1
|
Oba S, Hosoya T, Kawata D, Komiya Y, Iwai H, Koike R, Miyamoto S, Kanno T, Ainai A, Suzuki T, Hasegawa H, Yasuda S. Iguratimod, a promising therapeutic agent for COVID-19 that attenuates excessive inflammation in mouse models. Eur J Pharmacol 2025; 996:177537. [PMID: 40147575 DOI: 10.1016/j.ejphar.2025.177537] [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: 11/27/2024] [Revised: 03/18/2025] [Accepted: 03/20/2025] [Indexed: 03/29/2025]
Abstract
In severe COVID-19 patients, excessive inflammation can lead to multiorgan dysfunction. Current anti-inflammatory treatments like glucocorticoids partially improve the outcomes, while immune systems are compromised. We have identified that SARS-CoV-2-infected obese mice were a good model of the cytokine storm seen in COVID-19. Here, we revealed that iguratimod (IGU), an approved agent for rheumatoid arthritis, improved survival by attenuating inflammation with minimal immune suppression. In this study, C57BL/6 mice were fed a high-fat diet (HFD) or a normal-fat diet (NFD) for ten weeks before being infected with a mouse-adapted SARS-CoV-2. IGU significantly improved survival rates and reduced lung inflammation in HFD-fed mice, with minimal impact on interferon-induced genes and viral load. Meanwhile, dexamethasone (DEX) did not improve survival, while it suppressed various immune reactions with different mechanisms to IGU. Interestingly, IGU-treated mice had fewer SARS-CoV-2 positive cells in the lung, although viral replication was comparable to the control mice. Neither IGU nor DEX inhibited the SARS-CoV-2 infection in Vero-E6 cells, unlike the antiviral agent, remdesivir. Of note, IGU was effective prophylactically and therapeutically in HFD mice, and showed beneficial effects in NFD-fed mice with a lethal dose exposure of SARS-CoV-2. We demonstrated that IGU could be a promising treatment for severe COVID-19, especially in obese patients, by fine-tuning inflammation without compromising antiviral immunity. This study supports the possibility of drug repositioning for IGU COVID-19 beyond autoimmune diseases.
Collapse
Affiliation(s)
- Seiya Oba
- Department of Rheumatology, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Tokyo, Japan; Department of Pathology, National Institute of Infectious Diseases, Tokyo, 208-0011, Japan
| | - Tadashi Hosoya
- Department of Rheumatology, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Tokyo, Japan; Department of Pathology, National Institute of Infectious Diseases, Tokyo, 208-0011, Japan.
| | - Daisuke Kawata
- Department of Rheumatology, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Tokyo, Japan; Department of Pathology, National Institute of Infectious Diseases, Tokyo, 208-0011, Japan
| | - Yoji Komiya
- Department of Rheumatology, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Tokyo, Japan; Department of Pathology, National Institute of Infectious Diseases, Tokyo, 208-0011, Japan
| | - Hideyuki Iwai
- Department of Rheumatology, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Tokyo, Japan; Department of Pathology, National Institute of Infectious Diseases, Tokyo, 208-0011, Japan
| | - Ryuji Koike
- Department of Rheumatology, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Tokyo, Japan
| | - Sho Miyamoto
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, 208-0011, Japan
| | - Takayuki Kanno
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, 208-0011, Japan
| | - Akira Ainai
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, 208-0011, Japan
| | - Tadaki Suzuki
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, 208-0011, Japan
| | - Hideki Hasegawa
- WHO Collaborating Centre for Reference and Research on Influenza, Tokyo, Japan; Research Center for Influenza and Respiratory Virus, National Institute of Infectious Diseases, Tokyo, Japan
| | - Shinsuke Yasuda
- Department of Rheumatology, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Tokyo, Japan.
| |
Collapse
|
2
|
Xia L, Yan J, Chen Y, Robison TW, Chen T. Germline mutagenicity of molnupiravir and its active form, β-d-N4-hydroxycytidine, in Caenorhabditis elegans evaluated using whole-genome sequencing. Toxicol Lett 2025:S0378-4274(25)00093-1. [PMID: 40409569 DOI: 10.1016/j.toxlet.2025.05.007] [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: 03/24/2025] [Revised: 05/02/2025] [Accepted: 05/14/2025] [Indexed: 05/25/2025]
Abstract
Molnupiravir is a medication used to treat COVID-19 by introducing errors into the SARS-CoV-2 virus's genetic code, thereby preventing its replication. Previous studies, both in vitro and in vivo, have yielded conflicting results regarding its mutagenic potential. While most genotoxicity and mutagenicity tests for molnupiravir and its active form, β-d-N4-hydroxycytidine (NHC), were negative, a few in vitro tests showed positive results. Consequently, further investigation is necessary to evaluate various mutagenic endpoints of molnupiravir. In this study, acute toxicity was assessed by measuring the locomotive activity of Caenorhabditis elegans using a WMicroTracker to determine an appropriate dose range for the germline mutagenicity study. The C. elegans worms were treated with different concentrations of molnupiravir and NHC, along with vehicle controls and ethyl methanesulfonate (EMS) as a positive control. To assess germline mutagenicity, P0 worms from a single clone were exposed to selected concentrations of molnupiravir and NHC, as well as vehicle and positive controls, for 4h. Molnupiravir and NHC treatments had no significant effect on the locomotion of C. elegans worms after 1-, 2-, 3-, and 4-h exposures, compared to the vehicle control group. In contrast, EMS significantly reduced the worms' locomotive activity. Subsequent whole-genome sequencing of the F1 progeny from the treated P0 worms revealed that neither molnupiravir nor NHC increased the germline mutation frequency or altered mutation types, compared to the vehicle control. In contrast, EMS treatment significantly increased mutation frequency over the vehicle control, with a specific EMS mutational signature observed. These results suggest that molnupiravir and NHC are not mutagenic in C. elegans germ cells, aligning with previous findings that demonstrate the low mutagenicity of molnupiravir in clinical settings. Additionally, these findings highlight the utility of C. elegans as an alternative animal model for accelerating toxicity assessments and reducing the use of experimental animals.
Collapse
Affiliation(s)
- Li Xia
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR
| | - Jian Yan
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR
| | - Ying Chen
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR
| | - Timothy W Robison
- Office of New Drugs, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD
| | - Tao Chen
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR.
| |
Collapse
|
3
|
Pal S, Hanson QM, Ogden SC, Lee EM, Martinez NJ, Zakharov AV. Discovery of SARS-CoV-2 Nsp14-Methyltransferase (MTase) Inhibitors by Harnessing Scaffold-Centric Exploration of the Ultra Large Chemical Space. ACS Pharmacol Transl Sci 2025; 8:1366-1400. [PMID: 40370981 PMCID: PMC12070326 DOI: 10.1021/acsptsci.5c00111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 04/09/2025] [Accepted: 04/15/2025] [Indexed: 05/16/2025]
Abstract
The global impact of SARS-CoV-2 underscores the need for antiviral treatments beyond vaccines. This study targets Nsp14-MTase, a viral protein essential for replication. Initial quantitative high-throughput screening (qHTS) of ∼15,000 compounds from the selected NCATS in-house libraries identified 135 active hit molecules, reflecting a hit-rate of 1.04%. To enhance the search for promising antiviral agents, we expanded this screening campaign with two rounds of machine learning (ML)-based virtual screening of ∼130,000 compounds. The first iteration yielded 72 active compounds encompassing 27 chemotypes with an IC50 ranging from 1.45 μM to 33.27 μM, increasing the hit-rate 28-fold over the initial qHTS screen. Scaffold clustering of those hits revealed 27 chemotypes. The second iteration added 30 more hits (IC50: 2.18 μM-30.79 μM) across 12 new chemotypes. Initial structure-activity relationship (SAR) exploration around selected chemotypes identified NCGC00606183 (IC50: 0.41 μM) as the most potent hit. Hit-to-lead optimization using scaffold-centric exploration against the ultra large Enamine REAL Space (∼5.6 billion compounds) in HPC clusters identified 78 analogs, with 56 showing potent biochemical activity (IC50: 0.12 μM-18.23 μM) and cellular activity (0.27 μM-23.07 μM) in fully infectious SARS-CoV-2 live virus assays.
