1
|
Atatreh N, Mahgoub RE, Ghattas MA. Exploring covalent inhibitors of SARS-CoV-2 main protease: from peptidomimetics to novel scaffolds. J Enzyme Inhib Med Chem 2025; 40:2460045. [PMID: 39912405 PMCID: PMC11803818 DOI: 10.1080/14756366.2025.2460045] [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: 07/30/2024] [Revised: 01/16/2025] [Accepted: 01/24/2025] [Indexed: 02/07/2025] Open
Abstract
Peptidomimetic inhibitors mimic natural peptide substrates, employing electrophilic warheads to covalently interact with the catalytic Cys145 of Mpro. Examples include aldehydes, α-ketoamides, and aza-peptides, with discussions on their mechanisms of action, potency, and structural insights. Non-peptidomimetic inhibitors utilise diverse scaffolds and mechanisms, achieving covalent modification of Mpro.
Collapse
Affiliation(s)
- Noor Atatreh
- College of Pharmacy, Al Ain University, Abu Dhabi, United Arab Emirates
- AAU Health and Biomedical Research Centre, Al Ain University, Abu Dhabi, United Arab Emirates
| | - Radwa E. Mahgoub
- College of Pharmacy, Al Ain University, Abu Dhabi, United Arab Emirates
- AAU Health and Biomedical Research Centre, Al Ain University, Abu Dhabi, United Arab Emirates
| | - Mohammad A. Ghattas
- College of Pharmacy, Al Ain University, Abu Dhabi, United Arab Emirates
- AAU Health and Biomedical Research Centre, Al Ain University, Abu Dhabi, United Arab Emirates
| |
Collapse
|
2
|
Laczi D, Huamán SS, Andrews-Clark T, Laidlaw SM, Salah E, Dumjahn L, Lukacik P, Choudhry H, Walsh MA, Carroll MW, Schofield CJ, Brewitz L. Silaproline-bearing nirmatrelvir derivatives are potent inhibitors of the SARS-CoV-2 main protease highlighting the value of silicon-derivatives in structure-activity-relationship studies. Eur J Med Chem 2025; 291:117603. [PMID: 40220677 DOI: 10.1016/j.ejmech.2025.117603] [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: 12/12/2024] [Revised: 03/31/2025] [Accepted: 04/03/2025] [Indexed: 04/14/2025]
Abstract
Nirmatrelvir is a substrate-related inhibitor of the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) main protease (Mpro) that is clinically used in combination with ritonavir to treat COVID-19. Derivatives of nirmatrelvir, modified at the substrate P2-equivalent position, have been developed to fine-tune inhibitor properties and are now in clinical use. We report the synthesis of nirmatrelvir derivatives with a (R)-4,4-dimethyl-4-silaproline (silaproline) group at the P2-equivalent position. Mass spectrometry (MS)-based assays demonstrate that silaproline-bearing nirmatrelvir derivatives efficiently inhibit isolated recombinant Mpro, albeit with reduced potency compared to nirmatrelvir. Investigations with SARS-CoV-2 infected VeroE6 cells reveal that the silaproline-bearing inhibitors with a CF3 group at the P4-equivalent position inhibit viral progression, implying that incorporating silicon atoms into Mpro inhibitors can yield in vivo active inhibitors with appropriate optimization. MS and crystallographic studies show that the nucleophilic active site cysteine residue of Mpro (Cys145) reacts with the nitrile group of the silaproline-bearing inhibitors. Substituting the electrophilic nitrile group for a non-activated terminal alkyne shifts the inhibition mode from reversible covalent inhibition to irreversible covalent inhibition. One of the two prochiral silaproline methyl groups occupies space in the S2 pocket that is unoccupied in Mpro:nirmatrelvir complex structures, highlighting the value of sila-derivatives in structure-activity-relationship (SAR) studies. The combined results highlight the potential of silicon-containing molecules for inhibition of Mpro and, by implication, other nucleophilic cysteine enzymes.
Collapse
Affiliation(s)
- Dóra Laczi
- Chemistry Research Laboratory, Department of Chemistry and the Ineos Oxford Institute for Antimicrobial Research, University of Oxford, 12 Mansfield Road, OX1 3TA, Oxford, UK
| | - Sofia Schönbauer Huamán
- Chemistry Research Laboratory, Department of Chemistry and the Ineos Oxford Institute for Antimicrobial Research, University of Oxford, 12 Mansfield Road, OX1 3TA, Oxford, UK
| | - Taylah Andrews-Clark
- Chemistry Research Laboratory, Department of Chemistry and the Ineos Oxford Institute for Antimicrobial Research, University of Oxford, 12 Mansfield Road, OX1 3TA, Oxford, UK; Diamond Light Source Ltd., Harwell Science and Innovation Campus, Didcot, OX11 0DE, UK; Research Complex at Harwell, Harwell Science and Innovation Campus, Didcot, OX11 0FA, UK
| | - Stephen M Laidlaw
- Centre for Human Genetics & Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, OX3 7BN, Oxford, UK
| | - Eidarus Salah
- Chemistry Research Laboratory, Department of Chemistry and the Ineos Oxford Institute for Antimicrobial Research, University of Oxford, 12 Mansfield Road, OX1 3TA, Oxford, UK
| | - Leo Dumjahn
- Chemistry Research Laboratory, Department of Chemistry and the Ineos Oxford Institute for Antimicrobial Research, University of Oxford, 12 Mansfield Road, OX1 3TA, Oxford, UK
| | - Petra Lukacik
- Diamond Light Source Ltd., Harwell Science and Innovation Campus, Didcot, OX11 0DE, UK; Research Complex at Harwell, Harwell Science and Innovation Campus, Didcot, OX11 0FA, UK
| | - Hani Choudhry
- Department of Biochemistry, Center for Artificial Intelligence in Precision Medicines, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Martin A Walsh
- Diamond Light Source Ltd., Harwell Science and Innovation Campus, Didcot, OX11 0DE, UK; Research Complex at Harwell, Harwell Science and Innovation Campus, Didcot, OX11 0FA, UK
| | - Miles W Carroll
- Centre for Human Genetics & Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, OX3 7BN, Oxford, UK
| | - Christopher J Schofield
- Chemistry Research Laboratory, Department of Chemistry and the Ineos Oxford Institute for Antimicrobial Research, University of Oxford, 12 Mansfield Road, OX1 3TA, Oxford, UK.
| | - Lennart Brewitz
- Chemistry Research Laboratory, Department of Chemistry and the Ineos Oxford Institute for Antimicrobial Research, University of Oxford, 12 Mansfield Road, OX1 3TA, Oxford, UK.
| |
Collapse
|
3
|
Yang Z, Wang K, Zhang G, Jiang Y, Zeng R, Qiao J, Li Y, Deng X, Xia Z, Yao R, Zeng X, Zhang L, Zhao Y, Lei J, Chen R. A deep learning model for structure-based bioactivity optimization and its application in the bioactivity optimization of a SARS-CoV-2 main protease inhibitor. Eur J Med Chem 2025; 291:117602. [PMID: 40239482 DOI: 10.1016/j.ejmech.2025.117602] [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: 11/19/2024] [Revised: 04/02/2025] [Accepted: 04/03/2025] [Indexed: 04/18/2025]
Abstract
Bioactivity optimization is a crucial and technical task in the early stages of drug discovery, traditionally carried out through iterative substituent optimization, a process that is often both time-consuming and expensive. To address this challenge, we present Pocket-StrMod, a deep-learning model tailored for structure-based bioactivity optimization. Pocket-StrMod employs an autoregressive flow-based architecture, optimizing molecules within a specific protein binding pocket while explicitly incorporating chemical expertise. It synchronously optimizes all substituents by generating atoms and covalent bonds at designated sites within a molecular scaffold nestled inside a protein pocket. We applied this model to optimize the bioactivity of Hit1, an inhibitor of the SARS-CoV-2 main protease (Mpro) with initially poor bioactivity (IC50 : 34.56 μM). Following two rounds of optimization, six compounds were selected for synthesis and bioactivity testing. This led to the discovery of C5, a potent compound with an IC50 value of 33.6 nM, marking a remarkable 1028-fold improvement over Hit1. Furthermore, C5 demonstrated promising in vitro antiviral activity against SARS-CoV-2. Collectively, these findings underscore the great potential of deep learning in facilitating rapid and cost-effective bioactivity optimization in the early phases of drug development.
Collapse
Affiliation(s)
- Zhenyu Yang
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Kai Wang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Guo Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yuanyuan Jiang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Rui Zeng
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Jingxin Qiao
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yueyue Li
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Xinyue Deng
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Ziyi Xia
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Rui Yao
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Xiaoxi Zeng
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Liyun Zhang
- Lead Generation Unit, HitGen Inc., Tianfu International Bio-Town, Shuangliu District, Chengdu, Sichuan, 610200, China
| | - Yi Zhao
- Key Laboratory of Intelligent Information Processing, Advanced Computer Research Center, Institute of Computing Technology, Chinese Academy of Sciences, Beijing, 100190, China.
| | - Jian Lei
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China; National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China.
| | - Runsheng Chen
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China; Key Laboratory of RNA Biology, Center for Big Data Research in Health, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.
| |
Collapse
|
4
|
Hu S, Zhong Q, Xie X, Zhang S, Wang J, Liu H, Dai W. Research progress on critical viral protease inhibitors for coronaviruses and enteroviruses. Bioorg Med Chem Lett 2025; 122:130168. [PMID: 40074013 DOI: 10.1016/j.bmcl.2025.130168] [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: 10/29/2024] [Revised: 03/01/2025] [Accepted: 03/04/2025] [Indexed: 03/14/2025]
Abstract
Viral infectious diseases have been seriously affecting human life and health. SARS-CoV-2 was the pathogen that caused Coronavirus Disease 2019 (COVID-19), and the impact of COVID-19 is still existing. Enterovirus 71 (EV71) is the primary pathogen of hand, foot, and mouth disease (HFMD), and no effective direct-acting antiviral drugs targeting EV71 has been approved yet. Innate antiviral strategies play an important role in preventing virus infections depending on the powerful immune regulatory system of body, while viruses have evolved to exploit diverse methods to overcome immune response. Viral proteases, which are known in cleaving viral polyproteins, have also been found to modulate the innate immunity of host cells, thereby promoting viral proliferation. Herein, we reviewed the current development of SARS-CoV-2 3CLpro, PLpro, and EV71 3Cpro and 2Apro, mainly including structure, function, modulation of immune response, and inhibitors of these four proteases, to further deepen the understanding of viral pathogenesis and provide a new perspective for subsequent corresponding drug development.
Collapse
Affiliation(s)
- Shulei Hu
- China Pharmaceutical University, 639 Longmian Avenue, Jiangning District, Nanjing 211198, China; State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China
| | - Qiuyu Zhong
- China Pharmaceutical University, 639 Longmian Avenue, Jiangning District, Nanjing 211198, China; State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China
| | - Xiong Xie
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China
| | - Shurui Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China
| | - Jinlin Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China
| | - Hong Liu
- China Pharmaceutical University, 639 Longmian Avenue, Jiangning District, Nanjing 211198, China; State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China; School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, UCAS, Hangzhou 310024, China.
| | - Wenhao Dai
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China; School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, UCAS, Hangzhou 310024, China.
| |
Collapse
|
5
|
Alli VJ, Singh SK, Darna M, Suresh V, Sule SA, Jangam A, Kattula B, Pusarapu SL, Thomas J, Sardana Y, Gundla KP, Burra AG, Chandra Sekhar K, Patnaik SS, Reddi B, Muralidharan K, Bokara KK, Addlagatta A, Jadav SS. Development of acylhydrazone linked thiazoles as non-covalent dual inhibitors of SARS-CoV-2 proteases. Eur J Med Chem 2025; 290:117509. [PMID: 40132498 DOI: 10.1016/j.ejmech.2025.117509] [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: 11/20/2024] [Revised: 03/10/2025] [Accepted: 03/11/2025] [Indexed: 03/27/2025]
Abstract
SARS-CoV-2's papain-like protease (PLPro) and main protease (MPro) are essential for viral maturation and replication. Currently, Paxlovid is recommended to treat viral infections, but the emergence of Nirmatrelvir resistance new variants poses serious global risks. Dual targeting agents restrict viral replication, act on other crucial viral pathways, or exert simultaneous protease inhibition, increasing the complexity for the virus to develop resistance, and the design of dual inhibitors is an attractive strategy. Herein, we present research on new thiazole-aryl and thiazole-ester compounds that function as cysteine specific non-covalent competitive dual inhibitors of SARS-CoV-2's papain-like protease (PLPro) and main protease (MPro). Twelve of the 36 compounds demonstrated dual inhibition in the range of nanomolar to low micromolar concentrations while five others exhibit selective PLPro inhibition. Minimal cytotoxicity against two mammalian cell lines and no oral toxicity in rats (LD50 > 2000 mg/kg) were observed. SARS-CoV-2 viral load was successfully reduced by several compounds tested. N-acyl hydrazone (NAH)-thiazole core while forms π-π interactions with the catalytic histidine side chain (H41) in MPro active site, it exhibits a series of hydrophobic and hydrophilic interaction in the interface of BL2 loop and the active site of PLPro. Non-covalent dual inhibition demonstrated by novel NAH-thiazole derivatives in this study provides a path for the development of efficient antiviral agents against coronaviruses.
Collapse
Affiliation(s)
- Vidya Jyothi Alli
- Department of Natural Products and Medicinal Chemistry, CSIR-Indian Institute of Chemical Technology, Hyderabad, 500007, India; Academy of Scientific and Innovative Research, Ghaziabad 201002, India
| | - Shubham Kumar Singh
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, 500007, India; Academy of Scientific and Innovative Research, Ghaziabad 201002, India
| | - Mounika Darna
- Department of Natural Products and Medicinal Chemistry, CSIR-Indian Institute of Chemical Technology, Hyderabad, 500007, India; Academy of Scientific and Innovative Research, Ghaziabad 201002, India
| | - Vavilapalli Suresh
- Department of Organic Synthesis &Process Chemistry, CSIR-Indian Institute of Chemical Technology, Hyderabad-500007, India; Academy of Scientific and Innovative Research, Ghaziabad 201002, India
| | - Swapnil Anil Sule
- Department of Natural Products and Medicinal Chemistry, CSIR-Indian Institute of Chemical Technology, Hyderabad, 500007, India; Academy of Scientific and Innovative Research, Ghaziabad 201002, India
| | - Aruna Jangam
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, 500007, India; Academy of Scientific and Innovative Research, Ghaziabad 201002, India
| | - Bhavita Kattula
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, 500007, India; Academy of Scientific and Innovative Research, Ghaziabad 201002, India
| | - Sarva Lakshmi Pusarapu
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, 500007, India
| | - Jessie Thomas
- CSIR-Centre for Cellular and Molecular Biology, ANNEXE II, Medical Biotechnology Complex, Hyderabad, 500007, India; Academy of Scientific and Innovative Research, Ghaziabad 201002, India
| | - Yogesh Sardana
- CSIR-Centre for Cellular and Molecular Biology, ANNEXE II, Medical Biotechnology Complex, Hyderabad, 500007, India; Academy of Scientific and Innovative Research, Ghaziabad 201002, India
| | - Krishna Prasad Gundla
- Department of Organic Synthesis &Process Chemistry, CSIR-Indian Institute of Chemical Technology, Hyderabad-500007, India
| | - Amarender Goud Burra
- Department of Organic Synthesis &Process Chemistry, CSIR-Indian Institute of Chemical Technology, Hyderabad-500007, India; Academy of Scientific and Innovative Research, Ghaziabad 201002, India
| | - Kunta Chandra Sekhar
- Department of Organic Synthesis &Process Chemistry, CSIR-Indian Institute of Chemical Technology, Hyderabad-500007, India; Academy of Scientific and Innovative Research, Ghaziabad 201002, India
| | - Samata Sai Patnaik
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, 500007, India; Academy of Scientific and Innovative Research, Ghaziabad 201002, India
| | - Bharati Reddi
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, 500007, India; Academy of Scientific and Innovative Research, Ghaziabad 201002, India
| | - Kathirvel Muralidharan
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, 500007, India; Academy of Scientific and Innovative Research, Ghaziabad 201002, India
| | - Kiran Kumar Bokara
- CSIR-Centre for Cellular and Molecular Biology, ANNEXE II, Medical Biotechnology Complex, Hyderabad, 500007, India; Academy of Scientific and Innovative Research, Ghaziabad 201002, India.
| | - Anthony Addlagatta
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, 500007, India; Academy of Scientific and Innovative Research, Ghaziabad 201002, India.
| | - Surender Singh Jadav
- Department of Natural Products and Medicinal Chemistry, CSIR-Indian Institute of Chemical Technology, Hyderabad, 500007, India; Academy of Scientific and Innovative Research, Ghaziabad 201002, India.
| |
Collapse
|
6
|
Ho WS, Yogarajah T, Mohameed JBH, Fong DY, Cheong DHJ, Wong YH, Chu JJH, Chai CLL. Mutagenesis-Guided Target Identification Reveals the Protein-Binding Domain of Nsp14 in Coronaviruses as the Target of a Labdane-Oxindole Compound. ACS Infect Dis 2025; 11:1153-1166. [PMID: 40207883 DOI: 10.1021/acsinfecdis.4c00965] [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] [Indexed: 04/11/2025]
Abstract
The non-structural protein (nsp) 14 of coronaviruses plays an important role in maintaining the genomic stability of the virus during viral replication. This had garnered significant interest towards the identification and development of inhibitors against nsp14, specifically its exoribonuclease (ExoN) domain. However, no inhibitors have been successfully developed to date. The bioactivity of the nsp14-ExoN is governed through a complex formation with its co-factor nsp10. This provides opportunities to target the protein assembly as an antiviral modality. In this study, a labdane-oxindole compound (OX18) was identified as a promising new antiviral agent against coronaviruses. Through a combination of FRET- and BRET-based approaches, OX18 was found to target the nsp10-binding domain of nsp14. A key escape mutation to OX18 in nsp14 was also identified in our study, albeit compromising its exoribonuclease activity. To our knowledge, OX18 is the first small molecule to target the nsp14/10 protein assembly. As such, our work paves the way for the development of future inhibitors of the nsp14-ExoN with increased potency and complexity.
