1
|
Fürstner C, Ackerstaff J, Meier H, Straub A, Mittendorf J, Schamberger J, Schäfer M, Börngen K, Jörißen H, Zubov D, Zimmermann K, Tersteegen A, Geiss V, Hartmann E, Albrecht-Küpper B, D’Orléans-Juste P, Lapointe C, Vincent L, Heitmeier S, Tinel H. Discovery and Preclinical Characterization of Fulacimstat (BAY 1142524), a Potent and Selective Chymase Inhibitor As a New Profibrinolytic Approach for Safe Thrombus Resolution. J Med Chem 2025; 68:6108-6126. [PMID: 39541507 PMCID: PMC11956016 DOI: 10.1021/acs.jmedchem.4c01819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 10/11/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024]
Abstract
Chymase is a serine-protease produced by mast cells. In the past few decades, its role in fibrotic diseases triggered the search for orally available chymase inhibitors. Aiming at reducing adverse cardiac remodeling after myocardial infarction, our research efforts resulted in the discovery of fulacimstat (BAY 1142524). While clinical trials did not demonstrate efficacy in this indication, the recent discovery of a new unexpected biological role of chymase spurred a revival of interest in chymase inhibition: chymase was shown to inactivate plasmin within fibrin-rich clots. Chymase inhibitors are now considered as potential profibrinolytic drugs with low bleeding risk and therefore exceptional safety for the treatment of acute thrombosis settings such as stroke, pulmonary embolism, or venous thrombosis. This article describes the chemical optimization journey from a screening hit to the discovery of fulacimstat (BAY 1142524), a selective chymase inhibitor with a good safety profile, as well as its preclinical in vitro and in vivo characterization.
Collapse
Affiliation(s)
- Chantal Fürstner
- Bayer
AG, Pharmaceuticals, Research and Development, Aprather Weg 18a, 42113 Wuppertal, Germany
| | - Jens Ackerstaff
- Bayer
AG, Pharmaceuticals, Research and Development, Aprather Weg 18a, 42113 Wuppertal, Germany
| | - Heinrich Meier
- Bayer
AG, Pharmaceuticals, Research and Development, Aprather Weg 18a, 42113 Wuppertal, Germany
| | - Alexander Straub
- Bayer
AG, Pharmaceuticals, Research and Development, Aprather Weg 18a, 42113 Wuppertal, Germany
| | - Joachim Mittendorf
- Bayer
AG, Pharmaceuticals, Research and Development, Aprather Weg 18a, 42113 Wuppertal, Germany
| | - Jens Schamberger
- Bayer
AG, Pharmaceuticals, Research and Development, Aprather Weg 18a, 42113 Wuppertal, Germany
| | - Martina Schäfer
- Bayer
AG, Pharmaceuticals, Research and Development, Müllerstrasse 178, 13353 Berlin, Germany
| | - Kirsten Börngen
- Bayer
AG, Pharmaceuticals, Research and Development, Aprather Weg 18a, 42113 Wuppertal, Germany
| | - Hannah Jörißen
- Bayer
AG, Pharmaceuticals, Research and Development, Aprather Weg 18a, 42113 Wuppertal, Germany
| | - Dmitry Zubov
- Bayer
AG, Pharmaceuticals, Research and Development, Aprather Weg 18a, 42113 Wuppertal, Germany
| | - Katja Zimmermann
- Bayer
AG, Pharmaceuticals, Research and Development, Aprather Weg 18a, 42113 Wuppertal, Germany
| | - Adrian Tersteegen
- Bayer
AG, Pharmaceuticals, Research and Development, Aprather Weg 18a, 42113 Wuppertal, Germany
| | - Volker Geiss
- Bayer
AG, Pharmaceuticals, Research and Development, Aprather Weg 18a, 42113 Wuppertal, Germany
| | - Elke Hartmann
- Bayer
AG, Pharmaceuticals, Research and Development, Aprather Weg 18a, 42113 Wuppertal, Germany
| | - Barbara Albrecht-Küpper
- Bayer
AG, Pharmaceuticals, Research and Development, Aprather Weg 18a, 42113 Wuppertal, Germany
| | - Pedro D’Orléans-Juste
- Department
of Pharmacology and Physiology, Faculté de Médecine
et des Sciences de la Santé, Université
de Sherbrooke, 3001,
12e Avenue Nord, Sherbrooke, QC J1H 5N4, Canada
| | - Catherine Lapointe
- Department
of Pharmacology and Physiology, Faculté de Médecine
et des Sciences de la Santé, Université
de Sherbrooke, 3001,
12e Avenue Nord, Sherbrooke, QC J1H 5N4, Canada
| | - Laurence Vincent
- Department
of Pharmacology and Physiology, Faculté de Médecine
et des Sciences de la Santé, Université
de Sherbrooke, 3001,
12e Avenue Nord, Sherbrooke, QC J1H 5N4, Canada
| | - Stefan Heitmeier
- Bayer
AG, Pharmaceuticals, Research and Development, Aprather Weg 18a, 42113 Wuppertal, Germany
| | - Hanna Tinel
- Bayer
AG, Pharmaceuticals, Research and Development, Aprather Weg 18a, 42113 Wuppertal, Germany
| |
Collapse
|
2
|
von Nussbaum F, Testolin G. Opening the Door to a New Treatment Paradigm: The Re-emergence of Chymase Inhibitors. J Med Chem 2025; 68:6100-6103. [PMID: 39961794 DOI: 10.1021/acs.jmedchem.5c00413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/20/2025]
Abstract
Chymase is a serine protease that has been studied as a drug target in inflammatory diseases, so far with limited success. Fulacimstat is a novel drug candidate inhibiting chymase that has been investigated in clinical trials for the role of chymase in inflammation. Despite a good safety profile, further clinical development of fulacimstat was not pursued due to a lack of efficacy. Recently, a new role of chymase in blood clot biology has been identified, this resparks interest in chymase inhibitors, and positions fulacimstat as a potential innovative option to treat thrombotic conditions.