Collapse
Affiliation(s)
- Sourav Pal
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences
(NCATS), National Institutes of Health (NIH), Rockville, Maryland 20850, United States
| | - Quinlin M. Hanson
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences
(NCATS), National Institutes of Health (NIH), Rockville, Maryland 20850, United States
| | - Sarah C. Ogden
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences
(NCATS), National Institutes of Health (NIH), Rockville, Maryland 20850, United States
| | - Emily M. Lee
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences
(NCATS), National Institutes of Health (NIH), Rockville, Maryland 20850, United States
| | - Natalia J. Martinez
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences
(NCATS), National Institutes of Health (NIH), Rockville, Maryland 20850, United States
| | - Alexey V. Zakharov
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences
(NCATS), National Institutes of Health (NIH), Rockville, Maryland 20850, United States
| |
Collapse
|
4
|
Cheng Y, Zheng W, Dong X, Sun T, Xu M, Xiang L, Li J, Wang H, Jian X, Yu J, Li P, Hu T, Tian G, Jiang X, Zhang L, Aisa HA, Xie Y, Xiao G, Shen J. Design and Development of a Novel Oral 4'-Fluorouridine Double Prodrug VV261 against SFTSV. J Med Chem 2025; 68:9811-9826. [PMID: 40294286 DOI: 10.1021/acs.jmedchem.5c00626] [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: 04/30/2025]
Abstract
4'-Fluorouridine (4'-FU), despite demonstrating potent anti-SFTSV efficacy in vitro and in vivo, faces hindrances in its further development as a promising drug due to its weak chemical stability. Here, we report the discovery and development of VV261, a novel 4'-FU double prodrug with three isobutyryl groups on the ribose moiety and a nicotinoyloxymethyl group linked to the imide-nitrogen on the base moiety, exhibiting notable chemical stability and favorable pharmacokinetic properties. In SFTSV-infected mice, VV261 at 5 mg/kg/d for 7 days demonstrated complete protection against lethal SFTSV infection, prevented weight loss, and even a 2 day treatment significantly reduced both viral RNA copies and infectious virus titers in multiple organs, and notably alleviated splenic tissue lesions. After further preclinical evaluations, VV261, identified as a promising candidate drug for the treatment of SFTS, has entered Phase I clinical trials in China, the first such candidate to reach this stage for SFTS.
Collapse
Affiliation(s)
- Yong Cheng
- State Key Laboratory Basis of Xinjiang Indigenous Medicinal Plants Resource Utilization, Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences; Urumqi 830011, P. R. China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences; Shanghai 201203, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Wei Zheng
- Vigonvita Shanghai Co., Ltd., Shanghai 201210, P. R. China
| | - Xinru Dong
- State Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Tengxiao Sun
- State Key Laboratory Basis of Xinjiang Indigenous Medicinal Plants Resource Utilization, Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences; Urumqi 830011, P. R. China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences; Shanghai 201203, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Mengwei Xu
- State Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, P. R. China
| | - Li Xiang
- State Key Laboratory Basis of Xinjiang Indigenous Medicinal Plants Resource Utilization, Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences; Urumqi 830011, P. R. China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences; Shanghai 201203, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Jian Li
- State Key Laboratory Basis of Xinjiang Indigenous Medicinal Plants Resource Utilization, Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences; Urumqi 830011, P. R. China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences; Shanghai 201203, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Huilong Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences; Shanghai 201203, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Xiaoqin Jian
- State Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Jingjin Yu
- Vigonvita Shanghai Co., Ltd., Shanghai 201210, P. R. China
| | - Pengcheng Li
- Vigonvita Shanghai Co., Ltd., Shanghai 201210, P. R. China
| | - Tianwen Hu
- Vigonvita Shanghai Co., Ltd., Shanghai 201210, P. R. China
| | - Guanghui Tian
- Vigonvita Shanghai Co., Ltd., Shanghai 201210, P. R. China
| | - Xiangrui Jiang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences; Shanghai 201203, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Leike Zhang
- State Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
- Hubei Jiangxia Laboratory, Wuhan 430200, P. R. China
| | - Haji A Aisa
- State Key Laboratory Basis of Xinjiang Indigenous Medicinal Plants Resource Utilization, Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences; Urumqi 830011, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
- School of Pharmacy, Xinjiang Medical University, Urumqi 830054, P. R. China
| | - Yuanchao Xie
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences; Shanghai 201203, P. R. China
| | - Gengfu Xiao
- State Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Jingshan Shen
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences; Shanghai 201203, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| |
Collapse
|
5
|
Zadeh VR, Lew JM, Zahoor MA, Santer D, Feld JJ, Falzarano D. Combination therapy enhances the antiviral activity of IFN-λ against SARS-CoV-2 and MERS-CoV. Virus Res 2025; 355:199560. [PMID: 40113092 PMCID: PMC11994970 DOI: 10.1016/j.virusres.2025.199560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 02/07/2025] [Accepted: 03/17/2025] [Indexed: 03/22/2025]
Abstract
Therapeutic options against pathogenic human coronaviruses remain limited. In a recent clinical trial, we demonstrated the therapeutic efficacy of pegylated-IFN-λ in COVID-19 outpatients. However, the emergence of variants that have the potential to evade IFN-mediated antiviral responses raises concerns regarding the continued efficacy of this approach. In this work, we compared the sensitivity of SARS-CoV-2 variants and MERS-CoV to IFN-λ treatment in vitro and explored the potential of combination therapy with other FDA-authorized or approved antiviral agents. We observed that in contrast to the ancestral strain, all other SARS-CoV-2 lineages showed varying, but increased resistance to IFN-λ treatment, from a 5.7-fold increase in EC50 value for the P.1 strain to a 32.7-fold increase for the B.1.1.7 variant. We further show that combination treatment with remdesivir or nirmatrelvir enhanced the antiviral effect of IFN-λ against both SARS-CoV-2 and MERS-CoV. These findings justify the initiation of further in vivo testing that ultimately can help inform the development of more effective therapeutic guidelines against pathogenic coronaviruses.
Collapse
Affiliation(s)
- Vahid Rajabali Zadeh
- Vaccine and Infectious Disease Organization (VIDO), University of Saskatchewan, Saskatoon, SK, Canada
| | - Jocelyne M Lew
- Vaccine and Infectious Disease Organization (VIDO), University of Saskatchewan, Saskatoon, SK, Canada
| | - M Atif Zahoor
- Toronto Centre for Liver Disease, University of Toronto, Toronto, ON, Canada; University Health Network, University of Toronto, Toronto, ON, Canada
| | - Deanna Santer
- Department of Immunology, University of Manitoba, Winnipeg, MB, Canada
| | - Jordan J Feld
- Toronto Centre for Liver Disease, University of Toronto, Toronto, ON, Canada
| | - Darryl Falzarano
- Vaccine and Infectious Disease Organization (VIDO), University of Saskatchewan, Saskatoon, SK, Canada; Department of Veterinary Microbiology, University of Saskatchewan, Saskatoon, SK, Canada.