Collapse
Affiliation(s)
- Wei Shen Ho
- Department of Pharmacy and Pharmaceutical Sciences, Faculty of Science, National University of Singapore, 18 Science Drive 4, Singapore 117543, Singapore
| | - Thinesshwary Yogarajah
- Laboratory of Molecular RNA Virology and Antiviral Strategies, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University Health System, National University of Singapore, MD4 Level 5, 5 Science Drive 2, Singapore 117597, Singapore
| | - Jasmaadiyah Binte Habib Mohameed
- Laboratory of Molecular RNA Virology and Antiviral Strategies, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University Health System, National University of Singapore, MD4 Level 5, 5 Science Drive 2, Singapore 117597, Singapore
| | - Deborah Yuhui Fong
- Laboratory of Molecular RNA Virology and Antiviral Strategies, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University Health System, National University of Singapore, MD4 Level 5, 5 Science Drive 2, Singapore 117597, Singapore
| | - Dorothy Hui Juan Cheong
- Laboratory of Molecular RNA Virology and Antiviral Strategies, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University Health System, National University of Singapore, MD4 Level 5, 5 Science Drive 2, Singapore 117597, Singapore
- Infectious Disease Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
- NUSMed Biosafety Level 3 Core Facility, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
| | - Yi Hao Wong
- Laboratory of Molecular RNA Virology and Antiviral Strategies, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University Health System, National University of Singapore, MD4 Level 5, 5 Science Drive 2, Singapore 117597, Singapore
- Infectious Disease Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
- NUSMed Biosafety Level 3 Core Facility, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
| | - Justin Jang Hann Chu
- Laboratory of Molecular RNA Virology and Antiviral Strategies, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University Health System, National University of Singapore, MD4 Level 5, 5 Science Drive 2, Singapore 117597, Singapore
- Infectious Disease Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
- NUSMed Biosafety Level 3 Core Facility, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Collaborative and Translation Unit for HFMD, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos #06-05, Singapore 138673, Singapore
| | - Christina Li Lin Chai
- Department of Pharmacy and Pharmaceutical Sciences, Faculty of Science, National University of Singapore, 18 Science Drive 4, Singapore 117543, Singapore
| |
Collapse
|
7
|
Yang Q, Huang X, Zhang H, Sun J, Tang J, Chen Z, Liu L, Liu M, Sun Z, Tang Z, Wei D, Wang D, Wang Y, Yan M, Zhao L, Zhu A, Zhong Y, Yang H, Zhao Y, Dai J, Shi Y, Huang B, Zhang W, Zhao J, Chen X, Rao Z, Peng W. Expanding the utilization of binding pockets proves to be effective for noncovalent small molecule inhibitors against SARS-CoV-2 M pro. Eur J Med Chem 2025; 289:117497. [PMID: 40090296 DOI: 10.1016/j.ejmech.2025.117497] [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: 02/05/2025] [Revised: 02/27/2025] [Accepted: 03/08/2025] [Indexed: 03/18/2025]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has resulted in millions of deaths and continues to pose serious threats to global public health. The main protease (Mpro) of SARS-CoV-2 is crucial for viral replication and its conservation, making it an attractive drug target. Here, we employed a structure-based drug design strategy to develop and optimize novel inhibitors targeting SARS-CoV-2 Mpro. By fully exploring occupation of the S1, S2, and S3/S4 binding pockets, we identified eight promising inhibitors with half-maximal inhibitory concentration (IC50) values below 20 nM. The cocrystal structure of Mpro with compound 10 highlighted the crucial roles of the interactions within the S3/S4 pockets in inhibitor potency enhancement. These findings demonstrated that expanding the utilization of these binding pockets was an effective strategy for developing noncovalent small molecule inhibitors that target SARS-CoV-2 Mpro. Compound 4 demonstrated outstanding in vitro antiviral activity against wild-type SARS-CoV-2 with an EC50 of 9.4 nM. Moreover, oral treatment with compounds 1 and 9 exhibited excellent antiviral potency and substantially ameliorated virus-induced tissue damage in the lungs of Omicron BA.5-infected K18-human ACE2 (K18-hACE2) transgenic mice, indicating that these novel noncovalent inhibitors could be potential oral agents for the treatment of COVID-19.
Collapse
Affiliation(s)
- Qi Yang
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China; Guangzhou National Laboratory, Guangzhou, 510005, China
| | - Xupeng Huang
- Guangzhou National Laboratory, Guangzhou, 510005, China; Innovative Center for Pathogen Research, Guangzhou National Laboratory, Guangzhou, 510005, China
| | - Hongbo Zhang
- Beijing StoneWise Technology Co. Ltd., Beijing, 100080, China
| | - Jing Sun
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China; The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 511436, China
| | - Jielin Tang
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China; Guangzhou National Laboratory, Guangzhou, 510005, China
| | - Zhao Chen
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China; The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 511436, China
| | - Lijie Liu
- Guangzhou National Laboratory, Guangzhou, 510005, China; Innovative Center for Pathogen Research, Guangzhou National Laboratory, Guangzhou, 510005, China; Guangzhou Medical University, Guangzhou, 511436, China
| | - Man Liu
- Guangzhou National Laboratory, Guangzhou, 510005, China; Innovative Center for Pathogen Research, Guangzhou National Laboratory, Guangzhou, 510005, China; The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 511436, China
| | - Zeyun Sun
- Guangzhou National Laboratory, Guangzhou, 510005, China; Innovative Center for Pathogen Research, Guangzhou National Laboratory, Guangzhou, 510005, China; Guangzhou Medical University, Guangzhou, 511436, China
| | - Zhenhao Tang
- Guangzhou National Laboratory, Guangzhou, 510005, China; Innovative Center for Pathogen Research, Guangzhou National Laboratory, Guangzhou, 510005, China
| | - Dandan Wei
- Guangzhou National Laboratory, Guangzhou, 510005, China; Innovative Center for Pathogen Research, Guangzhou National Laboratory, Guangzhou, 510005, China
| | - Dong Wang
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China; The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 511436, China
| | - Yiliang Wang
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China; Guangzhou National Laboratory, Guangzhou, 510005, China; The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 511436, China
| | - Mengrong Yan
- Guangzhou National Laboratory, Guangzhou, 510005, China; Innovative Center for Pathogen Research, Guangzhou National Laboratory, Guangzhou, 510005, China
| | - Li Zhao
- Guangzhou National Laboratory, Guangzhou, 510005, China; Innovative Center for Pathogen Research, Guangzhou National Laboratory, Guangzhou, 510005, China; Guangzhou Medical University, Guangzhou, 511436, China
| | - Airu Zhu
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China; The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 511436, China
| | - Yihang Zhong
- Guangzhou National Laboratory, Guangzhou, 510005, China; Innovative Center for Pathogen Research, Guangzhou National Laboratory, Guangzhou, 510005, China
| | - Haitao Yang
- Shanghai Institute for Advanced Immunochemical Studies and School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Yao Zhao
- National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, Shenzhen, 518112, China
| | - Jun Dai
- Technology Centre, Guangzhou Customs, Guangzhou, 510623, China
| | - Yongxia Shi
- Technology Centre, Guangzhou Customs, Guangzhou, 510623, China
| | - Bo Huang
- Beijing StoneWise Technology Co. Ltd., Beijing, 100080, China.
| | - Wei Zhang
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China; Guangzhou National Laboratory, Guangzhou, 510005, China; Innovative Center for Pathogen Research, Guangzhou National Laboratory, Guangzhou, 510005, China.
| | - Jincun Zhao
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China; Guangzhou National Laboratory, Guangzhou, 510005, China; The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 511436, China.
| | - Xinwen Chen
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China; Guangzhou National Laboratory, Guangzhou, 510005, China; The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 511436, China.
| | - Zihe Rao
- Guangzhou National Laboratory, Guangzhou, 510005, China; Innovative Center for Pathogen Research, Guangzhou National Laboratory, Guangzhou, 510005, China; Laboratory of Structural Biology, School of Life Sciences and School of Medicine, Tsinghua University, Beijing, 100084, China; Shanghai Institute for Advanced Immunochemical Studies and School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China.
| | - Wei Peng
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China; Guangzhou National Laboratory, Guangzhou, 510005, China; Innovative Center for Pathogen Research, Guangzhou National Laboratory, Guangzhou, 510005, China; The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 511436, China; University of South China, Hengyang, 421001, China.
| |
Collapse
|
8
|
Zian D, Iaconis D, Nenci S, Crusco A, Tawde S, Sodano M, Vitalone R, Raje A, Palamini M, Carettoni D, Molteni A, Manelfi C, Tazzari V, Beccari AR, Malune P, Maloccu S, Paulis A, Corona A, Nieddu S, Coletti S, Scarabottolo L, Tramontano E, Esposito F, Catalani M. The efficiency of high-throughput screening (HTS) and in-silico data analysis during medical emergencies: Identification of effective antiviral 3CLpro inhibitors. Antiviral Res 2025; 237:106119. [PMID: 39978553 DOI: 10.1016/j.antiviral.2025.106119] [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: 11/18/2024] [Revised: 02/04/2025] [Accepted: 02/15/2025] [Indexed: 02/22/2025]
Abstract
The COVID-19 pandemic highlighted the importance of accelerating the drug discovery process. The 3-chymotrypsin-like protease (3CLpro) is a critical enzyme in the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) viral replication process and was quickly identified as a prime target for drug development. This study leverages High-Throughput Screening (HTS) to identify novel 3CLpro inhibitors. We screened a different library of 325,000 compounds, leading to the discovery of two new chemical scaffolds with selective inhibitory activity against 3CLpro. In-silico analysis and further experimental validation, elucidated the binding modes and mechanisms of action, revealing a covalent inhibitor targeting the catalytic pocket and two allosteric inhibitors affecting the monomer/dimer equilibrium of 3CLpro. The identified compounds demonstrated significant antiviral activity in vitro, reducing SARS-CoV-2 replication in VeroE6 and Calu-3 cell lines. This study highlights the potential of combining HTS and computational approaches to accelerate the discovery of effective antiviral agents, suggesting a workflow to support the research and the design of effective therapeutic strategies.
Collapse
Affiliation(s)
- Debora Zian
- Axxam SpA Openzone, Via Meucci 3, Bresso, 20091, Milan, Italy
| | - Daniela Iaconis
- EXSCALATE, Dompé Farmaceutici SpA, Via Tommaso De Amicis, 95, 80131, Napoli, Italy
| | - Simone Nenci
- Axxam SpA Openzone, Via Meucci 3, Bresso, 20091, Milan, Italy
| | | | | | | | - Rocco Vitalone
- Axxam SpA Openzone, Via Meucci 3, Bresso, 20091, Milan, Italy
| | - Ameya Raje
- Axxam SpA Openzone, Via Meucci 3, Bresso, 20091, Milan, Italy
| | | | | | - Angela Molteni
- Axxam SpA Openzone, Via Meucci 3, Bresso, 20091, Milan, Italy
| | - Candida Manelfi
- EXSCALATE, Dompé Farmaceutici SpA, Via Tommaso De Amicis, 95, 80131, Napoli, Italy
| | - Valerio Tazzari
- EXSCALATE, Dompé Farmaceutici SpA, Via Tommaso De Amicis, 95, 80131, Napoli, Italy
| | | | - Paolo Malune
- Department of Life and Environmental Sciences, University of Cagliari, S.P. 8 Monserrato, Sestu Km 0.700 - I, 09042, Monserrato, Italy
| | - Stefania Maloccu
- Department of Life and Environmental Sciences, University of Cagliari, S.P. 8 Monserrato, Sestu Km 0.700 - I, 09042, Monserrato, Italy
| | - Annalaura Paulis
- Department of Life and Environmental Sciences, University of Cagliari, S.P. 8 Monserrato, Sestu Km 0.700 - I, 09042, Monserrato, Italy
| | - Angela Corona
- Department of Life and Environmental Sciences, University of Cagliari, S.P. 8 Monserrato, Sestu Km 0.700 - I, 09042, Monserrato, Italy
| | - Salvatore Nieddu
- Department of Life and Environmental Sciences, University of Cagliari, S.P. 8 Monserrato, Sestu Km 0.700 - I, 09042, Monserrato, Italy
| | - Silvano Coletti
- Department of Engineering, University of Rome Guglielmo Marconi, Via Plinio 44, Rome, Italy
| | | | - Enzo Tramontano
- Department of Life and Environmental Sciences, University of Cagliari, S.P. 8 Monserrato, Sestu Km 0.700 - I, 09042, Monserrato, Italy
| | - Francesca Esposito
- Department of Life and Environmental Sciences, University of Cagliari, S.P. 8 Monserrato, Sestu Km 0.700 - I, 09042, Monserrato, Italy
| | | |
Collapse
|
9
|
Hasegawa M, Kijima S. MIDD in Japan- Implementations, challenges and opportunities. Adv Drug Deliv Rev 2025; 220:115553. [PMID: 40024482 DOI: 10.1016/j.addr.2025.115553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 02/12/2025] [Accepted: 02/28/2025] [Indexed: 03/04/2025]
Abstract
In Japan, there has been a growing adoption of Model Informed Drug Development (MIDD) approaches as the rationale for optimal dose decision and modeling outputs have supported better characterizing the risk-benefit profile of a drug by accelerating development period and regulatory-approval pathways. Three primary guidelines on pharmacometric analysis tools issued in Japan between 2019 and 2020 function as a shared communication medium between pharmaceutical companies and the regulatory agency in Japan and have contributed to increasing number of MIDD applications embedded into new drug application documents. This review article describes how the Pharmaceuticals and Medical Devices Agency have been promoting the adoption of modeling and simulation and how MIDD applications have been implemented in Japan by introducing multiple case studies. The intent is to share the knowledge and to promote the harmonization of regulations globally for accelerating the appropriate utilization of MIDD tools and implementation of various new technologies. (149 words).
Collapse
Affiliation(s)
| | - Shinichi Kijima
- Office of New Drug V, Pharmaceuticals and Medical Devices Agency (PMDA), Tokyo, Japan
| |
Collapse
|
10
|
Bernardi S, Roversi M, Torelli A, Musolino A, Nicastri E, Palma P, Rossi P, Vallesi L, Corsetti T, Lancella L, Lucarelli B, Galaverna F, Villani A, Perno CF, Raponi M. Safety of Nirmatrelvir-Ritonavir Administration in Children With Immunodeficiency and/or Comorbidities With SARS-CoV-2 Infection: A Retrospective Clinical Report. Pediatr Infect Dis J 2025; 44:442-448. [PMID: 39774678 DOI: 10.1097/inf.0000000000004657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
INTRODUCTION Despite the generally mild course of COVID-19 in children, immunocompromised patients may experience complications or severe infection. This study reports the clinical outcomes of pediatric patients treated with nirmatrelvir and ritonavir (N/R) for SARS-CoV-2 infection. METHODS We retrospectively reported the data of children with any immunodeficiency with COVID-19 who received N/R treatment between March 2022 and June 2023 at the Bambino Gesù Children's Hospital. Patients were treated with N/R for 5 days. We compared liver and kidney function before and after treatment with N/R and looked for a relationship between the duration of COVID-19 infection and the time from positivity to administration of N/R administration. RESULTS A total of 85 pediatric immunocompromised patients with COVID-19 were included in the study, with a mean age of 10.7 years (SD 4.8), mostly males (60%). We found a significant difference in the viral load before and after N/R administration. Four patients (4.7%) experienced adverse events related to N/R therapy. One of these had to discontinue N/R administration. Three patients (3.5%) experienced negative effects of drug interactions during N/R therapy, namely an increase of sirolimus and ciclosporin serum levels. A significant positive correlation was found between the time from SARS-CoV-2 positivity to N/R administration and the duration of SARS-CoV-2 swab positivity (R = 0.78, P < 0.001), suggesting that the earlier N/R is administered, the shorter the duration of COVID-19 in the study sample. CONCLUSION Our experience shows that N/R is reasonably safe in the pediatric population and could favor viral clearance, thus reducing the duration of infection.
Collapse
Affiliation(s)
- Stefania Bernardi
- From the Infectious Disease Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Marco Roversi
- PhD Program in Immunology, Molecular Medicine and Applied Biotechnology, University of Rome Tor Vergata, Rome, Italy
- Clinical Trial Area, Development and Implementation of Drugs, Vaccines, and Medical Devices for Pediatric Use, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Antonio Torelli
- Residency School of Pediatrics, University of Rome Tor Vergata, Rome, Italy
| | - Antonio Musolino
- Residency School of Pediatrics, University of Rome Tor Vergata, Rome, Italy
| | - Emanuele Nicastri
- Clinical Division of Infectious Diseases, National Institute for Infectious Diseases "Lazzaro Spallanzani," IRCCS, Rome, Italy
| | - Paolo Palma
- Clinical and Research Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
- Systems Medicine Department, University of Rome Tor Vergata, Rome, Italy
| | - Paolo Rossi
- Clinical Trial Area, Development and Implementation of Drugs, Vaccines, and Medical Devices for Pediatric Use, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
- Systems Medicine Department, University of Rome Tor Vergata, Rome, Italy
| | - Leonardo Vallesi
- Hospital Pharmacy Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Tiziana Corsetti
- Hospital Pharmacy Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Laura Lancella
- From the Infectious Disease Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Barbarella Lucarelli
- Research Area of Hematology and Oncology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome
| | - Federica Galaverna
- Research Area of Hematology and Oncology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome
| | - Alberto Villani
- Systems Medicine Department, University of Rome Tor Vergata, Rome, Italy
- General Pediatrics and ED 2nd Level, Bambino Gesù Children's Hospital IRCCS, Rome, Italy
| | - Carlo Federico Perno
- Microbiology and Diagnostic Immunology Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | | |
Collapse
|
11
|
Pan B, Mountford SJ, Kiso M, Anderson DE, Papadakis G, Jarman KE, Tilmanis DR, Maher B, Tran T, Shortt J, Yamayoshi S, Kawaoka Y, Thompson PE, Greenall SA, Warner N. Targeted protein degraders of SARS-CoV-2 Mpro are more active than enzymatic inhibition alone with activity against nirmatrelvir resistant virus. COMMUNICATIONS MEDICINE 2025; 5:140. [PMID: 40287552 PMCID: PMC12033352 DOI: 10.1038/s43856-025-00863-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 04/11/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND Effective antiviral therapy is lacking for most viral infections, and when available, is frequently compromised by the selection of resistance. Targeted protein degraders could provide an avenue to more effective antivirals, able to overcome the selection of resistance. The aim of this study was to determine whether adaptation of SARS-CoV-2 main protease (Mpro, also described as chymotrypsin-like protease (3CLpro) or non-structural protein 5 (Nsp5)) inhibitors into degraders leads to increased antiviral activity, including activity against resistant virus. METHODS We adapted the clinically approved Mpro inhibitor nirmatrelvir into a panel of degraders. Size-matched non-degrading controls were also synthesised to discriminate degradation activity from inhibition activity. Degrader activity was confirmed using an inducible Mpro-HiBiT tag expressing cell line. Antiviral activity against both wildtype and nirmatrelvir-resistant virus was performed using infection of susceptible cell lines. RESULTS Here we show three compounds, derived from nirmatrelvir and utilising VHL or IAP ubiquitin ligase recruiters, capable of degrading Mpro protein in a concentration, time and proteasome dependent fashion. These compounds also degrade nirmatrelvir-resistant mutant Mpro. The most potent of these compounds possesses enhanced antiviral activity against multiple wildtype SARS-CoV-2 strains and nirmatrelvir-resistant strains compared to non-degrading controls. CONCLUSIONS This work demonstrates the feasibility of generating degraders from viral protein inhibitors, and confirms that degraders possess higher antiviral potency and activity against resistant virus, compared to size matched non-degrading enzymatic inhibitors. These findings further support the development of targeted viral protein degraders as antiviral drugs, which may lead to more effective antiviral therapies for the future.
Collapse
Affiliation(s)
- Baolong Pan
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, USA
| | - Simon J Mountford
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Maki Kiso
- Division of Virology, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- The University of Tokyo Pandemic Preparedness, Infection and Advanced Research Center, Tokyo, Japan
| | - Danielle E Anderson
- Victorian Infectious Diseases Reference Laboratory, Royal Melbourne Hospital at The Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
- Department of Microbiology and Immunology, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Georgina Papadakis
- Victorian Infectious Diseases Reference Laboratory, Royal Melbourne Hospital at The Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Kate E Jarman
- The Walter and Eliza Hall Institute, Parkville, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Australia
| | - Danielle R Tilmanis
- The Walter and Eliza Hall Institute, Parkville, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Australia
| | - Belinda Maher
- Department of Medicine, School of Clinical Sciences, Monash University, Clayton, Australia
- Monash Haematology, Monash Health, Clayton, Australia
| | - Thomas Tran
- Victorian Infectious Diseases Reference Laboratory, Royal Melbourne Hospital at The Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Jake Shortt
- Department of Medicine, School of Clinical Sciences, Monash University, Clayton, Australia
- Monash Haematology, Monash Health, Clayton, Australia
| | - Seiya Yamayoshi
- Division of Virology, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- The University of Tokyo Pandemic Preparedness, Infection and Advanced Research Center, Tokyo, Japan
- International Research Center for Infectious Diseases, Institute of Medical Science, University of Tokyo, Tokyo, Japan
- The Research Center for Global Viral Diseases, National Center for Global Health and Medicine Research Institute, Tokyo, Japan
| | - Yoshihiro Kawaoka
- Division of Virology, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- The University of Tokyo Pandemic Preparedness, Infection and Advanced Research Center, Tokyo, Japan
- International Research Center for Infectious Diseases, Institute of Medical Science, University of Tokyo, Tokyo, Japan
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, USA
| | - Philip E Thompson
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Sam A Greenall
- Department of Medicine, School of Clinical Sciences, Monash University, Clayton, Australia
| | - Nadia Warner
- Victorian Infectious Diseases Reference Laboratory, Royal Melbourne Hospital at The Peter Doherty Institute for Infection and Immunity, Melbourne, Australia.
| |
Collapse
|
12
|
Detomasi TC, Degotte G, Huang S, Suryawanshi RK, Diallo A, Lizzadro L, Zaptero-Belinchón FJ, Taha TY, Li J, Richards AL, Hantz ER, Alam Z, Montano M, McCavitt-Malvido M, Gumpena R, Partridge JR, Correy GJ, Matsui Y, Charvat AF, Glenn IS, Rosecrans J, Revalde JL, Anderson D, Hultquist JF, Arkin MR, Neitz RJ, Swaney DL, Krogan NJ, Shoichet BK, Verba KA, Ott M, Renslo AR, Craik CS. Structure-based discovery of highly bioavailable, covalent, broad-spectrum coronavirus M Pro inhibitors with potent in vivo efficacy. SCIENCE ADVANCES 2025; 11:eadt7836. [PMID: 40267184 PMCID: PMC12017303 DOI: 10.1126/sciadv.adt7836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 03/19/2025] [Indexed: 04/25/2025]
Abstract
The main protease (MPro) of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a validated drug target. Starting with a lead-like dihydrouracil chemotype identified in a large-library docking campaign, we improved MPro inhibition >1000-fold by engaging additional MPro subsites and using a latent electrophile to engage Cys145. Advanced leads from this series show pan-coronavirus antiviral activity, low clearance in mice, and for AVI-4773, a rapid reduction in viral titers >1,000,000 after just three doses. Both compounds are well distributed in mouse tissues, including brain, where concentrations >1000× the 90% effective concentration are observed 8 hours after oral dosing for AVI-4773. AVI-4516 shows minimal inhibition of major cytochrome P450s and human proteases. AVI-4516 also exhibits synergy with the RNA-dependent RNA polymerase inhibitor, molnupiravir, in cellular infection models. Related analogs strongly inhibit nirmatrelvir-resistant MPro mutant virus. The properties of this chemotype are differentiated from existing clinical and preclinical MPro inhibitors and will advance therapeutic development against emerging SARS-CoV-2 variants and other coronaviruses.