Collapse
|
3
|
Qin H, Wei G, Lou Y, Zheng X, Bao M, Zhang Y, Huang P. K 2S 2O 8-mediated direct C-H heteroarylation/hydroxylation of indolin-2-ones with quinoxalin-2(1 H)-ones. Org Biomol Chem 2024; 22:279-283. [PMID: 38053489 DOI: 10.1039/d3ob01792c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2023]
Abstract
Herein, a K2S2O8-mediated direct heteroarylation and hydroxylation reaction between quinoxalin-2(1H)-ones with a C(sp2)-H bond and indolin-2-ones with a C(sp3)-H bond via an oxidative cross-coupling reaction has been reported. We have successfully established a feasible and concise reaction system that represents the first example of free-radical-promoted heteroarylation and hydroxylation reaction on the C-3 position of oxindole. A series of 3-substituted 3-hydroxyoxindoles are obtained in 0-83% yield using this methodology.
Collapse
Affiliation(s)
- Hui Qin
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China.
- Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Hangzhou, 310014, China
| | - Guoliang Wei
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China.
| | - Yutao Lou
- College of pharmacy, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Xiaowei Zheng
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China.
- Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Hangzhou, 310014, China
| | - Meihua Bao
- Academician Workstation, School of Stomatology, Changsha Medical University, Changsha, China
| | - Yiwen Zhang
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China.
- Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Hangzhou, 310014, China
| | - Ping Huang
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China.
- Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Hangzhou, 310014, China
| |
Collapse
|
4
|
Tan Y, Dai L, Huang W, Guo Y, Zheng S, Lei J, Chen H, Yang Y. DRlinker: Deep Reinforcement Learning for Optimization in Fragment Linking Design. J Chem Inf Model 2022; 62:5907-5917. [PMID: 36404642 DOI: 10.1021/acs.jcim.2c00982] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Fragment-based drug discovery is a widely used strategy for drug design in both academic and pharmaceutical industries. Although fragments can be linked to generate candidate compounds by the latest deep generative models, generating linkers with specified attributes remains underdeveloped. In this study, we presented a novel framework, DRlinker, to control fragment linking toward compounds with given attributes through reinforcement learning. The method has been shown to be effective for many tasks from controlling the linker length and log P, optimizing predicted bioactivity of compounds, to various multiobjective tasks. Specifically, our model successfully generated 91.0% and 93.9% of compounds complying with the desired linker length and log P and improved the 7.5 pChEMBL value in bioactivity optimization. Finally, a quasi-scaffold-hopping study revealed that DRlinker could generate nearly 30% molecules with high 3D similarity but low 2D similarity to the lead inhibitor, demonstrating the benefits and applicability of DRlinker in actual fragment-based drug design.
Collapse
Affiliation(s)
- Youhai Tan
- School of Computer Science and Engineering, Sun Yat-sen University, Guangzhou510006, China
| | - Lingxue Dai
- School of Computer Science and Engineering, Sun Yat-sen University, Guangzhou510006, China
| | - Weifeng Huang
- School of Pharmaceutical Science, Sun Yat-sen University, Guangzhou510006, China
| | - Yinfeng Guo
- School of Pharmaceutical Science, Sun Yat-sen University, Guangzhou510006, China
| | - Shuangjia Zheng
- School of Computer Science and Engineering, Sun Yat-sen University, Guangzhou510006, China.,Galixir Technologies, Beijing100083, China
| | - Jinping Lei
- School of Pharmaceutical Science, Sun Yat-sen University, Guangzhou510006, China
| | - Hongming Chen
- Guangzhou Laboratory, No. 9 XinDaoHuanBei Road, Guangzhou International Bio Island, Guangzhou510005, China
| | - Yuedong Yang
- School of Computer Science and Engineering, Sun Yat-sen University, Guangzhou510006, China
| |
Collapse
|
5
|
Golla S, Jalagam S, Poshala S, Kokatla HP. Transition metal-free functionalization of 2-oxindoles via sequential aldol and reductive aldol reactions using rongalite as a C1 reagent. Org Biomol Chem 2022; 20:4926-4932. [PMID: 35506377 DOI: 10.1039/d2ob00665k] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
A sequential one-pot classical aldol, transition-metal and hydride-free reductive aldol reaction is reported here for C(sp3)- H functionalization of 2-oxindoles using the multifaceted reagent rongalite. Here, rongalite functions as a hydride-free reducing agent and double C1 unit donor. This protocol enables the synthesis of a wide range of 3-methylindoline-2-ones and 3-(hydroxymethyl)-3-methylindolin-2-ones from 2-oxindoles (65-95% yields), which are the synthetic precursors for many natural products. Some of the important aspects of this synthetic approach include one-pot methylation and hydroxymethylation, low-cost rongalite (ca. $0.03 per 1 g), mild reaction conditions and applicability to gram-scale synthesis.
Collapse
Affiliation(s)
- Sivaparwathi Golla
- Department of Chemistry, National Institute of Technology Warangal, Warangal, Telangana-506004, India.
| | - Swathi Jalagam
- Department of Chemistry, National Institute of Technology Warangal, Warangal, Telangana-506004, India.
| | - Soumya Poshala
- Department of Chemistry, National Institute of Technology Warangal, Warangal, Telangana-506004, India.
| | - Hari Prasad Kokatla
- Department of Chemistry, National Institute of Technology Warangal, Warangal, Telangana-506004, India.
| |
Collapse
|
6
|
Smilova MD, Curran PR, Radoux CJ, von Delft F, Cole JC, Bradley AR, Marsden BD. Fragment Hotspot Mapping to Identify Selectivity-Determining Regions between Related Proteins. J Chem Inf Model 2022; 62:284-294. [PMID: 35020376 PMCID: PMC8790751 DOI: 10.1021/acs.jcim.1c00823] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
![]()
Selectivity is a
crucial property in small molecule development.