| |
Collapse
|
6
|
Qian B, Luo R, Shen B, Fan L, Zhang J, Zhang S, Sun Y, Deng X, Pang X, Zhong W, Gao Y. EIDD-2801 resists to infection and co-infection of SARS-CoV-2 and influenza virus. Virol J 2025; 22:126. [PMID: 40296172 PMCID: PMC12039283 DOI: 10.1186/s12985-025-02755-1] [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: 02/19/2025] [Accepted: 04/22/2025] [Indexed: 04/30/2025] Open
Abstract
BACKGROUND The coronavirus disease 2019 (COVID-19) pandemic has exerted a catastrophic impact on public health. Meanwhile, the seasonal influenza outbreak overlaps with the current pandemic wave. There is still an urgent need to develop effective therapeutic agents for the treatment of co-infection of multiple respiratory viruses. This study aimed to investigate antiviral effects of EIDD-2801, an orally bioavailable ribonucleoside analog, and its potent therapeutic effects in co-infection of multiple respiratory viruses. METHODS BALB/c mice and hamsters were infected with IFV or SARS-CoV-2, then were dosed orally with EIDD-2801 to measure the antiviral effects of EIDD-2801. Viral replication and mRNA transcription were evaluated by quantitative polymerase chain reaction (qPCR) and protein expression by Western Blot. Influenza viral titer was assessed using EID50 assay. RESULTS EIDD-2801 was found to be significantly effective against influenza A virus and influenza B virus. The antiviral activity against SARS-CoV-2 and further co-infection with influenza virus was also distinct. EIDD-2801 had potent antiviral effects against multiple respiratory viruses both in vitro and in vivo. CONCLUSION This study demonstrated that the small-molecule compound EIDD-2801, an orally available broad-spectrum antiviral agent, significantly inhibited the infection of influenza virus and SARS-CoV-2 and effectively protected animals from lethal influenza virus co-infection.
Collapse
Affiliation(s)
- Bingshuo Qian
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130122, China
- School of Pharmacy, Henan University, Kaifeng, 475004, China
| | - Rongbo Luo
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130122, China
| | - Beilei Shen
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130122, China
| | - Lingjun Fan
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130122, China
| | - Junkui Zhang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130122, China
- School of Pharmacy, Henan University, Kaifeng, 475004, China
| | - Shijun Zhang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130122, China
| | - Yan Sun
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130122, China
| | - Xiuwen Deng
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130122, China
| | - Xiaobin Pang
- School of Pharmacy, Henan University, Kaifeng, 475004, China.
| | - Wu Zhong
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China.
| | - Yuwei Gao
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130122, China.
| |
Collapse
|
7
|
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
|
8
|
Clark TM, Coggins SJ, Korman R, King J, Malik R. Treatment of feline infectious peritonitis in cats with molnupiravir: clinical observations and outcomes for 54 cases. Aust Vet J 2025. [PMID: 40234239 DOI: 10.1111/avj.13433] [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: 06/12/2024] [Revised: 02/13/2025] [Accepted: 02/21/2025] [Indexed: 04/17/2025]
Abstract
OBJECTIVE To evaluate the clinical applications and treatment outcomes using molnupiravir for the treatment of naturally occurring feline infectious peritonitis virus (FIPv). METHODS Ninety-two client-owned cats with confirmed or presumptive FIP were retrospectively recruited from 35 veterinary practices, primarily in Australia, between February 2023 and March 2024. Cats were categorised based on treatment received: Cohort A: Molnupiravir treatment: monotherapy, maintenance or rescue therapy; Cohort B: Remdesivir and/or GS-441524 treatment. Seventy-eight cats were enrolled. Molnupiravir was administered orally for a median of 84 days, at a median dose of 13.3 mg/kg BID. Remission was defined as the resolution of FIP-related signs with (i) normalisation of serum globulin concentrations and A:G ratio (≥0.6), or (ii) sustained clinical remission for at least 100 days after stopping anti-viral therapy. Cure rate was defined as the percentage of cats achieving sustained remission, without requiring rescue therapy or experiencing a relapsed disease. RESULTS Molnupiravir monotherapy resulted in a cure rate of 72% (13/18) while molnupiravir maintenance therapy achieved a cure rate of 86% (25/29), and molnupiravir utilised as a rescue therapy achieved a cure rate of 100% (7/7). Treatment with remdesivir/GS-441524 resulted in a cure rate of 71% (17/24 cats). Survival analysis revealed no difference in outcomes between cats treated with molnupiravir monotherapy and those treated with remdesivir/GS-441524. Adverse events associated with molnupiravir therapy included neutropenia, and transient elevations in hepatic enzymes. CONCLUSION Molnupiravir demonstrated comparable survival outcomes to remdesivir/GS-441524 for treating FIP and serves as an accessible, effective option across various presentations, including ocular and neurologic forms.
Collapse
Affiliation(s)
- T M Clark
- Veterinary Specialist Services, 24/34 Goggs Road, Jindalee, Queensland, 4074, Australia
| | - S J Coggins
- Sydney School of Veterinary Science, Faculty of Science, B14, The University of Sydney, Sydney, New South Wales, 2006, Australia
| | - R Korman
- Veterinary Specialist Services, 1-15 Lexington Road, Underwood, Queensland, 4119, Australia
| | - J King
- Veterinary Specialist Services, 24/34 Goggs Road, Jindalee, Queensland, 4074, Australia
| | - R Malik
- Sydney School of Veterinary Science, Faculty of Science, B14, The University of Sydney, Sydney, New South Wales, 2006, Australia
| |
Collapse
|
9
|
Nambi G, Alghadier M, Mohamed SHP, Vellaiyan A, Ebrahim EE, Eltabey Sobeh D, Aldhafian OR, Sirajudeen MS, Muthusamy H, Unnikrishnan R, Alshahrani NN, Albarakati AJA. Comparative effects of integrated physical training with a high protein diet versus a regular protein diet in post-COVID-19 older men with sarcopenia symptoms. BMJ Nutr Prev Health 2025:bmjnph-2024-001076. [DOI: 10.1136/bmjnph-2024-001076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2025] Open
Abstract
BackgroundSarcopenia has become a significant health issue, particularly as a common consequence of COVID-19 in older adults.ObjectiveThis study aimed to explore the clinical and psychological effects of integrated physical training with a high-protein diet compared with a regular protein diet in community-dwelling older men who had recovered from COVID-19 and exhibited symptoms of sarcopenia.MethodsThis is a single-blinded, randomised, controlled study conducted from March 2020 to December 2023 at the University hospital. The eligible participants were randomly assigned to two groups using the block randomisation method. The first group underwent integrated physical training with a high-protein diet (group A; n=38), with an average age of 64.1±3.8 years, while the second group underwent integrated physical training with a regular protein diet (group B; n=38), with an average age of 64.5±3.6 years over an 8-week period. Clinical parameters (handgrip strength and muscle mass—cross-sectional area CSA) and psychological measures (kinesiophobia and quality of life) were assessed at baseline, the fourth week, the eighth week and at a 6-month follow-up. The data were analysed using a 4×2 mixed model for repeated measures at different time points.ResultsDemographic characteristics such as age, height, weight and body mass index did not show any statistically significant differences between the groups (p>0.05). After the 8-week intervention and at the 6-month follow-up, handgrip strength decreased by −5.0 (95% CI −6.21 to −3.78), midthigh CSA decreased by −3.7 (95% CI −6.53 to −0.86), midcalf CSA decreased by −4.4 (95% CI −6.80 to −2.00), kinesiophobia level increased by 8.1 (95% CI 7.16 to 9.03) and quality of life decreased by −6.3 (95% CI −9.0 to −3.5). The findings indicated significantly greater improvement (p<0.001) in group A compared with group B, although there was no significant difference in muscle CSA in the arm region (p>0.05).ConclusionIntegrated physical training with a high-protein diet led to improvements in clinical (muscle strength and muscle mass) and psychological (kinesiophobia and quality of life) parameters compared with integrated physical training with a regular protein diet in older men recovering from COVID-19 and displaying symptoms of sarcopenia.