Collapse
Affiliation(s)
- Tyler C. Detomasi
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | - Gilles Degotte
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | - Sijie Huang
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | | | - Amy Diallo
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA
| | - Luca Lizzadro
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | | | - Taha Y. Taha
- Gladstone Institute of Virology, Gladstone Institutes, San Francisco, CA, USA
| | - Jiapeng Li
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | - Alicia L. Richards
- Gladstone Institute of Data Science and Biotechnology, Gladstone Institutes, San Francisco, CA, USA
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, USA
- Quantitative Biosciences Institute (QBI), University of California, San Francisco, San Francisco, CA, USA
| | - Eric R. Hantz
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | - Zain Alam
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | - Mauricio Montano
- Gladstone Institute of Virology, Gladstone Institutes, San Francisco, CA, USA
| | | | - Rajesh Gumpena
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA
| | - James R. Partridge
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA
| | - Galen J. Correy
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Yusuke Matsui
- Gladstone Institute of Virology, Gladstone Institutes, San Francisco, CA, USA
| | - Annemarie F. Charvat
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | - Isabella S. Glenn
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | - Julia Rosecrans
- Gladstone Institute of Virology, Gladstone Institutes, San Francisco, CA, USA
| | - Jezrael L. Revalde
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | - Dashiell Anderson
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | - Judd F. Hultquist
- Division of Infectious Diseases, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Michelle R. Arkin
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | - R. Jeffrey Neitz
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | - Danielle L. Swaney
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA
- Gladstone Institute of Data Science and Biotechnology, Gladstone Institutes, San Francisco, CA, USA
- Quantitative Biosciences Institute (QBI), University of California, San Francisco, San Francisco, CA, USA
| | - Nevan J. Krogan
- Gladstone Institute of Data Science and Biotechnology, Gladstone Institutes, San Francisco, CA, USA
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, USA
- Quantitative Biosciences Institute (QBI), University of California, San Francisco, San Francisco, CA, USA
| | - Brian K. Shoichet
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | - Kliment A. Verba
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA
- Quantitative Biosciences Institute (QBI), University of California, San Francisco, San Francisco, CA, USA
| | - Melanie Ott
- Gladstone Institute of Virology, Gladstone Institutes, San Francisco, CA, USA
| | - Adam R. Renslo
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | - Charles S. Craik
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
13
|
Liu S, Zhou J, Yu L, Liu Y, Huang Y, Ouyang Y, Liu GK, Xu XH, Shibata N. Nitrogen-Based Organofluorine Functional Molecules: Synthesis and Applications. Chem Rev 2025. [PMID: 40261821 DOI: 10.1021/acs.chemrev.4c00661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/24/2025]
Abstract
Fluorine and nitrogen form a successful partnership in organic synthesis, medicinal chemistry, and material sciences. Although fluorine-nitrogen chemistry has a long and rich history, this field has received increasing interest and made remarkable progress over the past two decades, driven by recent advancements in transition metal and organocatalysis and photochemistry. This review, emphasizing contributions from 2015 to 2023, aims to update the state of the art of the synthesis and applications of nitrogen-based organofluorine functional molecules in organic synthesis and medicinal chemistry. In dedicated sections, we first focus on fluorine-containing reagents organized according to the type of fluorine-containing groups attached to nitrogen, including N-F, N-RF, N-SRF, and N-ORF. This review also covers nitrogen-linked fluorine-containing building blocks, catalysts, pharmaceuticals, and agrochemicals, underlining these components' broad applicability and growing importance in modern chemistry.
Collapse
Affiliation(s)
- Shuai Liu
- College of Chemistry and Chemical Engineering, Donghua University, 2999 North Renmin Road, Shanghai 201620, China
- National Engineering Research Center for Carbohydrate Synthesis, Jiangxi Normal University, Nanchang 330022, China
| | - Jun Zhou
- Department of Nanopharmaceutical Sciences & Department of Life Science and Applied Chemistry, Nagoya Institute of Technology, Gokiso, Showa-ku, Nagoya 466-8555, Japan
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, China
| | - Lu Yu
- Key Laboratory of Fluorine and Nitrogen Chemistry and Advanced Materials, Shanghai Institute of Organic Chemistry, Chinese Academy of Science, 345 Lingling Lu, Shanghai 200032, China
| | - Yingle Liu
- School of Chemistry and Environmental Engineering, Sichuan University of Science&Engineering, 180 Xueyuan Street, Huixing Lu, Zigong, Sichuan 643000, China
| | - Yangen Huang
- College of Chemistry and Chemical Engineering, Donghua University, 2999 North Renmin Road, Shanghai 201620, China
| | - Yao Ouyang
- Key Laboratory of Fluorine and Nitrogen Chemistry and Advanced Materials, Shanghai Institute of Organic Chemistry, Chinese Academy of Science, 345 Lingling Lu, Shanghai 200032, China
| | - Guo-Kai Liu
- School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, China
| | - Xiu-Hua Xu
- Key Laboratory of Fluorine and Nitrogen Chemistry and Advanced Materials, Shanghai Institute of Organic Chemistry, Chinese Academy of Science, 345 Lingling Lu, Shanghai 200032, China
| | - Norio Shibata
- Department of Nanopharmaceutical Sciences & Department of Life Science and Applied Chemistry, Nagoya Institute of Technology, Gokiso, Showa-ku, Nagoya 466-8555, Japan
| |
Collapse
|
14
|
Bege M, Leiner K, Lovas M, Pető R, Bereczki I, Hodek J, Weber J, Kuczmog A, Borbás A. Synthesis of 3'-modified xylofuranosyl nucleosides bearing 5'-silyl or -butyryl groups and their antiviral effect against RNA viruses. Eur J Pharm Sci 2025; 209:107107. [PMID: 40268255 DOI: 10.1016/j.ejps.2025.107107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 04/18/2025] [Accepted: 04/21/2025] [Indexed: 04/25/2025]
Abstract
D-xylofuranosyl nucleoside analogues bearing alkylthio and glucosylthio substituents at the C3'-position were prepared by photoinitiated radical-mediated hydrothiolation reactions from the corresponding 2',5'-di-O-silyl-3'-exomethylene uridine. Sequential desilylation and 5'-O-butyrylation of the 3'-thiosubstituted molecules produced a 24-membered nucleoside series with diverse substitution patterns, and the compounds were evaluated for their in vitro antiviral activity against three dangerous human RNA viruses, SARS-CoV-2, SINV and CHIKV. Eight compounds exhibited SARS-CoV-2 activity with low micromolar EC50 values in Vero E6 cells, and two of them also inhibited virus growth in human Calu cells. The best anti-SARS-CoV-2 activity was exhibited by 2',5'-di-O-silylated 3'-C-alkylthio nucleosides. Twelve compounds showed in vitro antiviral activity against CHIKV and fourteen against SINV with low micromolar EC50 values, with the 5'-butyryl-2'-silyl-3'-alkylthio substitution pattern being the most favorable against both viruses. In the case of the tested nucleosides, removal of the 2'-O-silyl group completely abolished the antiviral activity of the compounds against all three viruses. Overall, the most potent antiviral agent was the disilylated 3'-glucosylthio xylonucleoside, which showed excellent and specific antiviral activity against SINV with an EC50 value of 3 μM and no toxic effect at the highest tested concentration of 120 μM.
Collapse
Affiliation(s)
- Miklós Bege
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Debrecen, Egyetem tér 1, Debrecen 4032, Hungary
| | - Krisztina Leiner
- National Laboratory of Virology, Szentágothai Research Centre, University of Pécs, Ifjúság útja 20, Pecs 7624, Hungary; Institute of Biology, Faculty of Sciences, University of Pécs, Ifjúság útja 6, Pécs 7624, Hungary
| | - Miklós Lovas
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Debrecen, Egyetem tér 1, Debrecen 4032, Hungary
| | - Réka Pető
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Debrecen, Egyetem tér 1, Debrecen 4032, Hungary
| | - Ilona Bereczki
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Debrecen, Egyetem tér 1, Debrecen 4032, Hungary; National Laboratory of Virology, Szentágothai Research Centre, University of Pécs, Ifjúság útja 20, Pecs 7624, Hungary
| | - Jan Hodek
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo náměstí 2, 16610 Prague 6, Czech Republic
| | - Jan Weber
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo náměstí 2, 16610 Prague 6, Czech Republic
| | - Anett Kuczmog
- National Laboratory of Virology, Szentágothai Research Centre, University of Pécs, Ifjúság útja 20, Pecs 7624, Hungary; Institute of Biology, Faculty of Sciences, University of Pécs, Ifjúság útja 6, Pécs 7624, Hungary
| | - Anikó Borbás
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Debrecen, Egyetem tér 1, Debrecen 4032, Hungary; National Laboratory of Virology, Szentágothai Research Centre, University of Pécs, Ifjúság útja 20, Pecs 7624, Hungary.
| |
Collapse
|
15
|
Saito-Tarashima N, Koma T, Hinotani N, Yoshida K, Ogasa M, Murai A, Inoue S, Kondo T, Doi N, Tsuneyama K, Nomaguchi M, Minakawa N. 3-Deazaguanosine inhibits SARS-CoV-2 viral replication and reduces the risk of COVID-19 pneumonia in hamster. iScience 2025; 28:112140. [PMID: 40171487 PMCID: PMC11960675 DOI: 10.1016/j.isci.2025.112140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 10/12/2024] [Accepted: 02/26/2025] [Indexed: 04/03/2025] Open
Abstract
The COVID-19 pandemic highlighted the serious threat that coronaviruses have on public health. Because coronavirus continuously undergoes cross-species transmission, additional therapeutic agents and targets are urgently needed. Here, we show that a 3-deazapurine ribonucleoside, 3-Deazaguanosine (C3Guo, 2), has potent antiviral activity against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Unexpectedly, C3Guo (2) does not act as an inhibitor of RNA-dependent RNA polymerase (RdRp), which is the therapeutic target of two key nucleoside/nucleotide inhibitors approved for the treatment of COVID-19 (Remdesivir and Molnupiravir); instead, it seems to function by targeting the capping machinery of viral RNA. In hamsters infected with SARS-CoV-2, administration of 2 markedly reduced infectious viral titers, and prevented the development of COVID-19 pneumonia better than Molnupiravir. The potency of 2 against SARS-CoV-2 underscores its potential as an effective therapeutic agent for COVID-19 and future zoonotic coronavirus infections and raises the possibility of antiviral nucleoside analogs with alternative therapeutic targets to RdRp.
Collapse
Affiliation(s)
- Noriko Saito-Tarashima
- Graduate School of Pharmaceutical Science, Tokushima University, 1-78-1 Shomachi, Tokushima, Tokushima 770-8505, Japan
| | - Takaaki Koma
- Department of Microbiology, Graduate School of Medicine, Tokushima University, 3-18-15 Kuramoto, Tokushima, Tokushima 770-8503, Japan
- Department of PostLED Photonics Research, Institute of PostLED Photonics, Tokushima University, 2-1 Minamijosanjima, Tokushima, Tokushima 770-8506, Japan
| | - Naoto Hinotani
- Graduate School of Pharmaceutical Science, Tokushima University, 1-78-1 Shomachi, Tokushima, Tokushima 770-8505, Japan
| | - Keigo Yoshida
- Graduate School of Pharmaceutical Science, Tokushima University, 1-78-1 Shomachi, Tokushima, Tokushima 770-8505, Japan
| | - Moka Ogasa
- Graduate School of Pharmaceutical Science, Tokushima University, 1-78-1 Shomachi, Tokushima, Tokushima 770-8505, Japan
| | - Akiho Murai
- Graduate School of Pharmaceutical Science, Tokushima University, 1-78-1 Shomachi, Tokushima, Tokushima 770-8505, Japan
| | - Syuya Inoue
- Graduate School of Pharmaceutical Science, Tokushima University, 1-78-1 Shomachi, Tokushima, Tokushima 770-8505, Japan
| | - Tomoyuki Kondo
- Department of Microbiology, Graduate School of Medicine, Tokushima University, 3-18-15 Kuramoto, Tokushima, Tokushima 770-8503, Japan
| | - Naoya Doi
- Department of Microbiology, Graduate School of Medicine, Tokushima University, 3-18-15 Kuramoto, Tokushima, Tokushima 770-8503, Japan
| | - Koichi Tsuneyama
- Department of PostLED Photonics Research, Institute of PostLED Photonics, Tokushima University, 2-1 Minamijosanjima, Tokushima, Tokushima 770-8506, Japan
- Department of Pathology and Laboratory Medicine, Graduate School of Medicine, Tokushima University, 3-18-15 Kuramoto, Tokushima, Tokushima 770-8503, Japan
| | - Masako Nomaguchi
- Department of Microbiology, Graduate School of Medicine, Tokushima University, 3-18-15 Kuramoto, Tokushima, Tokushima 770-8503, Japan
- Department of PostLED Photonics Research, Institute of PostLED Photonics, Tokushima University, 2-1 Minamijosanjima, Tokushima, Tokushima 770-8506, Japan
| | - Noriaki Minakawa
- Graduate School of Pharmaceutical Science, Tokushima University, 1-78-1 Shomachi, Tokushima, Tokushima 770-8505, Japan
| |
Collapse
|
16
|
Buchynskyi M, Kamyshna I, Halabitska I, Petakh P, Oksenych V, Kamyshnyi O. Genetic Predictors of Paxlovid Treatment Response: The Role of IFNAR2, OAS1, OAS3, and ACE2 in COVID-19 Clinical Course. J Pers Med 2025; 15:156. [PMID: 40278335 PMCID: PMC12028499 DOI: 10.3390/jpm15040156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 04/11/2025] [Accepted: 04/12/2025] [Indexed: 04/26/2025] Open
Abstract
Background: This study investigated the role of genetic polymorphisms in IFNAR2, OAS1, OAS3, and ACE2 as predictors of Paxlovid treatment response, specifically examining their influence on the clinical course and laboratory parameters of COVID-19 patients. Methods: We analyzed the impact of polymorphisms in genes associated with the interferon pathway (IFNAR2 rs2236757), antiviral response (OAS1 rs10774671, OAS3 rs10735079), and viral entry (ACE2 rs2074192) in individuals treated with Paxlovid. Results: Our findings suggest that genetic variations in these genes may modulate the immune response and coagulation pathways in the context of Paxlovid treatment during COVID-19 infection. Specifically, the IFNAR2 rs2236757 G allele was associated with alterations in inflammatory and coagulation markers, while polymorphisms in OAS1 and OAS3 influenced coagulation parameters. Furthermore, specific genotypes were linked to changes in clinical parameters such as oxygen saturation, leukocyte count, and liver function markers in Paxlovid-treated patients. Conclusions: These results highlight the potential of considering genetic factors in understanding individual responses to COVID-19 treatment with Paxlovid and informing future personalized approaches.
Collapse
Affiliation(s)
- Mykhailo Buchynskyi
- Department of Microbiology, Virology, and Immunology, I. Horbachevsky Ternopil National Medical University, 46001 Ternopil, Ukraine;
| | - Iryna Kamyshna
- Department of Medical Rehabilitation, I. Horbachevsky Ternopil National Medical University, 46001 Ternopil, Ukraine;
| | - Iryna Halabitska
- Department of Therapy and Family Medicine, I. Horbachevsky Ternopil National Medical University, Voli Square, 1, 46001 Ternopil, Ukraine;
| | - Pavlo Petakh
- Department of Biochemistry and Pharmacology, Uzhhorod National University, 88000 Uzhhorod, Ukraine;
| | - Valentyn Oksenych
- Broegelmann Research Laboratory, Department of Clinical Science, University of Bergen, 5020 Bergen, Norway
| | - Oleksandr Kamyshnyi
- Department of Microbiology, Virology, and Immunology, I. Horbachevsky Ternopil National Medical University, 46001 Ternopil, Ukraine;
| |
Collapse
|
17
|
Ballav N, Giri CK, Saha SN, Mane MV, Baidya M. Empowering Diastereoselective Cyclopropanation of Unactivated Alkenes with Sulfur Ylides through Nucleopalladation. J Am Chem Soc 2025; 147:13017-13026. [PMID: 40194297 DOI: 10.1021/jacs.5c03146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Regio- and stereoselective cyclopropanation of unactivated alkenes under mild conditions remains a challenging yet fundamental transformation. We present a versatile palladium(II)-catalyzed method for the diastereoselective cyclopropanation of alkenyl amines and alkenyl acids, which leverages the nucleopalladation mechanism and the unique ambiphilic reactivity of sulfur ylides. This Pd(II)/Pd(IV) catalytic protocol selectively delivers anti-cyclopropanes for allylamines with a removable isoquinoline-1-carboxamide auxiliary, while enabling excellent syn-selectivity for alkenyl acid derivatives containing a 2-(aminomethyl)pyridine derivative as a directing group. The protocol is operationally simple and scalable, features a wide substrate generality, and also remains effective in the presence of various medicinally relevant scaffolds. The cyclopropane products were further transformed into 1,2,3-trifunctionalized cyclopropanes and engaged in an aza-Piancatelli reaction, introducing additional molecular complexity. DFT studies were performed to shed light on the reaction mechanism and the origins of the observed stereoselectivity.
Collapse
Affiliation(s)
- Nityananda Ballav
- Department of Chemistry, Indian Institute of Technology Madras, Chennai 600036, India
| | - Chandan Kumar Giri
- Department of Chemistry, Indian Institute of Technology Madras, Chennai 600036, India
| | - Shib Nath Saha
- Department of Chemistry, Indian Institute of Technology Madras, Chennai 600036, India
| | - Manoj V Mane
- Centre for Nano and Material Sciences, Jain (Deemed-to-be University), Jain Global Campus, Kanakapura, Bangalore 562112, Karnataka, India
| | - Mahiuddin Baidya
- Department of Chemistry, Indian Institute of Technology Madras, Chennai 600036, India
| |
Collapse
|
18
|
Rebhan L, Fürst R, Schollmeyer D, Serafim RAM, Gehringer M. Go for Gold: Development of a Scalable Synthesis of [1-(Ethoxycarbonyl)cyclopropyl] Triphenylphosphonium Tetrafluoroborate, a Key Reagent to Explore Covalent Monopolar Spindle 1 Inhibitors. ChemistryOpen 2025:e2500106. [PMID: 40237105 DOI: 10.1002/open.202500106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 03/29/2025] [Indexed: 04/17/2025] Open
Abstract
Covalent approaches have resurged in drug discovery and chemical biology during the last decade. So-called targeted covalent inhibitors typically show a strong and persistent drug-target interaction as well as a high degree of selectivity. In our research group, RMS-07 (8), a First-in-Class covalent inhibitor of the protein kinase threonine tyrosine kinase (TTK)/monopolar spindle 1, which shows promising results in a variety of different solid cancer cell types and will be further optimized in terms of covalent binding kinetics, has recently been developed. However, synthetic accessibility is restricted by a high price and limited availability of [1-(ethoxycarbonyl)cyclopropyl] triphenylphosphonium tetrafluoroborate (10), a key reagent required to assemble the tricyclic core scaffold in a Wittig-type cyclization reaction. This reagent is also described as a valuable synthon for the synthesis of a range of ring systems with interesting applications in medicinal chemistry. However, reliable procedures for its large-scale synthesis are scarce. Only one prior report describes the synthesis of reagent 10, and it contains limited experimental details, making it challenging to reproduce and scale up. Herein, a concise and reproducible decigram-scale synthetic protocol for accessing key reagent 10 is described.
