51
|
Mondal M, Unver MY, Pal A, Bakker M, Berrier SP, Hirsch AKH. Fragment-Based Drug Design Facilitated by Protein-Templated Click Chemistry: Fragment Linking and Optimization of Inhibitors of the Aspartic Protease Endothiapepsin. Chemistry 2016; 22:14826-14830. [PMID: 27604032 PMCID: PMC5095814 DOI: 10.1002/chem.201603001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Indexed: 01/17/2023]
Abstract
There is an urgent need for the development of efficient methodologies that accelerate drug discovery. We demonstrate that the strategic combination of fragment linking/optimization and protein-templated click chemistry is an efficient and powerful method that accelerates the hit-identification process for the aspartic protease endothiapepsin. The best binder, which inhibits endothiapepsin with an IC50 value of 43 μm, represents the first example of triazole-based inhibitors of endothiapepsin. Our strategy could find application on a whole range of drug targets.
Collapse
Affiliation(s)
- Milon Mondal
- Stratingh Institute for Chemistry, University of Groningen, Nijenborgh 7, 9747 AG, Groningen, The Netherlands
| | - M Yagiz Unver
- Stratingh Institute for Chemistry, University of Groningen, Nijenborgh 7, 9747 AG, Groningen, The Netherlands
| | - Asish Pal
- Institute of Nano Science and Technology, Sector 64, Mohali, Punjab, 160062, India
| | - Matthijs Bakker
- Stratingh Institute for Chemistry, University of Groningen, Nijenborgh 7, 9747 AG, Groningen, The Netherlands
| | - Stephan P Berrier
- Stratingh Institute for Chemistry, University of Groningen, Nijenborgh 7, 9747 AG, Groningen, The Netherlands
| | - Anna K H Hirsch
- Stratingh Institute for Chemistry, University of Groningen, Nijenborgh 7, 9747 AG, Groningen, The Netherlands.
| |
Collapse
|
52
|
Song R, Han Z, He Q, Fan R. Amine-Mediated Transimination and Aromatization-Triggered Domino Reaction in the Synthesis of Polyfunctionalized 4-Aminoquinolines. Org Lett 2016; 18:5328-5331. [DOI: 10.1021/acs.orglett.6b02643] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Affiliation(s)
- Runzhe Song
- Department
of Chemistry, Fudan University, 220 Handan Road, Shanghai, 200433, China
| | - Zhaomeng Han
- Department
of Chemistry, Fudan University, 220 Handan Road, Shanghai, 200433, China
| | - Qiuqin He
- Department
of Chemistry, Fudan University, 220 Handan Road, Shanghai, 200433, China
| | - Renhua Fan
- Department
of Chemistry, Fudan University, 220 Handan Road, Shanghai, 200433, China
| |
Collapse
|
53
|
Hua Z, Fang L, Wu S, Wang L. Copper-Catalyzed Cross-Dehydrogenative Coupling ofN-Iminoquinolinium Ylides with Secondary Amines. European J Org Chem 2016. [DOI: 10.1002/ejoc.201600905] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Zerui Hua
- Key Laboratory of Advanced Materials and Institute of Fine Chemicals; East China University of Science and Technology; 130 Meilong Road 200237 Shanghai P. R. China
| | - Lei Fang
- Key Laboratory of Advanced Materials and Institute of Fine Chemicals; East China University of Science and Technology; 130 Meilong Road 200237 Shanghai P. R. China
| | - Shengying Wu
- Key Laboratory of Advanced Materials and Institute of Fine Chemicals; East China University of Science and Technology; 130 Meilong Road 200237 Shanghai P. R. China
| | - Limin Wang
- Key Laboratory of Advanced Materials and Institute of Fine Chemicals; East China University of Science and Technology; 130 Meilong Road 200237 Shanghai P. R. China
| |
Collapse
|
54
|
Jin H, Tian B, Song X, Xie J, Rudolph M, Rominger F, Hashmi ASK. Gold-katalysierte Synthese von Chinolinen aus Propargylsilylethern und Anthranilen über die Umpolung eines Goldcarben-Kohlenstoffatoms. Angew Chem Int Ed Engl 2016. [DOI: 10.1002/ange.201606043] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Hongming Jin
- Organisch-Chemisches Institut; Universität Heidelberg; Im Neuenheimer Feld 270, 6 9120 Heidelberg Deutschland
| | - Bin Tian
- Organisch-Chemisches Institut; Universität Heidelberg; Im Neuenheimer Feld 270, 6 9120 Heidelberg Deutschland
| | - Xinlong Song
- Organisch-Chemisches Institut; Universität Heidelberg; Im Neuenheimer Feld 270, 6 9120 Heidelberg Deutschland
| | - Jin Xie
- Organisch-Chemisches Institut; Universität Heidelberg; Im Neuenheimer Feld 270, 6 9120 Heidelberg Deutschland
| | - Matthias Rudolph
- Organisch-Chemisches Institut; Universität Heidelberg; Im Neuenheimer Feld 270, 6 9120 Heidelberg Deutschland
| | - Frank Rominger
- Organisch-Chemisches Institut; Universität Heidelberg; Im Neuenheimer Feld 270, 6 9120 Heidelberg Deutschland
| | - A. Stephen K. Hashmi
- Organisch-Chemisches Institut; Universität Heidelberg; Im Neuenheimer Feld 270, 6 9120 Heidelberg Deutschland
- Chemistry Department, Faculty of Science; King Abdulaziz University (KAU); 21589 Jeddah Saudi-Arabien
| |
Collapse
|
55
|
Jin H, Tian B, Song X, Xie J, Rudolph M, Rominger F, Hashmi ASK. Gold-Catalyzed Synthesis of Quinolines from Propargyl Silyl Ethers and Anthranils through the Umpolung of a Gold Carbene Carbon. Angew Chem Int Ed Engl 2016; 55:12688-92. [DOI: 10.1002/anie.201606043] [Citation(s) in RCA: 170] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Indexed: 12/17/2022]
Affiliation(s)
- Hongming Jin
- Organisch-Chemisches Institut; Universität Heidelberg; Im Neuenheimer Feld 270 69120 Heidelberg Germany
| | - Bin Tian
- Organisch-Chemisches Institut; Universität Heidelberg; Im Neuenheimer Feld 270 69120 Heidelberg Germany
| | - Xinlong Song
- Organisch-Chemisches Institut; Universität Heidelberg; Im Neuenheimer Feld 270 69120 Heidelberg Germany
| | - Jin Xie
- Organisch-Chemisches Institut; Universität Heidelberg; Im Neuenheimer Feld 270 69120 Heidelberg Germany
| | - Matthias Rudolph
- Organisch-Chemisches Institut; Universität Heidelberg; Im Neuenheimer Feld 270 69120 Heidelberg Germany
| | - Frank Rominger
- Organisch-Chemisches Institut; Universität Heidelberg; Im Neuenheimer Feld 270 69120 Heidelberg Germany
| | - A. Stephen K. Hashmi
- Organisch-Chemisches Institut; Universität Heidelberg; Im Neuenheimer Feld 270 69120 Heidelberg Germany
- Chemistry Department, Faculty of Science; King Abdulaziz University (KAU); 21589 Jeddah Saudi Arabia
| |
Collapse
|
56
|
Katsila T, Spyroulias GA, Patrinos GP, Matsoukas MT. Computational approaches in target identification and drug discovery. Comput Struct Biotechnol J 2016; 14:177-84. [PMID: 27293534 PMCID: PMC4887558 DOI: 10.1016/j.csbj.2016.04.004] [Citation(s) in RCA: 198] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2016] [Revised: 04/21/2016] [Accepted: 04/25/2016] [Indexed: 12/31/2022] Open
Abstract
In the big data era, voluminous datasets are routinely acquired, stored and analyzed with the aim to inform biomedical discoveries and validate hypotheses. No doubt, data volume and diversity have dramatically increased by the advent of new technologies and open data initiatives. Big data are used across the whole drug discovery pipeline from target identification and mechanism of action to identification of novel leads and drug candidates. Such methods are depicted and discussed, with the aim to provide a general view of computational tools and databases available. We feel that big data leveraging needs to be cost-effective and focus on personalized medicine. For this, we propose the interplay of information technologies and (chemo)informatic tools on the basis of their synergy.