Collapse
|
10
|
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
|
11
|
Zwicklbauer K, Bergmann M, Alberer M, von Both U, Hartmann K. [Feline infectious peritonitis - a current overview]. TIERARZTLICHE PRAXIS. AUSGABE K, KLEINTIERE/HEIMTIERE 2025; 53:96-102. [PMID: 40233794 DOI: 10.1055/a-2524-3760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Coronaviruses (CoVs) are positive, single-stranded RNA viruses that can infect various animal species as well as humans. Particularly relevant for cats is the feline coronavirus (FCoV), which is widespread in cat populations worldwide. Infection with FCoV is usually asymptomatic. However, in multi-cat households, approximately 5-12% of FCoV-infected cats develop feline infectious peritonitis (FIP) due to mutations in the spike gene. FIP is an immune-mediated disease that previously was always fatal. These mutations result in a tropism shift from enterocytes to monocytes and macrophages. The associated change in the virulence of FCoV leads to the characteristic granulomatous vasculitis and perivasculitis observed in FIP. Recently, significant advancements have been made in understanding FIP. Studies show that antiviral drugs used in human medicine, such as the nucleoside analog GS-441524, are effective against FIP and can provide affected cats with a survival chance of up to 100%. Additionally, a novel FCoV variant, FCoV-23, has been identified in cats from Cyprus. According to newest research, this virus arose through a recombination between FCoV and the highly virulent pantropic canine coronavirus; it can be directly transmitted from cat to cat and lead to FIP. Furthermore, increasing evidence suggests that FIP is frequently associated with myocarditis. This article provides an overview of the current knowledge on FIP, including its pathology, clinical signs, effective treatment options, and preventive measures.
Collapse
Affiliation(s)
| | | | - Martin Alberer
- Abteilung für Pädiatrische Infektionskrankheiten, Dr. von Haunersches Kinderspital, LMU München
| | - Ulrich von Both
- Abteilung für Pädiatrische Infektionskrankheiten, Dr. von Haunersches Kinderspital, LMU München
- Deutsches Zentrum für Infektionsforschung (DZIF), Partnerstandort München
| | | |
Collapse
|
12
|
Chawla A, Birger R, Maas BM, Cao Y, Wan H, Strizki J, Fridman A, Paschke A, de Anda C, Gao W, Rizk ML, Painter W, Holman W, Sardella S, Painter G, Stone JA. Comparison of Molnupiravir Exposure-Response Relationships for Virology Response and Mechanism of Action Biomarkers With Clinical Outcomes in Treatment of COVID-19. Clin Transl Sci 2025; 18:e70184. [PMID: 40138219 PMCID: PMC11939005 DOI: 10.1111/cts.70184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Accepted: 02/19/2025] [Indexed: 03/29/2025] Open
Abstract
Molnupiravir, an orally administered drug for the treatment of mild-to-moderate COVID-19, is a prodrug of the ribonucleoside β-D-N4-hydroxycytidine (NHC). NHC incorporation in the SARS-CoV-2 RNA strand causes an accumulation of deleterious errors in the genome, resulting in reduced viral infectivity and replication. Exposure-response (E-R) analyses for viral RNA mutation rate and virologic outcomes were conducted using data from three phase 2/3 studies of molnupiravir (P006, MOVe-IN, and MOVe-OUT). Three dose levels (200, 400, and 800 mg every 12 hours [Q12H]) and placebo were evaluated. E-R datasets were generated for SARS-CoV-2 RNA mutation and longitudinal SARS-CoV-2 RNA viral load. E-R models were defined for RNA mutation rate and viral load change from baseline at days 5 and 10. The models supported plasma NHC AUC0-12 as the appropriate pharmacokinetic driver for assessing E-R relationships. The highest percentage of participants with > 20 low-frequency nucleotide substitutions (LNS) per 10,000 bases, a measure of likely meaningful drug effect, was predicted in the 800 mg Q12H treatment group. A strong drug effect on the reduction of viral load was observed on days 5 and 10. E-R relationships were best represented by an Emax structural model with reasonable consistency in the estimated AUC50s (~2.3-fold), across the models, of 10,260 and 4390 nM*hr. for day 5 viral load change from baseline and LNS error rate, respectively. These biomarker E-R curves support the choice of 800 mg Q12H as providing near-maximal drug effect, consistent with findings from the previously published molnupiravir E-R model of clinical outcomes.
Collapse
Affiliation(s)
| | | | | | | | - Hong Wan
- Merck & Co., Inc.RahwayNew JerseyUSA
| | | | | | | | | | - Wei Gao
- Merck & Co., Inc.RahwayNew JerseyUSA
| | | | | | | | | | | | | |
Collapse
|
13
|
Lin SH, Liu JW, Yen YT, Chen MT, Wang JT, Tu YK, Fang CT, Chang SC. Effectiveness of molnupiravir as early treatment for COVID-19 to prevent mortality and hospitalisation in high-risk adults: A systematic review and meta-analysis of randomised trials and real-world studies involving 1,612,082 patients. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2025:S1684-1182(25)00077-5. [PMID: 40204602 DOI: 10.1016/j.jmii.2025.03.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 03/20/2025] [Accepted: 03/26/2025] [Indexed: 04/11/2025]
Abstract
BACKGROUND The efficacy of molnupiravir for COVID-19 treatment remains controversial due to substantial heterogeneity in dosage and study settings across randomised controlled trials (RCTs). METHOD We systematically searched Medline, PubMed, Embase, and the Cochrane Register of Clinical Trials up to February 3, 2025, for RCTs and real-world studies evaluating molnupiravir 800 mg twice daily as an early treatment for COVID-19 to prevent mortality and hospitalisation in high-risk adult outpatients. The primary outcomes were all-cause mortality and all-cause hospitalisation. Random-effects models were used to estimate pooled effect sizes. RESULTS Thirty-four studies were included, comprising 30,345 participants from 11 RCTs and 1,581,737 participants from 23 cohort studies. Molnupiravir reduced mortality risk by 55 %-65 % at 28 days (RCTs: risk ratio [RR] 0.35; 95 % CI 0.12-0.98, I2 0 %; cohort studies: RR 0.45; 95 % CI 0.27-0.73, I2 91 %). This benefit persisted at 3 months (RR 0.47; 95 % CI 0.23-0.95, I2 93 %) and 6 months (RR 0.62; 95 % CI 0.52-0.74, I2 0 %). The effectiveness in preventing 28-day hospitalisation varied by participants' mean age in both RCTs (35-45 vs. 45-57 years: RR 0.55; 95 % CI 0.36-0.84 vs. 1.06; 95 % CI 0.81-1.39, subgroup difference P = 0.01) and cohort studies (62-74 vs. 75-85 years: RR 0.88; 95 % CI 0.77-1.01 vs. 0.56; 95 % CI 0.44-0.72, subgroup difference P < 0.01). CONCLUSIONS Molnupiravir significantly reduces the risk of mortality. It also lowers the risk of hospitalisation in the oldest group (mean age ≥75 years) but not in younger groups (mean age 45-74 years).