Collapse
Affiliation(s)
- Leon Rebhan
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmaceutical Sciences, Eberhard Karls University Tübingen, 72076, Tübingen, Germany
- Department of Chemistry, Biochemistry and Pharmacy, University of Bern, 3012, Bern, Switzerland
| | - Rebekka Fürst
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmaceutical Sciences, Eberhard Karls University Tübingen, 72076, Tübingen, Germany
- Department for Medicinal Chemistry, Institute for Biomedical Engineering, Faculty of Medicine, Eberhard Karls University Tübingen, Auf der Morgenstelle 8, 72076, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) 'Image-Guided & Functionally Instructed Tumor Therapies', University of Tübingen, 72076, Tübingen, Germany
| | - Dieter Schollmeyer
- Department Chemie, Zentrale Analytik, Johannes Gutenberg-Universität Mainz, 55128, Mainz, Germany
| | - Ricardo A M Serafim
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmaceutical Sciences, Eberhard Karls University Tübingen, 72076, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) 'Image-Guided & Functionally Instructed Tumor Therapies', University of Tübingen, 72076, Tübingen, Germany
- Department of Organic and Pharmaceutical Chemistry, School of Engineering, Institut Químic de Sarrià (IQS), Universitat Ramon Llull (URL), Vía Augusta 390, 08017, Barcelona, Spain
| | - Matthias Gehringer
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmaceutical Sciences, Eberhard Karls University Tübingen, 72076, Tübingen, Germany
- Department for Medicinal Chemistry, Institute for Biomedical Engineering, Faculty of Medicine, Eberhard Karls University Tübingen, Auf der Morgenstelle 8, 72076, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) 'Image-Guided & Functionally Instructed Tumor Therapies', University of Tübingen, 72076, Tübingen, Germany
| |
Collapse
|
19
|
Zheng P, Li G, Chen Y, Li S, Yang S, Guo D, Zhou Q, Zhang X. Mini review: SHEN26, a novel oral antiviral drug for COVID-19 treatment. Bioorg Med Chem Lett 2025; 124:130243. [PMID: 40250816 DOI: 10.1016/j.bmcl.2025.130243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 04/11/2025] [Accepted: 04/15/2025] [Indexed: 04/20/2025]
Abstract
Over two years into the pandemic, global collaboration led to effective antiviral drugs targeting SARS-CoV-2's RdRp and 3CL protease. However, the virus continues to evolve, and certain low-virulence variants still circulate. Despite reduced virulence, ongoing transmission raises the risk of new mutations, underscoring the need for continued vigilance, research, and expansion of our antiviral and vaccine strategies. Our research team has developed SHEN26, a promising small-molecule antiviral drug for the treatment of COVID-19. This mini-review explores its development, including history, synthesis, preclinical evaluations, and findings from Phase I and II clinical trials. Data from each research phase further underscores SHEN26's potential as a safe and effective oral antiviral treatment for COVID-19, while also emphasizing its broader relevance in combating emerging RNA viral infections.
Collapse
Affiliation(s)
- Peisen Zheng
- Shenzhen Key Laboratory of Small Molecule Drug Discovery and Synthesis, Department of Chemistry, Shenzhen Grubbs Institute and Medi-X Pingshan, Southern University of Science and Technology, Shenzhen 518000, China
| | - Guanguan Li
- Shenzhen AntiV Pharma Co., Ltd., Shenzhen, Guangdong 518081, China
| | - Yuanguang Chen
- Shenzhen Key Laboratory of Small Molecule Drug Discovery and Synthesis, Department of Chemistry, Shenzhen Grubbs Institute and Medi-X Pingshan, Southern University of Science and Technology, Shenzhen 518000, China
| | - Shuo Li
- Shenzhen Key Laboratory of Small Molecule Drug Discovery and Synthesis, Department of Chemistry, Shenzhen Grubbs Institute and Medi-X Pingshan, Southern University of Science and Technology, Shenzhen 518000, China
| | - Sidi Yang
- Guangzhou National Laboratory, Guangzhou, Guangdong 510005, China
| | - Deyin Guo
- Guangzhou National Laboratory, Guangzhou, Guangdong 510005, China
| | - Qifan Zhou
- Shenzhen Key Laboratory of Small Molecule Drug Discovery and Synthesis, Department of Chemistry, Shenzhen Grubbs Institute and Medi-X Pingshan, Southern University of Science and Technology, Shenzhen 518000, China.
| | - Xumu Zhang
- Shenzhen Key Laboratory of Small Molecule Drug Discovery and Synthesis, Department of Chemistry, Shenzhen Grubbs Institute and Medi-X Pingshan, Southern University of Science and Technology, Shenzhen 518000, China.
| |
Collapse
|
20
|
Bao H, Meng H, Gong S, Gong Y, Tu G, Du Z, Wang Y, Wu J, Ma C, Ma Q, Yao X. Design, synthesis and activity evaluation of 4-(quinoline-2-yl)aniline derivatives as SARS-CoV‑2 main protease inhibitors. Bioorg Med Chem 2025; 121:118135. [PMID: 40024142 DOI: 10.1016/j.bmc.2025.118135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 02/19/2025] [Accepted: 02/22/2025] [Indexed: 03/04/2025]
Abstract
Since 2020, numerous compounds have been investigated for their potential use in treating SARS-CoV-2 infections. By identifying the molecular targets during the virus replication process, rationally designed anti-SARS-CoV-2 agents are developed. Among these targets, the main protease (Mpro) is a crucial enzyme required for virus replication, and its highly conserved characteristic make it an important drug target for the development of anti-SARS-CoV-2 drugs. Herein, we utilized warhead-based design strategy to conduct the structural optimization of M-1 developed through virtual screening, leading to a series of novel Mpro inhibitors with 4-(quinolin-2-yl)aniline scaffold. Among them, M-32 exhibited good SARS-CoV-2 Mpro inhibitory activity (IC50 = 5.2 μM) with a nearly 25-fold increase. Isothermal titration calorimetry (ITC) directly proved that M-32 binds directly to SARS-CoV-2 Mpro in an entropy-driven manner. Mass spectrometry (MS) further confirmed the covalent binding ability of M-32 to Mpro. Meanwhile, M-32 effectively inhibited the replication of SARS-CoV-2 in Vero E6 cells (EC50 = 5.29 μM).
Collapse
Affiliation(s)
- Honglei Bao
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, China
| | - Hui Meng
- Collaborative Innovation Centre of Henan Province for Green Manufacturing of Fine Chemicals, Key Laboratory of Green Chemical Media and Reactions, Ministry of Education, Henan Engineering Research Centre of Chiral Hydroxyl Pharmaceutical, Henan Engineering Laboratory of Chemical Pharmaceutical and Biomedical Materials, School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, Henan 453007, China
| | - Shilin Gong
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, China
| | - Yaguo Gong
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, China
| | - Gao Tu
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, China
| | - Zhenya Du
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, China; Centre for Artificial Intelligence Driven Drug Discovery, Faculty of Applied Sciences, Macao Polytechnic University, Macao 999078, China
| | - Yuwei Wang
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, China
| | - Jianlin Wu
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, China
| | - Chunhua Ma
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, China; Collaborative Innovation Centre of Henan Province for Green Manufacturing of Fine Chemicals, Key Laboratory of Green Chemical Media and Reactions, Ministry of Education, Henan Engineering Research Centre of Chiral Hydroxyl Pharmaceutical, Henan Engineering Laboratory of Chemical Pharmaceutical and Biomedical Materials, School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, Henan 453007, China.
| | - Qinhai Ma
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.
| | - Xiaojun Yao
- Centre for Artificial Intelligence Driven Drug Discovery, Faculty of Applied Sciences, Macao Polytechnic University, Macao 999078, China.
| |
Collapse
|
21
|
Roux H, Touret F, Coluccia A, Scio P, Bouzidi HS, di Giorgio C, Gattacceca F, Khoumeri O, Silvestri R, Vanelle P, Roche M. Design and synthesis of novel thioether analogs as promising antiviral agents: In vitro activity against enteroviruses of interest. Eur J Med Chem 2025; 288:117395. [PMID: 39986184 DOI: 10.1016/j.ejmech.2025.117395] [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: 12/27/2024] [Revised: 02/03/2025] [Accepted: 02/11/2025] [Indexed: 02/24/2025]
Abstract
The Enterovirus genus contains two major subgroups: rhinovirus (RV) species A-C and enterovirus (EV) ones A-D. While RV only infects the respiratory system, the EV can cause a wide variety of diseases, ranging from non-specific febrile illness to severe neurologic complications. To date, no curative treatments are commercially available. Our research team had recently developed EV-A71 inhibitors. To improve their activity and broaden their spectrum, we performed optimization of the structure following an iterative cycle of chemical modulations. As a result, we obtained two broad-spectrum inhibitors with micromolar activity against these 3 types of viruses (OM1260: EC50 (MRC-5, EV-A71) = 1.15 μM; EC50 (RD, EV-A71) = 4.38 μM; EC50 (MRC-5, E30) = 0.41 μM; EC50 (MRC-5, CVA24) = 1.15 μM; HR-568: EC50 (MRC-5, EV-A71) = 3.25 μM; EC50 (RD, EV-A71) = 1.53 μM; EC50 (MRC-5, E30) = 0.40 μM; EC50 (MRC-5, CVA24) = 1.22 μM). Docking studies shed light on structure-activity relationships, while time-of-drug addition assays confirmed their intervention during the early step of viral replication. Eventually, some pharmacokinetic modelling has been carried out to evaluate their druggability. All these results showed that OM1260 and HR-568 are promising candidates for further development.
Collapse
Affiliation(s)
- Hugo Roux
- Aix-Marseille Université, CNRS, ICR UMR 7273, PCR, Faculté de Pharmacie, 13005, Marseille, France.
| | - Franck Touret
- Unité des Virus Émergents (UVE: Aix-Marseille Univ, Università di Corsica, IRD 190, Inserm 1207, IRBA), France.
| | - Antonio Coluccia
- Department of Drug Chemistry and Technologies, Laboratory Affiliated with the Institute Pasteur Italy - Cenci Bolognetti Foundation, Sapienza University of Rome, Italy.
| | - Pietro Scio
- Department of Drug Chemistry and Technologies, Laboratory Affiliated with the Institute Pasteur Italy - Cenci Bolognetti Foundation, Sapienza University of Rome, Italy.
| | - Hawa Sophia Bouzidi
- Unité des Virus Émergents (UVE: Aix-Marseille Univ, Università di Corsica, IRD 190, Inserm 1207, IRBA), France.
| | - Carole di Giorgio
- Aix-Marseille Université, Avignon Université, CNRS, IRD, IMBE, Faculty of Pharmacy, Service of Environmental Mutagenesis, Marseille, France.
| | - Florence Gattacceca
- Aix-Marseille Université, COMPO INRIA-CRCM-INSERM-U1068, CNRS UMR7258, Marseille, France.
| | - Omar Khoumeri
- Aix-Marseille Université, CNRS, ICR UMR 7273, PCR, Faculté de Pharmacie, 13005, Marseille, France.
| | - Romano Silvestri
- Department of Drug Chemistry and Technologies, Laboratory Affiliated with the Institute Pasteur Italy - Cenci Bolognetti Foundation, Sapienza University of Rome, Italy.
| | - Patrice Vanelle
- Aix-Marseille Université, CNRS, ICR UMR 7273, PCR, Faculté de Pharmacie, 13005, Marseille, France.
| | - Manon Roche
- Aix-Marseille Université, CNRS, ICR UMR 7273, PCR, Faculté de Pharmacie, 13005, Marseille, France.
| |
Collapse
|
22
|
Cao L, Shi S, Zhang C, Zhao C. A phycobiliprotein-based reporter assay for the evaluation of SARS-CoV-2 main protease activity. Virology 2025; 608:110540. [PMID: 40233445 DOI: 10.1016/j.virol.2025.110540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Revised: 04/08/2025] [Accepted: 04/11/2025] [Indexed: 04/17/2025]
Abstract
SARS-CoV-2 Mpro is crucial for viral replication and transcription and is highly conserved. Therefore, it is an ideal target for developing broad-spectrum antiviral drugs. To address resistance to existing drugs caused by mutations, a simple and sensitive method for detecting the activity of Mpro is needed. Considering the excellent fluorescence properties of phycobiliproteins, this study developed a phycobiliprotein-based reporter assay to evaluate Mpro activity. An engineered lyase was generated by inserting the Mpro recognition sequence between the phycobiliprotein lyases CpcF and CpcE. To ensure that the binding of CpcE and CpcF depended on the linker, a series of truncated forms were constructed. Among them, the activity of CpcE/F-10 was significantly reduced in the presence of Mpro; however, both genetic and chemical inhibition of Mpro activity reversed these results. These data indicated that the fluorescence of phycobiliproteins was negatively correlated with Mpro activity. The reporter assay developed here will contribute to determining the impact of Mpro mutations and screening for new inhibitors.
Collapse
Affiliation(s)
- Le Cao
- College of Chemistry and Chemical Engineering, Taiyuan University of Technology, Taiyuan, 030024, People's Republic of China; College of Artificial Intelligence, Taiyuan University of Technology, Taiyuan, 030024, People's Republic of China
| | - Shuyuan Shi
- College of Chemistry and Chemical Engineering, Taiyuan University of Technology, Taiyuan, 030024, People's Republic of China; College of Artificial Intelligence, Taiyuan University of Technology, Taiyuan, 030024, People's Republic of China
| | - Chaofeng Zhang
- College of Chemistry and Chemical Engineering, Taiyuan University of Technology, Taiyuan, 030024, People's Republic of China.
| | - Cheng Zhao
- College of Chemistry and Chemical Engineering, Taiyuan University of Technology, Taiyuan, 030024, People's Republic of China.
| |
Collapse
|
23
|
Koehler MFT. The Sprint to Develop Coronavirus Antivirals. J Med Chem 2025; 68:6958-6960. [PMID: 40152255 DOI: 10.1021/acs.jmedchem.5c00578] [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: 03/29/2025]
Abstract
The preclinical work leading to the discovery of PF-07321332 (nirmatrelvir) is described in the Featured Article summarized in this Viewpoint. This work resulted in an Emergency Use Authorization (EUA) for Paxlovid within two years of the project's start. In addition to the medicinal chemistry approach taken, the authors describe how they achieved this remarkable speed.
Collapse
Affiliation(s)
- Michael F T Koehler
- Department of Discovery Chemistry, Genentech Inc., South San Francisco, California 94080, United States
| |
Collapse
|
24
|
Wang J, Nithianantham S, Chai SC, Jung YH, Yang L, Ong HW, Li Y, Zhang Y, Miller DJ, Chen T. Decoding the selective chemical modulation of CYP3A4. Nat Commun 2025; 16:3423. [PMID: 40210880 PMCID: PMC11985932 DOI: 10.1038/s41467-025-58749-8] [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: 07/02/2024] [Accepted: 04/01/2025] [Indexed: 04/12/2025] Open
Abstract
Drug-drug interactions associate with concurrent uses of multiple medications. Cytochrome P450 (CYP) 3A4 metabolizes a large portion of marketed drugs. To maintain the efficacy of drugs metabolized by CYP3A4, pan-CYP3A inhibitors such as ritonavir are often co-administered. Although selective CYP3A4 inhibitors have greater therapeutic benefits as they avoid inhibiting unintended CYPs and undesirable clinical consequences, the high homology between CYP3A4 and CYP3A5 has hampered the development of such selective inhibitors. Here, we report a series of selective CYP3A4 inhibitors with scaffolds identified by high-throughput screening. Structural, functional, and computational analyses reveal that the differential C-terminal loop conformations and two distinct ligand binding surfaces disfavor the binding of selective CYP3A4 inhibitors to CYP3A5. Structure-guided design of compounds validates the model and yields analogs that are selective for CYP3A4 versus other major CYPs. These findings demonstrate the feasibility to selectively inhibit CYP3A4 and provide guidance for designing better CYP3A4 selective inhibitors.
Collapse
Affiliation(s)
- Jingheng Wang
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Stanley Nithianantham
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Sergio C Chai
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Young-Hwan Jung
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Lei Yang
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Han Wee Ong
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Yong Li
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Yifan Zhang
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Darcie J Miller
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Taosheng Chen
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN, USA.
| |
Collapse
|
25
|
Tuttle JB, Allais C, Allerton CMN, Anderson AS, Arcari JT, Aschenbrenner LM, Avery M, Bellenger J, Berritt S, Boras B, Boscoe BP, Buzon LM, Cardin RD, Carlo AA, Coffman KJ, Dantonio A, Di L, Eng H, Farley KA, Ferre RA, Gajiwala KS, Gibson SA, Greasley SE, Hurst BL, Kadar EP, Kalgutkar AS, Lachapelle EA, Lanyon LF, Lee J, Lee J, Lian Y, Liu W, Martínez-Alsina LA, Mason SW, Noell S, Novak J, Obach RS, Ogilvie K, O'Neil SV, Ostner G, Owen DR, Patel NC, Pettersson M, Singh RS, Rai DK, Reese MR, Sakata S, Sammons MF, Sathish JG, Sharma R, Steppan CM, Stewart A, Updyke L, Verhoest PR, Wei L, Wright SW, Yang E, Yang Q, Zhu Y. Discovery of Nirmatrelvir (PF-07321332): A Potent, Orally Active Inhibitor of the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS CoV-2) Main Protease. J Med Chem 2025; 68:7003-7030. [PMID: 40019854 DOI: 10.1021/acs.jmedchem.4c02561] [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: 03/14/2025]
Abstract
In early 2020, severe acute respiratory syndrome coronavirus 2 (SARS CoV-2) infections leading to COVID-19 disease reached a global level leading to the World Health Organization (WHO) declaration of a pandemic. Scientists around the globe rapidly responded to try and discover novel therapeutics and repurpose extant drugs to treat the disease. This work describes the preclinical discovery efforts that led to the invention of PF-07321332 (nirmatrelvir, 14), a potent and orally active inhibitor of the SARS CoV-2 main protease (Mpro) enzyme. At the outset we focused on modifying PF-00835231 (1) discovered in 2004 as a potent inhibitor of the SARS CoV-1 Mpro with poor systemic exposure. Our effort was focused on modifying 1 with the goal of engineering in oral bioavailability by design, while maintaining cellular potency and low metabolic clearance. Modifications of 1 ultimately led to the invention of nirmatrelvir 14, the Mpro inhibitor component in PAXLOVID.