Collapse
Affiliation(s)
- Theodora Katsila
- University of Patras, School of Health Sciences, Department of Pharmacy, University Campus, Rion, Patras, Greece
| | - Georgios A. Spyroulias
- University of Patras, School of Health Sciences, Department of Pharmacy, University Campus, Rion, Patras, Greece
| | - George P. Patrinos
- University of Patras, School of Health Sciences, Department of Pharmacy, University Campus, Rion, Patras, Greece
- Department of Pathology, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates
| | - Minos-Timotheos Matsoukas
- University of Patras, School of Health Sciences, Department of Pharmacy, University Campus, Rion, Patras, Greece
| |
Collapse
|
57
|
Jordan JB, Whittington DA, Bartberger MD, Sickmier EA, Chen K, Cheng Y, Judd T. Fragment-Linking Approach Using 19F NMR Spectroscopy To Obtain Highly Potent and Selective Inhibitors of β-Secretase. J Med Chem 2016; 59:3732-49. [DOI: 10.1021/acs.jmedchem.5b01917] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- John B. Jordan
- Therapeutic Discovery, Amgen, Inc. One Amgen Center Drive, Thousand
Oaks, California 91320, United States
| | - Douglas A. Whittington
- Therapeutic Discovery, Amgen, Inc. One Amgen Center Drive, Thousand
Oaks, California 91320, United States
| | - Michael D. Bartberger
- Therapeutic Discovery, Amgen, Inc. One Amgen Center Drive, Thousand
Oaks, California 91320, United States
| | - E. Allen Sickmier
- Therapeutic Discovery, Amgen, Inc. One Amgen Center Drive, Thousand
Oaks, California 91320, United States
| | - Kui Chen
- Therapeutic Discovery, Amgen, Inc. One Amgen Center Drive, Thousand
Oaks, California 91320, United States
| | - Yuan Cheng
- Therapeutic Discovery, Amgen, Inc. One Amgen Center Drive, Thousand
Oaks, California 91320, United States
| | - Ted Judd
- Therapeutic Discovery, Amgen, Inc. One Amgen Center Drive, Thousand
Oaks, California 91320, United States
| |
Collapse
|
58
|
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
|
59
|
Zheng Q, Ding Q, Wang C, Chen W, Peng Y. Synthesis of 2-aminoquinolines via palladium-catalyzed intermolecular oxidative cyclization of 2-vinylanilines with isocyanides. Tetrahedron 2016. [DOI: 10.1016/j.tet.2015.12.060] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
60
|
Wyss DF, Cumming JN, Strickland CO, Stamford AW. BACE Inhibitors. FRAGMENT-BASED DRUG DISCOVERY LESSONS AND OUTLOOK 2016. [DOI: 10.1002/9783527683604.ch14] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
61
|
Ghosh AK, Cárdenas EL, Osswald HL. The Design, Development, and Evaluation of BACE1 Inhibitors for the Treatment of Alzheimer’s Disease. TOPICS IN MEDICINAL CHEMISTRY 2016. [DOI: 10.1007/7355_2016_16] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
62
|
Dhiman S, Saini HK, Nandwana NK, Kumar D, Kumar A. Copper-catalyzed synthesis of quinoline derivatives via tandem Knoevenagel condensation, amination and cyclization. RSC Adv 2016. [DOI: 10.1039/c6ra03798d] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
A novel regioselective synthesis of 2-aminoquinolines and 2-arylquinoline-3-carbonitriles is described via copper-mediated tandem reaction.
Collapse
Affiliation(s)
- Shiv Dhiman
- Department of Chemistry
- Birla Institute of Technology and Science
- Pilani
- India
| | - Hitesh Kumar Saini
- Department of Chemistry
- Birla Institute of Technology and Science
- Pilani
- India
| | | | - Dalip Kumar
- Department of Chemistry
- Birla Institute of Technology and Science
- Pilani
- India
| | - Anil Kumar
- Department of Chemistry
- Birla Institute of Technology and Science
- Pilani
- India
| |
Collapse
|
63
|
Rasina D, Otikovs M, Leitans J, Recacha R, Borysov OV, Kanepe-Lapsa I, Domraceva I, Pantelejevs T, Tars K, Blackman MJ, Jaudzems K, Jirgensons A. Fragment-Based Discovery of 2-Aminoquinazolin-4(3H)-ones As Novel Class Nonpeptidomimetic Inhibitors of the Plasmepsins I, II, and IV. J Med Chem 2015; 59:374-87. [PMID: 26670264 DOI: 10.1021/acs.jmedchem.5b01558] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
2-Aminoquinazolin-4(3H)-ones were identified as a novel class of malaria digestive vacuole plasmepsin inhibitors by using NMR-based fragment screening against Plm II. Initial fragment hit optimization led to a submicromolar inhibitor, which was cocrystallized with Plm II to produce an X-ray structure of the complex. The structure showed that 2-aminoquinazolin-4(3H)-ones bind to the open flap conformation of the enzyme and provided clues to target the flap pocket. Further improvement in potency was achieved via introduction of hydrophobic substituents occupying the flap pocket. Most of the 2-aminoquinazolin-4(3H)-one based inhibitors show a similar activity against digestive Plms I, II, and IV and >10-fold selectivity versus CatD, although varying the flap pocket substituent led to one Plm IV selective inhibitor. In cell-based assays, the compounds show growth inhibition of Plasmodium falciparum 3D7 with IC50 ∼ 1 μM. Together, these results suggest 2-aminoquinazolin-4(3H)-ones as perspective leads for future development of an antimalarial agent.
Collapse
Affiliation(s)
- Dace Rasina
- Latvian Institute of Organic Synthesis , Aizkraukles 21, Riga LV-1006, Latvia
| | - Martins Otikovs
- Latvian Institute of Organic Synthesis , Aizkraukles 21, Riga LV-1006, Latvia
| | - Janis Leitans
- Biomedical Research and Study Centre , Ratsupites 1, Riga LV-1067, Latvia
| | - Rosario Recacha
- Latvian Institute of Organic Synthesis , Aizkraukles 21, Riga LV-1006, Latvia
| | - Oleksandr V Borysov
- Latvian Institute of Organic Synthesis , Aizkraukles 21, Riga LV-1006, Latvia
| | - Iveta Kanepe-Lapsa
- Latvian Institute of Organic Synthesis , Aizkraukles 21, Riga LV-1006, Latvia
| | - Ilona Domraceva
- Latvian Institute of Organic Synthesis , Aizkraukles 21, Riga LV-1006, Latvia
| | - Teodors Pantelejevs
- Latvian Institute of Organic Synthesis , Aizkraukles 21, Riga LV-1006, Latvia
| | - Kaspars Tars
- Biomedical Research and Study Centre , Ratsupites 1, Riga LV-1067, Latvia
| | - Michael J Blackman
- The Francis Crick Institute, Mill Hill Laboratory , The Ridgeway, Mill Hill, London NW7 1AA, U.K
| | - Kristaps Jaudzems
- Latvian Institute of Organic Synthesis , Aizkraukles 21, Riga LV-1006, Latvia
| | - Aigars Jirgensons
- Latvian Institute of Organic Synthesis , Aizkraukles 21, Riga LV-1006, Latvia
| |
Collapse
|
64
|
Konno H, Sato T, Saito Y, Sakamoto I, Akaji K. Synthesis and evaluation of aminopyridine derivatives as potential BACE1 inhibitors. Bioorg Med Chem Lett 2015; 25:5127-32. [DOI: 10.1016/j.bmcl.2015.10.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Revised: 09/29/2015] [Accepted: 10/03/2015] [Indexed: 11/29/2022]
|
65
|
Ghosh AK, Osswald HL. BACE1 (β-secretase) inhibitors for the treatment of Alzheimer's disease. Chem Soc Rev 2015; 43:6765-813. [PMID: 24691405 DOI: 10.1039/c3cs60460h] [Citation(s) in RCA: 233] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
BACE1 (β-secretase, memapsin 2, Asp2) has emerged as a promising target for the treatment of Alzheimer's disease. BACE1 is an aspartic protease which functions in the first step of the pathway leading to the production and deposition of amyloid-β peptide (Aβ). Its gene deletion showed only mild phenotypes. BACE1 inhibition has direct implications in the Alzheimer's disease pathology without largely affecting viability. However, inhibiting BACE1 selectively in vivo has presented many challenges to medicinal chemists. Since its identification in 2000, inhibitors covering many different structural classes have been designed and developed. These inhibitors can be largely classified as either peptidomimetic or non-peptidic inhibitors. Progress in these fields resulted in inhibitors that contain many targeted drug-like characteristics. In this review, we describe structure-based design strategies and evolution of a wide range of BACE1 inhibitors including compounds that have been shown to reduce brain Aβ, rescue the cognitive decline in transgenic AD mice and inhibitor drug candidates that are currently in clinical trials.
Collapse
Affiliation(s)
- Arun K Ghosh
- Department of Chemistry and Department of Medicinal Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | | |
Collapse
|
66
|
Cheng Y, Brown J, Judd TC, Lopez P, Qian W, Powers TS, Chen JJ, Bartberger MD, Chen K, Dunn RT, Epstein O, Fremeau RT, Harried S, Hickman D, Hitchcock SA, Luo Y, Minatti AE, Patel VF, Vargas HM, Wahl RC, Weiss MM, Wen PH, White RD, Whittington DA, Zheng XM, Wood S. An Orally Available BACE1 Inhibitor That Affords Robust CNS Aβ Reduction without Cardiovascular Liabilities. ACS Med Chem Lett 2015; 6:210-5. [PMID: 25699151 DOI: 10.1021/ml500458t] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Accepted: 12/29/2014] [Indexed: 11/28/2022] Open
Abstract
BACE1 inhibition to prevent Aβ peptide formation is considered to be a potential route to a disease-modifying treatment for Alzheimer's disease. Previous efforts in our laboratory using a combined structure- and property-based approach have resulted in the identification of aminooxazoline xanthenes as potent BACE1 inhibitors. Herein, we report further optimization leading to the discovery of inhibitor 15 as an orally available and highly efficacious BACE1 inhibitor that robustly reduces CSF and brain Aβ levels in both rats and nonhuman primates. In addition, compound 15 exhibited low activity on the hERG ion channel and was well tolerated in an integrated cardiovascular safety model.