Collapse
Affiliation(s)
- Shen-Hua Lin
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan; Department of Pharmacy, Fu Jen Catholic University Hospital, Fu Jen Catholic University, New Taipei City, Taiwan.
| | - Jen-Wei Liu
- Department of Pharmacy, Fu Jen Catholic University Hospital, Fu Jen Catholic University, New Taipei City, Taiwan; School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan.
| | - Yi-Ti Yen
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan.
| | - Mong-Tan Chen
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan.
| | - Jann-Tay Wang
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan; National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan, Taiwan.
| | - Yu-Kang Tu
- Department of Dentistry, National Taiwan University Hospital, Taipei, Taiwan; Institute of Health Data Analytics & Statistics, College of Public Health, National Taiwan University, Taipei, Taiwan.
| | - Chi-Tai Fang
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan; Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan; National Taiwan University School of Medicine, Taipei, Taiwan; Master of Public Health Program, College of Public Health, National Taiwan University, Taipei, Taiwan.
| | - Shan-Chwen Chang
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan; National Taiwan University School of Medicine, Taipei, Taiwan.
| |
Collapse
|
14
|
Ma L, Lin Y. Orthogonal RNA replication enables directed evolution and Darwinian adaptation in mammalian cells. Nat Chem Biol 2025; 21:451-463. [PMID: 39753704 DOI: 10.1038/s41589-024-01783-2] [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: 04/04/2024] [Accepted: 10/31/2024] [Indexed: 01/31/2025]
Abstract
Directed evolution in mammalian cells offers a powerful approach for advancing synthetic biology applications. However, existing mammalian-based directed evolution methods face substantial bottlenecks, including host genome interference, small library size and uncontrolled mutagenesis. Here we engineered an orthogonal alphaviral RNA replication system to evolve RNA-based devices, enabling RNA replicase-assisted continuous evolution (REPLACE) in proliferating mammalian cells. This system generates a large, continuously diversified library of replicative RNAs through replicase-limited mode of replication and inducible mutagenesis. Using REPLACE, we engineered fluorescent proteins and transcription factors. Notably, cells equipped with REPLACE can undergo Darwinian adaptation, allowing them to evolve in response to both cell-extrinsic and cell-intrinsic challenges. Collectively, this work establishes a powerful platform for advancing mammalian synthetic biology and cell engineering applications through directed evolution.
Collapse
Affiliation(s)
- Liang Ma
- Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
- The MOE Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing, China
| | - Yihan Lin
- Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China.
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China.
- The MOE Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing, China.
- Chengdu Academy for Advanced Interdisciplinary Biotechnologies, Peking University, Chengdu, China.
| |
Collapse
|
15
|
Saha A, Choudhary S, Walia P, Kumar P, Tomar S. Transformative approaches in SARS-CoV-2 management: Vaccines, therapeutics and future direction. Virology 2025; 604:110394. [PMID: 39889481 DOI: 10.1016/j.virol.2025.110394] [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: 10/15/2024] [Revised: 12/24/2024] [Accepted: 12/28/2024] [Indexed: 02/03/2025]
Abstract
The global healthcare and economic challenges caused by the pandemic of COVID-19 reinforced the urgent demand for quick and effective therapeutic and preventative interventions. While vaccines served as the frontline of defense, antivirals emerged as adjunctive countermeasures, especially for people who developed infection, were immunocompromised, or were reluctant to be vaccinated. Beyond the serious complications of SARS-CoV-2 infection, the threats of long-COVID and the potential for zoonotic spillover continue to be significant health concerns that cannot be overlooked. Moreover, the incessant viral evolution, clinical safety issues, waning immune responses, and the emergence of drug-resistant variants pinpoint towards more severe viral threats in the future and call for broad-spectrum innovative therapies as a pre-pandemic preparedness measure. The present review provides a comprehensive up-to-date overview of the strategies utilized in the development of classical and next-generation vaccines against SARS-CoV-2, the clinical and experimental data obtained from clinical trials, while addressing safety risks that may arise. Besides vaccines, the review also covers recent breakthroughs in anti-SARS-CoV-2 drug discovery, emphasizing druggable viral and host targets, virus- and host-targeting antivirals, and highlighting mechanistically representative molecules that are either approved or are under clinical investigation. In conclusion, the integration of both vaccines and antiviral therapies, along with swift innovative strategies to address viral evolution and drug resistance is crucial to strengthen our preparedness against future viral outbreaks.
Collapse
Affiliation(s)
- Ankita Saha
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Uttarakhand, 247667, India
| | - Shweta Choudhary
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Uttarakhand, 247667, India
| | - Priyanshu Walia
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Uttarakhand, 247667, India
| | - Pravindra Kumar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Uttarakhand, 247667, India
| | - Shailly Tomar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Uttarakhand, 247667, India.
| |
Collapse
|
16
|
Mori Y, Yogo R, Kobayashi H, Katsuzaki H, Hirao Y, Kato S, Kotani H, Kawanishi S, Murata M, Oikawa S. Reactive oxygen species-mediated cytotoxic and DNA-damaging mechanism of N4-hydroxycytidine, a metabolite of the COVID-19 therapeutic drug molnupiravir. Free Radic Res 2025; 59:205-214. [PMID: 39973207 DOI: 10.1080/10715762.2025.2469738] [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: 08/21/2024] [Revised: 12/05/2024] [Accepted: 01/13/2025] [Indexed: 02/21/2025]
Abstract
Molnupiravir is a prodrug of the antiviral ribonucleoside analogue N4-hydroxycytidine (NHC), for use in the treatment of coronavirus disease 2019 (COVID-19). However, it is generally considered that NHC-triphosphate is incorporated into the host genome to induce mutations. In our previous preliminary report, we proposed oxidative DNA damage by NHC via cytidine deaminase (CDA)-mediated ROS formation. In the present study, we investigated cell viability using the HL-60 human leukemia cell line and its H2O2-resistant clone, HP100 cells. The survival rate was significantly reduced in HL-60 cells treated with NHC, but not in HP100 cells. LC-MS analysis revealed that uridine formation occurred from CDA-treated NHC, suggesting that CDA metabolizes NHC to uridine and hydroxylamine. We clarified mechanisms of CDA-mediated reactive oxygen species (ROS) generation and DNA damage by NHC using isolated DNA. CDA-treated NHC induced DNA damage in the presence of Cu(II). The DNA damage was enhanced by NADH addition and piperidine treatment. CDA-treated NHC and Cu(II) caused piperidine-labile sites at thymine, cytosine, and guanine, and the DNA cleavage pattern was similar to that of hydroxylamine. Catalase and bathocuproine inhibited the DNA damage, indicating the involvement of H2O2 and Cu(I). An indicator of oxidative DNA damage, 8-oxo-7,8-dihydro-2'-deoxyguanosine formation by CDA-treated NHC, was lower under hypoxic conditions than under normal conditions. Therefore, hydroxylamine, possibly produced from NHC treated with CDA, could induce metal-dependent H2O2 generation during the redox reactions, suggesting that oxidative DNA damage induced by ROS plays an important role in molnupiravir-related cytotoxicity and mutagenicity.