Collapse
Affiliation(s)
- Jamison B Tuttle
- Pfizer Research & Development, Cambridge, Massachusetts 02139, United States
| | - Christophe Allais
- Pfizer Research & Development, Groton, Connecticut 06340, United States
| | | | | | - Joel T Arcari
- Pfizer Research & Development, Groton, Connecticut 06340, United States
| | | | - Melissa Avery
- Pfizer Research & Development, Groton, Connecticut 06340, United States
| | - Justin Bellenger
- Pfizer Research & Development, Groton, Connecticut 06340, United States
| | - Simon Berritt
- Pfizer Research & Development, Groton, Connecticut 06340, United States
| | - Britton Boras
- Pfizer Research & Development, La Jolla, California 92121, United States
| | - Brian P Boscoe
- Pfizer Research & Development, Groton, Connecticut 06340, United States
| | - Leanne M Buzon
- Pfizer Research & Development, Groton, Connecticut 06340, United States
| | - Rhonda D Cardin
- Pfizer Research & Development, Pearl River, New York 10965, United States
| | - Anthony A Carlo
- Pfizer Research & Development, Groton, Connecticut 06340, United States
| | - Karen J Coffman
- Pfizer Research & Development, Groton, Connecticut 06340, United States
| | - Alyssa Dantonio
- Pfizer Research & Development, Groton, Connecticut 06340, United States
| | - Li Di
- Pfizer Research & Development, Groton, Connecticut 06340, United States
| | - Heather Eng
- Pfizer Research & Development, Groton, Connecticut 06340, United States
| | - Kathleen A Farley
- Pfizer Research & Development, Groton, Connecticut 06340, United States
| | - Rose Ann Ferre
- Pfizer Research & Development, La Jolla, California 92121, United States
| | - Ketan S Gajiwala
- Pfizer Research & Development, La Jolla, California 92121, United States
| | - Scott A Gibson
- Institute of Antiviral Research, Department of Animal, Dairy, and Veterinary Sciences, Utah State University, Logan, Utah 84322, United States
| | | | - Brett L Hurst
- Institute of Antiviral Research, Department of Animal, Dairy, and Veterinary Sciences, Utah State University, Logan, Utah 84322, United States
| | - Eugene P Kadar
- Pfizer Research & Development, Groton, Connecticut 06340, United States
| | - Amit S Kalgutkar
- Pfizer Research & Development, Cambridge, Massachusetts 02139, United States
| | - Erik A Lachapelle
- Pfizer Research & Development, Groton, Connecticut 06340, United States
| | - Lorraine F Lanyon
- Pfizer Research & Development, Groton, Connecticut 06340, United States
| | - Jisun Lee
- Pfizer Research & Development, Groton, Connecticut 06340, United States
| | - Jack Lee
- Pfizer Research & Development, Groton, Connecticut 06340, United States
| | - Yajing Lian
- Pfizer Research & Development, Groton, Connecticut 06340, United States
| | - Wei Liu
- Pfizer Research & Development, La Jolla, California 92121, United States
| | | | - Stephen W Mason
- Pfizer Research & Development, Pearl River, New York 10965, United States
| | - Stephen Noell
- Pfizer Research & Development, Groton, Connecticut 06340, United States
| | - Jonathan Novak
- Pfizer Research & Development, Cambridge, Massachusetts 02139, United States
| | - R Scott Obach
- Pfizer Research & Development, Groton, Connecticut 06340, United States
| | - Kevin Ogilvie
- Pfizer Research & Development, Groton, Connecticut 06340, United States
| | - Steven V O'Neil
- Pfizer Research & Development, Groton, Connecticut 06340, United States
| | - Gregory Ostner
- Pfizer Research & Development, Groton, Connecticut 06340, United States
| | - Dafydd R Owen
- Pfizer Research & Development, Cambridge, Massachusetts 02139, United States
| | - Nandini C Patel
- Pfizer Research & Development, Cambridge, Massachusetts 02139, United States
| | - Martin Pettersson
- Pfizer Research & Development, Cambridge, Massachusetts 02139, United States
| | - Ravi Shankar Singh
- Pfizer Research & Development, Cambridge, Massachusetts 02139, United States
| | - Devendra K Rai
- Pfizer Research & Development, Groton, Connecticut 06340, United States
| | - Matthew R Reese
- Pfizer Research & Development, Groton, Connecticut 06340, United States
| | - Sylvie Sakata
- Pfizer Research & Development, La Jolla, California 92121, United States
| | - Matthew F Sammons
- Pfizer Research & Development, Cambridge, Massachusetts 02139, United States
| | - Jean G Sathish
- Pfizer Research & Development, Pearl River, New York 10965, United States
| | - Raman Sharma
- Pfizer Research & Development, Groton, Connecticut 06340, United States
| | - Claire M Steppan
- Pfizer Research & Development, Groton, Connecticut 06340, United States
| | - Al Stewart
- Pfizer Research & Development, La Jolla, California 92121, United States
| | - Lawrence Updyke
- Pfizer Research & Development, Groton, Connecticut 06340, United States
| | - Patrick R Verhoest
- Pfizer Research & Development, Cambridge, Massachusetts 02139, United States
| | - Liuqing Wei
- Pfizer Research & Development, Groton, Connecticut 06340, United States
| | - Stephen W Wright
- Pfizer Research & Development, Groton, Connecticut 06340, United States
| | - Eddie Yang
- Pfizer Research & Development, Groton, Connecticut 06340, United States
| | - Qingyi Yang
- Pfizer Research & Development, Cambridge, Massachusetts 02139, United States
| | - Yuao Zhu
- Pfizer Research & Development, Pearl River, New York 10965, United States
| |
Collapse
|
26
|
Bath PM, Ball J, Boyd M, Gage H, Glover M, Godfrey M, Guthrie B, Hewitt J, Howard R, Jaki T, Juszczak E, Lasserson D, Leighton P, Leyland V, Shen Lim W, Logan P, Meakin G, Montgomery A, Ogollah R, Passmore P, Quinlan P, Rick C, Royal S, Shenkin SD, Upton C, Gordon AL. Lessons from the PROTECT-CH COVID-19 platform trial in care homes. Health Technol Assess 2025:1-26. [PMID: 40215170 PMCID: PMC12010236 DOI: 10.3310/mtrs8833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/23/2025] Open
Abstract
Background Coronavirus disease-2019 was associated with significant mortality and morbidity in care homes in 2020-1. Repurposed antiviral drugs might reduce morbidity and mortality through reducing viral transmission, infection, replication and inflammation. We aimed to compare the safety and efficacy of potential antiviral drugs in care home residents. Methods We designed a cluster-randomised, open-label, blinded end-point platform trial to test drugs in a postexposure prophylaxis paradigm. Participants aged 65+ years from United Kingdom care homes, with or without nursing, were eligible for participation. Care homes were to be allocated at random by computer to administer 42 days of antiviral agent (ciclesonide or niclosamide) plus standard care versus standard care alone to residents. The primary outcome at 60 days after randomisation comprised the most serious outcome, which was defined as all-cause mortality, all-cause hospitalisation, severe acute respiratory syndrome coronavirus 2 infection or no infection. Analysis would be by intention to treat using ordinal logistic regression. Other outcomes included individual components of the primary outcome, transmission, plus health economic and process evaluation outcomes. The planned sample size was 300 care homes corresponding to 9600 residents. With ~40% of care homes predicted to develop an outbreak during the trial, we needed to recruit 750 homes/24,000 residents. Results We initiated the trial including protocol, approvals, insurance, website, database, data algorithms, intervention selection and training materials. We built a network of principal investigators and staff (91) and care homes (299) to support the trial. However, we never contracted care homes or general practitioners since the trial was stopped in September 2021, as vaccination in care homes had significantly reduced infections. Multiple delays significantly delayed the start date, such as: (1) reduced prioritisation of pandemic trials in 2021; (2) cumbersome mechanisms for choosing the investigational medicinal products; (3) contracting between National Institute for Health and Care Research and the investigational medicinal product manufacturers; (4) publicising the investigational medicinal products; (5) identification of sufficient numbers of care homes; (6) identification and contracting with several thousand general practitioners; (7) limited research nurse availability and (8) identification of adequate insurance to cover care homes for research. Generic challenges included working across the four home nations with their different structures and regulations. Limitations The feasibility of contracting between the sponsor and the principal investigators, general practitioners and care homes; screening, consent and treatment of care home residents; data acquisition and the potential benefit of postexposure prophylaxis were never tested. Conclusions The success of vaccination meant that the role of postexposure prophylaxis of coronavirus disease-2019 in care home residents was not tested. Significant progress was made in developing the infrastructure and expertise necessary for a large-scale clinical trial of investigational medicinal products in United Kingdom care homes. Future work The role of postexposure prophylaxis of coronavirus disease-2019 in care home residents remains undefined. Significant logistical barriers to conducting research in care homes need to be removed urgently before future studies are possible. Further work is required to develop the infrastructure for clinical trials of investigational medicinal products in care homes. Serious consideration should be given to building and then hibernating a pandemic-ready platform trial suitable for care home research. Funding This article presents independent research funded by the National Institute for Health and Care Research (NIHR) Health Technology Assessment programme as award number NIHR133443.
Collapse
Affiliation(s)
- Philip M Bath
- Stroke Trials Unit, Mental Health & Clinical Neuroscience, University of Nottingham, Nottingham, UK
- Stroke, Nottingham University Hospitals NHS Trust, Queen's Medical Centre, Nottingham, UK
| | - Jonathan Ball
- Infections, Immunity and Microbes, School of Life Sciences, University of Nottingham, Nottingham, UK
| | - Matthew Boyd
- Division of Pharmacy Practice and Policy, School of Pharmacy, University of Nottingham, Nottingham, UK
| | - Heather Gage
- Department of Clinical and Experimental Medicine, Surrey Health Economics Centre, University of Surrey, Guildford, UK
| | - Matthew Glover
- Department of Clinical and Experimental Medicine, Surrey Health Economics Centre, University of Surrey, Guildford, UK
| | - Maureen Godfrey
- c/o Nottingham Clinical Trials Unit, University of Nottingham, Nottingham, UK
| | - Bruce Guthrie
- Advanced Care Research Centre, Usher Institute, University of Edinburgh, Edinburgh, UK
| | - Jonathan Hewitt
- Department of Population Medicine, Cardiff University, Cardiff, UK
| | - Robert Howard
- Division of Psychiatry, University College London, London, UK
| | - Thomas Jaki
- MRC Biostatistics Unit, University of Cambridge, Cambridge, UK
| | - Edmund Juszczak
- Nottingham Clinical Trials Unit, University of Nottingham, Nottingham, UK
| | - Daniel Lasserson
- Warwick Medical School, University of Warwick, Coventry, UK
- Acute Hospital at Home, Department of Geratology, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Paul Leighton
- Lifespan and Population Sciences, School of Medicine, University of Nottingham, Nottingham, UK
| | | | - Wei Shen Lim
- Respiratory Medicine, Nottingham University Hospitals NHS Trust, Nottingham, UK
| | - Pip Logan
- Unit of Injury, Inflammation and Recovery, School of Medicine, University of Nottingham, Nottingham, UK
- Nottingham City Care Partnership, Nottingham, UK
| | - Garry Meakin
- Nottingham Clinical Trials Unit, University of Nottingham, Nottingham, UK
| | - Alan Montgomery
- Nottingham Clinical Trials Unit, University of Nottingham, Nottingham, UK
| | - Reuben Ogollah
- Nottingham Clinical Trials Unit, University of Nottingham, Nottingham, UK
| | - Peter Passmore
- Centre for Public Health, Institute for Clinical Sciences, Queen's University Belfast, Belfast, UK
| | - Philip Quinlan
- Digital Health & Digital Research Service, University of Nottingham, Nottingham, UK
| | - Caroline Rick
- Nottingham Clinical Trials Unit, University of Nottingham, Nottingham, UK
| | - Simon Royal
- University of Nottingham Health Service, Cripps Health Centre, University Park, Nottingham, UK
| | - Susan D Shenkin
- Advanced Care Research Centre, Usher Institute, University of Edinburgh, Edinburgh, UK
- Ageing and Health, Usher Institute, University of Edinburgh, Edinburgh, UK
| | - Clare Upton
- Nottingham Clinical Trials Unit, University of Nottingham, Nottingham, UK
| | - Adam L Gordon
- NIHR Applied Research Collaboration-East Midlands (ARC-EM), Institute of Mental Health, Nottingham, UK
- Academic Unit of Injury, Recovery and Inflammation Sciences (IRIS), School of Medicine, University of Nottingham, Nottingham, UK
| |
Collapse
|
27
|
Su H, Wu G, Xiong M, Wang Y, Cao J, You M, Xiang Y, Nie T, Li M, Xiao G, Zhang L, Shao Q, Xu Y. Dynamic Cap-Mediated Substrate Access and Potent Inhibitor Design of Monkeypox Virus I7L Protease. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2501625. [PMID: 40193298 DOI: 10.1002/advs.202501625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Revised: 03/15/2025] [Indexed: 04/09/2025]
Abstract
Monkeypox virus (MPXV), an orthopoxvirus that has long been endemic in Africa, has posed a significant global health threat since 2022. The I7L protease, a highly conserved cysteine proteinase essential for orthopoxvirus replication, represents a promising target for broad-spectrum antiviral drug development. Here, the first crystal structure of MPXV I7L protease is reported, revealing its unique dimeric form and different conformations of a cap region nearby the active site. Molecular dynamics simulations and AlphaFold3 prediction of protease-substrate structures both suggest that this highly flexible cap acts as a conformational switch, regulating the substrate access to the active site. Additionally, the structural basis of substrate recognition and the catalytic mechanism of the protease are elucidated, mapping determinants of substrate specificity. These insights enable us to design covalent inhibitors to mimic the natural substrates and develop a fluorescence resonance energy transfer (FRET)-based protease assay to effectively assess the inhibitory activity, leading to the discovery of first-in-class inhibitors of MPXV I7L protease with nanomolar potency. Therefore, this work provides a comprehensive understanding of the MPXV I7L protease's structure, dynamics, and function, and presents an example of successful rational design of covalent peptidomimetic inhibitors, serving as a good starting point for drug development against MPXV.
Collapse
Affiliation(s)
- Haixia Su
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Guoqing Wu
- Lingang Laboratory, Shanghai, 200031, China
- School of Physical Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Muya Xiong
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
| | - Yuhang Wang
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Junyuan Cao
- CAS Key Laboratory of Special Pathogens, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430064, China
- Hubei Jiangxia Laboratory, Wuhan, 430200, China
| | - Mengyuan You
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | | | - Tianqing Nie
- Lingang Laboratory, Shanghai, 200031, China
- School of Physical Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Minjun Li
- Shanghai Synchrotron Radiation Facility, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai, 201204, China
| | - Gengfu Xiao
- CAS Key Laboratory of Special Pathogens, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430064, China
| | - Leike Zhang
- CAS Key Laboratory of Special Pathogens, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430064, China
- Hubei Jiangxia Laboratory, Wuhan, 430200, China
| | - Qiang Shao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yechun Xu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| |
Collapse
|
28
|
Gu L, Wang ZJ, Zhang XR, Liu Y, Zhao M, Jiang SZ, Pan J, Yuan Y, Cai H, Zhou T, Li T, Li TT, Xue W. Targeting the liquid-liquid phase separation of nucleocapsid broadly inhibits the replication of SARS-CoV-2 strains. Biochem Biophys Res Commun 2025; 756:151594. [PMID: 40086356 DOI: 10.1016/j.bbrc.2025.151594] [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: 02/26/2025] [Accepted: 03/05/2025] [Indexed: 03/16/2025]
Abstract
The COVID-19 pandemic, caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has led to a global public health crisis. The nucleocapsid (N) protein plays a pivotal role in a variety of biological processes in the life cycle of SARS-CoV-2, such as viral assembly. In this study, we investigated the liquid-liquid phase separation (LLPS) capacity of the N protein of seven SARS-CoV-2 strains, including the variants of concern (VOC) and interest (VOI), and its impact on viral replication. Using bioinformatic tools, we analyzed 11,433,558 complete genomes of SARS-CoV-2 and revealed a high degree of sequence conservation of N gene. While all the seven N proteins could undergo LLPS with RNA, the mutations in N impair its capacity of LLPS. With a SARS-CoV-2 trans-complementation system, we showed that SARS-CoV-2 variants carrying mutated N proteins exhibit impaired replication, highlighting the importance of LLPS of N in viral replication. We further demonstrated that (-)-gallocatechin gallate (GCG) efficiently inhibits the LLPS of N proteins and significantly suppresses the replication of different SARS-CoV-2 strains. Thus, our findings indicate that targeting the N-LLPS could be a viable strategy for the development of antiviral treatments against various SARS-CoV-2 strains, including those yet to emerge.
Collapse
Affiliation(s)
- Lin Gu
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100850, China
| | - Zheng-Jie Wang
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100850, China
| | - Xin-Ran Zhang
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100850, China
| | - Yu Liu
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100850, China
| | - Ming Zhao
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100850, China
| | - Shao-Zhen Jiang
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100850, China; School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Jie Pan
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100850, China
| | - Ying Yuan
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100850, China; Institute of Translational Medicine, Zhejiang University, Hangzhou, 310029, China
| | - Hong Cai
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100850, China
| | - Tao Zhou
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100850, China; Institute of Translational Medicine, Zhejiang University, Hangzhou, 310029, China
| | - Tao Li
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100850, China; School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China; Institute of Translational Medicine, Zhejiang University, Hangzhou, 310029, China
| | - Ting-Ting Li
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100850, China.
| | - Wen Xue
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100850, China.
| |
Collapse
|
29
|
Li G, Wu Y, Zhou F. Targeting SARS-CoV-2 NSP14 with TDI-015051: a new frontier in antiviral therapy. Sci Bull (Beijing) 2025:S2095-9273(25)00357-3. [PMID: 40263052 DOI: 10.1016/j.scib.2025.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/24/2025]
Affiliation(s)
- Guoxiang Li
- The First Affiliated Hospital, the Institutes of Biology and Medical Sciences, Suzhou Medical College, Soochow University, Suzhou 215000, China; Life Sciences Institute and State Key Laboratory of Transvascular Implantation Devices of the Second Affiliated Hospital of the Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310000, China
| | - Yiran Wu
- Life Sciences Institute and State Key Laboratory of Transvascular Implantation Devices of the Second Affiliated Hospital of the Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310000, China
| | - Fangfang Zhou
- The First Affiliated Hospital, the Institutes of Biology and Medical Sciences, Suzhou Medical College, Soochow University, Suzhou 215000, China.
| |
Collapse
|
30
|
M Bader S, Calleja DJ, Devine SM, Kuchel NW, Lu BGC, Wu X, Birkinshaw RW, Bhandari R, Loi K, Volpe R, Khakham Y, Au AE, Blackmore TR, Mackiewicz L, Dayton M, Schaefer J, Scherer L, Stock AT, Cooney JP, Schoffer K, Maluenda A, Kleeman EA, Davidson KC, Allison CC, Ebert G, Chen G, Katneni K, Klemm TA, Nachbur U, Georgy SR, Czabotar PE, Hannan AJ, Putoczki TL, Tanzer M, Pellegrini M, Lechtenberg BC, Charman SA, Call MJ, Mitchell JP, Lowes KN, Lessene G, Doerflinger M, Komander D. A novel PLpro inhibitor improves outcomes in a pre-clinical model of long COVID. Nat Commun 2025; 16:2900. [PMID: 40180914 PMCID: PMC11969009 DOI: 10.1038/s41467-025-57905-4] [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: 12/18/2024] [Accepted: 03/06/2025] [Indexed: 04/05/2025] Open
Abstract
The COVID-19 pandemic caused by the coronavirus SARS-CoV-2 has highlighted the vulnerability of a globally connected population to zoonotic viruses. The FDA-approved coronavirus antiviral Paxlovid targets the essential SARS-CoV-2 main protease, Mpro. Whilst effective in the acute phase of a COVID infection, Paxlovid cannot be used by all patients, can lead to viral recurrence, and does not protect against post-acute sequelae of COVID-19 (PASC), commonly known as long COVID, an emerging significant health burden that remains poorly understood and untreated. Alternative antivirals that are addressing broader patient needs are urgently required. We here report our drug discovery efforts to target PLpro, a further essential coronaviral protease, for which we report a novel chemical scaffold that targets SARS-CoV-2 PLpro with low nanomolar activity, and which exhibits activity against PLpro of other pathogenic coronaviruses. Our lead compound shows excellent in vivo efficacy in a mouse model of severe acute disease. Importantly, our mouse model recapitulates long-term pathologies matching closely those seen in PASC patients. Our lead compound offers protection against a range of PASC symptoms in this model, prevents lung pathology and reduces brain dysfunction. This provides proof-of-principle that PLpro inhibition may have clinical relevance for PASC prevention and treatment going forward.
Collapse
Affiliation(s)
- Stefanie M Bader
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Dale J Calleja
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Shane M Devine
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.