Collapse
Affiliation(s)
- Yuan Cheng
- Department of Medicinal Chemistry, ‡Department of Molecular
Structure, §Department of Neuroscience, ∥Department of HTS
and Molecular Pharmacology, and ⊥Department of Pharmacokinetics and Drug Metabolism, #Comparative Biology and
Safety Sciences, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
- Department of Medicinal Chemistry and ○Department of Molecular Structure, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
| | - James Brown
- Department of Medicinal Chemistry, ‡Department of Molecular
Structure, §Department of Neuroscience, ∥Department of HTS
and Molecular Pharmacology, and ⊥Department of Pharmacokinetics and Drug Metabolism, #Comparative Biology and
Safety Sciences, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
- Department of Medicinal Chemistry and ○Department of Molecular Structure, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Ted C. Judd
- Department of Medicinal Chemistry, ‡Department of Molecular
Structure, §Department of Neuroscience, ∥Department of HTS
and Molecular Pharmacology, and ⊥Department of Pharmacokinetics and Drug Metabolism, #Comparative Biology and
Safety Sciences, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
- Department of Medicinal Chemistry and ○Department of Molecular Structure, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Patricia Lopez
- Department of Medicinal Chemistry, ‡Department of Molecular
Structure, §Department of Neuroscience, ∥Department of HTS
and Molecular Pharmacology, and ⊥Department of Pharmacokinetics and Drug Metabolism, #Comparative Biology and
Safety Sciences, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
- Department of Medicinal Chemistry and ○Department of Molecular Structure, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Wenyuan Qian
- Department of Medicinal Chemistry, ‡Department of Molecular
Structure, §Department of Neuroscience, ∥Department of HTS
and Molecular Pharmacology, and ⊥Department of Pharmacokinetics and Drug Metabolism, #Comparative Biology and
Safety Sciences, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
- Department of Medicinal Chemistry and ○Department of Molecular Structure, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Timothy S. Powers
- Department of Medicinal Chemistry, ‡Department of Molecular
Structure, §Department of Neuroscience, ∥Department of HTS
and Molecular Pharmacology, and ⊥Department of Pharmacokinetics and Drug Metabolism, #Comparative Biology and
Safety Sciences, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
- Department of Medicinal Chemistry and ○Department of Molecular Structure, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Jian Jeffrey Chen
- Department of Medicinal Chemistry, ‡Department of Molecular
Structure, §Department of Neuroscience, ∥Department of HTS
and Molecular Pharmacology, and ⊥Department of Pharmacokinetics and Drug Metabolism, #Comparative Biology and
Safety Sciences, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
- Department of Medicinal Chemistry and ○Department of Molecular Structure, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Michael D. Bartberger
- Department of Medicinal Chemistry, ‡Department of Molecular
Structure, §Department of Neuroscience, ∥Department of HTS
and Molecular Pharmacology, and ⊥Department of Pharmacokinetics and Drug Metabolism, #Comparative Biology and
Safety Sciences, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
- Department of Medicinal Chemistry and ○Department of Molecular Structure, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Kui Chen
- Department of Medicinal Chemistry, ‡Department of Molecular
Structure, §Department of Neuroscience, ∥Department of HTS
and Molecular Pharmacology, and ⊥Department of Pharmacokinetics and Drug Metabolism, #Comparative Biology and
Safety Sciences, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
- Department of Medicinal Chemistry and ○Department of Molecular Structure, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Robert T. Dunn
- Department of Medicinal Chemistry, ‡Department of Molecular
Structure, §Department of Neuroscience, ∥Department of HTS
and Molecular Pharmacology, and ⊥Department of Pharmacokinetics and Drug Metabolism, #Comparative Biology and
Safety Sciences, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
- Department of Medicinal Chemistry and ○Department of Molecular Structure, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Oleg Epstein
- Department of Medicinal Chemistry, ‡Department of Molecular
Structure, §Department of Neuroscience, ∥Department of HTS
and Molecular Pharmacology, and ⊥Department of Pharmacokinetics and Drug Metabolism, #Comparative Biology and
Safety Sciences, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
- Department of Medicinal Chemistry and ○Department of Molecular Structure, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Robert T. Fremeau
- Department of Medicinal Chemistry, ‡Department of Molecular
Structure, §Department of Neuroscience, ∥Department of HTS
and Molecular Pharmacology, and ⊥Department of Pharmacokinetics and Drug Metabolism, #Comparative Biology and
Safety Sciences, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
- Department of Medicinal Chemistry and ○Department of Molecular Structure, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Scott Harried
- Department of Medicinal Chemistry, ‡Department of Molecular
Structure, §Department of Neuroscience, ∥Department of HTS
and Molecular Pharmacology, and ⊥Department of Pharmacokinetics and Drug Metabolism, #Comparative Biology and
Safety Sciences, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
- Department of Medicinal Chemistry and ○Department of Molecular Structure, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Dean Hickman
- Department of Medicinal Chemistry, ‡Department of Molecular
Structure, §Department of Neuroscience, ∥Department of HTS
and Molecular Pharmacology, and ⊥Department of Pharmacokinetics and Drug Metabolism, #Comparative Biology and
Safety Sciences, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
- Department of Medicinal Chemistry and ○Department of Molecular Structure, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Stephen A. Hitchcock
- Department of Medicinal Chemistry, ‡Department of Molecular
Structure, §Department of Neuroscience, ∥Department of HTS
and Molecular Pharmacology, and ⊥Department of Pharmacokinetics and Drug Metabolism, #Comparative Biology and
Safety Sciences, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
- Department of Medicinal Chemistry and ○Department of Molecular Structure, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Yi Luo
- Department of Medicinal Chemistry, ‡Department of Molecular
Structure, §Department of Neuroscience, ∥Department of HTS
and Molecular Pharmacology, and ⊥Department of Pharmacokinetics and Drug Metabolism, #Comparative Biology and
Safety Sciences, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
- Department of Medicinal Chemistry and ○Department of Molecular Structure, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Ana Elena Minatti
- Department of Medicinal Chemistry, ‡Department of Molecular
Structure, §Department of Neuroscience, ∥Department of HTS
and Molecular Pharmacology, and ⊥Department of Pharmacokinetics and Drug Metabolism, #Comparative Biology and
Safety Sciences, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
- Department of Medicinal Chemistry and ○Department of Molecular Structure, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Vinod F. Patel
- Department of Medicinal Chemistry, ‡Department of Molecular
Structure, §Department of Neuroscience, ∥Department of HTS
and Molecular Pharmacology, and ⊥Department of Pharmacokinetics and Drug Metabolism, #Comparative Biology and
Safety Sciences, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
- Department of Medicinal Chemistry and ○Department of Molecular Structure, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Hugo M. Vargas
- Department of Medicinal Chemistry, ‡Department of Molecular
Structure, §Department of Neuroscience, ∥Department of HTS
and Molecular Pharmacology, and ⊥Department of Pharmacokinetics and Drug Metabolism, #Comparative Biology and
Safety Sciences, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
- Department of Medicinal Chemistry and ○Department of Molecular Structure, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Robert C. Wahl
- Department of Medicinal Chemistry, ‡Department of Molecular
Structure, §Department of Neuroscience, ∥Department of HTS
and Molecular Pharmacology, and ⊥Department of Pharmacokinetics and Drug Metabolism, #Comparative Biology and
Safety Sciences, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
- Department of Medicinal Chemistry and ○Department of Molecular Structure, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Matthew M. Weiss
- Department of Medicinal Chemistry, ‡Department of Molecular
Structure, §Department of Neuroscience, ∥Department of HTS
and Molecular Pharmacology, and ⊥Department of Pharmacokinetics and Drug Metabolism, #Comparative Biology and
Safety Sciences, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
- Department of Medicinal Chemistry and ○Department of Molecular Structure, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Paul H. Wen
- Department of Medicinal Chemistry, ‡Department of Molecular
Structure, §Department of Neuroscience, ∥Department of HTS
and Molecular Pharmacology, and ⊥Department of Pharmacokinetics and Drug Metabolism, #Comparative Biology and
Safety Sciences, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
- Department of Medicinal Chemistry and ○Department of Molecular Structure, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Ryan D. White
- Department of Medicinal Chemistry, ‡Department of Molecular
Structure, §Department of Neuroscience, ∥Department of HTS
and Molecular Pharmacology, and ⊥Department of Pharmacokinetics and Drug Metabolism, #Comparative Biology and
Safety Sciences, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
- Department of Medicinal Chemistry and ○Department of Molecular Structure, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Douglas A. Whittington
- Department of Medicinal Chemistry, ‡Department of Molecular
Structure, §Department of Neuroscience, ∥Department of HTS
and Molecular Pharmacology, and ⊥Department of Pharmacokinetics and Drug Metabolism, #Comparative Biology and
Safety Sciences, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
- Department of Medicinal Chemistry and ○Department of Molecular Structure, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Xiao Mei Zheng
- Department of Medicinal Chemistry, ‡Department of Molecular
Structure, §Department of Neuroscience, ∥Department of HTS
and Molecular Pharmacology, and ⊥Department of Pharmacokinetics and Drug Metabolism, #Comparative Biology and
Safety Sciences, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
- Department of Medicinal Chemistry and ○Department of Molecular Structure, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Stephen Wood
- Department of Medicinal Chemistry, ‡Department of Molecular
Structure, §Department of Neuroscience, ∥Department of HTS
and Molecular Pharmacology, and ⊥Department of Pharmacokinetics and Drug Metabolism, #Comparative Biology and
Safety Sciences, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
- Department of Medicinal Chemistry and ○Department of Molecular Structure, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
| |
Collapse
|
67
|
Pirard B, Ertl P. Evaluation of a semi-automated workflow for fragment growing. J Chem Inf Model 2015; 55:180-93. [PMID: 25514394 DOI: 10.1021/ci5006355] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Intelligent Automatic Design (IADE) is an expert system developed at Novartis to identify nonclassical bioisosteres. In addition to bioisostere searching, one could also use IADE to grow a fragment bound to a protein. Here we report an evaluation of IADE as a tool for fragment growing. Three examples from the literature served as test cases. In all three cases, IADE generated close analogues of the published compounds and reproduced their crystallographic binding modes. This exercise validated the use of the IADE system for fragment growing. We have also gained experience in optimizing the performance of IADE for this type of application.
Collapse
Affiliation(s)
- Bernard Pirard
- Novartis Institutes for BioMedical Research , Novartis Campus, CH-4056 Basel, Switzerland
| | | |
Collapse
|
68
|
Mondal M, Groothuis DE, Hirsch AKH. Fragment growing exploiting dynamic combinatorial chemistry of inhibitors of the aspartic protease endothiapepsin. MEDCHEMCOMM 2015. [DOI: 10.1039/c5md00157a] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The novel combination of fragment growing and DCC is a powerful and efficient strategy to convert a fragment into a hit.
