1
|
Fang X, Zeng Y, Huang Y, Zhu Z, Lin S, Xu W, Zheng C, Hu X, Qiu Y, Ruan Z. Electrochemical synthesis of peptide aldehydes via C‒N bond cleavage of cyclic amines. Nat Commun 2024; 15:5181. [PMID: 38890290 PMCID: PMC11189564 DOI: 10.1038/s41467-024-49223-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 05/29/2024] [Indexed: 06/20/2024] Open
Abstract
Peptide aldehydes are crucial biomolecules essential to various biological systems, driving a continuous demand for efficient synthesis methods. Herein, we develop a metal-free, facile, and biocompatible strategy for direct electrochemical synthesis of unnatural peptide aldehydes. This electro-oxidative approach enabled a step- and atom-economical ring-opening via C‒N bond cleavage, allowing for homoproline-specific peptide diversification and expansion of substrate scope to include amides, esters, and cyclic amines of various sizes. The remarkable efficacy of the electro-synthetic protocol set the stage for the efficient modification and assembly of linear and macrocyclic peptides using a concise synthetic sequence with racemization-free conditions. Moreover, the combination of experiments and density functional theory (DFT) calculations indicates that different N-acyl groups play a decisive role in the reaction activity.
Collapse
Affiliation(s)
- Xinyue Fang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Yong Zeng
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Yawen Huang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Zile Zhu
- State Key Laboratory and Institute of Elemento-Organic Chemistry, Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, 94 Weijin Road, Tianjin, 300071, PR China
| | - Shengsheng Lin
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Wenyan Xu
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Chengwei Zheng
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Xinwei Hu
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, PR China.
| | - Youai Qiu
- State Key Laboratory and Institute of Elemento-Organic Chemistry, Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, 94 Weijin Road, Tianjin, 300071, PR China.
| | - Zhixiong Ruan
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, PR China.
| |
Collapse
|
2
|
Carney DW, Leffler AE, Bell JA, Chandrasinghe AS, Cheng C, Chang E, Dornford A, Dougan DR, Frye LL, Grimes ME, Knehans T, Knight JL, Komandla M, Lane W, Li H, Newman SR, Phimister K, Saikatendu KS, Silverstein H, Vafaei S. Exploiting high-energy hydration sites for the discovery of potent peptide aldehyde inhibitors of the SARS-CoV-2 main protease with cellular antiviral activity. Bioorg Med Chem 2024; 103:117577. [PMID: 38518735 DOI: 10.1016/j.bmc.2023.117577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 12/21/2023] [Accepted: 12/22/2023] [Indexed: 03/24/2024]
Abstract
Small-molecule antivirals that prevent the replication of the SARS-CoV-2 virus by blocking the enzymatic activity of its main protease (Mpro) are and will be a tenet of pandemic preparedness. However, the peptidic nature of such compounds often precludes the design of compounds within favorable physical property ranges, limiting cellular activity. Here we describe the discovery of peptide aldehyde Mpro inhibitors with potent enzymatic and cellular antiviral activity. This structure-activity relationship (SAR) exploration was guided by the use of calculated hydration site thermodynamic maps (WaterMap) to drive potency via displacement of waters from high-energy sites. Thousands of diverse compounds were designed to target these high-energy hydration sites and then prioritized for synthesis by physics- and structure-based Free-Energy Perturbation (FEP+) simulations, which accurately predicted biochemical potencies. This approach ultimately led to the rapid discovery of lead compounds with unique SAR that exhibited potent enzymatic and cellular activity with excellent pan-coronavirus coverage.
Collapse
Affiliation(s)
- Daniel W Carney
- Takeda Development Center Americas, Inc, 9625 Towne Centre Drive, San Diego, CA 92121, United States.
| | - Abba E Leffler
- Schrödinger, Inc, 1540 Broadway, New York, NY 10036, United States.
| | - Jeffrey A Bell
- Schrödinger, Inc, 1540 Broadway, New York, NY 10036, United States
| | | | - Cecilia Cheng
- Schrödinger, Inc, 9868 Scranton Road, Suite 3200, San Diego, CA 92121, United States
| | - Edcon Chang
- Takeda Development Center Americas, Inc, 9625 Towne Centre Drive, San Diego, CA 92121, United States
| | - Adam Dornford
- Schrödinger, Inc, 1 Main St, 11th Floor, Cambridge, MA 02142, United States
| | - Douglas R Dougan
- Takeda Development Center Americas, Inc, 9625 Towne Centre Drive, San Diego, CA 92121, United States
| | - Leah L Frye
- Schrödinger, Inc, 101 SW Main Street, Suite 1300, Portland, OR 97204, United States
| | - Mary E Grimes
- Schrödinger, Inc, 101 SW Main Street, Suite 1300, Portland, OR 97204, United States
| | - Tim Knehans
- Schrödinger GmbH, Glücksteinallee 25, 68163 Mannheim, Germany
| | | | - Mallareddy Komandla
- Takeda Development Center Americas, Inc, 9625 Towne Centre Drive, San Diego, CA 92121, United States
| | - Weston Lane
- Treeline Biosciences, 500 Arsenal Way, Watertown, MA 02472, United States
| | - Hubert Li
- Schrödinger, Inc, 9868 Scranton Road, Suite 3200, San Diego, CA 92121, United States
| | - Sophia R Newman
- Schrödinger, Inc, 101 SW Main Street, Suite 1300, Portland, OR 97204, United States
| | - Katalin Phimister
- Schrödinger Technologies Limited, 1st Floor West, Davidson House, Forbury Square, Reading RG1 3EU, United Kingdom
| | - Kumar S Saikatendu
- Takeda Development Center Americas, Inc, 9625 Towne Centre Drive, San Diego, CA 92121, United States
| | - Hercules Silverstein
- Schrödinger, Inc, 101 SW Main Street, Suite 1300, Portland, OR 97204, United States
| | | |
Collapse
|
3
|
Janin YL. On the origins of SARS-CoV-2 main protease inhibitors. RSC Med Chem 2024; 15:81-118. [PMID: 38283212 PMCID: PMC10809347 DOI: 10.1039/d3md00493g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 10/13/2023] [Indexed: 01/30/2024] Open
Abstract
In order to address the world-wide health challenge caused by the COVID-19 pandemic, the 3CL protease/SARS-CoV-2 main protease (SARS-CoV-2-Mpro) coded by its nsp5 gene became one of the biochemical targets for the design of antiviral drugs. In less than 3 years of research, 4 inhibitors of SARS-CoV-2-Mpro have actually been authorized for COVID-19 treatment (nirmatrelvir, ensitrelvir, leritrelvir and simnotrelvir) and more such as EDP-235, FB-2001 and STI-1558/Olgotrelvir or five undisclosed compounds (CDI-988, ASC11, ALG-097558, QLS1128 and H-10517) are undergoing clinical trials. This review is an attempt to picture this quite unprecedented medicinal chemistry feat and provide insights on how these cysteine protease inhibitors were discovered. Since many series of covalent SARS-CoV-2-Mpro inhibitors owe some of their origins to previous work on other proteases, we first provided a description of various inhibitors of cysteine-bearing human caspase-1 or cathepsin K, as well as inhibitors of serine proteases such as human dipeptidyl peptidase-4 or the hepatitis C protein complex NS3/4A. This is then followed by a description of the results of the approaches adopted (repurposing, structure-based and high throughput screening) to discover coronavirus main protease inhibitors.
Collapse
Affiliation(s)
- Yves L Janin
- Structure et Instabilité des Génomes (StrInG), Muséum National d'Histoire Naturelle, INSERM, CNRS, Alliance Sorbonne Université 75005 Paris France
| |
Collapse
|
4
|
Li X, Song Y. Structure and function of SARS-CoV and SARS-CoV-2 main proteases and their inhibition: A comprehensive review. Eur J Med Chem 2023; 260:115772. [PMID: 37659195 PMCID: PMC10529944 DOI: 10.1016/j.ejmech.2023.115772] [Citation(s) in RCA: 34] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 08/24/2023] [Accepted: 08/26/2023] [Indexed: 09/04/2023]
Abstract
Severe acute respiratory syndrome-associated coronavirus (SARS-CoV) identified in 2003 infected ∼8000 people in 26 countries with 800 deaths, which was soon contained and eradicated by syndromic surveillance and enhanced quarantine. A closely related coronavirus SARS-CoV-2, the causative agent of COVID-19 identified in 2019, has been dramatically more contagious and catastrophic. It has infected and caused various flu-like symptoms of billions of people in >200 countries, including >6 million people died of or with the virus. Despite the availability of several vaccines and antiviral drugs against SARS-CoV-2, finding new therapeutics is needed because of viral evolution and a possible emerging coronavirus in the future. The main protease (Mpro) of these coronaviruses plays important roles in their life cycle and is essential for the viral replication. This article represents a comprehensive review of the function, structure and inhibition of SARS-CoV and -CoV-2 Mpro, including structure-activity relationships, protein-inhibitor interactions and clinical trial status.