- Department of Medical Biology, University of Melbourne, Melbourne, Australia.
| | - Nathan W Kuchel
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Bernadine G C Lu
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Xinyu Wu
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Richard W Birkinshaw
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Reet Bhandari
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Katie Loi
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Rohan Volpe
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Yelena Khakham
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Amanda E Au
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Timothy R Blackmore
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Liana Mackiewicz
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Merle Dayton
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Jan Schaefer
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Lena Scherer
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Angus T Stock
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - James P Cooney
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Kael Schoffer
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Ana Maluenda
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Elizabeth A Kleeman
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Kathryn C Davidson
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Cody C Allison
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Gregor Ebert
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Gong Chen
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Kasiram Katneni
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Theresa A Klemm
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Ueli Nachbur
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Smitha Rose Georgy
- Anatomic Pathology - Veterinary Biosciences, Melbourne Veterinary School, University of Melbourne, Werribee, VIC, Australia
| | - Peter E Czabotar
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Anthony J Hannan
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
- Department of Anatomy and Physiology, University of Melbourne, Parkville, VIC, Australia
| | - Tracy L Putoczki
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
- Department of Surgery, University of Melbourne, Melbourne, Australia
| | - Maria Tanzer
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Marc Pellegrini
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
- Centenary Institute of Cancer Medicine and Cell Biology, Camperdown, NSW, Australia
| | - Bernhard C Lechtenberg
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Susan A Charman
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Melissa J Call
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Jeffrey P Mitchell
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Kym N Lowes
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Guillaume Lessene
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.
- Department of Medical Biology, University of Melbourne, Melbourne, Australia.
- Department of Biochemistry and Pharmacology, University of Melbourne, Melbourne, Australia.
| | - Marcel Doerflinger
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.
- Department of Medical Biology, University of Melbourne, Melbourne, Australia.
| | - David Komander
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.
- Department of Medical Biology, University of Melbourne, Melbourne, Australia.
| |
Collapse
|
31
|
Casalino L, Ramos-Guzmán CA, Amaro RE, Simmerling C, Lodola A, Mulholland AJ, Świderek K, Moliner V. A Reflection on the Use of Molecular Simulation to Respond to SARS-CoV-2 Pandemic Threats. J Phys Chem Lett 2025; 16:3249-3263. [PMID: 40118074 PMCID: PMC11973918 DOI: 10.1021/acs.jpclett.4c03654] [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: 12/22/2024] [Revised: 02/19/2025] [Accepted: 02/26/2025] [Indexed: 03/23/2025]
Abstract
Molecular simulations play important roles in understanding the lifecycle of the SARS-CoV-2 virus and contribute to the design and development of antiviral agents and diagnostic tests for COVID. Here, we discuss the insights that such simulations have provided and the challenges involved, focusing on the SARS-CoV-2 main protease (Mpro) and the spike glycoprotein. Mpro is the leading target for antivirals, while the spike glycoprotein is the target for vaccine design. Finally, we reflect on lessons from this pandemic for the simulation community. Data sharing initiatives and collaborations across the international research community contributed to advancing knowledge and should be built on to help in future pandemics and other global challenges such as antimicrobial resistance.
Collapse
Affiliation(s)
- Lorenzo Casalino
- Department
of Molecular Biology, University of California
San Diego, La Jolla, California 92093, United States
| | - Carlos A. Ramos-Guzmán
- Centre
for Computational Chemistry, School of Chemistry, University of Bristol, Bristol BS8 1TS, United
Kingdom
| | - Rommie E. Amaro
- Department
of Molecular Biology, University of California
San Diego, La Jolla, California 92093, United States
| | - Carlos Simmerling
- Department
of Chemistry and Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, New York 11794-3400, United States
| | - Alessio Lodola
- Dipartimento
di Scienze degli Alimenti e del Farmaco, Università degli Studi di Parma, I 43121 Parma, Italy
| | - Adrian J. Mulholland
- Centre
for Computational Chemistry, School of Chemistry, University of Bristol, Bristol BS8 1TS, United
Kingdom
| | - Katarzyna Świderek
- Biocomp
group, Institute of Advanced Materials (INAM), Universitat Jaume I, 12071 Castelló, Spain
| | - Vicent Moliner
- Biocomp
group, Institute of Advanced Materials (INAM), Universitat Jaume I, 12071 Castelló, Spain
| |
Collapse
|
32
|
Dong B, Chen Y, Wang X, Li J, Zhang S, Kang X, Li Y, Li B, Liao L, Zhang Z, Xiong J, Shao L, Huang S, Feng Y, Jiang T. Development of a highly sensitive luciferase assay for intracellular evaluation of coronavirus Mpro activity. Front Microbiol 2025; 16:1560251. [PMID: 40241735 PMCID: PMC12000094 DOI: 10.3389/fmicb.2025.1560251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 03/06/2025] [Indexed: 04/18/2025] Open
Abstract
COVID-19, caused by SARS-CoV-2 virus, has emerged as a global threat to human health. The main protease (Mpro) of SARS-CoV-2 is an excellent target for the development of antiviral drugs against COVID-19, and various protease biosensors have been developed to evaluate anti-coronavirus drugs. However, the application of these protease biosensors was limited due to high background fluorescence, poor signal-to-noise ratios, and constraints in enzyme activity thresholds for accessing live viruses. In this study, we rationally designed a highly conserved Mpro cleavage site sequence among different coronaviruses (CoVs) with high proteolytic activity, and described an intracellular coronavirus Mpro proteolytic (ICMP) reporter system that takes advantage of virus-encoded Mpro expressed in infected cells to reform the NanoBiT fluorescent protein. The system can be used to visualize and identify cells infected with coronavirus, and demonstrated high compatibility with various Mpro proteins from 13 different mammalian coronaviruses (covering α, β, γ, and δ CoVs), exhibiting at least a 1,030-fold increase in luminescence. Stronger Nluc signals were detectable with CoV 229E virus infection at a MOI of 0.001. Additionally, the system proved suitable for evaluating and screening of antiviral compounds, including lufotrelvir, GC376, Nirmatrelvir, X77, MG-101, and the potential inhibitor Cynaroside. The ICMP system is not only an invaluable tool for the detection of live coronaviruses, but also for the discovery of antivirals against current and future pandemic coronaviruses.
Collapse
Affiliation(s)
- Bao Dong
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| | - Yuehong Chen
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| | - Xin Wang
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, China
| | - Jing Li
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| | - Sen Zhang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| | - Xiaoping Kang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| | - Yuchang Li
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| | - Biao Li
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Liangning Liao
- School of Public Health, Mudanjiang Medical University, Mudanjiang, China
| | - Zhengwei Zhang
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Jiaqi Xiong
- School of Public Health, Mudanjiang Medical University, Mudanjiang, China
| | - Lele Shao
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Shenghai Huang
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Ye Feng
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| | - Tao Jiang
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
- School of Public Health, Mudanjiang Medical University, Mudanjiang, China
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| |
Collapse
|
33
|
Zhao L, Li C, Wang M, Zhou M, Jiang L, Zhang W, Yu J, Wang W, Zhou K, Pan K, Lam HY, Hung IFN, Chan KH, Liu L, Wang F, Zhao X, Chen Y. Potent antiviral activity of simnotrelvir against key epidemic SARS-CoV-2 variants with a high resistance barrier. Antimicrob Agents Chemother 2025; 69:e0155624. [PMID: 40062859 PMCID: PMC11963564 DOI: 10.1128/aac.01556-24] [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: 10/24/2024] [Accepted: 02/04/2025] [Indexed: 04/03/2025] Open
Abstract
Simnotrelvir is an oral small-molecule antiviral agent targeting the 3C-like protease (3CLpro) of SARS-CoV-2, proven effective against the Delta variant with favorable pharmacokinetics and safety in preclinical study. In this study, we further evaluated the antiviral efficacy of simnotrelvir against a range of emerging Omicron variants, including BA.1, BA.4, BA.5, CH.1.1, XBB.1.5, XBB.1.16, EG.5, and JN.1. In vitro assays with Vero E6 cells confirmed that simnotrelvir exhibited robust antiviral activity across these variants, comparable to the Food and Drug Administration (FDA)-approved drug nirmatrelvir. Additionally, simnotrelvir demonstrated effective inhibition against several nirmatrelvir-resistant SARS-CoV-2 3CLpro mutants, including A260V, Y54A, (T21I + S144A), F140A, H172Y, and E166V. Importantly, simnotrelvir showed better potency against the E166V mutation compared to nirmatrelvir. Resistance selection studies revealed that BA.5 developed reduced sensitivity after 5 and 10 passages, increasing the IC50 values by 3.2 and 4.5-fold, respectively, while HCoV-OC43 showed an 8.3-fold increase after 12 passages. Despite this, simnotrelvir's overall efficacy remains strong. Furthermore, clinical trials demonstrated that combining simnotrelvir with ritonavir significantly shortened symptom resolution in COVID-19 patients. Genomic analysis of treated patients found random nucleotide substitutions but no significant mutations linked to 3CLpro resistance. In conclusion, simnotrelvir shows strong antiviral activity against SARS-CoV-2 variants and maintains a high barrier to resistance, reinforcing its potential as an effective therapeutic option for current and future SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Liwei Zhao
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Chuang Li
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Mengyu Wang
- Jiangsu Simcere Pharmaceutical Company Limited, Nanjing, Jiangsu, China
- State Key Laboratory of Neurology and Oncology Drug Development, Nanjing, China
| | - Minyun Zhou
- Jiangsu Simcere Pharmaceutical Company Limited, Nanjing, Jiangsu, China
- State Key Laboratory of Neurology and Oncology Drug Development, Nanjing, China
| | - Lei Jiang
- Simcere Zaiming Pharmaceutical Company Limited, Shanghai, China
- Department of Chemical Engineering, Tsinghua University, Beijing, China
| | - Wanying Zhang
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jie Yu
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Wei Wang
- Jiangsu Simcere Pharmaceutical Company Limited, Nanjing, Jiangsu, China
| | - Kangping Zhou
- Hubei Provincial Center for Disease Control and Prevention, Wuhan, Hubei, China
| | - Kai Pan
- Hubei Provincial Center for Disease Control and Prevention, Wuhan, Hubei, China
| | - Hoi-Yan Lam
- Department of Microbiology, State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection,The University of Hong Kong, Hong Kong SAR, China
| | - Ivan Fan-Ngai Hung
- Department of Microbiology, State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection,The University of Hong Kong, Hong Kong SAR, China
- Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong SAR, China
| | - Kwok-Hung Chan
- Department of Microbiology, State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection,The University of Hong Kong, Hong Kong SAR, China
- Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong SAR, China
| | - Lian Liu
- Jiangsu Simcere Pharmaceutical Company Limited, Nanjing, Jiangsu, China
- State Key Laboratory of Neurology and Oncology Drug Development, Nanjing, China
| | - Feng Wang
- Jiangsu Simcere Pharmaceutical Company Limited, Nanjing, Jiangsu, China
- State Key Laboratory of Neurology and Oncology Drug Development, Nanjing, China
| | - Xiaofeng Zhao
- Jiangsu Simcere Pharmaceutical Company Limited, Nanjing, Jiangsu, China
- State Key Laboratory of Neurology and Oncology Drug Development, Nanjing, China
| | - Yuxin Chen
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing, Jiangsu, China
| |
Collapse
|
34
|
Yu H, Chen T, Li J, Zhang X, Wu Y. Effectiveness of nirmatrelvir/ritonavir in hospitalized haematological malignancy patients with mild-to-moderate COVID-19: A retrospective study. Br J Haematol 2025; 206:1077-1085. [PMID: 40032259 DOI: 10.1111/bjh.20039] [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: 11/10/2024] [Accepted: 02/21/2025] [Indexed: 03/05/2025]
Abstract
Patients with haematological malignancies (HMs) are highly vulnerable to COVID-19 due to their immunocompromised status, which leads to prolonged viral clearance and severe outcomes. Nirmatrelvir/ritonavir has shown efficacy in reducing severity and mortality in high-risk COVID-19 outpatients, but its effectiveness in hospitalized HM patients remains unclear. We conducted a retrospective study to assess the effectiveness of nirmatrelvir/ritonavir on mortality and viral clearance in hospitalized HM patients with mild-to-moderate COVID-19 during China's first COVID-19 surge. Mortality rate and viral clearance time were the primary end-points. Cox proportional hazards models were used to detect factors associated with mortality and viral clearance. A total of 116 HM patients, with a median age of 47.2 years, hospitalized for a minimum of 5 days with mild-to-moderate COVID-19, were included in this study. There was no difference in the 90-day mortality rate between HM patients treated with nirmatrelvir/ritonavir within 5 days and those not treated (4.9% vs. 5.3%, p = 1.000). Nirmatrelvir/ritonavir use within 5 days reduced the time to viral clearance (hazard ratio [HR] = 1.59, 95% confidence interval [CI] 1.04-2.42). Nirmatrelvir/ritonavir use within 5 days in hospitalized HM patients with mild-to-moderate COVID-19 does not reduce mortality but accelerates viral clearance.
Collapse
Affiliation(s)
- Hongbin Yu
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Tian Chen
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Jiawei Li
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Xin Zhang
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Yu Wu
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
35
|
Van Damme E, Abeywickrema P, Yin Y, Xie J, Jacobs S, Mann MK, Doijen J, Miller R, Piassek M, Marsili S, Subramanian M, Gottlieb L, Abdelnabi R, Van Gool M, Van den Broeck N, De Pauw I, Diels A, Vermeulen P, Temmerman K, Scobey T, Mattocks M, Schäfer A, Jochmans D, De Jonghe S, Leyssen P, Chiu W, Diosa Toro M, Zwaagstra M, Leijs AA, De Gruyter HLM, Buyck C, Van Den Heede K, Jacobs F, Van den Eynde C, Thijs L, Raeymaekers V, Miller S, Del Rosario A, Neyts J, Peeters D, Baric RS, van Kuppeveld FJM, Snijder EJ, van Hemert MJ, Monshouwer M, Sharma S, Draghia-Akli R, Koul A, Van Loock M. A small-molecule SARS-CoV-2 inhibitor targeting the membrane protein. Nature 2025; 640:506-513. [PMID: 40140563 PMCID: PMC11981937 DOI: 10.1038/s41586-025-08651-6] [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: 02/21/2024] [Accepted: 01/15/2025] [Indexed: 03/28/2025]
Abstract
The membrane (M) protein of betacoronaviruses is well conserved and has a key role in viral assembly1,2. Here we describe the identification of JNJ-9676, a small-molecule inhibitor targeting the coronavirus M protein. JNJ-9676 demonstrates in vitro nanomolar antiviral activity against SARS-CoV-2, SARS-CoV and sarbecovirus strains from bat and pangolin zoonotic origin. Using cryogenic electron microscopy (cryo-EM), we determined a binding pocket of JNJ-9676 formed by the transmembrane domains of the M protein dimer. Compound binding stabilized the M protein dimer in an altered conformational state between its long and short forms, preventing the release of infectious virus. In a pre-exposure Syrian golden hamster model, JNJ-9676 (25 mg per kg twice per day) showed excellent efficacy, illustrated by a significant reduction in viral load and infectious virus in the lung by 3.5 and 4 log10-transformed RNA copies and 50% tissue culture infective dose (TCID50) per mg lung, respectively. Histopathology scores at this dose were reduced to the baseline. In a post-exposure hamster model, JNJ-9676 was efficacious at 75 mg per kg twice per day even when added at 48 h after infection, when peak viral loads were observed. The M protein is an attractive antiviral target to block coronavirus replication, and JNJ-9676 represents an interesting chemical series towards identifying clinical candidates addressing the current and future coronavirus pandemics.
Collapse
Affiliation(s)
- Ellen Van Damme
- Global Public Health R&D, Janssen Pharmaceutica, Beerse, Belgium
| | - Pravien Abeywickrema
- Discovery Technologies & Molecular Pharmacology, Janssen Research & Development, Spring House, PA, USA
| | - Yanting Yin
- Discovery Technologies & Molecular Pharmacology, Janssen Research & Development, Spring House, PA, USA
| | - Jiexiong Xie
- Global Public Health R&D, Janssen Pharmaceutica, Beerse, Belgium
| | - Sofie Jacobs
- Global Public Health R&D, Janssen Pharmaceutica, Beerse, Belgium
| | - Mandeep Kaur Mann
- Global Public Health R&D, Janssen Research & Development, Spring House, PA, USA
| | - Jordi Doijen
- Global Public Health R&D, Janssen Pharmaceutica, Beerse, Belgium
| | - Robyn Miller
- Discovery Technologies & Molecular Pharmacology, Janssen Research & Development, Spring House, PA, USA
| | - Madison Piassek
- Discovery Technologies & Molecular Pharmacology, Janssen Research & Development, Spring House, PA, USA
| | | | - Murali Subramanian
- Translational PK/PD & Investigative Toxicology (TPPIT), Janssen Research & Development, Beerse, Belgium
- Gilead Sciences, Foster City, CA, USA
| | - Leah Gottlieb
- Discovery Technologies & Molecular Pharmacology, Janssen Research & Development, Spring House, PA, USA
- Red Nucleus, Philadelphia, PA, USA
| | - Rana Abdelnabi
- Virology, Antiviral Drug & Vaccine Research Group, Department of Microbiology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
- VirusBank Platform, Leuven, Belgium
| | | | | | | | - Annick Diels
- Discovery Technologies & Molecular Pharmacology, Janssen Research & Development, Beerse, Belgium
| | - Peter Vermeulen
- Discovery Technologies & Molecular Pharmacology, Janssen Research & Development, Beerse, Belgium
| | - Koen Temmerman
- Discovery Technologies & Molecular Pharmacology, Janssen Research & Development, Beerse, Belgium
| | - Trevor Scobey
- Department of Epidemiology, University of North Carolina at Chapel Hill Gillings School of Global Public Health, Chapel Hill, NC, USA
| | - Melissa Mattocks
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
| | - Alexandra Schäfer
- Department of Epidemiology, University of North Carolina at Chapel Hill Gillings School of Global Public Health, Chapel Hill, NC, USA
| | - Dirk Jochmans
- Virology, Antiviral Drug & Vaccine Research Group, Department of Microbiology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Steven De Jonghe
- Virology, Antiviral Drug & Vaccine Research Group, Department of Microbiology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Pieter Leyssen
- Virology, Antiviral Drug & Vaccine Research Group, Department of Microbiology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Winston Chiu
- Virology, Antiviral Drug & Vaccine Research Group, Department of Microbiology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Mayra Diosa Toro
- Virology Section, Division of Infectious Diseases and Immunology, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
- Eurofins BioPharma Product Testing, Leiden, The Netherlands
| | - Marleen Zwaagstra
- Virology Section, Division of Infectious Diseases and Immunology, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Anouk A Leijs
- Molecular Virology Laboratory, Leiden University Center of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Heidi L M De Gruyter
- Molecular Virology Laboratory, Leiden University Center of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Christophe Buyck
- In Silico Discovery (ISD), Computer-Aided Drug Design (CADD), Janssen Pharmaceutica, Beerse, Belgium
| | - Klaas Van Den Heede
- Global Public Health R&D, Janssen Pharmaceutica, Beerse, Belgium
- Independent Researcher, Mechelen, Belgium
| | - Frank Jacobs
- Translational PK/PD & Investigative Toxicology (TPPIT), Janssen Research & Development, Beerse, Belgium
| | | | | | | | - Seth Miller
- Discovery Technologies & Molecular Pharmacology, Janssen Research & Development, Spring House, PA, USA
- Spark Therapeutics, Philadelphia, PA, USA
| | - Amanda Del Rosario
- Discovery Technologies & Molecular Pharmacology, Janssen Research & Development, Spring House, PA, USA
| | - Johan Neyts
- Virology, Antiviral Drug & Vaccine Research Group, Department of Microbiology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
- VirusBank Platform, Leuven, Belgium
| | - Danielle Peeters
- Discovery Technologies & Molecular Pharmacology, Janssen Research & Development, Beerse, Belgium
| | - Ralph S Baric
- Department of Epidemiology, University of North Carolina at Chapel Hill Gillings School of Global Public Health, Chapel Hill, NC, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
| | - Frank J M van Kuppeveld
- Virology Section, Division of Infectious Diseases and Immunology, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Eric J Snijder
- Molecular Virology Laboratory, Leiden University Center of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Martijn J van Hemert
- Molecular Virology Laboratory, Leiden University Center of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Mario Monshouwer
- Translational PK/PD & Investigative Toxicology (TPPIT), Janssen Research & Development, Beerse, Belgium
| | - Sujata Sharma
- Discovery Technologies & Molecular Pharmacology, Janssen Research & Development, Spring House, PA, USA
| | - Ruxandra Draghia-Akli
- Global Public Health R&D, Janssen Research & Development, Spring House, PA, USA.