Collapse
Affiliation(s)
- Milon Mondal
- Stratingh Institute for Chemistry
- University of Groningen
- 9747 AG Groningen
- The Netherlands
| | - Daphne E. Groothuis
- Stratingh Institute for Chemistry
- University of Groningen
- 9747 AG Groningen
- The Netherlands
| | - Anna K. H. Hirsch
- Stratingh Institute for Chemistry
- University of Groningen
- 9747 AG Groningen
- The Netherlands
| |
Collapse
|
69
|
Sadigh-Eteghad S, Sabermarouf B, Majdi A, Talebi M, Farhoudi M, Mahmoudi J. Amyloid-beta: a crucial factor in Alzheimer's disease. Med Princ Pract 2014; 24:1-10. [PMID: 25471398 PMCID: PMC5588216 DOI: 10.1159/000369101] [Citation(s) in RCA: 330] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Accepted: 10/15/2014] [Indexed: 12/15/2022] Open
Abstract
Alzheimer's disease (AD) is the most prevalent form of dementia which affects people older than 60 years of age. In AD, the dysregulation of the amyloid-beta (Aβ) level leads to the appearance of senile plaques which contain Aβ depositions. Aβ is a complex biological molecule which interacts with many types of receptors and/or forms insoluble assemblies and, eventually, its nonphysiological depositions alternate with the normal neuronal conditions. In this situation, AD signs appear and the patients experience marked cognitional disabilities. In general, intellect, social skills, personality, and memory are influenced by this disease and, in the long run, it leads to a reduction in quality of life and life expectancy. Due to the pivotal role of Aβ in the pathobiology of AD, a great deal of effort has been made to reveal its exact role in neuronal dysfunctions and to finding efficacious therapeutic strategies against its adverse neuronal outcomes. Hence, the determination of its different molecular assemblies and the mechanisms underlying its pathological effects are of interest. In the present paper, some of the well-established structural forms of Aβ, its interactions with various receptors and possible molecular and cellular mechanisms underlying its neurotoxicity are discussed. In addition, several Aβ-based rodent models of AD are reviewed.
Collapse
Affiliation(s)
| | | | | | | | | | - Javad Mahmoudi
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
70
|
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
|
71
|
Zhang L, Zheng L, Guo B, Hua R. One-Pot Synthesis of Multisubstituted 2-Aminoquinolines from Annulation of 1-Aryl Tetrazoles with Internal Alkynes via Double C–H Activation and Denitrogenation. J Org Chem 2014; 79:11541-8. [DOI: 10.1021/jo502192b] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Lei Zhang
- Department of Chemistry, Tsinghua University, Key Laboratory of Organic Optoelectronics & Molecular Engineering of Ministry of Education, Beijing 100084, China
| | - Liyao Zheng
- Department of Chemistry, Tsinghua University, Key Laboratory of Organic Optoelectronics & Molecular Engineering of Ministry of Education, Beijing 100084, China
| | - Biao Guo
- Department of Chemistry, Tsinghua University, Key Laboratory of Organic Optoelectronics & Molecular Engineering of Ministry of Education, Beijing 100084, China
| | - Ruimao Hua
- Department of Chemistry, Tsinghua University, Key Laboratory of Organic Optoelectronics & Molecular Engineering of Ministry of Education, Beijing 100084, China
| |
Collapse
|
72
|
Epstein O, Bryan MC, Cheng AC, Derakhchan K, Dineen TA, Hickman D, Hua Z, Human JB, Kreiman C, Marx IE, Weiss MM, Wahl RC, Wen PH, Whittington DA, Wood S, Zheng XM, Fremeau RT, White RD, Patel VF. Lead optimization and modulation of hERG activity in a series of aminooxazoline xanthene β-site amyloid precursor protein cleaving enzyme (BACE1) inhibitors. J Med Chem 2014; 57:9796-810. [PMID: 25389560 DOI: 10.1021/jm501266w] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The optimization of a series of aminooxazoline xanthene inhibitors of β-site amyloid precursor protein cleaving enzyme 1 (BACE1) is described. An early lead compound showed robust Aβ lowering activity in a rat pharmacodynamic model, but advancement was precluded by a low therapeutic window to QTc prolongation in cardiovascular models consistent with in vitro activity on the hERG ion channel. While the introduction of polar groups was effective in reducing hERG binding affinity, this came at the expense of higher than desired Pgp-mediated efflux. A balance of low Pgp efflux and hERG activity was achieved by lowering the polar surface area of the P3 substituent while retaining polarity in the P2' side chain. The introduction of a fluorine in position 4 of the xanthene ring improved BACE1 potency (5-10-fold). The combination of these optimized fragments resulted in identification of compound 40, which showed robust Aβ reduction in a rat pharmacodynamic model (78% Aβ reduction in CSF at 10 mg/kg po) and also showed acceptable cardiovascular safety in vivo.
Collapse
Affiliation(s)
- Oleg Epstein
- Departments of Therapeutic Discovery, ‡Neuroscience, §Molecular Structure and Characterization, ∥Pharmacokinetics and Drug Metabolism, and ⊥Comparative Biology and Safety Sciences, Amgen Inc. , 360 Binney Street, Cambridge, Massachusetts 02142, One Amgen Center Drive, Thousand Oaks, California 91320, and 1120 Veterans Boulevard, South San Francisco, California 94080, United States
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
73
|
Barman A, Prabhakar R. Computational Insights into Substrate and Site Specificities, Catalytic Mechanism, and Protonation States of the Catalytic Asp Dyad of β -Secretase. SCIENTIFICA 2014; 2014:598728. [PMID: 25309776 PMCID: PMC4189502 DOI: 10.1155/2014/598728] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Accepted: 06/21/2014] [Indexed: 06/04/2023]
Abstract
In this review, information regarding substrate and site specificities, catalytic mechanism, and protonation states of the catalytic Asp dyad of β-secretase (BACE1) derived from computational studies has been discussed. BACE1 catalyzes the rate-limiting step in the generation of Alzheimer amyloid beta peptide through the proteolytic cleavage of the amyloid precursor protein. Due to its biological functioning, this enzyme has been considered as one of the most important targets for finding the cure for Alzheimer's disease. Molecular dynamics (MD) simulations suggested that structural differences in the key regions (inserts A, D, and F and the 10s loop) of the enzyme are responsible for the observed difference in its activities towards the WT- and SW-substrates. The modifications in the flap, third strand, and insert F regions were found to be involved in the alteration in the site specificity of the glycosylphosphatidylinositol bound form of BACE1. Our QM and QM/MM calculations suggested that BACE1 hydrolyzed the SW-substrate more efficiently than the WT-substrate and that cleavage of the peptide bond occurred in the rate-determining step. The results from molecular docking studies showed that the information concerning a single protonation state of the Asp dyad is not enough to run an in silico screening campaign.
Collapse
Affiliation(s)
- Arghya Barman
- Department of Chemistry, University of Miami, 1301 Memorial Drive, Coral Gables, FL 33146, USA
| | - Rajeev Prabhakar
- Department of Chemistry, University of Miami, 1301 Memorial Drive, Coral Gables, FL 33146, USA
| |
Collapse
|
74
|
Chen X, Li X, Qu Z, Ke D, Qu L, Duan L, Mai W, Yuan J, Chen J, Zhao Y. H-Phosphonate-Mediated Amination of QuinolineN-Oxides with Tertiary Amines: A Mild and Metal-Free Synthesis of 2-Dialkylaminoquinolines. Adv Synth Catal 2014. [DOI: 10.1002/adsc.201301065] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
75
|
Kumar GR, Kumar YK, Kant R, Reddy MS. Tandem Cu-catalyzed ketenimine formation and intramolecular nucleophile capture: Synthesis of 1,2-dihydro-2-iminoquinolines from 1-(o-acetamidophenyl)propargyl alcohols. Beilstein J Org Chem 2014; 10:1255-1260. [PMID: 24991276 PMCID: PMC4077525 DOI: 10.3762/bjoc.10.125] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Accepted: 04/30/2014] [Indexed: 12/18/2022] Open
Abstract
The copper-catalyzed ketenimine formation reaction of 1-(o-acetamidophenyl)propargyl alcohols with various sulfonyl azides is found to undergo a concomitant intramolecular nucleophile attack to generate 1,2-dihydro-2-iminoquinolines after aromatization (via elimination of acetyl and hydroxy groups) and tautomerization. The reaction produces 4-substituted and 3,4-unsubstituted title compounds in moderate to good yields under mild reaction conditions.