Binding site comparisons within a protein family are a key piece of
information when aiming to modulate the selectivity profile of a compound.
Binding site differences can be exploited to confer selectivity for
a specific target, while shared areas can provide insights into polypharmacology.
As the quantity of structural data grows, automated methods are needed
to process, summarize, and present these data to users. We present
a computational method that provides quantitative and data-driven
summaries of the available binding site information from an ensemble
of structures of the same protein. The resulting ensemble maps identify
the key interactions important for ligand binding in the ensemble.
The comparison of ensemble maps of related proteins enables the identification
of selectivity-determining regions within a protein family. We applied
the method to three examples from the well-researched human bromodomain
and kinase families, demonstrating that the method is able to identify
selectivity-determining regions that have been used to introduce selectivity
in past drug discovery campaigns. We then illustrate how the resulting
maps can be used to automate comparisons across a target protein family.
Collapse
Affiliation(s)
- Mihaela D Smilova
- Centre for Medicines Discovery, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Headington, Oxford OX3 7DQ, U.K
| | - Peter R Curran
- The Cambridge Crystallographic Data Centre (CCDC), Cambridge CB2 1EZ, U.K.,Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, U.K
| | - Chris J Radoux
- Exscientia Ltd., The Schrödinger Building, Oxford Science Park, Oxford OX4 4GE, U.K
| | - Frank von Delft
- Centre for Medicines Discovery, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Headington, Oxford OX3 7DQ, U.K.,Diamond Light Source Ltd., Harwell Science and Innovation Campus, Didcot OX11 0DE, U.K.,Research Complex at Harwell. Harwell Science and Innovation Campus, Didcot OX11 0FA, U.K.,Department of Biochemistry, University of Johannesburg, Auckland Park 2006, South Africa
| | - Jason C Cole
- The Cambridge Crystallographic Data Centre (CCDC), Cambridge CB2 1EZ, U.K
| | - Anthony R Bradley
- Exscientia Ltd., The Schrödinger Building, Oxford Science Park, Oxford OX4 4GE, U.K
| | - Brian D Marsden
- Centre for Medicines Discovery, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Headington, Oxford OX3 7DQ, U.K.,Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford OX3 7DQ, U.K
| |
Collapse
|
7
|
Silva RAL, da Silva Filho DA, Moberg ME, Pappenfus TM, Janzen DE. Halogen Interactions in Halogenated Oxindoles: Crystallographic and Computational Investigations of Intermolecular Interactions. Molecules 2021; 26:molecules26185487. [PMID: 34576963 PMCID: PMC8464904 DOI: 10.3390/molecules26185487] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 09/01/2021] [Accepted: 09/06/2021] [Indexed: 11/16/2022] Open
Abstract
X-ray structural determinations and computational studies were used to investigate halogen interactions in two halogenated oxindoles. Comparative analyses of the interaction energy and the interaction properties were carried out for Br···Br, C-H···Br, C-H···O and N-H···O interactions. Employing Møller-Plesset second-order perturbation theory (MP2) and density functional theory (DFT), the basis set superposition error (BSSE) corrected interaction energy (Eint(BSSE)) was determined using a supramolecular approach. The Eint(BSSE) results were compared with interaction energies obtained by Quantum Theory of Atoms in Molecules (QTAIM)-based methods. Reduced Density Gradient (RDG), QTAIM and Natural bond orbital (NBO) calculations provided insight into possible pathways for the intermolecular interactions examined. Comparative analysis employing the electron density at the bond critical points (BCP) and molecular electrostatic potential (MEP) showed that the interaction energies and the relative orientations of the monomers in the dimers may in part be understood in light of charge redistribution in these two compounds.
Collapse
Affiliation(s)
- Rodrigo A. Lemos Silva
- Institute of Physics, University of Brasilia, Brasilia 70910-900, Brazil; (R.A.L.S.); (D.A.d.S.F.)
| | - Demetrio A. da Silva Filho
- Institute of Physics, University of Brasilia, Brasilia 70910-900, Brazil; (R.A.L.S.); (D.A.d.S.F.)
- International Center for Condensed Matter Physics, University of Brasilia, CP 04455, Brasilia 70919-970, Brazil
| | - Megan E. Moberg
- Department of Chemistry & Biochemistry, St. Catherine University, St. Paul, MN 55105, USA;
| | - Ted M. Pappenfus
- Division of Science and Mathematics, University of Minnesota, Morris, MN 56267, USA;
| | - Daron E. Janzen
- Department of Chemistry & Biochemistry, St. Catherine University, St. Paul, MN 55105, USA;
- Correspondence:
| |
Collapse
|
8
|
Taylor SJ, Abeywardane A, Liang S, Xiong Z, Proudfoot JR, Farmer BS, Gao DA, Heim-Riether A, Smith-Keenan LL, Muegge I, Yu Y, Zhang Q, Souza D, Panzenbeck M, Goldberg D, Hill-Drzewi M, Margarit M, Collins B, Li JX, Zuvela-Jelaska L, Li J, Farrow NA. Indole Inhibitors of MMP-13 for Arthritic Disorders. ACS OMEGA 2021; 6:18635-18650. [PMID: 34337203 PMCID: PMC8319936 DOI: 10.1021/acsomega.1c01320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 06/30/2021] [Indexed: 06/13/2023]
Abstract
Here, we described the design, by fragment merging and multiparameter optimization, of selective MMP-13 inhibitors that display an appropriate balance of potency and physicochemical properties to qualify as tool compounds suitable for in vivo testing. Optimization of potency was guided by structure-based insights, specifically to replace an ester moiety and introduce polar directional hydrogen bonding interactions in the core of the molecule. By introducing polar enthalpic interactions in this series of inhibitors, the overall beneficial physicochemical properties were maintained. These physicochemical properties translated to excellent drug-like properties beyond potency. In a murine model of rheumatoid arthritis, treatment of mice with selective inhibitors of MMP-13 resulted in a statistically significant reduction in the mean arthritic score vs control when dosed over a 14 day period.