- Research & Development, Novavax Inc., Gaithersburg, MD, USA.
| | - Anil Koul
- Global Public Health R&D, Janssen Pharmaceutica, Beerse, Belgium.
| | - Marnix Van Loock
- Global Public Health R&D, Janssen Pharmaceutica, Beerse, Belgium.
| |
Collapse
|
36
|
Böger CR, Martens-Lobenhoffer J, Worthmann H, Stichtenoth DO, Brod T. Therapeutic Drug Monitoring of Nirmatrelvir/Ritonavir (Paxlovid) in Patients Treated for COVID-19: Results From a Prospective Multicenter Observational Study. Ther Drug Monit 2025; 47:258-264. [PMID: 39625118 DOI: 10.1097/ftd.0000000000001290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 07/05/2024] [Indexed: 03/15/2025]
Abstract
BACKGROUND Paxlovid is a combination of the antiviral agents nirmatrelvir and ritonavir indicated for the oral treatment of high-risk, symptomatic patients with coronavirus disease 2019 (COVID-19). As real-world data on the plasma concentrations of nirmatrelvir/ritonavir (Paxlovid) are limited, the aim of this study was to investigate nirmatrelvir/ritonavir plasma trough levels in a clinical setting using therapeutic drug monitoring. METHODS A prospective, noninterventional, multicenter, observational clinical study was conducted in which the plasma trough levels of nirmatrelvir/ritonavir were simultaneously determined by using liquid chromatography tandem mass spectrometry in patients with symptomatic COVID-19. The blood samples were collected on days 1, 3, and 5 after the first full-dose day (day 0), and patient data such as sex, height, weight, renal function, liver enzymes, and concomitant (co-) medications were obtained to describe the plasma levels with respect to potential influencing factors. RESULTS A total of 46 blood samples from 21 patients were analyzed. The geometric mean C min was 4997 ng/mL for nirmatrelvir and 529.4 ng/mL for ritonavir. The plasma concentrations covered a wide range, the highest being observed in patients with advanced age and renally excreted comedications. Patients older than 65 years had a significantly higher risk of achieving excessive plasma trough concentrations above 8840 ng/mL for nirmatrelvir and 1440 ng/mL for ritonavir compared with younger patients (odds ratio 11.2, 95% confidence interval 1.04-120.4). CONCLUSIONS The plasma trough concentrations of nirmatrelvir and ritonavir in patients treated for symptomatic COVID-19 were higher than the reference values of 2210 ng/mL for nirmatrelvir and 360 ng/mL for ritonavir stated in the product characteristics. Advanced age and renally eliminated comedication were identified as possible influencing factors that warrant further investigation.
Collapse
Affiliation(s)
- Corinna R Böger
- Institute of Clinical Pharmacology, Hannover Medical School, Hannover, Germany
| | | | - Hans Worthmann
- Department of Neurology, Hannover Medical School, Hannover, Germany ; and
| | - Dirk O Stichtenoth
- Institute of Clinical Pharmacology, Hannover Medical School, Hannover, Germany
| | - Torben Brod
- Department of Respiratory Medicine, Hannover Medical School, Hannover, Germany
| |
Collapse
|
37
|
Petcherski A, Tingley BM, Martin A, Adams S, Brownstein AJ, Steinberg RA, Shabane B, Ngo J, Osto C, Garcia G, Veliova M, Arumugaswami V, Colby AH, Shirihai OS, Grinstaff MW. Endolysosome-targeted nanoparticle delivery of antiviral therapy for coronavirus infections. Life Sci Alliance 2025; 8:e202403182. [PMID: 39900438 PMCID: PMC11790838 DOI: 10.26508/lsa.202403182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 01/22/2025] [Accepted: 01/22/2025] [Indexed: 02/05/2025] Open
Abstract
SARS-CoV-2 can infect cells through endocytic uptake, a process that is targeted by inhibition of lysosomal proteases. However, clinically this approach to treat viral infections has afforded mixed results, with some studies detailing an oral regimen of hydroxychloroquine accompanied by significant off-target toxicities. We rationalized that an organelle-targeted approach will avoid toxicity while increasing the concentration of the drug at the target. Here, we describe a lysosome-targeted, mefloquine-loaded poly(glycerol monostearate-co-ε-caprolactone) nanoparticle (MFQ-NP) for pulmonary delivery via inhalation. Mefloquine is a more effective inhibitor of viral endocytosis than hydroxychloroquine in cellular models of COVID-19. MFQ-NPs are less toxic than molecular mefloquine, are 100-150 nm in diameter, and possess a negative surface charge, which facilitates uptake via endocytosis allowing inhibition of lysosomal proteases. MFQ-NPs inhibit coronavirus infection in mouse MHV-A59 and human OC43 coronavirus model systems and inhibit SARS-CoV-2 WA1 and its Omicron variant in a human lung epithelium model. Organelle-targeted delivery is an effective means to inhibit viral infection.
Collapse
Affiliation(s)
- Anton Petcherski
- Department of Medicine, Division of Endocrinology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Brett M Tingley
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Andrew Martin
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Sarah Adams
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Alexandra J Brownstein
- Department of Medicine, Division of Endocrinology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- Molecular Cellular Integrative Physiology, University of California Los Angeles, Los Angeles, CA, USA
| | - Ross A Steinberg
- Department of Medicine, Division of Endocrinology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Byourak Shabane
- Department of Medicine, Division of Endocrinology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Jennifer Ngo
- Department of Medicine, Division of Endocrinology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Corey Osto
- Department of Medicine, Division of Endocrinology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Gustavo Garcia
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Michaela Veliova
- Department of Medicine, Division of Endocrinology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Vaithilingaraja Arumugaswami
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Aaron H Colby
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Orian S Shirihai
- Department of Medicine, Division of Endocrinology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Mark W Grinstaff
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
- Department of Chemistry, Boston University, Boston, MA, USA
| |
Collapse
|
38
|
Hennecker C, Venegas F, Wang G, Stille J, Milaczewska A, Moitessier N, Mittermaier A. Mechanistic Characterization of Covalent Enzyme Inhibition by Isothermal Titration Calorimetry Kinetic Competition (ITC-KC). Anal Chem 2025; 97:6368-6381. [PMID: 40106584 DOI: 10.1021/acs.analchem.4c04003] [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: 03/22/2025]
Abstract
Covalent enzyme inhibitors can offer high potency and specificity and are increasingly sought after in drug discovery. They typically inhibit in two steps: noncovalent binding followed by covalent bond formation. Rational optimization requires quantitative information on both steps. Current methods for measuring these steps are technically demanding, time-consuming, and are not well suited for routine insertion into drug discovery pipelines. We have developed a new approach, using isothermal titration calorimetry kinetic competition (ITC-KC), that overcomes many of these challenges. ITC-KC measures enzyme activity directly, via the heat flow generated during catalysis, making it a sensitive and nearly universal approach. We performed extensive numerical simulations in which ITC-KC outperformed current methods with 3- to 10-fold greater accuracy. We applied ITC-KC to a library of 19 inhibitors of the protease 3CLpro from SARS-CoV-2 and found that the reactive warheads and noncovalent binding portions of these molecules influenced the two-step inhibition mechanism in complex and unpredictable ways. This highlights the need for detailed mechanistic information in the development of covalent inhibitors, information that ITC-KC can provide rapidly, accurately, and essentially universally.
Collapse
Affiliation(s)
- Christopher Hennecker
- Department of Chemistry, McGill University.801 Sherbrooke St. W. Montreal, Montreal H3A 0B8, Canada
| | - Felipe Venegas
- Department of Chemistry, McGill University.801 Sherbrooke St. W. Montreal, Montreal H3A 0B8, Canada
| | - Guanyu Wang
- Department of Chemistry, McGill University.801 Sherbrooke St. W. Montreal, Montreal H3A 0B8, Canada
| | - Julia Stille
- Department of Chemistry, McGill University.801 Sherbrooke St. W. Montreal, Montreal H3A 0B8, Canada
| | - Anna Milaczewska
- Jerzy Haber Institute of Catalysis and Surface Chemistry, Polish Academy of Sciences, Niezapominajek 8, Kraków 30-239, Poland
| | - Nicolas Moitessier
- Department of Chemistry, McGill University.801 Sherbrooke St. W. Montreal, Montreal H3A 0B8, Canada
| | - Anthony Mittermaier
- Department of Chemistry, McGill University.801 Sherbrooke St. W. Montreal, Montreal H3A 0B8, Canada
| |
Collapse
|
39
|
Ng TM, Wang Z, Chan ECY. Physiologically-based pharmacokinetic modelling guided dose evaluations of nirmatrelvir/ritonavir in renal impairment for the management of COVID-19. Br J Clin Pharmacol 2025; 91:1041-1048. [PMID: 38616514 DOI: 10.1111/bcp.16074] [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: 12/20/2023] [Revised: 03/06/2024] [Accepted: 03/21/2024] [Indexed: 04/16/2024] Open
Abstract
We aimed to address factors contributing to the pharmacokinetic changes of nirmatrelvir/ritonavir in renal impaired (RI) patients and recommend dosing adjustment via a physiologically-based pharmacokinetic (PBPK) modelling approach. A PBPK model of nirmatrelvir/ritonavir was developed via Simcyp® Simulator. Sensitivity analysis of the influence of hepatic CYP3A4 intrinsic clearance and abundance, as well as hepatic non-CYP3A4 metabolism (other human liver microsomes [HLM] CLint) was performed to evaluate the effects of RI on oral clearance of nirmatrelvir. Other HLM CLint, the most sensitive parameter, was adjusted, and the simulated plasma concentration profiles of nirmatrelvir in severe RI subjects were within the therapeutic index of 292-10 000 ng/mL for dosing regimens of loading doses of 300/100 mg followed by 150/100 mg or 75/100 mg twice daily of nirmatrelvir/ritonavir. Considering that nirmatrelvir is available as a 150 mg tablet, we recommend 300/100 mg followed by 150/100 mg twice daily as the dosing regimen to be investigated in severe RI.
Collapse
Affiliation(s)
- Tat Ming Ng
- Department of Pharmacy and Pharmaceutical Sciences, Faculty of Science, National University of Singapore, Singapore, Singapore
- Division of Pharmacy, Tan Tock Seng Hospital, Novena, Singapore
| | - Ziteng Wang
- Department of Pharmacy and Pharmaceutical Sciences, Faculty of Science, National University of Singapore, Singapore, Singapore
| | - Eric Chun Yong Chan
- Department of Pharmacy and Pharmaceutical Sciences, Faculty of Science, National University of Singapore, Singapore, Singapore
| |
Collapse
|
40
|
Petersen EF, Rasmussen CLM, Prabhala BK, Heidtmann CV, Nielsen P, Nielsen CU. The oral bioavailability of a pleuromutilin antibiotic candidate is increased after co-administration with the CYP3A4 inhibitor ritonavir and the P-gp inhibitor zosuquidar formulated as amorphous solid dispersions. Int J Pharm 2025; 673:125397. [PMID: 40010527 DOI: 10.1016/j.ijpharm.2025.125397] [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: 01/10/2025] [Revised: 02/18/2025] [Accepted: 02/21/2025] [Indexed: 02/28/2025]
Abstract
The aim of the present work was to investigate if CYP-mediated metabolism or P-gp recognition were the main limitations to developing oral formulations of the pleuromutilin drug candidate CVH-174, 16, and to subsequently increase the bioavailability through a formulation design based on amorphous solid dispersions (ASDs) containing either a CYP3A inhibitor or a P-gp inhibitor or both. ASDs were produced using HPMC-5 with ritonavir and zosuquidar as CYP3A4 and P-gp inhibitors, respectively, through freeze-drying. The ASDs were characterized using XRPD over time to assess the stability of the formulations. The oral bioavailability was investigated in Sprague-Dawley rats following either oral or intravenous (IV) dosing. The results showed that ritonavir could be supersaturated when formulated in an HPMC-5-based ASD, whereas HPMC-5-based ASDs could not increase the solubility of CVH-174 and zosuquidar. The ASD formulations remained stable for the period covering the experiments. In vivo IV dosing showed that CVH-174 was metabolized fast with a half-life of 0.15 h. The oral bioavailability of CVH-174 was low ∼ 1 % and could not be increased by co-dosing with a P-gp inhibitor alone, whereas the CYP3A4 inhibitor ritonavir did increase the bioavailability. The combined co-administration of ritonavir- and zosuquidar-containing ASDs surprisingly increased CVH-174 bioavailability to around 18 %. In conclusion, the oral bioavailability of CVH-174 can be significantly increased through a formulation design encompassing an inhibitor of the CYP3A4 enzyme, and this holds great potential for the future development of an inherent metabolic labile pleuromutilin drug class.
Collapse
Affiliation(s)
- Emilie Fynbo Petersen
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, DK-5230 Odense M, Denmark
| | | | - Bala Krishna Prabhala
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, DK-5230 Odense M, Denmark
| | - Christoffer Vogsen Heidtmann
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, DK-5230 Odense M, Denmark
| | - Poul Nielsen
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, DK-5230 Odense M, Denmark
| | - Carsten Uhd Nielsen
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, DK-5230 Odense M, Denmark.
| |
Collapse
|
41
|
Wang W, Zhou X, Li W, Zeng P, Guo L, Wang Q, Li J. Inhibitory efficacy and structural insights of Bofutrelvir against SARS-CoV-2 M pro mutants and MERS-CoV M pro. Commun Biol 2025; 8:493. [PMID: 40133408 PMCID: PMC11937426 DOI: 10.1038/s42003-025-07929-9] [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: 10/06/2024] [Accepted: 03/11/2025] [Indexed: 03/27/2025] Open
Abstract
The COVID-19 pandemic has caused significant global health and economic disruption. Mutations E166N, E166R, E166N, S144A and His163A in the SARS-CoV-2 main protease (Mpro) have been implicated in reducing the efficacy of certain antiviral treatments. Bofutrelvir, a promising inhibitor, has shown effectiveness against SARS-CoV-2 Mpro. This study aims to evaluate the inhibitory effects of Bofutrelvir on the E166N, E166R, His163A, E166V and S144A mutants of SARS-CoV-2 Mpro, as well as on MERS-CoV Mpro. Our findings indicate a substantial reduction in the inhibitory potency of Bofutrelvir against these mutants and MERS-CoV, with IC50 values significantly higher than those for the wild-type SARS-CoV-2 Mpro. Specifically, the E166N, E166R, E166V, S144A, and H163A mutations significantly reduce the binding affinity and inhibitory effectiveness of Bofutrelvir due to disrupted hydrogen bonds, altered binding site stability, and reduced enzyme activity. Structural analysis of the crystal complexes showed that changes in interactions at the S1 subsite in the mutants and the loss of hydrogen bonds at the S4 subsite in MERS-CoV Mpro are critical factors contributing to the diminished inhibitory activity. These insights reveal the necessity of ongoing structural analysis to adapt therapeutic strategies.
Collapse
Affiliation(s)
- Weiwei Wang
- Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai, China
| | - Xuelan Zhou
- Jiangxi Province Key Laboratory of Pharmacology of Traditional Chinese Medicine, School of Pharmacy, Gannan Medical University, Ganzhou, China
| | - Wenwen Li
- Jiangxi Province Key Laboratory of Pharmacology of Traditional Chinese Medicine, School of Pharmacy, Gannan Medical University, Ganzhou, China
| | - Pei Zeng
- Shenzhen Crystalo Biopharmaceutical Co, Ltd, Shenzhen, China
| | - Li Guo
- Jiangxi Jmerry Biopharmaceutical Co, Ltd, Ganzhou, China
| | - Qisheng Wang
- Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai, China.
| | - Jian Li
- Jiangxi Province Key Laboratory of Pharmacology of Traditional Chinese Medicine, School of Pharmacy, Gannan Medical University, Ganzhou, China.
| |
Collapse
|
42
|
Torri D, Bering L, Yates LRL, Angiolini SM, Xu G, Cuesta‐Hoyos S, Shepherd SA, Micklefield J. Enzymatic Cascades for Stereoselective and Regioselective Amide Bond Assembly. Angew Chem Int Ed Engl 2025; 64:e202422185. [PMID: 39792621 PMCID: PMC11933536 DOI: 10.1002/anie.202422185] [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: 11/14/2024] [Revised: 12/24/2024] [Accepted: 01/10/2025] [Indexed: 01/12/2025]
Abstract
Amide bond formation is fundamental in nature and is widely used in the synthesis of pharmaceuticals and other valuable products. Current methods for amide synthesis are often step and atom inefficient, requiring the use of protecting groups, deleterious reagents and organic solvents that create significant waste. The development of cleaner and more efficient catalytic methods for amide synthesis remains an urgent unmet need. Herein, we present novel biocatalytic cascade reactions for synthesising various amides under mild aqueous conditions from readily available organic nitriles combining nitrile hydrolysing enzymes and amide bond synthetase enzymes. These cooperative biocatalytic cascades enable kinetic resolution of racemic nitriles and provide a highly enantioselective biocatalytic extension of the Strecker reaction. The regioselective non-directed C-H bond amidation of simple arenes was demonstrated through the incorporation of photoredox catalysis to the front end of the cascade. C-H bond amidation of simple aromatic precursors was also achieved via a CO2 fixation cascade combining enzymatic carboxylation and amide bond synthesis in one-pot.