Collapse
Affiliation(s)
- Gadi Ranjith Kumar
- Medicinal & Process Chemistry Division, CSIR-Central Drug Research Institute, BS-10/1, Sector 10, Jankipuram extension, Sitapur Road, P.O. Box 173, Lucknow 226031, India.,Academy of Scientific and Innovative Research, New Delhi 110001, India
| | - Yalla Kiran Kumar
- Academy of Scientific and Innovative Research, New Delhi 110001, India
| | - Ruchir Kant
- Molecular & Structural Biology Division, CSIR-Central Drug Research Institute,BS-10/1, Sector 10, Jankipuram extension, Sitapur Road, P.O. Box 173, Lucknow 226031, India
| | - Maddi Sridhar Reddy
- Medicinal & Process Chemistry Division, CSIR-Central Drug Research Institute, BS-10/1, Sector 10, Jankipuram extension, Sitapur Road, P.O. Box 173, Lucknow 226031, India.,Academy of Scientific and Innovative Research, New Delhi 110001, India
| |
Collapse
|
76
|
Yin X, Scalia A, Leroy L, Cuttitta CM, Polizzo GM, Ericson DL, Roessler CG, Campos O, Ma MY, Agarwal R, Jackimowicz R, Allaire M, Orville AM, Sweet RM, Soares AS. Hitting the target: fragment screening with acoustic in situ co-crystallization of proteins plus fragment libraries on pin-mounted data-collection micromeshes. ACTA CRYSTALLOGRAPHICA. SECTION D, BIOLOGICAL CRYSTALLOGRAPHY 2014; 70:1177-89. [PMID: 24816088 PMCID: PMC4014116 DOI: 10.1107/s1399004713034603] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2013] [Accepted: 12/24/2013] [Indexed: 11/17/2022]
Abstract
Acoustic droplet ejection (ADE) is a powerful technology that supports crystallographic applications such as growing, improving and manipulating protein crystals. A fragment-screening strategy is described that uses ADE to co-crystallize proteins with fragment libraries directly on MiTeGen MicroMeshes. Co-crystallization trials can be prepared rapidly and economically. The high speed of specimen preparation and the low consumption of fragment and protein allow the use of individual rather than pooled fragments. The Echo 550 liquid-handling instrument (Labcyte Inc., Sunnyvale, California, USA) generates droplets with accurate trajectories, which allows multiple co-crystallization experiments to be discretely positioned on a single data-collection micromesh. This accuracy also allows all components to be transferred through small apertures. Consequently, the crystallization tray is in equilibrium with the reservoir before, during and after the transfer of protein, precipitant and fragment to the micromesh on which crystallization will occur. This strict control of the specimen environment means that the crystallography experiments remain identical as the working volumes are decreased from the few microlitres level to the few nanolitres level. Using this system, lysozyme, thermolysin, trypsin and stachydrine demethylase crystals were co-crystallized with a small 33-compound mini-library to search for fragment hits. This technology pushes towards a much faster, more automated and more flexible strategy for structure-based drug discovery using as little as 2.5 nl of each major component.
Collapse
Affiliation(s)
- Xingyu Yin
- Office of Educational Programs, Brookhaven National Laboratory, Upton, NY 11973-5000, USA
- Department of Biochemistry and Cell Biology, Stony Brook University, NY 11794-5215, USA
- Nanjing University, Nanjing, Jiangsu, People’s Republic of China
| | - Alexander Scalia
- Office of Educational Programs, Brookhaven National Laboratory, Upton, NY 11973-5000, USA
- Department of Biological Sciences, Binghamton University, 4400 Vestal Parkway East, NY 13902, USA
| | - Ludmila Leroy
- Office of Educational Programs, Brookhaven National Laboratory, Upton, NY 11973-5000, USA
- CAPES Foundation, Ministry of Education of Brazil, 70040-020 Brasilia-DF, Brazil
- Universidade Federal de Minas Gerais, 6627 Av. Antonio Carlos, 31270-901 Belo Horizonte-MG, Brazil
| | - Christina M. Cuttitta
- Office of Educational Programs, Brookhaven National Laboratory, Upton, NY 11973-5000, USA
- Center for Developmental Neuroscience and Department of Biology, College of Staten Island, The City University of New York, 2800 Victory Boulevard, Staten Island, NY 10314, USA
| | - Gina M. Polizzo
- Office of Educational Programs, Brookhaven National Laboratory, Upton, NY 11973-5000, USA
- St Joseph’s College, 155 West Roe Boulevard, East Patchogue, NY 11772, USA
| | - Daniel L. Ericson
- Office of Educational Programs, Brookhaven National Laboratory, Upton, NY 11973-5000, USA
- Department of Biomedical Engineering, University at Buffalo, SUNY, 12 Capen Hall, Buffalo, NY 14260, USA
| | - Christian G. Roessler
- Photon Sciences Directorate, Brookhaven National Laboratory, Upton, NY 11973-5000, USA
| | - Olven Campos
- Office of Educational Programs, Brookhaven National Laboratory, Upton, NY 11973-5000, USA
- Department of Biological Science, Florida Atlantic University, 777 Glades Road, Boca Raton, FL 33414, USA
| | - Millie Y. Ma
- Office of Educational Programs, Brookhaven National Laboratory, Upton, NY 11973-5000, USA
- Comsewogue High School, 565 Bicycle Path, Port Jefferson Station, NY 11776, USA
| | - Rakhi Agarwal
- Biosciences Department, Brookhaven National Laboratory, Upton, NY 11973-5000, USA
| | - Rick Jackimowicz
- Photon Sciences Directorate, Brookhaven National Laboratory, Upton, NY 11973-5000, USA
| | - Marc Allaire
- Photon Sciences Directorate, Brookhaven National Laboratory, Upton, NY 11973-5000, USA
| | - Allen M. Orville
- Photon Sciences Directorate, Brookhaven National Laboratory, Upton, NY 11973-5000, USA
- Biosciences Department, Brookhaven National Laboratory, Upton, NY 11973-5000, USA
| | - Robert M. Sweet
- Photon Sciences Directorate, Brookhaven National Laboratory, Upton, NY 11973-5000, USA
| | - Alexei S. Soares
- Photon Sciences Directorate, Brookhaven National Laboratory, Upton, NY 11973-5000, USA
| |
Collapse
|
77
|
4-Oxo-1,4-dihydro-quinoline-3-carboxamides as BACE-1 inhibitors: Synthesis, biological evaluation and docking studies. Eur J Med Chem 2014; 79:413-21. [DOI: 10.1016/j.ejmech.2014.04.025] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Revised: 04/07/2014] [Accepted: 04/07/2014] [Indexed: 11/18/2022]
|
78
|
Kalászi A, Szisz D, Imre G, Polgár T. Screen3D: a novel fully flexible high-throughput shape-similarity search method. J Chem Inf Model 2014; 54:1036-49. [PMID: 24568118 DOI: 10.1021/ci400620f] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
3D shape- or volume-based virtual screening is a broadly used approach in drug discovery. In recent years a large number of publications have appeared in which these tools were compared not only to competitive methods but to docking studies as well. Studies often showed that the effectiveness of docking could be highly variable due to a large number of possible confounding factors, while ligand-based, shape-based approaches were more consistent. Here, we describe a novel, fully flexible shape-based virtual screening algorithm that does not require previous 3D conformation or conformer generation. Due to its solid consistency it can easily be used on desktop computers by non-expert scientists. The algorithm is demonstrated in a study for the investigation of β-secretase inhibitors and benchmarked on the Directory of Useful Decoys data set.
Collapse
Affiliation(s)
- Adrián Kalászi
- ChemAxon Ltd., Graphisoft park, Zahony u. 7, Budapest, Hungary , 1037
| | | | | | | |
Collapse
|
79
|
Cinelli MA, Li H, Chreifi G, Martásek P, Roman LJ, Poulos TL, Silverman RB. Simplified 2-aminoquinoline-based scaffold for potent and selective neuronal nitric oxide synthase inhibition. J Med Chem 2014; 57:1513-30. [PMID: 24472039 PMCID: PMC3954451 DOI: 10.1021/jm401838x] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
![]()
Since high levels of nitric oxide
(NO) are implicated in neurodegenerative
disorders, inhibition of the neuronal isoform of nitric oxide synthase
(nNOS) and reduction of NO levels are therapeutically desirable. Nonetheless,
many nNOS inhibitors mimic l-arginine and are poorly bioavailable.
2-Aminoquinoline-based scaffolds were designed with the hope that
they could (a) mimic aminopyridines as potent, isoform-selective arginine
isosteres and (b) possess chemical properties more conducive to oral
bioavailability and CNS penetration. A series of these compounds was
synthesized and assayed against purified nNOS enzymes, endothelial
NOS (eNOS), and inducible NOS (iNOS). Several compounds built on a
7-substituted 2-aminoquinoline core are potent and isoform-selective;
X-ray crystallography indicates that aminoquinolines exert inhibitory
effects by mimicking substrate interactions with the conserved active
site glutamate residue. The most potent and selective compounds, 7 and 15, were tested in a Caco-2 assay and showed
good permeability and low efflux, suggesting high potential for oral
bioavailability.
Collapse
Affiliation(s)
- Maris A Cinelli
- Departments of Chemistry and Molecular Biosciences, Chemistry of Life Processes Institute, Center for Molecular Innovation and Drug Discovery, Northwestern University , 2145 Sheridan Road, Evanston, Illinois 60208-3113, United States
| | | | | | | | | | | | | |
Collapse
|
80
|
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
|
81
|
Aillerie A, Pellegrini S, Bousquet T, Pélinski L. In situ generation of ammonia for the copper-catalyzed synthesis of primary aminoquinolines. NEW J CHEM 2014. [DOI: 10.1039/c4nj00046c] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Primary aminoquinolines were synthesized from iodoquinolines in the presence of a copper(i) iodide catalyst and formamide as both solvent and source of ammonia generated in situ.
Collapse
Affiliation(s)
- Alexandre Aillerie
- Université Lille Nord de France
- F-59000 Lille, France
- Université de Lille 1
- Unité de Catalyse et de Chimie du Solide
- UMR CNRS 8181
| | - Sylvain Pellegrini
- Université Lille Nord de France
- F-59000 Lille, France
- Université de Lille 1
- Unité de Catalyse et de Chimie du Solide
- UMR CNRS 8181
| | - Till Bousquet
- Université Lille Nord de France
- F-59000 Lille, France
- Université de Lille 1
- Unité de Catalyse et de Chimie du Solide
- UMR CNRS 8181
| | - Lydie Pélinski
- Université Lille Nord de France
- F-59000 Lille, France
- Université de Lille 1
- Unité de Catalyse et de Chimie du Solide
- UMR CNRS 8181
| |
Collapse
|
82
|
Jiang B, Hu L, Gui W. Facile synthesis of 2-amino-3-bromoquinolines by palladium-catalyzed isocyanide insertion and cyclization of gem-dibromovinylanilines. RSC Adv 2014. [DOI: 10.1039/c4ra00821a] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
A novel and efficient synthesis of 2-amino-3-bromoquinolines through palladium-catalyzed isocyanide insertion followed by intramolecular cyclization of gem-dibromovinylanilines was developed.