Collapse
Affiliation(s)
- Yurie Mori
- Department of Environmental and Molecular Medicine, Mie University Graduate School of Medicine, Tsu, Japan
| | - Rinya Yogo
- Department of Environmental and Molecular Medicine, Mie University Graduate School of Medicine, Tsu, Japan
- Department of Forensic Medicine and Sciences, Mie University Graduate School of Medicine, Tsu, Japan
| | - Hatasu Kobayashi
- Department of Environmental and Molecular Medicine, Mie University Graduate School of Medicine, Tsu, Japan
| | - Hirotaka Katsuzaki
- Department of Life Sciences, Graduate School of Bioresources, Mie University, Tsu, Japan
| | - Yuichiro Hirao
- Department of Environmental and Molecular Medicine, Mie University Graduate School of Medicine, Tsu, Japan
| | - Shinya Kato
- Radioisotope Experimental Facility, Advanced Science Research Promotion Center, Mie University, Tsu, Japan
| | - Hirokazu Kotani
- Department of Forensic Medicine and Sciences, Mie University Graduate School of Medicine, Tsu, Japan
| | - Shosuke Kawanishi
- Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Suzuka, Japan
| | - Mariko Murata
- Department of Environmental and Molecular Medicine, Mie University Graduate School of Medicine, Tsu, Japan
| | - Shinji Oikawa
- Department of Environmental and Molecular Medicine, Mie University Graduate School of Medicine, Tsu, Japan
| |
Collapse
|
17
|
Grimes SL, Heaton BE, Anderson ML, Burke K, Stevens L, Lu X, Heaton NS, Denison MR, Anderson-Daniels J. The coronavirus nsp14 exoribonuclease interface with the cofactor nsp10 is essential for efficient virus replication and enzymatic activity. J Virol 2025; 99:e0170824. [PMID: 39791922 PMCID: PMC11852845 DOI: 10.1128/jvi.01708-24] [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: 09/25/2024] [Accepted: 11/27/2024] [Indexed: 01/12/2025] Open
Abstract
Coronaviruses (CoVs) encode non-structural proteins (nsp's) 1-16, which assemble to form replication-transcription complexes that function in viral RNA synthesis. All CoVs encode a proofreading 3'-5' exoribonuclease in non-structural protein 14 (nsp14-ExoN) that mediates proofreading and high-fidelity replication and is critical for other roles in replication and pathogenesis. The in vitro enzymatic activity of nsp14-ExoN is enhanced in the presence of the cofactor nsp10. We introduced alanine substitutions in nsp14 of murine hepatitis virus (MHV) at the nsp14-nsp10 interface and recovered mutant viruses with a range of impairments in replication and in vitro biochemical exonuclease activity. Two of these substitutions, nsp14 K7A and D8A, had impairments intermediate between wild type-MHV nsp14 and the known ExoN(-) D89A/E91A nsp14 catalytic inactivation mutant. All introduced nsp14-nsp10 interface alanine substitutions impaired in vitro exonuclease activity. Passage of the K7A and D8A mutant viruses selected second-site non-synonymous mutations in nsp14 associated with improved mutant virus replication and exonuclease activity. These results confirm the essential role of the nsp14-nsp10 interaction for efficient enzymatic activity and virus replication, identify proximal and long-distance determinants of nsp14-nsp10 interaction, and support targeting the nsp14-nsp10 interface for viral inhibition and attenuation.IMPORTANCECoronavirus replication requires assembly of a replication transcription complex composed of nsp's, including polymerase, helicase, exonuclease, capping enzymes, and non-enzymatic cofactors. The coronavirus nsp14 exoribonuclease mediates several functions in the viral life cycle including genomic and subgenomic RNA synthesis, RNA recombination, RNA proofreading and high-fidelity replication, and native resistance to many nucleoside analogs. The nsp-14 exonuclease activity in vitro requires the non-enzymatic cofactor nsp10, but the determinants and importance of the nsp14-nsp10 interactions during viral replication have not been defined. Here we show that for the coronavirus murine hepatitis virus, nsp14 residues at the nsp14-nsp10 interface are essential for efficient viral replication and in vitro exonuclease activity. These results shed new light on the requirements for protein interactions within the coronavirus replication transcription complex, and they may reveal novel non-active-site targets for virus inhibition and attenuation.
Collapse
Affiliation(s)
- Samantha L. Grimes
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Brook E. Heaton
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Mackenzie L. Anderson
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Katie Burke
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Laura Stevens
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Xiaotao Lu
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Nicholas S. Heaton
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Mark R. Denison
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | | |
Collapse
|
18
|
Sims AC, Schäfer A, Okuda K, Leist SR, Kocher JF, Cockrell AS, Hawkins PE, Furusho M, Jensen KL, Kyle JE, Burnum-Johnson KE, Stratton KG, Lamar NC, Niccora CD, Weitz KK, Smith RD, Metz TO, Waters KM, Boucher RC, Montgomery SA, Baric RS, Sheahan TP. Dysregulation of lung epithelial cell homeostasis and immunity contributes to Middle East respiratory syndrome coronavirus disease severity. mSphere 2025; 10:e0095124. [PMID: 39882872 PMCID: PMC11853001 DOI: 10.1128/msphere.00951-24] [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: 12/10/2024] [Accepted: 01/15/2025] [Indexed: 01/31/2025] Open
Abstract
Coronaviruses (CoV) emerge suddenly from animal reservoirs to cause novel diseases in new hosts. Discovered in 2012, the Middle East respiratory syndrome coronavirus (MERS-CoV) is endemic in camels in the Middle East and is continually causing local outbreaks and epidemics. While all three newly emerging human CoVs from the past 20 years (SARS-CoV, SARS-CoV-2, and MERS-CoV) cause respiratory disease, each CoV has unique host interactions that drive differential pathogeneses. To better understand the virus and host interactions driving lethal MERS-CoV infection, we performed a longitudinal multi-omics analysis of sublethal and lethal MERS-CoV infection in mice. Significant differences were observed in body weight loss, virus titers, and acute lung injury among lethal and sub-lethal virus doses. Virus-induced apoptosis of type I and II alveolar epithelial cells suggests that loss or dysregulation of these key cell populations was a major driver of severe disease. Omics analysis suggested differential pathogenesis was multi-factorial with clear differences among innate and adaptive immune pathways as well as those that regulate lung epithelial homeostasis. Infection of mice lacking functional T and B cells showed that adaptive immunity was important in controlling viral replication but also increased pathogenesis. In summary, we provide a high-resolution host response atlas for MERS-CoV infection and disease severity. Multi-omics studies of viral pathogenesis offer a unique opportunity to not only better understand the molecular mechanisms of disease but also to identify genes and pathways that can be exploited for therapeutic intervention all of which is important for our future pandemic preparedness.IMPORTANCEEmerging coronaviruses like SARS-CoV, SARS-CoV-2, and MERS-CoV cause a range of disease outcomes in humans from an asymptomatic, moderate, and severe respiratory disease that can progress to death but the factors causing these disparate outcomes remain unclear. Understanding host responses to mild and life-threatening infections provides insight into virus-host networks within and across organ systems that contribute to disease outcomes. We used multi-omics approaches to comprehensively define the host response to moderate and severe MERS-CoV infection. Severe respiratory disease was associated with dysregulation of the immune response. Key lung epithelial cell populations that are essential for lung function get infected and die. Mice lacking key immune cell populations experienced greater virus replication but decreased disease severity implicating the immune system in both protective and pathogenic roles in response to MERS-CoV. These data could be utilized to design new therapeutic strategies targeting specific pathways that contribute to severe disease.
Collapse
Affiliation(s)
- Amy C. Sims
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Alexandra Schäfer
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Kenichi Okuda
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Sarah R. Leist
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Jacob F. Kocher
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Adam S. Cockrell
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Padraig E. Hawkins
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Minako Furusho
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Kara L. Jensen
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Jennifer E. Kyle
- Biological Sciences Division, Pacific Northwest National Laboratories, Richland, Washington, USA
| | | | - Kelly G. Stratton
- Biological Sciences Division, Pacific Northwest National Laboratories, Richland, Washington, USA
| | - Natalie C. Lamar
- AI & Data Analytics Division, Pacific Northwest National Laboratories, Richland, Washington, USA
| | - Carrie D. Niccora
- Biological Sciences Division, Pacific Northwest National Laboratories, Richland, Washington, USA
| | - Karl K. Weitz
- Biological Sciences Division, Pacific Northwest National Laboratories, Richland, Washington, USA
| | - Richard D. Smith
- Biological Sciences Division, Pacific Northwest National Laboratories, Richland, Washington, USA
| | - Thomas O. Metz
- Biological Sciences Division, Pacific Northwest National Laboratories, Richland, Washington, USA
| | - Katrina M. Waters
- Biological Sciences Division, Pacific Northwest National Laboratories, Richland, Washington, USA
| | - Richard C. Boucher
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Stephanie A. Montgomery
- Department of Pathology & Laboratory Medicine, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Ralph S. Baric
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Timothy P. Sheahan
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
19
|
Waters MD, Warren SG. A tale of two drugs: Molnupiravir and Paxlovid. MUTATION RESEARCH. REVIEWS IN MUTATION RESEARCH 2025; 795:108533. [PMID: 39920989 DOI: 10.1016/j.mrrev.2025.108533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 01/24/2025] [Accepted: 01/27/2025] [Indexed: 02/10/2025]
Abstract
The orally administered antiviral drug Lagevrio or molnupiravir (MOV) and the combination antiviral drug nirmatrelvir/ritonavir or Paxlovid (PAX) have been shown to reduce the likelihood of hospitalization and death for high-risk patients with COVID-19. Clinical studies, including those comparing PAX and MOV, were reviewed; both drugs are effective in reducing morbidity and mortality in COVID patients, although PAX generally appears to be more efficacious. Both drugs received Emergency Use Authorization in the United States for mild to moderate COVID-19 infection, while only PAX has subsequently been given full FDA approval. The principal disadvantage of PAX is that it interacts with many commonly used drugs, while MOV does not. The purpose of this review is to summarize current information and knowledge about these two drugs. The two drugs have completely different mechanisms of action. PAX inhibits viral replication while MOV induces viral replication errors that are expected to lead to viral inactivation. There is, however, the potential that MOV also could mutate host DNA and cause the virus to mutate into variants with new features. The package insert for MOV states that patients should be notified of relevant toxicity issues before administration. Sensitive mutation detection/analysis studies, such as error corrected Next Generation Sequencing (ecNGS) or HPRT mutation detection assays, in MOV-treated patients are needed to establish the safety of MOV.