Collapse
Affiliation(s)
- Daniele Torri
- Department of Chemistry and Manchester Institute of BiotechnologyThe University of ManchesterManchesterM1 7DNUK
| | - Luis Bering
- Department of Chemistry and Manchester Institute of BiotechnologyThe University of ManchesterManchesterM1 7DNUK
| | - Luke R. L. Yates
- Department of Chemistry and Manchester Institute of BiotechnologyThe University of ManchesterManchesterM1 7DNUK
| | - Stuart M. Angiolini
- Department of Chemistry and Manchester Institute of BiotechnologyThe University of ManchesterManchesterM1 7DNUK
| | - Guangcai Xu
- Department of Chemistry and Manchester Institute of BiotechnologyThe University of ManchesterManchesterM1 7DNUK
| | - Sebastian Cuesta‐Hoyos
- Department of Chemistry and Manchester Institute of BiotechnologyThe University of ManchesterManchesterM1 7DNUK
| | - Sarah A. Shepherd
- Department of Chemistry and Manchester Institute of BiotechnologyThe University of ManchesterManchesterM1 7DNUK
| | - Jason Micklefield
- Department of Chemistry and Manchester Institute of BiotechnologyThe University of ManchesterManchesterM1 7DNUK
- Department of Chemistry, Molecular Science Research HubImperial CollegeLondonW12 0BZUK
| |
Collapse
|
43
|
Yu W, Krishnan MK, Weekly M, Shanker RM, Doshi P, Ragan JA, Greene RA, Gampper B, Caron S, McKillop A, Ludwig J, Dolsten M. The unprecedented Paxlovid journey from milligrams to millions of patient doses during the Covid-19 pandemic. COMMUNICATIONS MEDICINE 2025; 5:80. [PMID: 40113988 PMCID: PMC11926161 DOI: 10.1038/s43856-025-00798-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 03/07/2025] [Indexed: 03/22/2025] Open
Affiliation(s)
- Weili Yu
- Pfizer Research & Development, Pharmaceutical Sciences, Pfizer Inc, Groton, CT, USA
| | - Mahesh K Krishnan
- Pfizer Research & Development, Pharmaceutical Sciences, Pfizer Inc, Groton, CT, USA.
| | - Matt Weekly
- Pfizer Research & Development, Pharmaceutical Sciences, Pfizer Inc, Groton, CT, USA
| | - Ravi M Shanker
- Pfizer Research & Development, Pharmaceutical Sciences, Pfizer Inc, Groton, CT, USA
| | - Pankaj Doshi
- Pfizer Research & Development, Pharmaceutical Sciences, Pfizer Inc, Groton, CT, USA
| | - John A Ragan
- Pfizer Research & Development, Pharmaceutical Sciences, Pfizer Inc, Groton, CT, USA
| | | | | | - Stéphane Caron
- Pfizer Research & Development, Pharmaceutical Sciences, Pfizer Inc, Groton, CT, USA
| | - Andrew McKillop
- Pfizer Research & Development, Pharmaceutical Sciences, Pfizer Inc, Groton, CT, USA
| | - John Ludwig
- Pfizer Research & Development, Pfizer Inc, Chesterfield, MO, USA
| | - Mikael Dolsten
- Pfizer Research, Development & Medical, Pfizer Inc, New York, NY, USA
| |
Collapse
|
44
|
Chen J, Zhang W, Li Y, Liu C, Dong T, Chen H, Wu C, Su J, Li B, Zhang W, Hu B, Jia J, Ma CB, Zhu Y, He X, Li A, Pan K, Lin H, Guo Z, Li C, Zhang L, Yan H, Zhou P, Peng W, Shi ZL. Bat-infecting merbecovirus HKU5-CoV lineage 2 can use human ACE2 as a cell entry receptor. Cell 2025; 188:1729-1742.e16. [PMID: 39970913 DOI: 10.1016/j.cell.2025.01.042] [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: 09/17/2024] [Revised: 12/16/2024] [Accepted: 01/29/2025] [Indexed: 02/21/2025]
Abstract
Merbecoviruses comprise four viral species with remarkable genetic diversity: MERS-related coronavirus, Tylonycteris bat coronavirus HKU4, Pipistrellus bat coronavirus HKU5, and Hedgehog coronavirus 1. However, the potential human spillover risk of animal merbecoviruses remains to be investigated. Here, we reported the discovery of HKU5-CoV lineage 2 (HKU5-CoV-2) in bats that efficiently utilize human angiotensin-converting enzyme 2 (ACE2) as a functional receptor and exhibits a broad host tropism. Cryo-EM analysis of HKU5-CoV-2 receptor-binding domain (RBD) and human ACE2 complex revealed an entirely distinct binding mode compared with other ACE2-utilizing merbecoviruses with RBD footprint largely shared with ACE2-using sarbecoviruses and NL63. Structural and functional analyses indicate that HKU5-CoV-2 has a better adaptation to human ACE2 than lineage 1 HKU5-CoV. Authentic HKU5-CoV-2 infected human ACE2-expressing cell lines and human respiratory and enteric organoids. This study reveals a distinct lineage of HKU5-CoVs in bats that efficiently use human ACE2 and underscores their potential zoonotic risk.
Collapse
Affiliation(s)
- Jing Chen
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Wei Zhang
- Guangzhou National Laboratory, No. 9 XingDaoHuanBei Road, Guangzhou International Bio Island, Guangzhou, China; The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Yang Li
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Chen Liu
- State Key Laboratory of Virology and Biosafety, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
| | - Tianyi Dong
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China; University of Chinese Academy of Sciences, Beijing, China
| | - Huiyu Chen
- Guangzhou National Laboratory, No. 9 XingDaoHuanBei Road, Guangzhou International Bio Island, Guangzhou, China; The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Chunguang Wu
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China; University of Chinese Academy of Sciences, Beijing, China
| | - Jia Su
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China; University of Chinese Academy of Sciences, Beijing, China
| | - Bei Li
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Wei Zhang
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Ben Hu
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Jingkun Jia
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China; University of Chinese Academy of Sciences, Beijing, China
| | - Cheng-Bao Ma
- State Key Laboratory of Virology and Biosafety, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
| | - Yan Zhu
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Xiangyang He
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou, China
| | - Ang Li
- Guangzhou National Laboratory, No. 9 XingDaoHuanBei Road, Guangzhou International Bio Island, Guangzhou, China; The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Kaiyi Pan
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China; University of Chinese Academy of Sciences, Beijing, China
| | - Haofeng Lin
- The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Zishuo Guo
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China; University of Chinese Academy of Sciences, Beijing, China
| | - Cong Li
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Libiao Zhang
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou, China.
| | - Huan Yan
- State Key Laboratory of Virology and Biosafety, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, China.
| | - Peng Zhou
- Guangzhou National Laboratory, No. 9 XingDaoHuanBei Road, Guangzhou International Bio Island, Guangzhou, China; The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China.
| | - Wei Peng
- Guangzhou National Laboratory, No. 9 XingDaoHuanBei Road, Guangzhou International Bio Island, Guangzhou, China; The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China.
| | - Zheng-Li Shi
- Guangzhou National Laboratory, No. 9 XingDaoHuanBei Road, Guangzhou International Bio Island, Guangzhou, China.
| |
Collapse
|
45
|
Dhaka P, Mahto JK, Singh A, Kumar P, Tomar S. Structural insights into the RNA binding inhibitors of the C-terminal domain of the SARS-CoV-2 nucleocapsid. J Struct Biol 2025; 217:108197. [PMID: 40113149 DOI: 10.1016/j.jsb.2025.108197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 01/26/2025] [Accepted: 03/13/2025] [Indexed: 03/22/2025]
Abstract
The SARS-CoV-2 nucleocapsid (N) protein is an essential structural element of the virion, playing a crucial role in enclosing the viral genome into a ribonucleoprotein (RNP) assembly, as well as viral replication and transmission. The C-terminal domain of the N-protein (N-CTD) is essential for encapsidation, contributing to the stabilization of the RNP complex. In a previous study, three inhibitors (ceftriaxone, cefuroxime, and ampicillin) were screened for their potential to disrupt the RNA packaging process by targeting the N-protein. However, the binding efficacy, mechanism of RNA binding inhibition, and molecular insights of binding with N-CTD remain unclear. In this study, we evaluated the binding efficacy of these inhibitors using isothermal titration calorimetry (ITC), revealing the affinity of ceftriaxone (18 ± 1.3 μM), cefuroxime (55 ± 4.2 μM), and ampicillin (28 ± 1.2 μM) with the N-CTD. Further inhibition assay and fluorescence polarisation assay demonstrated RNA binding inhibition, with IC50 ranging from ∼ 12 to 18 μM and KD values between 24 μM to 32 μM for the inhibitors, respectively. Additionally, we also determined the inhibitor-bound complex crystal structures of N-CTD-Ceftriaxone (2.0 Å) and N-CTD-Ampicillin (2.2 Å), along with the structure of apo N-CTD (1.4 Å). These crystal structures revealed previously unobserved interaction sites involving residues K261, K266, R293, Q294, and W301 at the oligomerization interface and the predicted RNA-binding region of N-CTD. These findings provide valuable molecular insights into the inhibition of N-CTD, highlighting its potential as an underexplored but promising target for the development of novel antiviral agents against coronaviruses.
Collapse
Affiliation(s)
- Preeti Dhaka
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand 247667, India
| | - Jai Krishna Mahto
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand 247667, India
| | - Ankur Singh
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand 247667, India
| | - Pravindra Kumar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand 247667, India.
| | - Shailly Tomar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand 247667, India.
| |
Collapse
|
46
|
Adediji A, Sroithongmoon A, Suroengrit A, Wilasluck P, Deetanya P, Sanachai K, Karnchanapandh K, Boonyasuppayakorn S, Wangkanont K, Rungrotmongkol T, Khotavivattana T. Design, synthesis, and antiviral activity of fragmented-lapatinib aminoquinazoline analogs towards SARS-CoV-2 inhibition. Eur J Med Chem 2025; 286:117303. [PMID: 39879938 DOI: 10.1016/j.ejmech.2025.117303] [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: 07/30/2024] [Revised: 01/16/2025] [Accepted: 01/18/2025] [Indexed: 01/31/2025]
Abstract
The severe impact of COVID-19 on global health and economies highlights the critical need for innovative treatments. Recently, lapatinib, a drug initially used for breast cancer, has been identified as a potential inhibitor of the main protease (Mpro) of SARS-CoV-2, meriting further investigation. Utilizing rational design strategies and guided by MD simulations, we developed novel aminoquinazoline analogs based on fragmented lapatinib's structure. Preliminary computational screenings identified promising candidates, which were synthesized using a concise 3-4 step process. In vitro assays demonstrated notable antiviral efficacy against SARS-CoV-2-infected cells for all analogs, with Bb1 showing an EC50 of 1.10 μM and significantly lower toxicity (13.55 % at 50 μM) compared to lapatinib. Further studies confirmed that these analogs effectively inhibit SARS-CoV-2 Mpro, with Bb7 displaying the highest activity. MD simulations revealed that Bb7 achieves stability within the Mpro binding pocket through interactions with specific residues. These findings indicate that aminoquinazoline analogs hold significant promise as therapeutic candidates for COVID-19.
Collapse
Affiliation(s)
- Ayomide Adediji
- Center of Excellence in Natural Products, Department of Chemistry, Faculty of Science, Chulalongkorn University, Pathumwan, Bangkok, 10330, Thailand
| | - Akeanan Sroithongmoon
- Center of Excellence in Natural Products, Department of Chemistry, Faculty of Science, Chulalongkorn University, Pathumwan, Bangkok, 10330, Thailand
| | - Aphinya Suroengrit
- Center of Excellence in Applied Medical Virology, Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Patcharin Wilasluck
- Center of Excellence for Molecular Biology and Genomics of Shrimp, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand; Center of Excellence for Molecular Crop, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Peerapon Deetanya
- Center of Excellence for Molecular Biology and Genomics of Shrimp, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand; Center of Excellence for Molecular Crop, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Kamonpan Sanachai
- Department of Biochemistry, Faculty of Science, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Kun Karnchanapandh
- Program in Bioinformatics and Computational Biology, Graduate School, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Siwaporn Boonyasuppayakorn
- Center of Excellence in Applied Medical Virology, Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Kittikhun Wangkanont
- Center of Excellence for Molecular Biology and Genomics of Shrimp, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand; Center of Excellence for Molecular Crop, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Thanyada Rungrotmongkol
- Program in Bioinformatics and Computational Biology, Graduate School, Chulalongkorn University, Bangkok, 10330, Thailand; Center of Excellence in Biocatalyst and Sustainable Biotechnology, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Tanatorn Khotavivattana
- Center of Excellence in Natural Products, Department of Chemistry, Faculty of Science, Chulalongkorn University, Pathumwan, Bangkok, 10330, Thailand.
| |
Collapse
|
47
|
Li K, Jadhav P, Wen Y, Tan H, Wang J. Development of a Fluorescence Polarization Assay for the SARS-CoV-2 Papain-like Protease. ACS Pharmacol Transl Sci 2025; 8:774-784. [PMID: 40109744 PMCID: PMC11915184 DOI: 10.1021/acsptsci.4c00642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 02/11/2025] [Accepted: 02/14/2025] [Indexed: 03/22/2025]
Abstract
The COVID-19 pandemic has caused significant losses to the global community. Although effective vaccination and antiviral therapeutics provide primary defense, SARS-CoV-2 remains a public health threat, given the emerging resistant variants. The SARS-CoV-2 papain-like protease (PLpro) is essential for viral replication and is a promising drug target. We recently designed a series of biarylphenyl PLpro inhibitors with a representative lead Jun12682 showing potent antiviral efficacy in a SARS-CoV-2 infection mouse model. In this study, we designed a fluorescein-labeled biarylphenyl probe Jun12781 and used it to optimize a fluorescence polarization (FP) assay. The FP assay is suitable for high-throughput screening with a Z' factor of 0.69. In addition, we found a positive correlation between the FP binding affinity and the enzymatic inhibitory potency of PLpro inhibitors, suggesting that the FP assay is valid in characterizing the binding affinity of PLpro inhibitors.
Collapse
Affiliation(s)
- Kan Li
- Department of Medicinal Chemistry, Ernest Mario School of Pharmacy, Rutgers, the State University of New Jersey, Piscataway, New Jersey 08854, United States
| | - Prakash Jadhav
- Department of Medicinal Chemistry, Ernest Mario School of Pharmacy, Rutgers, the State University of New Jersey, Piscataway, New Jersey 08854, United States
| | - Yu Wen
- Department of Medicinal Chemistry, Ernest Mario School of Pharmacy, Rutgers, the State University of New Jersey, Piscataway, New Jersey 08854, United States
| | - Haozhou Tan
- Department of Medicinal Chemistry, Ernest Mario School of Pharmacy, Rutgers, the State University of New Jersey, Piscataway, New Jersey 08854, United States
| | - Jun Wang
- Department of Medicinal Chemistry, Ernest Mario School of Pharmacy, Rutgers, the State University of New Jersey, Piscataway, New Jersey 08854, United States
| |
Collapse
|
48
|
Handa T, Saha A, Narayanan A, Ronzier E, Kumar P, Singla J, Tomar S. Structural Virology: The Key Determinants in Development of Antiviral Therapeutics. Viruses 2025; 17:417. [PMID: 40143346 PMCID: PMC11945554 DOI: 10.3390/v17030417] [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: 01/12/2025] [Revised: 03/07/2025] [Accepted: 03/10/2025] [Indexed: 03/28/2025] Open
Abstract
Structural virology has emerged as the foundation for the development of effective antiviral therapeutics. It is pivotal in providing crucial insights into the three-dimensional frame of viruses and viral proteins at atomic-level or near-atomic-level resolution. Structure-based assessment of viral components, including capsids, envelope proteins, replication machinery, and host interaction interfaces, is instrumental in unraveling the multiplex mechanisms of viral infection, replication, and pathogenesis. The structural elucidation of viral enzymes, including proteases, polymerases, and integrases, has been essential in combating viruses like HIV-1 and HIV-2, SARS-CoV-2, and influenza. Techniques including X-ray crystallography, Nuclear Magnetic Resonance spectroscopy, Cryo-electron Microscopy, and Cryo-electron Tomography have revolutionized the field of virology and significantly aided in the discovery of antiviral therapeutics. The ubiquity of chronic viral infections, along with the emergence and reemergence of new viral threats necessitate the development of novel antiviral strategies and agents, while the extensive structural diversity of viruses and their high mutation rates further underscore the critical need for structural analysis of viral proteins to aid antiviral development. This review highlights the significance of structure-based investigations for bridging the gap between structure and function, thus facilitating the development of effective antiviral therapeutics, vaccines, and antibodies for tackling emerging viral threats.
Collapse
Affiliation(s)
- Tanuj Handa
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247667, India; (T.H.); (A.S.); (P.K.); (J.S.)
| | - Ankita Saha
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247667, India; (T.H.); (A.S.); (P.K.); (J.S.)
| | - Aarthi Narayanan
- Department of Biology, College of Science, George Mason University, Fairfax, VA 22030, USA;
| | - Elsa Ronzier
- Biomedical Research Laboratory, Institute for Biohealth Innovation, George Mason University, Fairfax, VA 22030, USA;
| | - Pravindra Kumar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247667, India; (T.H.); (A.S.); (P.K.); (J.S.)
| | - Jitin Singla
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247667, India; (T.H.); (A.S.); (P.K.); (J.S.)
| | - Shailly Tomar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247667, India; (T.H.); (A.S.); (P.K.); (J.S.)
| |
Collapse
|
49
|
Hu Y, Li H, Wang K, Wu D, Zhang H, Ding Y, Wu J, Ye S, Peng Y, Liu L. Single- and multiple-dose pharmacokinetics and safety of the SARS-CoV-2 3CL protease inhibitor RAY1216: a phase 1 study in healthy participants. Antimicrob Agents Chemother 2025; 69:e0145024. [PMID: 39887246 PMCID: PMC11881559 DOI: 10.1128/aac.01450-24] [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: 09/25/2024] [Accepted: 01/15/2025] [Indexed: 02/01/2025] Open
Abstract
Coronavirus disease 2019, which leads to pneumonia, is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). RAY1216 is a 3C-like protease inhibitor that targets SARS-CoV-2. The aim of our study was to assess the pharmacokinetics (PK) and safety of RAY1216 in healthy volunteers. This was a randomized, placebo-controlled, double-blind study consisting of four components: a single ascending dose study, a drug-drug interaction study, a multiple ascending dose study, and a food-effect study. All participants were randomly assigned to receive either a single dose or multiple doses of RAY1216 or placebo. A total of 88 healthy adult participants (male-to-female ratio of 1:1) aged 18-50 years were enrolled. A total of 37 participants (42%) experienced at least one adverse event (AE). All AEs were mild or moderate and were resolved without additional treatment. The most commonly reported adverse drug reactions were hypertriglyceridemia, hyperuricemia, and elevated serum creatinine levels. RAY1216 was well-absorbed after administration with exposure increasing in a dose-dependent manner. Food appeared to increase exposure and delay the absorption of RAY1216. Ritonavir significantly inhibited drug metabolism, and increased drug exposure increased the associated safety risks. RAY1216 was found to be well tolerated and safe in healthy participants. On the basis of preclinical results, PK characteristics, and the safety profile of RAY1216, a dosage of 400 mg three times daily was selected, thereby establishing a foundation for future research and for the clinical application of RAY1216.CLINICAL TRIALSThis study is registered with ClinicalTrials.gov as NCT05829551.
Collapse
Affiliation(s)
- Yue Hu
- Phase I Clinical Research Center, First Hospital of Jilin University, Changchun, China
| | - Haijun Li
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, China
- Guangdong Raynovent Biotech Co., Ltd., Guangzhou, China
| | - Kun Wang
- Shanghai Qiangshi Information Technology Co., Ltd., Shanghai, China
| | - Dandan Wu
- Phase I Clinical Research Center, First Hospital of Jilin University, Changchun, China
| | - Hong Zhang
- Phase I Clinical Research Center, First Hospital of Jilin University, Changchun, China
| | - Yanhua Ding
- Phase I Clinical Research Center, First Hospital of Jilin University, Changchun, China
| | - Junyan Wu
- Phase I Clinical Trial Centre, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Suiwen Ye
- Phase I Clinical Trial Centre, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yun Peng
- Guangdong Raynovent Biotech Co., Ltd., Guangzhou, China
| | - Li Liu
- Department of Pediatrics, First Hospital of Jilin University, Changchun, China
| |
Collapse
|
50
|
Hernandez G, Osinski A, Majumdar A, Eitson JL, Antczak M, Pawłowski K, Niederstrasser H, Servage KA, Posner B, Schoggins JW, Ready JM, Tagliabracci VS. Covalent inhibition of the SARS-CoV-2 NiRAN domain via an active-site cysteine. J Biol Chem 2025; 301:108378. [PMID: 40049411 PMCID: PMC12013494 DOI: 10.1016/j.jbc.2025.108378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 02/07/2025] [Accepted: 02/25/2025] [Indexed: 03/12/2025] Open
Abstract
The kinase-like nidovirus RdRp-associated nucleotidyl transferase (NiRAN) domain of nsp12 in SARS-CoV-2 catalyzes the formation of the 5' RNA cap structure. This activity is required for viral replication, offering a new target for the development of antivirals. Here, we develop a high-throughput assay to screen for small molecule inhibitors targeting the SARS-CoV-2 NiRAN domain. We identified NiRAN covalent inhibitor 2 (NCI-2), a compound with a reactive chloromethyl group that covalently binds to an active site cysteine (Cys53) in the NiRAN domain, inhibiting its activity. NCI-2 can enter cells, bind to, and inactivate ectopically expressed nsp12. A cryo-EM reconstruction of the SARS-CoV-2 replication-transcription complex bound to NCI-2 offers a detailed structural blueprint for rational drug design. Although NCI-2 showed limited potency against SARS-CoV-2 replication in cells, our work lays the groundwork for developing more potent and selective inhibitors targeting the NiRAN domain. This approach presents a promising therapeutic strategy for effectively combating COVID-19 and potentially mitigating future coronavirus outbreaks.
Collapse
Affiliation(s)
- Genaro Hernandez
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Adam Osinski
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA; Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Abir Majumdar
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA; Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Jennifer L Eitson
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Monika Antczak
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Krzysztof Pawłowski
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA; Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | | | - Kelly A Servage
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA; Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Bruce Posner
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - John W Schoggins
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Joseph M Ready
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Vincent S Tagliabracci
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA; Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, Texas, USA; Harold C. Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, Texas, USA; Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA.
| |
Collapse
|