Collapse
Affiliation(s)
- Baishan Jiang
- Institute of Chemical Biology
- Guangzhou Institutes of Biomedicine and Health
- Chinese Academy of Sciences
- Guangzhou 510530, China
| | - Langxi Hu
- Institute of Chemical Biology
- Guangzhou Institutes of Biomedicine and Health
- Chinese Academy of Sciences
- Guangzhou 510530, China
| | - Weijun Gui
- Institute of Chemical Biology
- Guangzhou Institutes of Biomedicine and Health
- Chinese Academy of Sciences
- Guangzhou 510530, China
| |
Collapse
|
83
|
Hu L, Gui W, Liu Z, Jiang B. Synthesis of 3-aryl-2-aminoquinolines: palladium-catalyzed cascade reactions of gem-dibromovinylanilines with tert-butyl isocyanide and arylboronic acids. RSC Adv 2014. [DOI: 10.1039/c4ra05670a] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
A three-component cascade reaction involving gem-dibromovinylanilines, tert-butyl isocyanide and arylboronic acids for the efficient synthesis of 3-aryl-2-aminoquinolines has been developed.
Collapse
Affiliation(s)
- Langxi Hu
- Institute of Chemical Biology
- Guangzhou Institutes of Biomedicine and Health
- Chinese Academy of Sciences
- Guangzhou 510530, China
| | - Weijun Gui
- Institute of Chemical Biology
- Guangzhou Institutes of Biomedicine and Health
- Chinese Academy of Sciences
- Guangzhou 510530, China
| | - Zichen Liu
- Shenzhen University Health Science Center
- Shenzhen University
- China
| | - Baishan Jiang
- Institute of Chemical Biology
- Guangzhou Institutes of Biomedicine and Health
- Chinese Academy of Sciences
- Guangzhou 510530, China
| |
Collapse
|
84
|
Konno H, Endo H, Ise S, Miyazaki K, Aoki H, Sanjoh A, Kobayashi K, Hattori Y, Akaji K. Synthesis and evaluation of curcumin derivatives toward an inhibitor of beta-site amyloid precursor protein cleaving enzyme 1. Bioorg Med Chem Lett 2014; 24:685-90. [DOI: 10.1016/j.bmcl.2013.11.039] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Revised: 11/13/2013] [Accepted: 11/15/2013] [Indexed: 11/24/2022]
|
85
|
Could MM-GBSA be accurate enough for calculation of absolute protein/ligand binding free energies? J Mol Graph Model 2013; 46:41-51. [DOI: 10.1016/j.jmgm.2013.09.005] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Revised: 09/06/2013] [Accepted: 09/07/2013] [Indexed: 11/20/2022]
|
86
|
Liu B, Gao H, Yu Y, Wu W, Jiang H. Palladium-Catalyzed Intermolecular Aerobic Oxidative Cyclization of 2-Ethynylanilines with Isocyanides: Regioselective Synthesis of 4-Halo-2-aminoquinolines. J Org Chem 2013; 78:10319-28. [DOI: 10.1021/jo401707j] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Bifu Liu
- School
of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou, Guangdong 510640, PR China
| | - Hanling Gao
- School
of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou, Guangdong 510640, PR China
| | - Yue Yu
- School
of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou, Guangdong 510640, PR China
| | - Wanqing Wu
- School
of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou, Guangdong 510640, PR China
| | - Huanfeng Jiang
- School
of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou, Guangdong 510640, PR China
| |
Collapse
|
87
|
Stamford A, Strickland C. Inhibitors of BACE for treating Alzheimer's disease: a fragment-based drug discovery story. Curr Opin Chem Biol 2013; 17:320-8. [PMID: 23683349 DOI: 10.1016/j.cbpa.2013.04.016] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2013] [Revised: 04/02/2013] [Accepted: 04/15/2013] [Indexed: 11/27/2022]
Abstract
Several fragment-based methods have been applied to the discovery of new lead sources for inhibitors of BACE1, an important therapeutic target for Alzheimer's disease. Among the most common fragment hits were various amidine-containing molecules in which the amidine engaged in discrete H-bond donor-acceptor interaction with the BACE1 catalytic dyad. Structure and medicinal chemistry knowledge-based optimization with emphasis on ligand efficiency resulted in identification of a key pharmacophore comprising a non-planar cyclic amidine scaffold directly attached to a phenyl group projecting into S1. This key pharmacophore is a common feature of known clinical candidates and has dominated the recent patent literature. A structural comparison of the non-planar cyclic amidine motif with other BACE1 pharmacophores highlights its uniqueness and distinct advantages.
Collapse
Affiliation(s)
- Andrew Stamford
- Merck Research Laboratories, 126 East Lincoln Avenue, Rahway, NJ 07065, United States.
| | | |
Collapse
|
88
|
Hunt KW, Cook AW, Watts RJ, Clark CT, Vigers G, Smith D, Metcalf AT, Gunawardana IW, Burkard M, Cox AA, Geck Do MK, Dutcher D, Thomas AA, Rana S, Kallan NC, DeLisle RK, Rizzi JP, Regal K, Sammond D, Groneberg R, Siu M, Purkey H, Lyssikatos JP, Marlow A, Liu X, Tang TP. Spirocyclic β-site amyloid precursor protein cleaving enzyme 1 (BACE1) inhibitors: from hit to lowering of cerebrospinal fluid (CSF) amyloid β in a higher species. J Med Chem 2013; 56:3379-403. [PMID: 23537249 DOI: 10.1021/jm4002154] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
A hallmark of Alzheimer's disease is the brain deposition of amyloid beta (Aβ), a peptide of 36-43 amino acids that is likely a primary driver of neurodegeneration. Aβ is produced by the sequential cleavage of APP by BACE1 and γ-secretase; therefore, inhibition of BACE1 represents an attractive therapeutic target to slow or prevent Alzheimer's disease. Herein we describe BACE1 inhibitors with limited molecular flexibility and molecular weight that decrease CSF Aβ in vivo, despite efflux. Starting with spirocycle 1a, we explore structure-activity relationships of core changes, P3 moieties, and Asp binding functional groups in order to optimize BACE1 affinity, cathepsin D selectivity, and blood-brain barrier (BBB) penetration. Using wild type guinea pig and rat, we demonstrate a PK/PD relationship between free drug concentrations in the brain and CSF Aβ lowering. Optimization of brain exposure led to the discovery of (R)-50 which reduced CSF Aβ in rodents and in monkey.
Collapse
Affiliation(s)
- Kevin W Hunt
- Array BioPharma, 3200 Walnut Street, Boulder, CO 80301, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
89
|
Abstract
INTRODUCTION Alzheimer's disease (AD), which is characterized by progressive intellectual deterioration, is the most common cause of dementia. β-Secretase (or BACE1) expression is a trigger for amyloid β peptide formation, a cause of AD, and thus is a molecular target for the development of drugs against AD. Many BACE1 inhibitors have been identified by academic and pharmaceutical research groups and a number of advanced technologies in drug discovery have been applied to the drug discovery. AREAS COVERED The purpose of this review is to present and discuss the methodologies used for BACE1 inhibitor drug discovery via substrate- and structure-based design, high-throughput screening and fragment-based drug design. The authors also review the advantages and disadvantages of these methodologies. EXPERT OPINION Many BACE1 inhibitors have been designed using X-ray crystal structure-based drug design as well as through in silico screening. Nevertheless, there are serious problems with regards to deciding the best X-ray crystal structure for designing BACE1 inhibitors through computational approaches. There are two prominent configurations of BACE1 but there is still room for improvement. Future developments may make it possible to identify BACE1 inhibitors as potential drug candidates.
Collapse
Affiliation(s)
- Yoshio Hamada
- Kobe Gakuin University, Faculty of Pharmaceutical Sciences, Minatojima, Chuo-ku, Kobe 650-8586, Japan
| | | |
Collapse
|
90
|
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: 9.4] [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
|
91
|
Zhu T, Lee H, Lei H, Jones C, Patel K, Johnson ME, Hevener KE. Fragment-based drug discovery using a multidomain, parallel MD-MM/PBSA screening protocol. J Chem Inf Model 2013; 53:560-72. [PMID: 23432621 PMCID: PMC3752004 DOI: 10.1021/ci300502h] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
We have developed a rigorous computational screening protocol to identify novel fragment-like inhibitors of N(5)-CAIR mutase (PurE), a key enzyme involved in de novo purine synthesis that represents a novel target for the design of antibacterial agents. This computational screening protocol utilizes molecular docking, graphics processing unit (GPU)-accelerated molecular dynamics, and Molecular Mechanics/Poisson-Boltzmann Surface Area (MM/PBSA) free energy estimations to investigate the binding modes and energies of fragments in the active sites of PurE. PurE is a functional octamer comprised of identical subunits. The octameric structure, with its eight active sites, provided a distinct advantage in these studies because, for a given simulation length, we were able to place eight separate fragment compounds in the active sites to increase the throughput of the MM/PBSA analysis. To validate this protocol, we have screened an in-house fragment library consisting of 352 compounds. The theoretical results were then compared with the results of two experimental fragment screens, Nuclear Magnetic Resonance (NMR) and Surface Plasmon Resonance (SPR) binding analyses. In these validation studies, the protocol was able to effectively identify the competitive binders that had been independently identified by experimental testing, suggesting the potential utility of this method for the identification of novel fragments for future development as PurE inhibitors.