Collapse
Affiliation(s)
- Xin Li
- Department of Pharmacology and Chemical Biology, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, USA; Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, USA.
| | - Yongcheng Song
- Department of Pharmacology and Chemical Biology, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, USA; Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, USA.
| |
Collapse
|
5
|
Wu C, Zhang H, Kong N, Wu B, Lin X, Wang H. Dynamic Control of Cyclic Peptide Assembly to Form Higher-Order Assemblies. Angew Chem Int Ed Engl 2023; 62:e202303455. [PMID: 37409642 DOI: 10.1002/anie.202303455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 07/02/2023] [Accepted: 07/05/2023] [Indexed: 07/07/2023]
Abstract
Chirality correction, asymmetry, ring-chain tautomerism and hierarchical assemblies are fundamental phenomena in nature. They are geometrically related and may impact the biological roles of a protein or other supermolecules. It is challenging to study those behaviors within an artificial system due to the complexity of displaying these features. Herein, we design an alternating D,L peptide to recreate and validate the naturally occurring chirality inversion prior to cyclization in water. The resulting asymmetrical cyclic peptide containing a 4-imidazolidinone ring provides an excellent platform to study the ring-chain tautomerism, thermostability and dynamic assembly of the nanostructures. Different from traditional cyclic D,L peptides, the formation of 4-imidazolidinone promotes the formation of intertwined nanostructures. Analysis of the nanostructures confirmed the left-handedness, representing chirality induced self-assembly. This proves that a rationally designed peptide can mimic multiple natural phenomena and could promote the development of functional biomaterials, catalysts, antibiotics, and supermolecules.
Collapse
Affiliation(s)
- Chongyang Wu
- Key Laboratory of Precise Synthesis of Functional Molecules of Zhejiang Province, Department of Chemistry, School of Science, Westlake University, Institute of Natural Sciences, Westlake Institute for Advanced Study, No. 600 Dunyu Road, Hangzhou, 310024, Zhejiang Province, China
| | - Hongyue Zhang
- Key Laboratory of Precise Synthesis of Functional Molecules of Zhejiang Province, Department of Chemistry, School of Science, Westlake University, Institute of Natural Sciences, Westlake Institute for Advanced Study, No. 600 Dunyu Road, Hangzhou, 310024, Zhejiang Province, China
| | - Nan Kong
- Key Laboratory of Precise Synthesis of Functional Molecules of Zhejiang Province, Department of Chemistry, School of Science, Westlake University, Institute of Natural Sciences, Westlake Institute for Advanced Study, No. 600 Dunyu Road, Hangzhou, 310024, Zhejiang Province, China
| | - Bihan Wu
- Key Laboratory of Precise Synthesis of Functional Molecules of Zhejiang Province, Department of Chemistry, School of Science, Westlake University, Institute of Natural Sciences, Westlake Institute for Advanced Study, No. 600 Dunyu Road, Hangzhou, 310024, Zhejiang Province, China
| | - Xinhui Lin
- Key Laboratory of Precise Synthesis of Functional Molecules of Zhejiang Province, Department of Chemistry, School of Science, Westlake University, Institute of Natural Sciences, Westlake Institute for Advanced Study, No. 600 Dunyu Road, Hangzhou, 310024, Zhejiang Province, China
| | - Huaimin Wang
- Key Laboratory of Precise Synthesis of Functional Molecules of Zhejiang Province, Department of Chemistry, School of Science, Westlake University, Institute of Natural Sciences, Westlake Institute for Advanced Study, No. 600 Dunyu Road, Hangzhou, 310024, Zhejiang Province, China
- Westlake Laboratory of Life Sciences and Biomedicine, School of Life Sciences, Westlake University, Hangzhou, 310024, Zhejiang, China
| |
Collapse
|
6
|
Bugatti K. A Brief Guide to Preparing a Peptide-Drug Conjugate. Chembiochem 2023; 24:e202300254. [PMID: 37288718 DOI: 10.1002/cbic.202300254] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 06/06/2023] [Accepted: 06/08/2023] [Indexed: 06/09/2023]
Abstract
Peptide-drug conjugates (PDCs) have recently emerged as interesting hybrid constructs not only for targeted therapy, but also for the early diagnosis of different pathologies. In most cases, the crucial step in the PDC synthesis is the final conjugation step, where a specific drug is bound to a particular peptide-/peptidomimetic-targeting unit. Thus, this concept paper aims to give a short guide to determining the finest conjugation reaction, by considering in particular the reaction conditions, the stability of the linker and the major pros and cons of each reaction. Based on the recent PDCs reported in literature, the most common and efficient conjugation methods will be systematically presented and compared, generating a short guide to consult while planning the synthesis of a novel peptide-drug conjugate.
Collapse
Affiliation(s)
- Kelly Bugatti
- Department of Food and Drug, University of Parma, Parco Area delle Scienze 27/A, 43124, Parma, Italy
| |
Collapse
|
7
|
Tang B, He F, Liu D, He F, Wu T, Fang M, Niu Z, Wu Z, Xu D. AI-Aided Design of Novel Targeted Covalent Inhibitors against SARS-CoV-2. Biomolecules 2022. [PMID: 35740872 DOI: 10.1101/2020.03.03.972133v1.full] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2023] Open
Abstract
The drug repurposing of known approved drugs (e.g., lopinavir/ritonavir) has failed to treat SARS-CoV-2-infected patients. Therefore, it is important to generate new chemical entities against this virus. As a critical enzyme in the lifecycle of the coronavirus, the 3C-like main protease (3CLpro or Mpro) is the most attractive target for antiviral drug design. Based on a recently solved structure (PDB ID: 6LU7), we developed a novel advanced deep Q-learning network with a fragment-based drug design (ADQN-FBDD) for generating potential lead compounds targeting SARS-CoV-2 3CLpro. We obtained a series of derivatives from the lead compounds based on our structure-based optimization policy (SBOP). All of the 47 lead compounds obtained directly with our AI model and related derivatives based on the SBOP are accessible in our molecular library. These compounds can be used as potential candidates by researchers to develop drugs against SARS-CoV-2.
Collapse
Affiliation(s)
- Bowen Tang
- Department of Electrical Engineering and Computer Science, Informatics Institute, Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361000, China
- MindRank AI Ltd., Hangzhou 310000, China
| | - Fengming He
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361000, China
| | - Dongpeng Liu
- Department of Electrical Engineering and Computer Science, Informatics Institute, Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA
| | - Fei He
- Department of Electrical Engineering and Computer Science, Informatics Institute, Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA
- School of Information Science and Technology, Northeast Normal University, Changchun 130117, China
| | - Tong Wu
- Department of Electrical Engineering and Computer Science, Informatics Institute, Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA
- Department of Epidemiology and Statistics, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing 100006, China
| | - Meijuan Fang
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361000, China
| | | | - Zhen Wu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361000, China
| | - Dong Xu
- Department of Electrical Engineering and Computer Science, Informatics Institute, Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA
| |
Collapse
|
8
|
Tang B, He F, Liu D, He F, Wu T, Fang M, Niu Z, Wu Z, Xu D. AI-Aided Design of Novel Targeted Covalent Inhibitors against SARS-CoV-2. Biomolecules 2022; 12:746. [PMID: 35740872 PMCID: PMC9220321 DOI: 10.3390/biom12060746] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 05/17/2022] [Accepted: 05/20/2022] [Indexed: 02/04/2023] Open
Abstract
The drug repurposing of known approved drugs (e.g., lopinavir/ritonavir) has failed to treat SARS-CoV-2-infected patients. Therefore, it is important to generate new chemical entities against this virus. As a critical enzyme in the lifecycle of the coronavirus, the 3C-like main protease (3CLpro or Mpro) is the most attractive target for antiviral drug design. Based on a recently solved structure (PDB ID: 6LU7), we developed a novel advanced deep Q-learning network with a fragment-based drug design (ADQN-FBDD) for generating potential lead compounds targeting SARS-CoV-2 3CLpro. We obtained a series of derivatives from the lead compounds based on our structure-based optimization policy (SBOP). All of the 47 lead compounds obtained directly with our AI model and related derivatives based on the SBOP are accessible in our molecular library. These compounds can be used as potential candidates by researchers to develop drugs against SARS-CoV-2.
Collapse
Affiliation(s)
- Bowen Tang
- Department of Electrical Engineering and Computer Science, Informatics Institute, Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA; (B.T.); (D.L.); (F.H.); (T.W.)
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361000, China; (F.H.); (M.F.)
- MindRank AI Ltd., Hangzhou 310000, China;
| | - Fengming He
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361000, China; (F.H.); (M.F.)
| | - Dongpeng Liu
- Department of Electrical Engineering and Computer Science, Informatics Institute, Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA; (B.T.); (D.L.); (F.H.); (T.W.)
| | - Fei He
- Department of Electrical Engineering and Computer Science, Informatics Institute, Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA; (B.T.); (D.L.); (F.H.); (T.W.)
- School of Information Science and Technology, Northeast Normal University, Changchun 130117, China
| | - Tong Wu
- Department of Electrical Engineering and Computer Science, Informatics Institute, Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA; (B.T.); (D.L.); (F.H.); (T.W.)