Collapse
Affiliation(s)
- Steven J. Taylor
- Boehringer
Ingelheim Pharmaceuticals Inc., 900 Ridgebury Road, Ridgefield, Connecticut 06877-0368, United States
| | - Asitha Abeywardane
- Boehringer
Ingelheim Pharmaceuticals Inc., 900 Ridgebury Road, Ridgefield, Connecticut 06877-0368, United States
| | - Shuang Liang
- Boehringer
Ingelheim Pharmaceuticals Inc., 900 Ridgebury Road, Ridgefield, Connecticut 06877-0368, United States
| | - Zhaoming Xiong
- Boehringer
Ingelheim Pharmaceuticals Inc., 900 Ridgebury Road, Ridgefield, Connecticut 06877-0368, United States
| | - John R. Proudfoot
- Boehringer
Ingelheim Pharmaceuticals Inc., 900 Ridgebury Road, Ridgefield, Connecticut 06877-0368, United States
| | - Bennett Sandy Farmer
- Boehringer
Ingelheim Pharmaceuticals Inc., 900 Ridgebury Road, Ridgefield, Connecticut 06877-0368, United States
| | - Donghong A. Gao
- Boehringer
Ingelheim Pharmaceuticals Inc., 900 Ridgebury Road, Ridgefield, Connecticut 06877-0368, United States
| | - Alexander Heim-Riether
- Boehringer
Ingelheim Pharmaceuticals Inc., 900 Ridgebury Road, Ridgefield, Connecticut 06877-0368, United States
| | - Lana Louise Smith-Keenan
- Boehringer
Ingelheim Pharmaceuticals Inc., 900 Ridgebury Road, Ridgefield, Connecticut 06877-0368, United States
| | - Ingo Muegge
- Boehringer
Ingelheim Pharmaceuticals Inc., 900 Ridgebury Road, Ridgefield, Connecticut 06877-0368, United States
| | - Yang Yu
- Boehringer
Ingelheim Pharmaceuticals Inc., 900 Ridgebury Road, Ridgefield, Connecticut 06877-0368, United States
| | - Qiang Zhang
- Boehringer
Ingelheim Pharmaceuticals Inc., 900 Ridgebury Road, Ridgefield, Connecticut 06877-0368, United States
| | - Donald Souza
- Boehringer
Ingelheim Pharmaceuticals Inc., 900 Ridgebury Road, Ridgefield, Connecticut 06877-0368, United States
| | - Mark Panzenbeck
- Boehringer
Ingelheim Pharmaceuticals Inc., 900 Ridgebury Road, Ridgefield, Connecticut 06877-0368, United States
| | - Daniel Goldberg
- Boehringer
Ingelheim Pharmaceuticals Inc., 900 Ridgebury Road, Ridgefield, Connecticut 06877-0368, United States
| | - Melissa Hill-Drzewi
- Boehringer
Ingelheim Pharmaceuticals Inc., 900 Ridgebury Road, Ridgefield, Connecticut 06877-0368, United States
| | - Mariana Margarit
- Boehringer
Ingelheim Pharmaceuticals Inc., 900 Ridgebury Road, Ridgefield, Connecticut 06877-0368, United States
| | - Brandon Collins
- Boehringer
Ingelheim Pharmaceuticals Inc., 900 Ridgebury Road, Ridgefield, Connecticut 06877-0368, United States
| | - John Xiang Li
- Boehringer
Ingelheim Pharmaceuticals Inc., 900 Ridgebury Road, Ridgefield, Connecticut 06877-0368, United States
| | - Ljiljana Zuvela-Jelaska
- Boehringer
Ingelheim Pharmaceuticals Inc., 900 Ridgebury Road, Ridgefield, Connecticut 06877-0368, United States
| | - Jun Li
- Boehringer
Ingelheim Pharmaceuticals Inc., 900 Ridgebury Road, Ridgefield, Connecticut 06877-0368, United States
| | - Neil A. Farrow
- Boehringer
Ingelheim Pharmaceuticals Inc., 900 Ridgebury Road, Ridgefield, Connecticut 06877-0368, United States
| |
Collapse
|
9
|
Robinson H, Oatley SA, Rowedder JE, Slade P, Macdonald SJF, Argent SP, Hirst JD, McInally T, Moody CJ. Late-Stage Functionalization by Chan-Lam Amination: Rapid Access to Potent and Selective Integrin Inhibitors. Chemistry 2020; 26:7678-7684. [PMID: 32129907 DOI: 10.1002/chem.202001059] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Indexed: 11/07/2022]
Abstract
A late-stage functionalization of the aromatic ring in amino acid derivatives is described. The key step is a copper-catalysed diversification of a boronate ester by amination (Chan-Lam reaction) that can be carried out on a complex β-aryl-β-amino acid scaffold. This not only considerably extends the substrate scope of amination partners, but also delivers an array of potent and selective integrin inhibitors as potential treatment agents of idiopathic pulmonary fibrosis (IPF). This versatile chemical strategy, which is amenable to high-throughput-array protocols, allows the installation of pharmaceutically valuable heteroaromatic fragments at a late stage by direct coupling to NH heterocycles, leading to compounds with drug-like attributes. It thus constitutes a useful addition to the medicinal chemist's repertoire.