Collapse
Affiliation(s)
- Michael D Waters
- Michael Waters Consulting USA, 210 N Wake Street, Hillsborough, NC 27278, United States.
| | - Stafford G Warren
- Anne Arundel Medical Center, 2001 Medical Parkway, Annapolis, MD 21401, United States
| |
Collapse
|
20
|
Zhang W, Xia L, Yuan Z, Liu M, Jiao Y, Wang Z. Simultaneous determination of nirmatrelvir, ritonavir, and beta-D-N4-hydroxycytidine in human plasma and epithelial lining fluid using LC-MS/MS and its clinical application to compare rates of achieving effective concentrations. Heliyon 2025; 11:e41737. [PMID: 39882486 PMCID: PMC11774765 DOI: 10.1016/j.heliyon.2025.e41737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 12/28/2024] [Accepted: 01/06/2025] [Indexed: 01/31/2025] Open
Abstract
Currently, the trials found that the clinical efficacy of molnupiravir is lower than ritonavir-boosted nirmatrelvir. An explanation for these different efficacies in clinical treatments is still limited. The analysis method was developed and validated to simultaneously quantify nirmatrelvir, ritonavir, and beta-D-N4-hydroxycytidine (NHC) in human plasma and bronchoalveolar lavage fluid (BALF) by electrospray ionization mass spectrometry. Our method was validated over a linear range of 30-10000 ng/mL for both matrices, with precision and accuracy within 15 % across four concentrations. Recovery rates for both analytes from plasma and BALF were between 90.7-102.2 % and 90.5-107.7 %, respectively. The analytical method was then applied to monitor therapeutic drug concentrations in 59 plasma samples from 23 patients receiving ritonavir-boosted nirmatrelvir or molnupiravir. By setting target plasma concentrations of 292 ng/mL for nirmatrelvir and 1205 ng/mL for NHC, based on in vitro antiviral 90 % virus inhibitory concentrations (EC90), the drug's molecular weight and its binding to human plasma proteins, we observed that ritonavir-boosted nirmatrelvir had substantially greater rates of achieving target plasma concentrations. Additionally, we monitored epithelial lining fluid in 4 BALF samples from 4 patients and observed that NHC exhibited higher permeability in lung tissue (approximately 20 % higher than nirmatrelvir). However, subtherapeutic antiviral concentrations of NHC were also present in epithelial lining fluid. These findings highlight the importance of considering these factors in determining the efficacy of these drugs in treating coronavirus disease 2019 (COVID-19).
Collapse
Affiliation(s)
- Wenjing Zhang
- Department of Pharmacy, Shanghai Changhai Hospital, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Lin Xia
- Department of Pharmacy, Shanghai Changhai Hospital, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Zhilong Yuan
- Department of Pharmacy, Shanghai Changhai Hospital, The First Affiliated Hospital of Naval Medical University, Shanghai, China
- School of Pharmacy, Bengbu Medical College, Bengbu, 233004, China
| | - Mengdan Liu
- Department of Pharmacy, Shanghai Changhai Hospital, The First Affiliated Hospital of Naval Medical University, Shanghai, China
- College of Life Sciences and Biopharmaceuticals, Shenyang Pharmaceutical University, Shenyang, China
| | - Yang Jiao
- Department of Respiratory and Critical Care Medicine, Shanghai Changhai Hospital, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Zhuo Wang
- Department of Pharmacy, Shanghai Changhai Hospital, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| |
Collapse
|
21
|
Ling-Hu T, Simons LM, Rios-Guzman E, Carvalho AM, Agnes MFR, Alisoltanidehkordi A, Ozer EA, Lorenzo-Redondo R, Hultquist JF. The impact of remdesivir on SARS-CoV-2 evolution in vivo. JCI Insight 2025; 10:e182376. [PMID: 39836474 PMCID: PMC11949014 DOI: 10.1172/jci.insight.182376] [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: 04/24/2024] [Accepted: 01/13/2025] [Indexed: 01/23/2025] Open
Abstract
The impact of remdesivir on SARS-CoV-2 diversity and evolution in vivo has remained unclear. In this single-center, retrospective cohort study, we assessed SARS-CoV-2 diversification and diversity over time in a cohort of hospitalized patients who did or did not receive remdesivir. Whole-genome sequencing was performed on 98 paired specimens collected from 49 patients before and after remdesivir administration. The genetic divergence between paired specimens was not significantly different in this cohort compared with that in a control group of patients who did not receive the drug. However, when we focused on minority variants, several positions showed preferential diversification after remdesivir treatment, some of which were associated with specific variants of concern. Most notably, remdesivir administration resulted in strong selection for a nonsynonymous mutation in nsp12, G671S, previously associated with enhanced viral fitness. This same mutation was found to be enriched in a second cohort of 143 inpatients with specimens collected after remdesivir administration compared with controls. Only one other mutation previously implicated in remdesivir resistance (nsp12:V792I) was found to be preferentially selected for after remdesivir administration. These data suggest that SARS-CoV-2 variants with enhanced replicative fitness may be selected for in the presence of antiviral therapy as an indirect means to overcome this selective pressure.