- Department of Epidemiology and Statistics, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing 100006, China
| | - Meijuan Fang
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361000, China; (F.H.); (M.F.)
| | | | - Zhen Wu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361000, China; (F.H.); (M.F.)
| | - Dong Xu
- Department of Electrical Engineering and Computer Science, Informatics Institute, Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA; (B.T.); (D.L.); (F.H.); (T.W.)
| |
Collapse
|
9
|
Zhu J, Zhang H, Lin Q, Lyu J, Lu L, Chen H, Zhang X, Zhang Y, Chen K. Progress on SARS-CoV-2 3CLpro Inhibitors: Inspiration from SARS-CoV 3CLpro Peptidomimetics and Small-Molecule Anti-Inflammatory Compounds. Drug Des Devel Ther 2022; 16:1067-1082. [PMID: 35450403 PMCID: PMC9015912 DOI: 10.2147/dddt.s359009] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 03/26/2022] [Indexed: 11/23/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) currently poses a threat to human health. 3C-like proteinase (3CLpro) plays an important role in the viral life cycle. Hence, it is considered an attractive antiviral target protein. Whole-genome sequencing showed that the sequence homology between SARS-CoV-2 3CLpro and SARS-CoV 3CLpro is 96.08%, with high similarity in the substrate-binding region. Thus, assessing peptidomimetic inhibitors of SARS-CoV 3CLpro could accelerate the development of peptidomimetic inhibitors for SARS-CoV-2 3CLpro. Accordingly, we herein discuss progress on SARS-CoV-2 3CLpro peptidomimetic inhibitors. Inflammation plays a major role in the pathophysiological process of COVID-19. Small-molecule compounds targeting 3CLpro with both antiviral and anti-inflammatory effects are also briefly discussed in this paper.
Collapse
Affiliation(s)
- Jiajie Zhu
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, People’s Republic of China
| | - Haiyan Zhang
- Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China
| | - Qinghong Lin
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, People’s Republic of China
| | - Jingting Lyu
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, People’s Republic of China
| | - Lu Lu
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, People’s Republic of China
| | - Hanxi Chen
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, People’s Republic of China
| | - Xuning Zhang
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, People’s Republic of China
| | - Yanjun Zhang
- Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, People’s Republic of China
| | - Keda Chen
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, People’s Republic of China
- Correspondence: Keda Chen, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, People’s Republic of China, Tel +8615068129828, Email ; Yanjun Zhang, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, People’s Republic of China, Tel +8613858115856, Fax +86057188280783, Email
| |
Collapse
|
10
|
Mirzaie S, Abdi F, GhavamiNejad A, Lu B, Wu XY. Covalent Antiviral Agents. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1322:285-312. [PMID: 34258745 DOI: 10.1007/978-981-16-0267-2_11] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
Nowadays, many viral infections have emerged and are taking a huge toll on human lives globally. Meanwhile, viral resistance to current drugs has drastically increased. Hence, there is a pressing need to design potent broad-spectrum antiviral agents to treat a variety of viral infections and overcome viral resistance. Covalent inhibitors have the potential to achieve both goals owing to their biochemical efficiency, prolonged duration of action, and the capability to inhibit shallow, solvent-exposed substrate-binding domains. In this chapter, we review the structures, activities, and inhibition mechanisms of covalent inhibitors against severe acute respiratory syndrome coronavirus 2, dengue virus, enterovirus, hepatitis C virus, human immunodeficiency virus, and influenza viruses. We also discuss the application of in silico study in covalent inhibitor design.
Collapse
Affiliation(s)
- Sako Mirzaie
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie L. Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada.
| | - Fatemeh Abdi
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie L. Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada
| | - Amin GhavamiNejad
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie L. Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada
| | - Brian Lu
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie L. Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada
| | - Xiao Yu Wu
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie L. Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
11
|
Tauber C, Wamser R, Arkona C, Tügend M, Abdul Aziz UB, Pach S, Schulz R, Jochmans D, Wolber G, Neyts J, Rademann J. Chemische Evolution antiviraler Wirkstoffe gegen Enterovirus D68 durch Proteintemplat‐gesteuerte Knoevenagelreaktionen. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202102074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Carolin Tauber
- Fachbereich Biologie, Chemie and Pharmazie Institut für Pharmazie Medizinische Chemie Freie Universität Berlin Königin-Luise-Str. 2+4 14195 Berlin Deutschland
| | - Rebekka Wamser
- Fachbereich Biologie, Chemie and Pharmazie Institut für Pharmazie Medizinische Chemie Freie Universität Berlin Königin-Luise-Str. 2+4 14195 Berlin Deutschland
| | - Christoph Arkona
- Fachbereich Biologie, Chemie and Pharmazie Institut für Pharmazie Medizinische Chemie Freie Universität Berlin Königin-Luise-Str. 2+4 14195 Berlin Deutschland
| | - Marisa Tügend
- Fachbereich Biologie, Chemie and Pharmazie Institut für Pharmazie Medizinische Chemie Freie Universität Berlin Königin-Luise-Str. 2+4 14195 Berlin Deutschland
| | - Umer Bin Abdul Aziz
- Fachbereich Biologie, Chemie and Pharmazie Institut für Pharmazie Medizinische Chemie Freie Universität Berlin Königin-Luise-Str. 2+4 14195 Berlin Deutschland
| | - Szymon Pach
- Fachbereich Biologie, Chemie and Pharmazie Institut für Pharmazie Medizinische Chemie Freie Universität Berlin Königin-Luise-Str. 2+4 14195 Berlin Deutschland
| | - Robert Schulz
- Fachbereich Biologie, Chemie and Pharmazie Institut für Pharmazie Medizinische Chemie Freie Universität Berlin Königin-Luise-Str. 2+4 14195 Berlin Deutschland
| | - Dirk Jochmans
- Department of Microbiology, Immunology and Transplantation Rega Institute KU Leuven Leuven Belgien
| | - Gerhard Wolber
- Fachbereich Biologie, Chemie and Pharmazie Institut für Pharmazie Medizinische Chemie Freie Universität Berlin Königin-Luise-Str. 2+4 14195 Berlin Deutschland
| | - Johan Neyts
- Department of Microbiology, Immunology and Transplantation Rega Institute KU Leuven Leuven Belgien
| | - Jörg Rademann
- Fachbereich Biologie, Chemie and Pharmazie Institut für Pharmazie Medizinische Chemie Freie Universität Berlin Königin-Luise-Str. 2+4 14195 Berlin Deutschland
| |
Collapse
|
12
|
Tauber C, Wamser R, Arkona C, Tügend M, Abdul Aziz UB, Pach S, Schulz R, Jochmans D, Wolber G, Neyts J, Rademann J. Chemical Evolution of Antivirals Against Enterovirus D68 through Protein-Templated Knoevenagel Reactions. Angew Chem Int Ed Engl 2021; 60:13294-13301. [PMID: 33749121 PMCID: PMC8252737 DOI: 10.1002/anie.202102074] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Indexed: 02/06/2023]
Abstract
The generation of bioactive molecules from inactive precursors is a crucial step in the chemical evolution of life, however, mechanistic insights into this aspect of abiogenesis are scarce. Here, we investigate the protein-catalyzed formation of antivirals by the 3C-protease of enterovirus D68. The enzyme induces aldol condensations yielding inhibitors with antiviral activity in cells. Kinetic and thermodynamic analyses reveal that the bioactivity emerges from a dynamic reaction system including inhibitor formation, alkylation of the protein target by the inhibitors, and competitive addition of non-protein nucleophiles to the inhibitors. The most active antivirals are slowly reversible inhibitors with elongated target residence times. The study reveals first examples for the chemical evolution of bio-actives through protein-catalyzed, non-enzymatic C-C couplings. The discovered mechanism works under physiological conditions and might constitute a native process of drug development.
Collapse
Affiliation(s)
- Carolin Tauber
- Department of Biology, Chemistry and PharmacyInstitute of PharmacyMedicinal ChemistryFreie Universität BerlinKönigin-Luise-Str. 2+414195BerlinGermany
| | - Rebekka Wamser
- Department of Biology, Chemistry and PharmacyInstitute of PharmacyMedicinal ChemistryFreie Universität BerlinKönigin-Luise-Str. 2+414195BerlinGermany
| | - Christoph Arkona
- Department of Biology, Chemistry and PharmacyInstitute of PharmacyMedicinal ChemistryFreie Universität BerlinKönigin-Luise-Str. 2+414195BerlinGermany
| | - Marisa Tügend
- Department of Biology, Chemistry and PharmacyInstitute of PharmacyMedicinal ChemistryFreie Universität BerlinKönigin-Luise-Str. 2+414195BerlinGermany
| | - Umer Bin Abdul Aziz
- Department of Biology, Chemistry and PharmacyInstitute of PharmacyMedicinal ChemistryFreie Universität BerlinKönigin-Luise-Str. 2+414195BerlinGermany
| | - Szymon Pach
- Department of Biology, Chemistry and PharmacyInstitute of PharmacyMedicinal ChemistryFreie Universität BerlinKönigin-Luise-Str. 2+414195BerlinGermany
| | - Robert Schulz
- Department of Biology, Chemistry and PharmacyInstitute of PharmacyMedicinal ChemistryFreie Universität BerlinKönigin-Luise-Str. 2+414195BerlinGermany
| | - Dirk Jochmans
- Department of Microbiology, Immunology and TransplantationRega InstituteKU LeuvenLeuvenBelgium
| | - Gerhard Wolber
- Department of Biology, Chemistry and PharmacyInstitute of PharmacyMedicinal ChemistryFreie Universität BerlinKönigin-Luise-Str. 2+414195BerlinGermany
| | - Johan Neyts
- Department of Microbiology, Immunology and TransplantationRega InstituteKU LeuvenLeuvenBelgium
| | - Jörg Rademann
- Department of Biology, Chemistry and PharmacyInstitute of PharmacyMedicinal ChemistryFreie Universität BerlinKönigin-Luise-Str. 2+414195BerlinGermany
| |
Collapse
|
13
|
CAVIAR: a method for automatic cavity detection, description and decomposition into subcavities. J Comput Aided Mol Des 2021; 35:737-750. [PMID: 34050420 DOI: 10.1007/s10822-021-00390-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 05/11/2021] [Indexed: 10/21/2022]
Abstract
The accurate description of protein binding sites is essential to the determination of similarity and the application of machine learning methods to relate the binding sites to observed functions. This work describes CAVIAR, a new open source tool for generating descriptors for binding sites, using protein structures in PDB and mmCIF format as well as trajectory frames from molecular dynamics simulations as input. The applicability of CAVIAR descriptors is showcased by computing machine learning predictions of binding site ligandability. The method can also automatically assign subcavities, even in the absence of a bound ligand. The defined subpockets mimic the empirical definitions used in medicinal chemistry projects. It is shown that the experimental binding affinity scales relatively well with the number of subcavities filled by the ligand, with compounds binding to more than three subcavities having nanomolar or better affinities to the target. The CAVIAR descriptors and methods can be used in any machine learning-based investigations of problems involving binding sites, from protein engineering to hit identification. The full software code is available on GitHub and a conda package is hosted on Anaconda cloud.