Collapse
Affiliation(s)
- Henry Robinson
- School of Chemistry, GSK Carbon Neutral Laboratories for Sustainable Chemistry, University of Nottingham, Jubilee Campus, Triumph Road, Nottingham, NG7 2TU, UK
| | - Steven A Oatley
- School of Chemistry, University of Nottingham, University Park, Nottingham, NG7 2RD, UK
| | - James E Rowedder
- Medicinal Science & Technology, GlaxoSmithKline Medicines Research Centre, Gunnels Wood Road, Stevenage, SG1 2NY, UK
| | - Pawel Slade
- Medicinal Science & Technology, GlaxoSmithKline Medicines Research Centre, Gunnels Wood Road, Stevenage, SG1 2NY, UK
| | - Simon J F Macdonald
- Medicinal Science & Technology, GlaxoSmithKline Medicines Research Centre, Gunnels Wood Road, Stevenage, SG1 2NY, UK
| | - Stephen P Argent
- School of Chemistry, University of Nottingham, University Park, Nottingham, NG7 2RD, UK
| | - Jonathan D Hirst
- School of Chemistry, University of Nottingham, University Park, Nottingham, NG7 2RD, UK
| | - Thomas McInally
- School of Chemistry, GSK Carbon Neutral Laboratories for Sustainable Chemistry, University of Nottingham, Jubilee Campus, Triumph Road, Nottingham, NG7 2TU, UK
| | - Christopher J Moody
- School of Chemistry, University of Nottingham, University Park, Nottingham, NG7 2RD, UK
| |
Collapse
|
10
|
Bancet A, Raingeval C, Lomberget T, Le Borgne M, Guichou JF, Krimm I. Fragment Linking Strategies for Structure-Based Drug Design. J Med Chem 2020; 63:11420-11435. [DOI: 10.1021/acs.jmedchem.0c00242] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Alexandre Bancet
- EA 4446 Bioactive Molecules and Medicinal Chemistry, Faculté de Pharmacie, ISPB, SFR Santé Lyon-Est CNRS UMS3453, INSERM US7, Université de Lyon, Université Claude Bernard Lyon 1, 69373 Lyon Cedex 8, France
- Centre de RMN à Très Hauts Champs, Université de Lyon, CNRS, Université Claude Bernard Lyon 1, ENS, 5 Rue de la Doua, F-69100 Villeurbanne, France
| | - Claire Raingeval
- Centre de RMN à Très Hauts Champs, Université de Lyon, CNRS, Université Claude Bernard Lyon 1, ENS, 5 Rue de la Doua, F-69100 Villeurbanne, France
| | - Thierry Lomberget
- EA 4446 Bioactive Molecules and Medicinal Chemistry, Faculté de Pharmacie, ISPB, SFR Santé Lyon-Est CNRS UMS3453, INSERM US7, Université de Lyon, Université Claude Bernard Lyon 1, 69373 Lyon Cedex 8, France
| | - Marc Le Borgne
- EA 4446 Bioactive Molecules and Medicinal Chemistry, Faculté de Pharmacie, ISPB, SFR Santé Lyon-Est CNRS UMS3453, INSERM US7, Université de Lyon, Université Claude Bernard Lyon 1, 69373 Lyon Cedex 8, France
| | | | - Isabelle Krimm
- Centre de RMN à Très Hauts Champs, Université de Lyon, CNRS, Université Claude Bernard Lyon 1, ENS, 5 Rue de la Doua, F-69100 Villeurbanne, France
- Centre de Recherche en Cancérologie de Lyon, Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, 69008 Lyon, France
| |
Collapse
|
11
|
Li CY, Yap K, Swedberg JE, Craik DJ, de Veer SJ. Binding Loop Substitutions in the Cyclic Peptide SFTI-1 Generate Potent and Selective Chymase Inhibitors. J Med Chem 2020; 63:816-826. [PMID: 31855419 DOI: 10.1021/acs.jmedchem.9b01811] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Chymase is a serine protease that is predominantly expressed by mast cells and has key roles in immune defense and the cardiovascular system. This enzyme has also emerged as a therapeutic target for cardiovascular disease due to its ability to remodel cardiac tissue and generate angiotensin II. Here, we used the nature-derived cyclic peptide sunflower trypsin inhibitor-1 (SFTI-1) as a template for designing novel chymase inhibitors. The key binding contacts of SFTI-1 were optimized by combining a peptide substrate library screen with structure-based design, which yielded several variants with potent activity. The lead variant was further modified by replacing the P1 Tyr residue with para-substituted Phe derivatives, generating new inhibitors with improved potency (Ki = 1.8 nM) and higher selectivity over closely related enzymes. Several variants were shown to block angiotensin I cleavage in vitro, highlighting their potential for further development and future evaluation as pharmaceutical leads.