Collapse
Affiliation(s)
- Tian Zhu
- Center for Pharmaceutical Biotechnology, University of Illinois at Chicago, 900 S Ashland Ave., Suite 3100, Chicago, IL 60607-7173 (USA)
| | - Hyun Lee
- Center for Pharmaceutical Biotechnology, University of Illinois at Chicago, 900 S Ashland Ave., Suite 3100, Chicago, IL 60607-7173 (USA)
| | - Hao Lei
- Center for Pharmaceutical Biotechnology, University of Illinois at Chicago, 900 S Ashland Ave., Suite 3100, Chicago, IL 60607-7173 (USA)
| | - Christopher Jones
- Center for Pharmaceutical Biotechnology, University of Illinois at Chicago, 900 S Ashland Ave., Suite 3100, Chicago, IL 60607-7173 (USA)
| | - Kavankumar Patel
- Center for Pharmaceutical Biotechnology, University of Illinois at Chicago, 900 S Ashland Ave., Suite 3100, Chicago, IL 60607-7173 (USA)
| | - Michael E. Johnson
- Center for Pharmaceutical Biotechnology, University of Illinois at Chicago, 900 S Ashland Ave., Suite 3100, Chicago, IL 60607-7173 (USA)
| | - Kirk E. Hevener
- Center for Pharmaceutical Biotechnology, University of Illinois at Chicago, 900 S Ashland Ave., Suite 3100, Chicago, IL 60607-7173 (USA)
| |
Collapse
|
92
|
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: 2.8] [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
|
93
|
Bisignano P, Lambruschini C, Bicego M, Murino V, Favia AD, Cavalli A. In silico deconstruction of ATP-competitive inhibitors of glycogen synthase kinase-3β. J Chem Inf Model 2012. [PMID: 23198830 DOI: 10.1021/ci300355p] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Fragment-based methods have emerged in the last two decades as alternatives to traditional high throughput screenings for the identification of chemical starting points in drug discovery. One arguable yet popular assumption about fragment-based design is that the fragment binding mode remains conserved upon chemical expansion. For instance, the question of the binding conservation upon fragmentation of a molecule is still unclear. A number of papers have challenged this hypothesis by means of experimental techniques, with controversial results, "underlining" the idea that a simple generalization, maybe, is not possible. From a computational standpoint, the issue has been rarely addressed and mostly to test novel protocols on limited data sets. To fill this gap, we here report on a computational retrospective study concerned with the in silico deconstruction of leadlike compounds, active on the pharmaceutically relevant enzyme glycogen synthase kinase-3β.
Collapse
Affiliation(s)
- Paola Bisignano
- Department of Drug Discovery and Development, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genova, Italy
| | | | | | | | | | | |
Collapse
|
94
|
Folk DS, Torosian JC, Hwang S, McCafferty DG, Franz KJ. Monitoring β-secretase activity in living cells with a membrane-anchored FRET probe. Angew Chem Int Ed Engl 2012; 51:10795-9. [PMID: 23023944 PMCID: PMC3495557 DOI: 10.1002/anie.201206673] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2012] [Indexed: 11/09/2022]
Affiliation(s)
| | | | | | | | - Katherine J. Franz
- Department of Chemistry, Duke University 124 Science Dr., Durham, NC 27708, USA
| |
Collapse
|
95
|
Yonezawa S, Yamamoto T, Yamakawa H, Muto C, Hosono M, Hattori K, Higashino K, Yutsudo T, Iwamoto H, Kondo Y, Sakagami M, Togame H, Tanaka Y, Nakano T, Takemoto H, Arisawa M, Shuto S. Conformational Restriction Approach to β-Secretase (BACE1) Inhibitors: Effect of a Cyclopropane Ring To Induce an Alternative Binding Mode. J Med Chem 2012; 55:8838-58. [PMID: 22998419 DOI: 10.1021/jm3011405] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Shuji Yonezawa
- Shionogi Innovation Center for Drug Discovery, Shionogi & Co., Ltd., Kita-21 Nishi-11 Kita-ku, Sapporo 001-0021, Japan
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo
060-0812, Japan
| | - Takahiko Yamamoto
- Pharmaceutical Research Division, Medicinal Research Laboratories, and Innovative Drug Discovery Research Laboratories, Shionogi & Co., Ltd., 1-1, Futaba-cho 3-chome, Toyonaka, Osaka 561-0825, Japan
| | - Hidekuni Yamakawa
- Pharmaceutical Research Division, Medicinal Research Laboratories, and Innovative Drug Discovery Research Laboratories, Shionogi & Co., Ltd., 1-1, Futaba-cho 3-chome, Toyonaka, Osaka 561-0825, Japan
| | - Chie Muto
- Pharmaceutical Research Division, Medicinal Research Laboratories, and Innovative Drug Discovery Research Laboratories, Shionogi & Co., Ltd., 1-1, Futaba-cho 3-chome, Toyonaka, Osaka 561-0825, Japan
| | - Motoko Hosono
- Pharmaceutical Research Division, Medicinal Research Laboratories, and Innovative Drug Discovery Research Laboratories, Shionogi & Co., Ltd., 1-1, Futaba-cho 3-chome, Toyonaka, Osaka 561-0825, Japan
| | - Kazunari Hattori
- Pharmaceutical Research Division, Medicinal Research Laboratories, and Innovative Drug Discovery Research Laboratories, Shionogi & Co., Ltd., 1-1, Futaba-cho 3-chome, Toyonaka, Osaka 561-0825, Japan
| | - Kenichi Higashino
- Shionogi Innovation Center for Drug Discovery, Shionogi & Co., Ltd., Kita-21 Nishi-11 Kita-ku, Sapporo 001-0021, Japan
| | - Takashi Yutsudo
- Pharmaceutical Research Division, Medicinal Research Laboratories, and Innovative Drug Discovery Research Laboratories, Shionogi & Co., Ltd., 1-1, Futaba-cho 3-chome, Toyonaka, Osaka 561-0825, Japan
| | - Hideo Iwamoto
- Shionogi Techno Advance Research
Co., Ltd., 1-1, Futaba-cho 3-chome, Toyonaka, Osaka 561-0825, Japan
| | - Yutaka Kondo
- Shionogi Techno Advance Research
Co., Ltd., 1-1, Futaba-cho 3-chome, Toyonaka, Osaka 561-0825, Japan
| | - Masahiro Sakagami
- Pharmaceutical Research Division, Medicinal Research Laboratories, and Innovative Drug Discovery Research Laboratories, Shionogi & Co., Ltd., 1-1, Futaba-cho 3-chome, Toyonaka, Osaka 561-0825, Japan
| | - Hiroko Togame
- Quality, Safety and Regulatory Affairs Management Division, Shionogi & Co., Ltd., 1-8, Doshomachi 3-chome, Chuo-ku, Osaka 541-0045, Japan
| | - Yoshikazu Tanaka
- Shionogi Innovation Center for Drug Discovery, Shionogi & Co., Ltd., Kita-21 Nishi-11 Kita-ku, Sapporo 001-0021, Japan
| | - Toru Nakano
- Shionogi Innovation Center for Drug Discovery, Shionogi & Co., Ltd., Kita-21 Nishi-11 Kita-ku, Sapporo 001-0021, Japan
| | - Hiroshi Takemoto
- Pharmaceutical Research Division, Medicinal Research Laboratories, and Innovative Drug Discovery Research Laboratories, Shionogi & Co., Ltd., 1-1, Futaba-cho 3-chome, Toyonaka, Osaka 561-0825, Japan
| | - Mitsuhiro Arisawa
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo
060-0812, Japan
| | - Satoshi Shuto
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo
060-0812, Japan
| |
Collapse
|
96
|
Mandal M, Zhu Z, Cumming JN, Liu X, Strickland C, Mazzola RD, Caldwell JP, Leach P, Grzelak M, Hyde L, Zhang Q, Terracina G, Zhang L, Chen X, Kuvelkar R, Kennedy ME, Favreau L, Cox K, Orth P, Buevich A, Voigt J, Wang H, Kazakevich I, McKittrick BA, Greenlee W, Parker EM, Stamford AW. Design and Validation of Bicyclic Iminopyrimidinones As Beta Amyloid Cleaving Enzyme-1 (BACE1) Inhibitors: Conformational Constraint to Favor a Bioactive Conformation. J Med Chem 2012; 55:9331-45. [DOI: 10.1021/jm301039c] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Mihirbaran Mandal
- Department
of Medicinal Chemistry, ‡Department of Neuroscience, §Global Structural Chemistry, ∥Department of Analytical
Chemistry, ⊥Department of Basic Pharmaceutical Sciences, and #Department of Exploratory Drug Metabolism, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Zhaoning Zhu
- Department
of Medicinal Chemistry, ‡Department of Neuroscience, §Global Structural Chemistry, ∥Department of Analytical
Chemistry, ⊥Department of Basic Pharmaceutical Sciences, and #Department of Exploratory Drug Metabolism, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Jared N. Cumming
- Department
of Medicinal Chemistry, ‡Department of Neuroscience, §Global Structural Chemistry, ∥Department of Analytical
Chemistry, ⊥Department of Basic Pharmaceutical Sciences, and #Department of Exploratory Drug Metabolism, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Xiaoxiang Liu
- Department
of Medicinal Chemistry, ‡Department of Neuroscience, §Global Structural Chemistry, ∥Department of Analytical
Chemistry, ⊥Department of Basic Pharmaceutical Sciences, and #Department of Exploratory Drug Metabolism, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Corey Strickland
- Department
of Medicinal Chemistry, ‡Department of Neuroscience, §Global Structural Chemistry, ∥Department of Analytical
Chemistry, ⊥Department of Basic Pharmaceutical Sciences, and #Department of Exploratory Drug Metabolism, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Robert D. Mazzola
- Department
of Medicinal Chemistry, ‡Department of Neuroscience, §Global Structural Chemistry, ∥Department of Analytical
Chemistry, ⊥Department of Basic Pharmaceutical Sciences, and #Department of Exploratory Drug Metabolism, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - John P. Caldwell
- Department
of Medicinal Chemistry, ‡Department of Neuroscience, §Global Structural Chemistry, ∥Department of Analytical
Chemistry, ⊥Department of Basic Pharmaceutical Sciences, and #Department of Exploratory Drug Metabolism, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Prescott Leach
- Department
of Medicinal Chemistry, ‡Department of Neuroscience, §Global Structural Chemistry, ∥Department of Analytical