Collapse
|
14
|
Citarella A, Scala A, Piperno A, Micale N. SARS-CoV-2 M pro: A Potential Target for Peptidomimetics and Small-Molecule Inhibitors. Biomolecules 2021; 11:607. [PMID: 33921886 PMCID: PMC8073203 DOI: 10.3390/biom11040607] [Citation(s) in RCA: 94] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 04/13/2021] [Accepted: 04/16/2021] [Indexed: 12/17/2022] Open
Abstract
The uncontrolled spread of the COVID-19 pandemic caused by the new coronavirus SARS-CoV-2 during 2020-2021 is one of the most devastating events in the history, with remarkable impacts on the health, economic systems, and habits of the entire world population. While some effective vaccines are nowadays approved and extensively administered, the long-term efficacy and safety of this line of intervention is constantly under debate as coronaviruses rapidly mutate and several SARS-CoV-2 variants have been already identified worldwide. Then, the WHO's main recommendations to prevent severe clinical complications by COVID-19 are still essentially based on social distancing and limitation of human interactions, therefore the identification of new target-based drugs became a priority. Several strategies have been proposed to counteract such viral infection, including the repurposing of FDA already approved for the treatment of HIV, HCV, and EBOLA, inter alia. Among the evaluated compounds, inhibitors of the main protease of the coronavirus (Mpro) are becoming more and more promising candidates. Mpro holds a pivotal role during the onset of the infection and its function is intimately related with the beginning of viral replication. The interruption of its catalytic activity could represent a relevant strategy for the development of anti-coronavirus drugs. SARS-CoV-2 Mpro is a peculiar cysteine protease of the coronavirus family, responsible for the replication and infectivity of the parasite. This review offers a detailed analysis of the repurposed drugs and the newly synthesized molecules developed to date for the treatment of COVID-19 which share the common feature of targeting SARS-CoV-2 Mpro, as well as a brief overview of the main enzymatic and cell-based assays to efficaciously screen such compounds.
Collapse
Affiliation(s)
| | | | | | - Nicola Micale
- Department of Chemical, Biological, Pharmaceutical, and Environmental Sciences, University of Messina, 98166 Messina, Italy; (A.C.); (A.S.); (A.P.)
| |
Collapse
|
15
|
Ullrich S, Nitsche C. The SARS-CoV-2 main protease as drug target. Bioorg Med Chem Lett 2020; 30:127377. [PMID: 32738988 PMCID: PMC7331567 DOI: 10.1016/j.bmcl.2020.127377] [Citation(s) in RCA: 461] [Impact Index Per Article: 115.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 06/27/2020] [Accepted: 06/29/2020] [Indexed: 01/08/2023]
Abstract
The unprecedented pandemic of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is threatening global health. The virus emerged in late 2019 and can cause a severe disease associated with significant mortality. Several vaccine development and drug discovery campaigns are underway. The SARS-CoV-2 main protease is considered a promising drug target, as it is dissimilar to human proteases. Sequence and structure of the main protease are closely related to those from other betacoronaviruses, facilitating drug discovery attempts based on previous lead compounds. Covalently binding peptidomimetics and small molecules are investigated. Various compounds show antiviral activity in infected human cells.
Collapse
Affiliation(s)
- Sven Ullrich
- Research School of Chemistry, Australian National University, Canberra, ACT 2601, Australia
| | - Christoph Nitsche
- Research School of Chemistry, Australian National University, Canberra, ACT 2601, Australia.
| |
Collapse
|
16
|
Hosseini-Zare MS, Thilagavathi R, Selvam C. Targeting severe acute respiratory syndrome-coronavirus (SARS-CoV-1) with structurally diverse inhibitors: a comprehensive review. RSC Adv 2020; 10:28287-28299. [PMID: 35519094 PMCID: PMC9055768 DOI: 10.1039/d0ra04395h] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Accepted: 07/13/2020] [Indexed: 12/13/2022] Open
Abstract
Coronaviruses, which were discovered in 1968, can lead to some human viral disorders, like severe acute respiratory syndrome (SARS), Middle East respiratory syndrome-related (MERS), and, recently, coronavirus disease 2019 (COVID-19). The coronavirus that leads to COVID-19 is rapidly spreading all over the world and is the reason for the deaths of thousands of people. Recent research has revealed that there is about 80% sequence homology between the coronaviruses that cause SARS and COVID-19. Considering this fact, we decided to collect the maximum available information on targets, structures, and inhibitors reported so far for SARS-CoV-1 that could be useful for researchers who work on closely related COVID-19. There are vital proteases, like papain-like protease 2 (PL2pro) and 3C-like protease (3CLpro), or main protease (Mpro), that are involved in and are essential for the replication of SARS coronavirus and so are valuable targets for the treatment of patients affected by this type of virus. SARS-CoV-1 NTPase/helicase plays an important role in the release of several non-structural proteins (nsps), so it is another essential target relating to the viral life cycle. In this paper, we provide extensive information about diverse molecules with anti-SARS activity. In addition to traditional medicinal chemistry outcomes, HTS, virtual screening efforts, and structural insights for better understanding inhibitors and SARS-CoV-1 target complexes are also discussed. This study covers a wide range of anti-SARS agents, particularly SARS-CoV-1 inhibitors, and provides new insights into drug design for the deadly SARS-CoV-2 virus.
Collapse
Affiliation(s)
- Maryam S Hosseini-Zare
- Department of Pharmaceutical and Environmental Health Sciences, College of Pharmacy and Health Sciences, Texas Southern University Houston TX-77004 USA +1-713-313-7552
| | - Ramasamy Thilagavathi
- Department of Biotechnology, Faculty of Engineering, Karpagam Academy of Higher Education Coimbatore India
| | - Chelliah Selvam
- Department of Pharmaceutical and Environmental Health Sciences, College of Pharmacy and Health Sciences, Texas Southern University Houston TX-77004 USA +1-713-313-7552
| |
Collapse
|
17
|
Eguida M, Rognan D. A Computer Vision Approach to Align and Compare Protein Cavities: Application to Fragment-Based Drug Design. J Med Chem 2020; 63:7127-7142. [DOI: 10.1021/acs.jmedchem.0c00422] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Merveille Eguida
- UMR 7200 CNRS-Université de Strasbourg, Laboratoire d’Innovation Thérapeutique, 67400 Illkirch, France
| | - Didier Rognan
- UMR 7200 CNRS-Université de Strasbourg, Laboratoire d’Innovation Thérapeutique, 67400 Illkirch, France
| |
Collapse
|
18
|
Tang B, He F, Liu D, Fang M, Wu Z, Xu D. AI-aided design of novel targeted covalent inhibitors against SARS-CoV-2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2020. [PMID: 32511346 DOI: 10.1101/2020.03.03.972133] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The focused drug repurposing of known approved drugs (such as lopinavir/ritonavir) has been reported failed for curing SARS-CoV-2 infected patients. It is urgent to generate new chemical entities against this virus. As a key enzyme in the life-cycle of coronavirus, the 3C-like main protease (3CL pro or M pro ) is the most attractive for antiviral drug design. Based on a recently solved structure (PDB ID: 6LU7), we developed a novel advanced deep Q-learning network with the fragment-based drug design (ADQN-FBDD) for generating potential lead compounds targeting SARS-CoV-2 3CL pro . We obtained a series of derivatives from those lead compounds by our structure-based optimization policy (SBOP). All the 47 lead compounds directly from our AI-model and related derivatives based on SBOP are accessible in our molecular library at https://github.com/tbwxmu/2019-nCov . These compounds can be used as potential candidates for researchers in their development of drugs against SARS-CoV-2.