Collapse
Affiliation(s)
- Choi Yi Li
- Institute for Molecular Bioscience , The University of Queensland , Brisbane , QLD 4072 , Australia
| | - Kuok Yap
- Institute for Molecular Bioscience , The University of Queensland , Brisbane , QLD 4072 , Australia
| | - Joakim E Swedberg
- Institute for Molecular Bioscience , The University of Queensland , Brisbane , QLD 4072 , Australia
| | - David J Craik
- Institute for Molecular Bioscience , The University of Queensland , Brisbane , QLD 4072 , Australia
| | - Simon J de Veer
- Institute for Molecular Bioscience , The University of Queensland , Brisbane , QLD 4072 , Australia
| |
Collapse
|
12
|
Qin H, Li Q, Xu J, Zhang J, Qu W, Liu W, Feng F, Sun H. A Mild and Direct C(sp 3)-S Cross-Coupling of Oxindoles with Thiols: Synthesis of Unsymmetrical 3-Thiooxindoles. J Org Chem 2019; 84:14342-14348. [PMID: 31580081 DOI: 10.1021/acs.joc.9b02205] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Herein, an operationally simple and mild strategy to construct sulfenation of oxindoles with a series of thiols in the absence of transition metals was developed. This methodology provides an efficient way to directly form a C-S bond at the C-3 position of oxindoles under mild reaction conditions with a cheap and common solvent and base in moderate to good yields.
Collapse
Affiliation(s)
- Hui Qin
- Department of Natural Medicinal Chemistry , China Pharmaceutical University , Nanjing 211198 , China
| | - Qi Li
- Department of Medicinal Chemistry , China Pharmaceutical University , Nanjing 210009 , China
| | - Jian Xu
- Department of Natural Medicinal Chemistry , China Pharmaceutical University , Nanjing 211198 , China
| | - Jie Zhang
- Department of Natural Medicinal Chemistry , China Pharmaceutical University , Nanjing 211198 , China
| | - Wei Qu
- Department of Natural Medicinal Chemistry , China Pharmaceutical University , Nanjing 211198 , China.,Key Laboratory of Biomedical Functional Materials , China Pharmaceutical University , Nanjing 211198 , China
| | - Wenyuan Liu
- Department of Pharmaceutical Analysis , China Pharmaceutical University , Nanjing 210009 , China
| | - Feng Feng
- Department of Natural Medicinal Chemistry , China Pharmaceutical University , Nanjing 211198 , China.,Key Laboratory of Biomedical Functional Materials , China Pharmaceutical University , Nanjing 211198 , China.,Jiangsu Food and Pharmaceutical Science College , Huai'an 223003 , China
| | - Haopeng Sun
- Department of Medicinal Chemistry , China Pharmaceutical University , Nanjing 210009 , China
| |
Collapse
|
13
|
Ozawa SI, Takahashi M, Yamaotsu N, Hirono S. Structure-based virtual screening for novel chymase inhibitors by in silico fragment mapping. J Mol Graph Model 2019; 89:102-108. [PMID: 30884446 DOI: 10.1016/j.jmgm.2019.03.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 03/06/2019] [Accepted: 03/07/2019] [Indexed: 01/22/2023]
Abstract
The term chymase refers to a family of chymotrypsin-like serine proteases stored within the secretory granules of mast cells. Recently, a variety of small molecule inhibitors for chymase have been developed with a primary focus on the treatment of cardiovascular diseases. Despite the expected therapeutic benefit of these chymase inhibitors, they have not been used clinically. Here, we attempted to identify new chymase inhibitors using a multistep structure-based virtual screening protocol combined with our knowledge-based in silico fragment mapping technique. The mapping procedure identified fragments with novel modes of interaction at the oxyanion hole of chymase. Next, we constructed a three-dimensional (3D) pharmacophore model and retrieved eight candidate chymase inhibitors from a commercial database that included approximately five million compounds. This selection was achieved using a multistep virtual screening protocol, which combined a 3D pharmacophore-based search, docking calculations, and analyses of binding free energy. One of the eight compounds exhibited concentration-dependent chymase inhibitory activity, which could be further optimized to develop more potent chymase inhibitors.
Collapse
Affiliation(s)
- Shin-Ichiro Ozawa
- Department of Pharmaceutical Sciences, School of Pharmacy, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo, 108-8641, Japan.
| | - Miki Takahashi
- Department of Pharmaceutical Sciences, School of Pharmacy, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo, 108-8641, Japan
| | - Noriyuki Yamaotsu
- Department of Pharmaceutical Sciences, School of Pharmacy, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo, 108-8641, Japan
| | - Shuichi Hirono
- Department of Pharmaceutical Sciences, School of Pharmacy, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo, 108-8641, Japan
| |
Collapse
|
14
|
Qin H, Miao Y, Zhang K, Xu J, Sun H, Liu W, Feng F, Qu W. A convenient cyclopropanation process of oxindoles via bromoethylsulfonium salt. Tetrahedron 2018. [DOI: 10.1016/j.tet.2018.09.042] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
15
|
Ahmad S, Ferrario CM. Chymase inhibitors for the treatment of cardiac diseases: a patent review (2010-2018). Expert Opin Ther Pat 2018; 28:755-764. [PMID: 30278800 DOI: 10.1080/13543776.2018.1531848] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Chymase is primarily found in mast cells (MCs), fibroblasts, and vascular endothelial cells. MC chymase is released into the extracellular interstitium in response to inflammatory signals, tissue injury, and cellular stress. Among many functions, chymase is a major extravascular source for angiotensin II (Ang II) generation. Several recent pre-clinical and a few clinical studies point to the relatively unrecognized fact that chymase inhibition may have significant therapeutic advantages over other treatments in halting progression of cardiac and vascular disease. AREAS COVERED The present review covers patent literature on chymase inhibitors for the treatment of cardiac diseases registered between 2010 and 2018. EXPERT OPINION Increase in cardiac MC number in various cardiac diseases has been found in pathological tissues of human and experimental animals. Meta-analysis data from large clinical trials employing angiotensin-converting enzyme (ACE) inhibitors show a relatively small risk reduction of clinical cardiovascular endpoints. The disconnect between the expected benefit associated with Ang II blockade of synthesis or activity underscores a greater participation of chymase compared to ACE in forming Ang II in humans. Emerging literature and a reconsideration of previous studies provide lucid arguments to reconsider chymase as a primary Ang II forming enzyme in human heart and vasculature.
