifying Approach Against Alzheimer's Disease. ANNUAL REPORTS IN MEDICINAL CHEMISTRY 2012. [DOI: 10.1016/b978-0-12-396492-2.00005-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register]
|
37
|
Delgado O, Monteagudo A, Van Gool M, Trabanco AA, Fustero S. A practical entry to β-aryl-β-alkyl amino alcohols: application to the synthesis of a potent BACE1 inhibitor. Org Biomol Chem 2012; 10:6758-66. [DOI: 10.1039/c2ob25845e] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|