Collapse
|
19
|
Ehrt C, Brinkjost T, Koch O. A benchmark driven guide to binding site comparison: An exhaustive evaluation using tailor-made data sets (ProSPECCTs). PLoS Comput Biol 2018; 14:e1006483. [PMID: 30408032 PMCID: PMC6224041 DOI: 10.1371/journal.pcbi.1006483] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Accepted: 09/02/2018] [Indexed: 11/24/2022] Open
Abstract
The automated comparison of protein-ligand binding sites provides useful insights into yet unexplored site similarities. Various stages of computational and chemical biology research can benefit from this knowledge. The search for putative off-targets and the establishment of polypharmacological effects by comparing binding sites led to promising results for numerous projects. Although many cavity comparison methods are available, a comprehensive analysis to guide the choice of a tool for a specific application is wanting. Moreover, the broad variety of binding site modeling approaches, comparison algorithms, and scoring metrics impedes this choice. Herein, we aim to elucidate strengths and weaknesses of binding site comparison methodologies. A detailed benchmark study is the only possibility to rationalize the selection of appropriate tools for different scenarios. Specific evaluation data sets were developed to shed light on multiple aspects of binding site comparison. An assembly of all applied benchmark sets (ProSPECCTs–Protein Site Pairs for the Evaluation of Cavity Comparison Tools) is made available for the evaluation and optimization of further and still emerging methods. The results indicate the importance of such analyses to facilitate the choice of a methodology that complies with the requirements of a specific scientific challenge. Binding site similarities are useful in the context of promiscuity prediction, drug repurposing, the analysis of protein-ligand and protein-protein complexes, function prediction, and further fields of general interest in chemical biology and biochemistry. Many years of research have led to the development of a multitude of methods for binding site analysis and comparison. On the one hand, their availability supports research. On the other hand, the huge number of methods hampers the efficient selection of a specific tool. Our research is dedicated to the analysis of different cavity comparison tools. We use several binding site data sets to establish guidelines which can be applied to ensure a successful application of comparison methods by circumventing potential pitfalls.
Collapse
Affiliation(s)
- Christiane Ehrt
- Faculty of Chemistry and Chemical Biology, TU Dortmund University, Dortmund, Germany
| | - Tobias Brinkjost
- Faculty of Chemistry and Chemical Biology, TU Dortmund University, Dortmund, Germany
- Department of Computer Science, TU Dortmund University, Dortmund, Germany
| | - Oliver Koch
- Faculty of Chemistry and Chemical Biology, TU Dortmund University, Dortmund, Germany
- * E-mail: ,
| |
Collapse
|
20
|
Pillaiyar T, Manickam M, Namasivayam V, Hayashi Y, Jung SH. An Overview of Severe Acute Respiratory Syndrome-Coronavirus (SARS-CoV) 3CL Protease Inhibitors: Peptidomimetics and Small Molecule Chemotherapy. J Med Chem 2016; 59:6595-628. [PMID: 26878082 PMCID: PMC7075650 DOI: 10.1021/acs.jmedchem.5b01461] [Citation(s) in RCA: 524] [Impact Index Per Article: 65.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2015] [Indexed: 01/17/2023]
Abstract
Severe acute respiratory syndrome (SARS) is caused by a newly emerged coronavirus that infected more than 8000 individuals and resulted in more than 800 (10-15%) fatalities in 2003. The causative agent of SARS has been identified as a novel human coronavirus (SARS-CoV), and its viral protease, SARS-CoV 3CL(pro), has been shown to be essential for replication and has hence been recognized as a potent drug target for SARS infection. Currently, there is no effective treatment for this epidemic despite the intensive research that has been undertaken since 2003 (over 3500 publications). This perspective focuses on the status of various efficacious anti-SARS-CoV 3CL(pro) chemotherapies discovered during the last 12 years (2003-2015) from all sources, including laboratory synthetic methods, natural products, and virtual screening. We describe here mainly peptidomimetic and small molecule inhibitors of SARS-CoV 3CL(pro). Attempts have been made to provide a complete description of the structural features and binding modes of these inhibitors under many conditions.
Collapse
Affiliation(s)
- Thanigaimalai Pillaiyar
- Pharmaceutical
Institute, Pharmaceutical Chemistry I, University
of Bonn, An der Immenburg
4, D-53121 Bonn, Germany
| | - Manoj Manickam
- College
of Pharmacy and Institute of Drug Research and Development, Chungnam National University, Daejeon 34134, South Korea
| | - Vigneshwaran Namasivayam
- Pharmaceutical
Institute, Pharmaceutical Chemistry I, University
of Bonn, An der Immenburg
4, D-53121 Bonn, Germany
| | - Yoshio Hayashi
- Department
of Medicinal Chemistry, Tokyo University
of Pharmacy and Life Sciences, Tokyo 192-0392, Japan
| | - Sang-Hun Jung
- College
of Pharmacy and Institute of Drug Research and Development, Chungnam National University, Daejeon 34134, South Korea
| |
Collapse
|
21
|
Ehrt C, Brinkjost T, Koch O. Impact of Binding Site Comparisons on Medicinal Chemistry and Rational Molecular Design. J Med Chem 2016; 59:4121-51. [PMID: 27046190 DOI: 10.1021/acs.jmedchem.6b00078] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Modern rational drug design not only deals with the search for ligands binding to interesting and promising validated targets but also aims to identify the function and ligands of yet uncharacterized proteins having impact on different diseases. Additionally, it contributes to the design of inhibitors with distinct selectivity patterns and the prediction of possible off-target effects. The identification of similarities between binding sites of various proteins is a useful approach to cope with those challenges. The main scope of this perspective is to describe applications of different protein binding site comparison approaches to outline their applicability and impact on molecular design. The article deals with various substantial application domains and provides some outstanding examples to show how various binding site comparison methods can be applied to promote in silico drug design workflows. In addition, we will also briefly introduce the fundamental principles of different protein binding site comparison methods.
Collapse
Affiliation(s)
- Christiane Ehrt
- Faculty of Chemistry and Chemical Biology, TU Dortmund University , Otto-Hahn-Straße 6, 44227 Dortmund, Germany
| | - Tobias Brinkjost
- Faculty of Chemistry and Chemical Biology, TU Dortmund University , Otto-Hahn-Straße 6, 44227 Dortmund, Germany.,Department of Computer Science, TU Dortmund University , Otto-Hahn-Straße 14, 44224 Dortmund, Germany
| | - Oliver Koch
- Faculty of Chemistry and Chemical Biology, TU Dortmund University , Otto-Hahn-Straße 6, 44227 Dortmund, Germany
| |
Collapse
|
22
|
Catalytic activation of pre-substrates via dynamic fragment assembly on protein templates. Nat Commun 2014; 5:5170. [PMID: 25403979 DOI: 10.1038/ncomms6170] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Accepted: 09/08/2014] [Indexed: 11/09/2022] Open
Abstract
Sensitive detection of small molecule fragments binding to defined sites of biomacromolecules is still a considerable challenge. Here we demonstrate that protein-binding fragments are able to induce enzymatic reactions on the protein surface via dynamic fragment ligation. Fragments binding to the S1 pocket of serine proteases containing a nitrogen, oxygen or sulphur nucleophile are found to activate electrophilic pre-substrates through a reversible, covalent ligation reaction. The dynamic ligation reaction positions the pre-substrate molecule at the active site of the protein thereby inducing its enzymatic cleavage. Catalytic activation of pre-substrates is confirmed by fluorescence spectroscopy and by high-performance liquid chromatography. The approach is investigated with 3 pre-substrates and 14 protein-binding fragments and the specific activation and the templating effect exerted by the enzyme is quantified for each protease-fragment-pre-substrate combination. The described approach enables the site-specific identification of protein-binding fragments, the functional characterization of enzymatic sites and the quantitative analysis of protein template-assisted ligation reactions.
Collapse
|
23
|
Hilgenfeld R. From SARS to MERS: crystallographic studies on coronaviral proteases enable antiviral drug design. FEBS J 2014; 281:4085-96. [PMID: 25039866 PMCID: PMC7163996 DOI: 10.1111/febs.12936] [Citation(s) in RCA: 451] [Impact Index Per Article: 45.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Revised: 07/07/2014] [Accepted: 07/15/2014] [Indexed: 01/08/2023]
Abstract
This review focuses on the important contributions that macromolecular crystallography has made over the past 12 years to elucidating structures and mechanisms of the essential proteases of coronaviruses, the main protease (M(pro) ) and the papain-like protease (PL(pro) ). The role of X-ray crystallography in structure-assisted drug discovery against these targets is discussed. Aspects dealt with in this review include the emergence of the SARS coronavirus in 2002-2003 and of the MERS coronavirus 10 years later and the origins of these viruses. The crystal structure of the free SARS coronavirus M(pro) and its dependence on pH is discussed, as are efforts to design inhibitors on the basis of these structures. The mechanism of maturation of the enzyme from the viral polyprotein is still a matter of debate. The crystal structure of the SARS coronavirus PL(pro) and its complex with ubiquitin is also discussed, as is its orthologue from MERS coronavirus. Efforts at predictive structure-based inhibitor development for bat coronavirus M(pro) s to increase the preparedness against zoonotic transmission to man are described as well. The paper closes with a brief discussion of structure-based discovery of antivirals in an academic setting.