Collapse
Affiliation(s)
- Sarfaraz Ahmad
- a Department of Surgery , Wake Forest School of Medicine , Winston Salem , NC , USA
| | - Carlos M Ferrario
- a Department of Surgery , Wake Forest School of Medicine , Winston Salem , NC , USA.,b Department of Physiology-Pharmacology , Wake Forest School of Medicine , Winston Salem , NC , USA.,c Department of Social Sciences, Division of Public Health , Wake Forest School of Medicine , Winston Salem , NC , USA
| |
Collapse
|
16
|
Jiang X, Zheng C, Lei L, Lin K, Yu C. Synthesis of 2-Oxindoles from Substituted Indoles by Hypervalent-Iodine Oxidation. European J Org Chem 2018. [DOI: 10.1002/ejoc.201701807] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Xinpeng Jiang
- College of Pharmaceutical Sciences; Zhejiang University of Technology; Hangzhou P. R. China
| | - Cong Zheng
- College of Pharmaceutical Sciences; Zhejiang University of Technology; Hangzhou P. R. China
| | - Lijun Lei
- College of Pharmaceutical Sciences; Zhejiang University of Technology; Hangzhou P. R. China
| | - Kai Lin
- College of Pharmaceutical Sciences; Zhejiang University of Technology; Hangzhou P. R. China
| | - Chuanming Yu
- College of Pharmaceutical Sciences; Zhejiang University of Technology; Hangzhou P. R. China
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals; Zhejiang University of Technology; Hangzhou P. R. China
| |
Collapse
|
17
|
Bigatti M, Dal Corso A, Vanetti S, Cazzamalli S, Rieder U, Scheuermann J, Neri D, Sladojevich F. Impact of a Central Scaffold on the Binding Affinity of Fragment Pairs Isolated from DNA-Encoded Self-Assembling Chemical Libraries. ChemMedChem 2017; 12:1748-1752. [PMID: 28944578 DOI: 10.1002/cmdc.201700569] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Indexed: 12/19/2022]
Abstract
The screening of encoded self-assembling chemical libraries allows the identification of fragment pairs that bind to adjacent pockets on target proteins of interest. For practical applications, it is necessary to link these ligand pairs into discrete organic molecules, devoid of any nucleic acid component. Here we describe the discovery of a synergistic binding pair for acid alpha-1 glycoprotein and a chemical strategy for the identification of optimal linkers, connecting the two fragments. The procedure yielded a set of small organic ligands, the best of which exhibited a dissociation constant of 9.9 nm, as measured in solution by fluorescence polarization.
Collapse
Affiliation(s)
| | - Alberto Dal Corso
- Institute of Pharmaceutical Sciences, ETH Zürich, 8093, Zürich, Switzerland
| | | | - Samuele Cazzamalli
- Institute of Pharmaceutical Sciences, ETH Zürich, 8093, Zürich, Switzerland
| | | | - Jörg Scheuermann
- Institute of Pharmaceutical Sciences, ETH Zürich, 8093, Zürich, Switzerland
| | - Dario Neri
- Institute of Pharmaceutical Sciences, ETH Zürich, 8093, Zürich, Switzerland
| | | |
Collapse
|
18
|
|
19
|
Abstract
After 20 years of sometimes quiet growth, fragment-based drug discovery (FBDD) has become mainstream. More than 30 drug candidates derived from fragments have entered the clinic, with two approved and several more in advanced trials. FBDD has been widely applied in both academia and industry, as evidenced by the large number of papers from universities, non-profit research institutions, biotechnology companies and pharmaceutical companies. Moreover, FBDD draws on a diverse range of disciplines, from biochemistry and biophysics to computational and medicinal chemistry. As the promise of FBDD strategies becomes increasingly realized, now is an opportune time to draw lessons and point the way to the future. This Review briefly discusses how to design fragment libraries, how to select screening techniques and how to make the most of information gleaned from them. It also shows how concepts from FBDD have permeated and enhanced drug discovery efforts.
Collapse
|
20
|
Ohtera A, Miyamae Y, Yoshida K, Maejima K, Akita T, Kakizuka A, Irie K, Masuda S, Kambe T, Nagao M. Identification of a New Type of Covalent PPARγ Agonist using a Ligand-Linking Strategy. ACS Chem Biol 2015; 10:2794-804. [PMID: 26414848 DOI: 10.1021/acschembio.5b00628] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Peroxisome proliferator-activated receptor γ (PPARγ) is a ligand-activated transcription factor that plays an important role in adipogenesis and glucose metabolism. The ligand-binding pocket (LBP) of PPARγ has a large Y-shaped cavity with multiple subpockets where multiple ligands can simultaneously bind and cooperatively activate PPARγ. Focusing on this unique property of the PPARγ LBP, we describe a novel two-step cell-based strategy to develop PPARγ ligands. First, a combination of ligands that cooperatively activates PPARγ was identified using a luciferase reporter assay. Second, hybrid ligands were designed and synthesized. For proof of concept, we focused on covalent agonists, which activate PPARγ through a unique activation mechanism regulated by a covalent linkage with the Cys285 residue in the PPARγ LBP. Despite their biological significance and pharmacological potential, few covalent PPARγ agonists are known except for endogenous fatty acid metabolites. With our strategy, we determined that plant-derived cinnamic acid derivatives cooperatively activated PPARγ by combining with GW9662, an irreversible antagonist. GW9662 covalently reacts with the Cys285 residue. A docking study predicted that a cinnamic acid derivative can bind to the open cavity in GW9662-bound PPARγ LBP. On the basis of the putative binding mode, structures of both ligands were linked successfully to create a potent PPARγ agonist, which enhanced the transactivation potential of PPARγ at submicromolar levels through covalent modification of Cys285. Our approach could lead to the discovery of novel high-potency PPARγ agonists.