Chemistry, ⊥Department of Basic Pharmaceutical Sciences, and #Department of Exploratory Drug Metabolism, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Michael Grzelak
- Department
of Medicinal Chemistry, ‡Department of Neuroscience, §Global Structural Chemistry, ∥Department of Analytical
Chemistry, ⊥Department of Basic Pharmaceutical Sciences, and #Department of Exploratory Drug Metabolism, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Lynn Hyde
- Department
of Medicinal Chemistry, ‡Department of Neuroscience, §Global Structural Chemistry, ∥Department of Analytical
Chemistry, ⊥Department of Basic Pharmaceutical Sciences, and #Department of Exploratory Drug Metabolism, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Qi Zhang
- Department
of Medicinal Chemistry, ‡Department of Neuroscience, §Global Structural Chemistry, ∥Department of Analytical
Chemistry, ⊥Department of Basic Pharmaceutical Sciences, and #Department of Exploratory Drug Metabolism, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Giuseppe Terracina
- Department
of Medicinal Chemistry, ‡Department of Neuroscience, §Global Structural Chemistry, ∥Department of Analytical
Chemistry, ⊥Department of Basic Pharmaceutical Sciences, and #Department of Exploratory Drug Metabolism, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Lili Zhang
- Department
of Medicinal Chemistry, ‡Department of Neuroscience, §Global Structural Chemistry, ∥Department of Analytical
Chemistry, ⊥Department of Basic Pharmaceutical Sciences, and #Department of Exploratory Drug Metabolism, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Xia Chen
- Department
of Medicinal Chemistry, ‡Department of Neuroscience, §Global Structural Chemistry, ∥Department of Analytical
Chemistry, ⊥Department of Basic Pharmaceutical Sciences, and #Department of Exploratory Drug Metabolism, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Reshma Kuvelkar
- Department
of Medicinal Chemistry, ‡Department of Neuroscience, §Global Structural Chemistry, ∥Department of Analytical
Chemistry, ⊥Department of Basic Pharmaceutical Sciences, and #Department of Exploratory Drug Metabolism, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Matthew E. Kennedy
- Department
of Medicinal Chemistry, ‡Department of Neuroscience, §Global Structural Chemistry, ∥Department of Analytical
Chemistry, ⊥Department of Basic Pharmaceutical Sciences, and #Department of Exploratory Drug Metabolism, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Leonard Favreau
- Department
of Medicinal Chemistry, ‡Department of Neuroscience, §Global Structural Chemistry, ∥Department of Analytical
Chemistry, ⊥Department of Basic Pharmaceutical Sciences, and #Department of Exploratory Drug Metabolism, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Kathleen Cox
- Department
of Medicinal Chemistry, ‡Department of Neuroscience, §Global Structural Chemistry, ∥Department of Analytical
Chemistry, ⊥Department of Basic Pharmaceutical Sciences, and #Department of Exploratory Drug Metabolism, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Peter Orth
- Department
of Medicinal Chemistry, ‡Department of Neuroscience, §Global Structural Chemistry, ∥Department of Analytical
Chemistry, ⊥Department of Basic Pharmaceutical Sciences, and #Department of Exploratory Drug Metabolism, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Alexei Buevich
- Department
of Medicinal Chemistry, ‡Department of Neuroscience, §Global Structural Chemistry, ∥Department of Analytical
Chemistry, ⊥Department of Basic Pharmaceutical Sciences, and #Department of Exploratory Drug Metabolism, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Johannes Voigt
- Department
of Medicinal Chemistry, ‡Department of Neuroscience, §Global Structural Chemistry, ∥Department of Analytical
Chemistry, ⊥Department of Basic Pharmaceutical Sciences, and #Department of Exploratory Drug Metabolism, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Hongwu Wang
- Department
of Medicinal Chemistry, ‡Department of Neuroscience, §Global Structural Chemistry, ∥Department of Analytical
Chemistry, ⊥Department of Basic Pharmaceutical Sciences, and #Department of Exploratory Drug Metabolism, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Irina Kazakevich
- Department
of Medicinal Chemistry, ‡Department of Neuroscience, §Global Structural Chemistry, ∥Department of Analytical
Chemistry, ⊥Department of Basic Pharmaceutical Sciences, and #Department of Exploratory Drug Metabolism, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Brian A. McKittrick
- Department
of Medicinal Chemistry, ‡Department of Neuroscience, §Global Structural Chemistry, ∥Department of Analytical
Chemistry, ⊥Department of Basic Pharmaceutical Sciences, and #Department of Exploratory Drug Metabolism, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - William Greenlee
- Department
of Medicinal Chemistry, ‡Department of Neuroscience, §Global Structural Chemistry, ∥Department of Analytical
Chemistry, ⊥Department of Basic Pharmaceutical Sciences, and #Department of Exploratory Drug Metabolism, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Eric M. Parker
- Department
of Medicinal Chemistry, ‡Department of Neuroscience, §Global Structural Chemistry, ∥Department of Analytical
Chemistry, ⊥Department of Basic Pharmaceutical Sciences, and #Department of Exploratory Drug Metabolism, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Andrew W. Stamford
- Department
of Medicinal Chemistry, ‡Department of Neuroscience, §Global Structural Chemistry, ∥Department of Analytical
Chemistry, ⊥Department of Basic Pharmaceutical Sciences, and #Department of Exploratory Drug Metabolism, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| |
Collapse
|
97
|
Folk DS, Torosian JC, Hwang S, McCafferty DG, Franz KJ. Monitoring β-Secretase Activity in Living Cells with a Membrane-Anchored FRET Probe. Angew Chem Int Ed Engl 2012. [DOI: 10.1002/ange.201206673] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
98
|
Swahn BM, Kolmodin K, Karlström S, von Berg S, Söderman P, Holenz J, Berg S, Lindström J, Sundström M, Turek D, Kihlström J, Slivo C, Andersson L, Pyring D, Rotticci D, Öhberg L, Kers A, Bogar K, von Kieseritzky F, Bergh M, Olsson LL, Janson J, Eketjäll S, Georgievska B, Jeppsson F, Fälting J. Design and Synthesis of β-Site Amyloid Precursor Protein Cleaving Enzyme (BACE1) Inhibitors with in Vivo Brain Reduction of β-Amyloid Peptides. J Med Chem 2012; 55:9346-61. [DOI: 10.1021/jm3009025] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Lise-Lotte Olsson
- Discovery Sciences, AstraZeneca R&D Mölndal, SE-43183 Mölndal, Sweden
| | | | | | | | | | | |
Collapse
|
99
|
Barman A, Prabhakar R. Protonation states of the catalytic dyad of β-secretase (BACE1) in the presence of chemically diverse inhibitors: a molecular docking study. J Chem Inf Model 2012; 52:1275-87. [PMID: 22545704 DOI: 10.1021/ci200611t] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
In this molecular docking study, the protonation states of the catalytic Asp dyad of the beta-secretase (BACE1) enzyme in the presence of eight chemically diverse inhibitors have been predicted. BACE1 catalyzes the rate-determining step in the generation of Alzheimer amyloid beta peptides and is widely considered as a promising therapeutic target. All the inhibitors were redocked into their corresponding X-ray structures using a combination of eight different protonation states of the Asp dyad for each inhibitor. Five inhibitors were primarily found to favor two different monoprotonated states, and the remaining three favor a dideprotonated state. In addition, five of them exhibited secondary preference for a diprotonated state. These results show that the knowledge of a single protonation state of the Asp dyad is not sufficient to search for the novel inhibitors of BACE1 and the most plausible state for each inhibitor must be determined prior to conducting in-silico screening.
Collapse
Affiliation(s)
- Arghya Barman
- Department of Chemistry, University of Miami, 1301 Memorial Drive, Coral Gables, Florida 33146, USA
| | | |
Collapse
|
100
|
Wang K, Herdtweck E, Dömling A. Cyanoacetamides (IV): versatile one-pot route to 2-quinoline-3-carboxamides. ACS COMBINATORIAL SCIENCE 2012; 14:316-22. [PMID: 22486416 DOI: 10.1021/co3000133] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Cyanoacetic acid derivatives are the starting materials for a plethora of multicomponent reaction (MCR) scaffolds. Herein, we describe scope of a valuable general protocol for the synthesis of arrays of 2-aminoquinoline-3-carboxamides from cyanoacetamides and 2-aminobenzaldehydes or heterocyclic derivatives via a Friedländer reaction variation. In many cases, the reactions involve a very convenient work up by simple precipitation and filtration. More than 40 new products are described. We foresee our protocol and the resulting derivatives becoming very valuable to greatly expanding the scaffold space of cyanoacetamide derivatives.
Collapse
Affiliation(s)
- Kan Wang
- University of Pittsburgh, Drug Discovery Institute,
Biomedical Science Tower 3, 3501 Fifth
Avenue, Pittsburgh, Pennsylvania 15261, United States
| | | | - Alexander Dömling
- University of Pittsburgh, Drug Discovery Institute,
Biomedical Science Tower 3, 3501 Fifth
Avenue, Pittsburgh, Pennsylvania 15261, United States
- University of Groningen, 9700 AB Groningen, the Netherlands
| |
Collapse
|