Collapse
Affiliation(s)
- Rolf Hilgenfeld
- Institute of Biochemistry, Center for Structural and Cell Biology in Medicine, and German Center for Infection Research, University of Lübeck, Germany
| |
Collapse
|
24
|
Synthesis, modification and docking studies of 5-sulfonyl isatin derivatives as SARS-CoV 3C-like protease inhibitors. Bioorg Med Chem 2013; 22:292-302. [PMID: 24316352 PMCID: PMC7111328 DOI: 10.1016/j.bmc.2013.11.028] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Revised: 11/12/2013] [Accepted: 11/13/2013] [Indexed: 11/21/2022]
Abstract
The Severe Acute Respiratory Syndrome (SARS) is a serious life-threatening and strikingly mortal respiratory illness caused by SARS-CoV. SARS-CoV which contains a chymotrypsin-like main protease analogous to that of the main picornavirus protease, 3CL(pro). 3CL(pro) plays a pivotal role in the viral replication cycle and is a potential target for SARS inhibitor development. A series of isatin derivatives as possible SARS-CoV 3CL(pro) inhibitors was designed, synthesized, and evaluated by in vitro protease assay using fluorogenic substrate peptide, in which several showed potent inhibition against the 3CL(pro). Structure-activity relationship was analyzed, and possible binding interaction modes were proposed by molecular docking studies. Among all compounds, 8k₁ showed most potent inhibitory activity against 3CL(pro) (IC₅₀=1.04 μM). These results indicated that these inhibitors could be potentially developed into anti-SARS drugs.
Collapse
|
25
|
Fernández-Bachiller MI, Horatscheck A, Lisurek M, Rademann J. Alzheimer's disease: identification and development of β-secretase (BACE-1) binding fragments and inhibitors by dynamic ligation screening (DLS). ChemMedChem 2013; 8:1041-56. [PMID: 23757181 DOI: 10.1002/cmdc.201300078] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Revised: 04/25/2013] [Indexed: 11/12/2022]
Abstract
The application of dynamic ligation screening (DLS), a methodology for fragment-based drug discovery (FBDD), to the aspartic protease β-secretase (BACE-1) is reported. For this purpose, three new fluorescence resonance energy transfer (FRET) substrates were designed and synthesized. Their kinetic parameters (Vmax , KM , and kcat ) were determined and compared with a commercial substrate. Secondly, a peptide aldehyde was designed as a chemically reactive inhibitor (CRI) based on the Swedish mutation substrate sequence. Incubation of this CRI with the protease, a FRET substrate, and one amine per well taken from an amine library, which was assembled by a maximum common substructure (MCS) approach, revealed the fragment 3-(3-aminophenyl)-2H-chromen-2-one (1) to be a competitive BACE-1 inhibitor that enhanced the activity of the CRI. Irreversibly formed fragment combination products of 1 with the initial peptide sequence were active and confirmed the targeting of the active site through the ethane-1,2-diamine isostere. Finally, structure-assisted combination of fragment 1 with secondary fragments that target the S1 site in hit optimization yielded novel, entirely fragment-based BACE-1 inhibitors with up to 30-fold improved binding affinity. Interactions with the protein were explained by molecular modeling studies, which indicate that the new fragment combinations interact with the catalytic aspartic acid dyad, as well as with the adjacent binding sites required for potency.
Collapse
Affiliation(s)
- María Isabel Fernández-Bachiller
- Medicinal Chemistry Department, Leibniz Institut für Molekulare Pharmakologie (FMP) Campus Berlin-Buch, Robert-Rössle Str. 10, 13125 Berlin, Germany
| | | | | | | |
Collapse
|
26
|
Ede NJ, Hill J, Joy JK, Ede AM, Koppens ML. Solid-phase synthesis and screening of a library of C-terminal arginine peptide aldehydes against Murray Valley encephalitis virus protease. J Pept Sci 2012; 18:661-8. [PMID: 22991186 PMCID: PMC7167811 DOI: 10.1002/psc.2450] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2012] [Revised: 07/13/2012] [Accepted: 08/05/2012] [Indexed: 02/02/2023]
Abstract
Murray Valley encephalitis virus is a member of the flavivirus group, a large family of single‐stranded RNA viruses, which cause serious disease in all regions of the world. Unfortunately, no suitable antivirals are available, and there are commercial vaccines for only three flaviviruses. The solid‐phase synthesis of a library of 400 C‐terminal arginine peptide aldehydes and their screening against Murray Valley encephalitis virus protease are demonstrated. The library was utilised to elucidate several tripeptide sequences that can be used as inhibitors in further SAR studies. Copyright © 2012 European Peptide Society and John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Nicholas J Ede
- Consegna Group Ltd, 7/21 Northumberland St, Collingwood 3066, Victoria, Australia.
| | | | | | | | | |
Collapse
|
27
|
Saavedra CJ, Boto A, Hernández R. Preparation of modified peptides: direct conversion of α-amino acids into β-amino aldehydes. Org Biomol Chem 2012; 10:4448-61. [DOI: 10.1039/c2ob25433f] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
28
|
Akaji K, Konno H, Mitsui H, Teruya K, Shimamoto Y, Hattori Y, Ozaki T, Kusunoki M, Sanjoh A. Structure-based design, synthesis, and evaluation of peptide-mimetic SARS 3CL protease inhibitors. J Med Chem 2011; 54:7962-73. [PMID: 22014094 DOI: 10.1021/jm200870n] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The design and evaluation of low molecular weight peptide-based severe acute respiratory syndrome (SARS) chymotrypsin-like protease (3CL) protease inhibitors are described. A substrate-based peptide aldehyde was selected as a starting compound, and optimum side-chain structures were determined, based on a comparison of inhibitory activities with Michael type inhibitors. For the efficient screening of peptide aldehydes containing a specific C-terminal residue, a new approach employing thioacetal to aldehyde conversion mediated by N-bromosuccinimide was devised. Structural optimization was carried out based on X-ray crystallographic analyses of the R188I SARS 3CL protease in a complex with each inhibitor to provide a tetrapeptide aldehyde with an IC(50) value of 98 nM. The resulting compound carried no substrate sequence, except for a P(3) site directed toward the outside of the protease. X-ray crystallography provided insights into the protein-ligand interactions.
Collapse
Affiliation(s)
- Kenichi Akaji
- Department of Medicinal Chemistry, Kyoto Pharmaceutical University, Yamashina-ku, Kyoto 607-8412, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Zhu L, George S, Schmidt MF, Al-Gharabli SI, Rademann J, Hilgenfeld R. Peptide aldehyde inhibitors challenge the substrate specificity of the SARS-coronavirus main protease. Antiviral Res 2011; 92:204-12. [PMID: 21854807 PMCID: PMC7114241 DOI: 10.1016/j.antiviral.2011.08.001] [Citation(s) in RCA: 96] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2011] [Accepted: 08/03/2011] [Indexed: 02/08/2023]
Abstract
SARS coronavirus main protease (SARS-CoV M(pro)) is essential for the replication of the virus and regarded as a major antiviral drug target. The enzyme is a cysteine protease, with a catalytic dyad (Cys-145/His-41) in the active site. Aldehyde inhibitors can bind reversibly to the active-site sulfhydryl of SARS-CoV M(pro). Previous studies using peptidic substrates and inhibitors showed that the substrate specificity of SARS-CoV M(pro) requires glutamine in the P1 position and a large hydrophobic residue in the P2 position. We determined four crystal structures of SARS-CoV M(pro) in complex with pentapeptide aldehydes (Ac-ESTLQ-H, Ac-NSFSQ-H, Ac-DSFDQ-H, and Ac-NSTSQ-H). Kinetic data showed that all of these aldehydes exhibit inhibitory activity towards SARS-CoV M(pro), with K(i) values in the μM range. Surprisingly, the X-ray structures revealed that the hydrophobic S2 pocket of the enzyme can accommodate serine and even aspartic-acid side-chains in the P2 positions of the inhibitors. Consequently, we reassessed the substrate specificity of the enzyme by testing the cleavage of 20 different tetradecapeptide substrates with varying amino-acid residues in the P2 position. The cleavage efficiency for the substrate with serine in the P2 position was 160-times lower than that for the original substrate (P2=Leu); furthermore, the substrate with aspartic acid in the P2 position was not cleaved at all. We also determined a crystal structure of SARS-CoV M(pro) in complex with aldehyde Cm-FF-H, which has its P1-phenylalanine residue bound to the relatively hydrophilic S1 pocket of the enzyme and yet exhibits a high inhibitory activity against SARS-CoV M(pro), with K(i)=2.24±0.58 μM. These results show that the stringent substrate specificity of the SARS-CoV M(pro) with respect to the P1 and P2 positions can be overruled by the highly electrophilic character of the aldehyde warhead, thereby constituting a deviation from the dogma that peptidic inhibitors need to correspond to the observed cleavage specificity of the target protease.
Collapse
Affiliation(s)
- Lili Zhu
- Institute of Biochemistry, Center for Structural and Cell Biology in Medicine, University of Lübeck, Ratzeburger Allee 160, 23538 Lübeck, Germany
| | | | | | | | | | | |
Collapse
|
30
|
Structure-based antivirals for emerging and neglected RNA viruses: an emerging field for medicinal chemistry in academia. Future Med Chem 2011; 2:1061-7. [PMID: 21426155 DOI: 10.4155/fmc.10.211] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
"Many [neglected viruses] predominantly hit developing countries in tropical and subtropical regions of the world (40% of the world's population are now at risk of contracting dengue fever), but developed countries are by no means immune to their impact."