Collapse
Affiliation(s)
| | | | | | | | - Toru Akita
- Nippon Shinyaku CO., LTD., Kyoto 601-8550, Japan
| | | | | | | | | | | |
Collapse
|
21
|
Andreoli F, Kaid-Slimane R, Coppola F, Farran D, Roussel C, Vanthuyne N. Access to N-Thioalkenyl and N-(o-Thio)aryl-benzimidazol-2-ones by Ring Opening of Thiazolobenzimidazolium and Benzimidazobenzothiazolium Salts and C–O Bond Cleavage of an Alkoxide. J Org Chem 2015; 80:3233-41. [DOI: 10.1021/acs.joc.5b00221] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
- Federico Andreoli
- Aix Marseille Université, Centrale Marseille, CNRS, iSm2 UMR 7313, 13397 Marseille, France
| | - Radia Kaid-Slimane
- Laboratoire
de synthèse organique appliquée, Département
de Chimie, Faculté des Sciences, Université d’Oran (Es Sénia), B.P. 1524, El M’naouer
Oran, Algérie
| | - Fabien Coppola
- Aix Marseille Université, Centrale Marseille, CNRS, iSm2 UMR 7313, 13397 Marseille, France
| | - Daniel Farran
- Aix Marseille Université, Centrale Marseille, CNRS, iSm2 UMR 7313, 13397 Marseille, France
| | - Christian Roussel
- Aix Marseille Université, Centrale Marseille, CNRS, iSm2 UMR 7313, 13397 Marseille, France
| | - Nicolas Vanthuyne
- Aix Marseille Université, Centrale Marseille, CNRS, iSm2 UMR 7313, 13397 Marseille, France
| |
Collapse
|
22
|
(3Z)-3-(2-[4-(aryl)-1,3-thiazol-2-yl]hydrazin-1-ylidene)-2,3-dihydro-1H-indol-2-one derivatives as dual inhibitors of HIV-1 reverse transcriptase. Eur J Med Chem 2015; 93:452-60. [PMID: 25728026 DOI: 10.1016/j.ejmech.2015.02.032] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Revised: 01/13/2015] [Accepted: 02/19/2015] [Indexed: 11/24/2022]
Abstract
The HIV-1 Reverse Transcriptase (RT) is a validated and deeply explored biological target for the treatment of AIDS. However, only drugs targeting the RT-associated DNA polymerase (DP) function have been approved for clinical use. We designed and synthesised a new generation of HIV-1 RT inhibitors, based on the (3Z)-3-(2-[4-(aryl)-1,3-thiazol-2-yl]hydrazin-1-ylidene)-2,3-dihydro-1H-indol-2-one scaffold. These compounds are active towards both RT-associated functions, DNA polymerase and ribonuclease H. The structure, biological activity and mode of action of the new derivatives have been investigated. In particular, the nature of the aromatic group in the position 4 of the thiazole ring plays a key role in the modulation of the activity towards the two RT-associated functions.
Collapse
|
23
|
Keri RS, Hiremathad A, Budagumpi S, Nagaraja BM. Comprehensive Review in Current Developments of Benzimidazole-Based Medicinal Chemistry. Chem Biol Drug Des 2014; 86:19-65. [PMID: 25352112 DOI: 10.1111/cbdd.12462] [Citation(s) in RCA: 214] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Accepted: 10/12/2014] [Indexed: 12/13/2022]
Abstract
The properties of benzimidazole and its derivatives have been studied over more than one hundred years. Benzimidazole derivatives are useful intermediates/subunits for the development of molecules of pharmaceutical or biological interest. Substituted benzimidazole derivatives have found applications in diverse therapeutic areas such as antiulcer, anticancer agents, and anthelmintic species to name just a few. This work systematically gives a comprehensive review in current developments of benzimidazole-based compounds in the whole range of medicinal chemistry as anticancer, antibacterial, antifungal, anti-inflammatory, analgesic agents, anti-HIV, antioxidant, anticonvulsant, antitubercular, antidiabetic, antileishmanial, antihistaminic, antimalarial agents, and other medicinal agents. This review will further be helpful for the researcher on the basis of substitution pattern around the nucleus with an aim to help medicinal chemists for developing an SAR on benzimidazole drugs/compounds.
Collapse
Affiliation(s)
- Rangappa S Keri
- Centre for Nano and Material Sciences, Jain University, Jain Global Campus, Bangalore, Karnataka, 562112, India
| | - Asha Hiremathad
- Centre for Nano and Material Sciences, Jain University, Jain Global Campus, Bangalore, Karnataka, 562112, India
| | - Srinivasa Budagumpi
- Centre for Nano and Material Sciences, Jain University, Jain Global Campus, Bangalore, Karnataka, 562112, India
| | - Bhari Mallanna Nagaraja
- Centre for Nano and Material Sciences, Jain University, Jain Global Campus, Bangalore, Karnataka, 562112, India
| |
Collapse
|
24
|
Fei X, Yuan Y, Lee YM, Jeong KW, Seo SY. Synthesis, Biological Evaluation of SPF-32629A-Based 2- and 4-Pyridone Analogs as Chymase Inhibitors. B KOREAN CHEM SOC 2014. [DOI: 10.5012/bkcs.2014.35.8.2547] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
25
|
A unique one-pot reaction via CC cleavage from aminomethylene benzimidazoles to access benzimidazolones with wide potentiality. Tetrahedron Lett 2014. [DOI: 10.1016/j.tetlet.2014.05.113] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|