Collapse
|
31
|
Haebich D, Hillisch A, El Sheikh S. A practical total synthesis of the microbial alkaline proteinase inhibitor (MAPI). ChemMedChem 2009; 4:2054-9. [PMID: 19816894 DOI: 10.1002/cmdc.200900346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Diverse serine and cysteine proteases as well as alkaline proteinases and elastases play a crucial role in numerous biological processes. Natural peptide aldehydes such as the "microbial alkaline proteinase inhibitor" (MAPI, 1) are valuable tools to characterize novel enzymes and to study their function in nature. Within a drug discovery program we wanted to design and explore non-natural MAPI congeners with novel biological profiles. To that end we devised a simple, practical, and scalable synthesis of MAPI 1 from readily available amino acid building blocks. The modular nature of our approach allows convenient structural modification of the MAPI backbone.
Collapse
Affiliation(s)
- Dieter Haebich
- Bayer Schering Pharma AG, Medicinal Chemistry, 42096 Wuppertal, Germany.
| | | | | |
Collapse
|
32
|
Schmidt MF, Rademann J. Dynamic template-assisted strategies in fragment-based drug discovery. Trends Biotechnol 2009; 27:512-21. [PMID: 19679363 PMCID: PMC7114311 DOI: 10.1016/j.tibtech.2009.06.001] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2009] [Revised: 06/16/2009] [Accepted: 06/17/2009] [Indexed: 11/27/2022]
Abstract
Fragment-based methods for drug discovery are increasingly popular because they provide drug leads with greater ligand efficiency than conventional high-throughput screening. However, established methods for fragment detection do not address the central question in fragment-based ligand discovery: how can a primary ligand be optimally extended by a secondary fragment? Dynamic screening methods solve this issue by using a protein target as a template for ligand assembly, thus yielding high-affinity binders from low-affinity fragments. This review summarizes recent work on dynamic screening methodology, which resulted in the development of several high-affinity binders for various targets. Strengths and limitations of the published approaches are discussed and possible contributions of dynamic screening methodology to the drug discovery process are highlighted.
Collapse
Affiliation(s)
- Marco F Schmidt
- Department of Medicinal Chemistry, Leibniz-Institute of Molecular Pharmacology, Robert-Rössle-Strasse 10, 13125 Berlin, Germany
| | | |
Collapse
|
33
|
Lead Discovery Using Virtual Screening. TOPICS IN MEDICINAL CHEMISTRY 2009. [PMCID: PMC7176223 DOI: 10.1007/7355_2009_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The practice of virtual screening (VS) to identify chemical leads to known or novel targets is becoming a core function of the computational chemist within industry. By employing a range of techniques, when attempting to identify compounds with activity against a biological target, a small focused subset of a larger collection of compounds can be identified and tested, often with results much better than selecting a similar number of compounds at random. We will review the key methods available, their relative success, and provide practical insights into best practices and key gaps. We will also argue that the capability of VS methods has grown to a point where fuller integration with experimental methods, including HTS, could increase the effectiveness of both.
Collapse
|
34
|
Deceuninck A, Madder A. From DNA cross-linking to peptide labeling: on the versatility of the furan-oxidation–conjugation strategy. Chem Commun (Camb) 2009:340-2. [DOI: 10.1039/b817447d] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
35
|
Akaji K, Konno H, Onozuka M, Makino A, Saito H, Nosaka K. Evaluation of peptide-aldehyde inhibitors using R188I mutant of SARS 3CL protease as a proteolysis-resistant mutant. Bioorg Med Chem 2008; 16:9400-8. [PMID: 18845442 PMCID: PMC7126698 DOI: 10.1016/j.bmc.2008.09.057] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2008] [Revised: 09/22/2008] [Accepted: 09/23/2008] [Indexed: 12/01/2022]
Abstract
The 3C-like (3CL) protease of the severe acute respiratory syndrome (SARS) coronavirus is a key enzyme for the virus maturation. We found for the first time that the mature SARS 3CL protease is subject to degradation at 188Arg/189Gln. Replacing Arg with Ile at position 188 rendered the protease resistant to proteolysis. The R188I mutant digested a conserved undecapeptide substrate with a K(m) of 33.8 microM and k(cat) of 4753 s(-1). Compared with the value reported for the mature protease containing a C-terminal His-tag, the relative activity of the mutant was nearly 10(6). Novel peptide-aldehyde derivatives containing a side-chain-protected C-terminal Gln efficiently inhibited the catalytic activity of the R188I mutant. The results indicated for the first time that the tetrapeptide sequence is enough for inhibitory activities of peptide-aldehyde derivatives.
Collapse
Affiliation(s)
- Kenichi Akaji
- Department of Chemistry, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kita-ku, Kyoto 603-8334, Japan.
| | | | | | | | | | | |
Collapse
|
36
|
Verschueren KHG, Pumpor K, Anemüller S, Chen S, Mesters JR, Hilgenfeld R. A structural view of the inactivation of the SARS coronavirus main proteinase by benzotriazole esters. ACTA ACUST UNITED AC 2008; 15:597-606. [PMID: 18559270 PMCID: PMC7110992 DOI: 10.1016/j.chembiol.2008.04.011] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2007] [Revised: 04/16/2008] [Accepted: 04/17/2008] [Indexed: 01/04/2023]
Abstract
The main proteinase (Mpro) of the severe acute respiratory syndrome (SARS) coronavirus is a principal target for the design of anticoronaviral compounds. Benzotriazole esters have been reported as potent nonpeptidic inhibitors of the enzyme, but their exact mechanism of action remains unclear. Here we present crystal structures of SARS-CoV Mpro, the active-site cysteine of which has been acylated by benzotriazole esters that act as suicide inhibitors. In one of the structures, the thioester product has been hydrolyzed and benzoic acid is observed to bind to the hydrophobic S2 pocket. This structure also features the enzyme with a shortened N-terminal segment (“amputated N finger”). The results further the understanding of the important role of the N finger for catalysis as well as the design of benzotriazole inhibitors with improved specificity.
Collapse
Affiliation(s)
- Koen H G Verschueren
- Institute of Biochemistry, Center for Structural and Cell Biology in Medicine, University of Lübeck, Lübeck, Germany
| | | | | | | | | | | |
Collapse
|
37
|
Recent antiviral strategies against human coronavirus-related respiratory illnesses. Curr Opin Pulm Med 2008; 14:248-53. [PMID: 18427249 DOI: 10.1097/mcp.0b013e3282f7646f] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
PURPOSE OF REVIEW The main purpose of this review is to summarize the current research (2006-2007) concerning the development of novel anticoronaviral strategies and compounds. RECENT FINDINGS Recent research led to the identification of several novel agents inhibiting coronaviral replication. The most promising compounds include carbohydrate-binding agents, neutralizing antibodies and drugs targeting a coronaviral envelope protein. SUMMARY Although initial outbreaks of coronavirus that causes severe acute respiratory syndrome (SARS-CoV) were controlled by public health measures, the development of vaccines and antiviral agents for SARS-CoV is essential for improving control and treatment of future outbreaks. Four years after the SARS-CoV epidemic, several compounds with an anticoronaviral activity have been identified.
Collapse
|
38
|
Schmidt MF, Isidro-Llobet A, Lisurek M, El-Dahshan A, Tan J, Hilgenfeld R, Rademann J. Sensitized detection of inhibitory fragments and iterative development of non-peptidic protease inhibitors by dynamic ligation screening. Angew Chem Int Ed Engl 2008; 47:3275-8. [PMID: 18348134 PMCID: PMC7159527 DOI: 10.1002/anie.200704594] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- Marco Florian Schmidt
- Leibniz Institute for Molecular Pharmacology, Robert-Rössle-Strasse 10, 13125 Berlin, Germany
| | | | | | | | | | | | | |
Collapse
|
39
|
Current World Literature. Curr Opin Pulm Med 2008; 14:266-73. [DOI: 10.1097/mcp.0b013e3282ff8c19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
40
|
Schmidt M, Isidro-Llobet A, Lisurek M, El-Dahshan A, Tan J, Hilgenfeld R, Rademann J. Sensibilisierte Detektion inhibitorischer Fragmente und iterative Entwicklung nicht-peptidischer Proteaseinhibitoren durch dynamisches Ligationsscreening. Angew Chem Int Ed Engl 2008. [DOI: 10.1002/ange.200704594] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
41
|
Mukherjee P, Desai P, Ross L, White EL, Avery MA. Structure-based virtual screening against SARS-3CL(pro) to identify novel non-peptidic hits. Bioorg Med Chem 2008; 16:4138-49. [PMID: 18343121 PMCID: PMC7127700 DOI: 10.1016/j.bmc.2008.01.011] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2007] [Revised: 01/04/2008] [Accepted: 01/07/2008] [Indexed: 01/01/2023]
Abstract
Severe acute respiratory syndrome is a highly infectious upper respiratory tract disease caused by SARS-CoV, a previously unidentified human coronavirus. SARS-3CL(pro) is a viral cysteine protease critical to the pathogen's life cycle and hence a therapeutic target of importance. The recently elucidated crystal structures of this enzyme provide an opportunity for the discovery of inhibitors through rational drug design. In the current study, Gold docking program was utilized to conduct extensive docking studies against the target crystal structure to develop a robust and predictive docking protocol. The validated docking protocol was used to conduct a structure-based virtual screening of the Asinex Platinum collection. Biological evaluation of a screened selection of compounds was carried out to identify novel inhibitors of the viral protease.
Collapse
Affiliation(s)
- Prasenjit Mukherjee
- Department of Medicinal Chemistry, School of Pharmacy, University of Mississippi, Faser 417, University, MS 38677, USA
| | | | | | | | | |
Collapse
|
42
|
Shao Y, Yang W, Peng H, Hsu M, Tsai K, Kuo T, Wang AH, Liang P, Lin C, Yang A, Wong C. Structure-based design and synthesis of highly potent SARS-CoV 3CL protease inhibitors. Chembiochem 2008; 8:1654-7. [PMID: 17722121 PMCID: PMC7162026 DOI: 10.1002/cbic.200700254] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Affiliation(s)
- Yi‐Ming Shao
- Genomics Research Center, Academia Sinica, No. 128, Sec. 2, Academia Rd., Taipei City 115, Taiwan, R.O.C., Fax: (+886) 2‐2789‐8771
- Institute of Biological Chemistry, Academia Sinica, No. 128, Sec. 2, Academia Rd., Taipei City 115, Taiwan, R.O.C
- Institute of Biochemical Sciences, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei City 106, Taiwan, R.O.C
| | - Wen‐Bin Yang
- Genomics Research Center, Academia Sinica, No. 128, Sec. 2, Academia Rd., Taipei City 115, Taiwan, R.O.C., Fax: (+886) 2‐2789‐8771
| | - Hung‐Pin Peng
- Genomics Research Center, Academia Sinica, No. 128, Sec. 2, Academia Rd., Taipei City 115, Taiwan, R.O.C., Fax: (+886) 2‐2789‐8771
| | - Min‐Feng Hsu
- Institute of Biological Chemistry, Academia Sinica, No. 128, Sec. 2, Academia Rd., Taipei City 115, Taiwan, R.O.C
| | - Keng‐Chang Tsai
- Genomics Research Center, Academia Sinica, No. 128, Sec. 2, Academia Rd., Taipei City 115, Taiwan, R.O.C., Fax: (+886) 2‐2789‐8771
| | - Tun‐Hsun Kuo
- Institute of Biological Chemistry, Academia Sinica, No. 128, Sec. 2, Academia Rd., Taipei City 115, Taiwan, R.O.C
| | - Andrew H.‐J. Wang
- Genomics Research Center, Academia Sinica, No. 128, Sec. 2, Academia Rd., Taipei City 115, Taiwan, R.O.C., Fax: (+886) 2‐2789‐8771
- Institute of Biological Chemistry, Academia Sinica, No. 128, Sec. 2, Academia Rd., Taipei City 115, Taiwan, R.O.C
- Institute of Biochemical Sciences, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei City 106, Taiwan, R.O.C
| | - Po‐Huang Liang
- Genomics Research Center, Academia Sinica, No. 128, Sec. 2, Academia Rd., Taipei City 115, Taiwan, R.O.C., Fax: (+886) 2‐2789‐8771
- Institute of Biological Chemistry, Academia Sinica, No. 128, Sec. 2, Academia Rd., Taipei City 115, Taiwan, R.O.C
- Institute of Biochemical Sciences, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei City 106, Taiwan, R.O.C
| | - Chun‐Hung Lin
- Genomics Research Center, Academia Sinica, No. 128, Sec. 2, Academia Rd., Taipei City 115, Taiwan, R.O.C., Fax: (+886) 2‐2789‐8771
- Institute of Biological Chemistry, Academia Sinica, No. 128, Sec. 2, Academia Rd., Taipei City 115, Taiwan, R.O.C
- Institute of Biochemical Sciences, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei City 106, Taiwan, R.O.C
| | - An‐Suei Yang
- Genomics Research Center, Academia Sinica, No. 128, Sec. 2, Academia Rd., Taipei City 115, Taiwan, R.O.C., Fax: (+886) 2‐2789‐8771
| | - Chi‐Huey Wong
- Genomics Research Center, Academia Sinica, No. 128, Sec. 2, Academia Rd., Taipei City 115, Taiwan, R.O.C., Fax: (+886) 2‐2789‐8771
- Institute of Biological Chemistry, Academia Sinica, No. 128, Sec. 2, Academia Rd., Taipei City 115, Taiwan, R.O.C
- Institute of Biochemical Sciences, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei City 106, Taiwan, R.O.C
- Department of Chemistry, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| |
Collapse
|
43
|
Gerlach C, Münzel M, Baum B, Gerber HD, Craan T, Diederich W, Klebe G. KNOBLE: Ein wissensbasierter Ansatz zu Design und Synthese einfach herstellbarer Liganden als Ausgangspunkt für eine strukturbasierte Leitstrukturoptimierung. Angew Chem Int Ed Engl 2007. [DOI: 10.1002/ange.200703323] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
44
|
Gerlach C, Münzel M, Baum B, Gerber HD, Craan T, Diederich W, Klebe G. KNOBLE: A Knowledge-Based Approach for the Design and Synthesis of Readily Accessible Small-Molecule Chemical Probes To Test Protein Binding. Angew Chem Int Ed Engl 2007; 46:9105-9. [DOI: 10.1002/anie.200703323] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
45
|
Mesters JR, Tan J, Hilgenfeld R. Viral enzymes. Curr Opin Struct Biol 2006; 16:776-86. [PMID: 17085042 PMCID: PMC7127120 DOI: 10.1016/j.sbi.2006.10.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2006] [Revised: 10/16/2006] [Accepted: 10/24/2006] [Indexed: 01/09/2023]
Abstract
Viral genomes show unequalled diversity, ranging from single-stranded DNA to double-stranded RNA. Moreover, viruses can quickly adapt to the host's immune response and drug treatment. Although they tend to make optimal use of the host cell's reservoir of proteins, viruses need to carry some enzymatic functions with them, as they may not be available or accessible in the infected cell. Recently, progress has been made in our structural understanding of viral enzymes involved in all stages of the viral life cycle, which includes entry, hijack, replication and exit stages.
Collapse
Affiliation(s)
- Jeroen R Mesters
- Institute of Biochemistry, Center for Structural and Cell Biology in Medicine, University of Lübeck, Ratzeburger Allee 160, 23538 Lübeck, Germany
| | | | | |
Collapse
|
46
|
Kuhn D, Weskamp N, Schmitt S, Hüllermeier E, Klebe G. From the similarity analysis of protein cavities to the functional classification of protein families using cavbase. J Mol Biol 2006; 359:1023-44. [PMID: 16697007 PMCID: PMC7094329 DOI: 10.1016/j.jmb.2006.04.024] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2006] [Revised: 03/31/2006] [Accepted: 04/06/2006] [Indexed: 02/05/2023]
Abstract
In this contribution, the classification of protein binding sites using the physicochemical properties exposed to their pockets is presented. We recently introduced Cavbase, a method for describing and comparing protein binding pockets on the basis of the geometrical and physicochemical properties of their active sites. Here, we present algorithmic and methodological enhancements in the Cavbase property description and in the cavity comparison step. We give examples of the Cavbase similarity analysis detecting pronounced similarities in the binding sites of proteins unrelated in sequence. A similarity search using SARS M(pro) protease subpockets as queries retrieved ligands and ligand fragments accommodated in a physicochemical environment similar to that of the query. This allowed the characterization of the protease recognition pockets and the identification of molecular building blocks that can be incorporated into novel antiviral compounds. A cluster analysis procedure for the functional classification of binding pockets was implemented and calibrated using a diverse set of enzyme binding sites. Two relevant protein families, the alpha-carbonic anhydrases and the protein kinases, are used to demonstrate the scope of our cluster approach. We propose a relevant classification of both protein families, on the basis of the binding motifs in their active sites. The classification provides a new perspective on functional properties across a protein family and is able to highlight features important for potency and selectivity. Furthermore, this information can be used to identify possible cross-reactivities among proteins due to similarities in their binding sites.
Collapse
Key Words
- protein binding pockets
- classification of protein binding pockets
- cluster analysis of protein binding pockets
- protein kinases
- sars protease
- sam, s-adenosyl-methionine
- fad, flavine adenine dinucleotide
- sars, severe acute respiratory syndrome
- cov, coronavirus
- tgev, transmissible gastroenteritis virus
- ca, carbonic anhydrase
- cml, chronic myelogenous leukemia
- map, mitogen-activated protein kinases
- cdks, cyclin-dependent protein kinases
- hb, hydrogen bond
- rmsd, root-mean-square deviations
- upgma, unweighted pair group method with arithmetic mean
- ec, enzyme classification
Collapse
Affiliation(s)
- Daniel Kuhn
- Department of Pharmaceutical Chemistry, University of Marburg, Marbacher Weg 6, D-35032 Marburg, Germany
| | | | | | | | | |
Collapse
|