Collapse
Affiliation(s)
- Ted Ling-Hu
- Division of Infectious Diseases, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Center for Pathogen Genomics and Microbial Evolution, Northwestern University Havey Institute for Global Health, Chicago, Illinois, USA
| | - Lacy M. Simons
- Division of Infectious Diseases, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Center for Pathogen Genomics and Microbial Evolution, Northwestern University Havey Institute for Global Health, Chicago, Illinois, USA
| | - Estefany Rios-Guzman
- Division of Infectious Diseases, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Center for Pathogen Genomics and Microbial Evolution, Northwestern University Havey Institute for Global Health, Chicago, Illinois, USA
| | - Alexandre Machado Carvalho
- Division of Infectious Diseases, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Center for Pathogen Genomics and Microbial Evolution, Northwestern University Havey Institute for Global Health, Chicago, Illinois, USA
| | - Maria Francesca R. Agnes
- Division of Infectious Diseases, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Center for Pathogen Genomics and Microbial Evolution, Northwestern University Havey Institute for Global Health, Chicago, Illinois, USA
| | - Arghavan Alisoltanidehkordi
- Division of Infectious Diseases, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Center for Pathogen Genomics and Microbial Evolution, Northwestern University Havey Institute for Global Health, Chicago, Illinois, USA
| | - Egon A. Ozer
- Division of Infectious Diseases, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Center for Pathogen Genomics and Microbial Evolution, Northwestern University Havey Institute for Global Health, Chicago, Illinois, USA
| | - Ramon Lorenzo-Redondo
- Division of Infectious Diseases, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Center for Pathogen Genomics and Microbial Evolution, Northwestern University Havey Institute for Global Health, Chicago, Illinois, USA
| | - Judd F. Hultquist
- Division of Infectious Diseases, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Center for Pathogen Genomics and Microbial Evolution, Northwestern University Havey Institute for Global Health, Chicago, Illinois, USA
| |
Collapse
|
22
|
Palla S, Palla SR, Liu JJ, Chao TL, Lee TH, Kavala V, Liu IC, Wang LHC, Chang SY, Yao CF, Liang PH. Green Synthesis of Tetrahydropyrazino[2,1-a:5,4-a']diisoquinolines as SARS-CoV-2 Entry Inhibitors. ACS OMEGA 2025; 10:1164-1176. [PMID: 39829498 PMCID: PMC11740144 DOI: 10.1021/acsomega.4c08640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 12/10/2024] [Accepted: 12/16/2024] [Indexed: 01/22/2025]
Abstract
A class of tetrahydropyrazino[2,1-a:5,4-a']diisoquinoline derivatives were synthesized under environmentally friendly conditions using water as the solvent. The 3-D structures of some synthesized compounds were determined by X-ray diffraction. Since naturally occurring isoquinoline alkaloids have significant antiviral activities against a wide range of viruses, including coronaviruses, the synthesized compounds were assayed for their inhibitory activities against SARS-CoV-2. Our results showed that the active compounds 50 and 96 blocked the delta SARS-CoV-2 entry into VeroE6 cells to display EC50 of 26.5 ± 6.9 and 17.0 ± 3.7 μM, respectively, by inhibiting the interaction between SARS-CoV-2 Spike's receptor binding domain (RBD) and human receptor angiotensin-converting enzyme 2 (ACE2), and CC50 greater than 100 μM. This study provides a green synthesis method of tetrahydropyrazinodiisoquinoline for antiviral or other applications.
Collapse
Affiliation(s)
- Sowndarya Palla
- Department
of Chemistry, National Taiwan Normal University, Taipei 11677, Taiwan
| | - Srinivasa Rao Palla
- Institute
of Biochemical Sciences, National Taiwan
University, Taipei 10617, Taiwan
- Taiwan
International Graduate Program, Academia
Sinica, Taipei 11529, Taiwan
| | - Jia-Jin Liu
- Institute
of Biochemical Sciences, National Taiwan
University, Taipei 10617, Taiwan
| | - Tai-Ling Chao
- Department
of Clinical Laboratory Sciences and Medical Biotechnology, National Taiwan University, Taipei 10048, Taiwan
| | - Ting-Hui Lee
- Department
of Life Science, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Veerababurao Kavala
- Department
of Chemistry, National Taiwan Normal University, Taipei 11677, Taiwan
| | - I-Chen Liu
- Institute
of Biochemical Sciences, National Taiwan
University, Taipei 10617, Taiwan
| | - Lily Hui-Ching Wang
- Institute
of Molecular and Cellular Biology, National
Tsing Hua University, Hsinchu 30013, Taiwan
| | - Sui-Yuan Chang
- Department
of Clinical Laboratory Sciences and Medical Biotechnology, National Taiwan University, Taipei 10048, Taiwan
- Department
of Laboratory Medicine, National Taiwan
University Hospital, Taipei 10002, Taiwan
| | - Ching-Fa Yao
- Department
of Chemistry, National Taiwan Normal University, Taipei 11677, Taiwan
| | - Po-Huang Liang
- Institute
of Biological Chemistry, Academia Sinica, Taipei 11529, Taiwan
- Institute
of Biochemical Sciences, National Taiwan
University, Taipei 10617, Taiwan
- Taiwan
International Graduate Program, Academia
Sinica, Taipei 11529, Taiwan
| |
Collapse
|
23
|
Anglero-Rodriguez YI, Lempp FA, Subramanian M, McIninch J, Schlegel MK, Bohan D, Wong E, Brown CR, Foster DJ, Castoreno AB, Nguyen T, Cuffe D, Montiel-Ruiz M, Kaiser H, Sahakyan A, Spreafico R, Morskaya SS, Barry JD, Berman D, Zhang L, Lefebvre S, Kasper A, Racie T, Weddle D, Mobley M, Wassarman K, Bisbe A, Zlatev I, Rogers A, Nechev L, Dybowski J, Chong S, Nair J, Simon A, Sloan K, Hwang S, Virgin HW, Fitzgerald K, Maier MA, Hinkle G, Hebner CM, Akinc A, Jadhav V. High resistance barrier and prophylactic protection in preclinical models of SARS-CoV-2 with two siRNA combination. Nucleic Acids Res 2025; 53:gkae1195. [PMID: 39657790 PMCID: PMC11724309 DOI: 10.1093/nar/gkae1195] [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: 09/11/2024] [Revised: 11/13/2024] [Accepted: 11/18/2024] [Indexed: 12/12/2024] Open
Abstract
RNA interference is a natural antiviral mechanism that could be harnessed to combat SARS-CoV-2 infection by targeting and destroying the viral RNA. We identified potent lipophilic small interfering RNA (siRNA) conjugates targeting highly conserved regions of SARS-CoV-2 outside of the spike-encoding region capable of achieving ≥3-log viral reduction. Serial passaging studies demonstrated that a two-siRNA combination prevented development of resistance compared to a single siRNA approach. Viral resistance to single siRNA treatment occurred due to emergence of point mutations at critical positions required for siRNA-mediated target binding and cleavage, which led to a loss of siRNA efficacy. With a two-siRNA combination, emergence of mutations within the siRNA binding site was abolished. When delivered intranasally, two-siRNA combination protected Syrian hamsters from weight loss and lung pathology by viral infection upon prophylactic administration but not following onset of infection. Together, the data support potential utility of RNAi as a prophylactic approach with high resistance barrier to counteract SARS-CoV-2 emergent variants and complement vaccination. Most importantly, given that the siRNAs can be rapidly developed from a new pathogen sequence, this strategy has implications as a new type of preventive medicine that may protect against future coronavirus pandemics.
Collapse
Affiliation(s)
| | | | | | | | | | - Dana Bohan
- Vir Biotechnology Inc.; San Francisco, CA 94158, USA
| | - Emily Wong
- Vir Biotechnology Inc.; San Francisco, CA 94158, USA
| | | | | | | | - Tuyen Nguyen
- Alnylam Pharmaceuticals; Cambridge, MA 002142, USA
| | - Dara Cuffe
- Alnylam Pharmaceuticals; Cambridge, MA 002142, USA
| | | | - Hannah Kaiser
- Vir Biotechnology Inc.; San Francisco, CA 94158, USA
| | - Anna Sahakyan
- Vir Biotechnology Inc.; San Francisco, CA 94158, USA
| | | | | | | | | | - Ligang Zhang
- Alnylam Pharmaceuticals; Cambridge, MA 002142, USA
| | | | - Anne Kasper
- Alnylam Pharmaceuticals; Cambridge, MA 002142, USA
| | | | - Diann Weddle
- Alnylam Pharmaceuticals; Cambridge, MA 002142, USA
| | | | | | - Anna Bisbe
- Alnylam Pharmaceuticals; Cambridge, MA 002142, USA
| | - Ivan Zlatev
- Alnylam Pharmaceuticals; Cambridge, MA 002142, USA
| | - Arlin Rogers
- Alnylam Pharmaceuticals; Cambridge, MA 002142, USA
| | | | | | - Saeho Chong
- Alnylam Pharmaceuticals; Cambridge, MA 002142, USA
| | | | - Amy Simon
- Alnylam Pharmaceuticals; Cambridge, MA 002142, USA
| | - Kevin Sloan
- Alnylam Pharmaceuticals; Cambridge, MA 002142, USA
| | | | - Herbert W Virgin
- Vir Biotechnology Inc.; San Francisco, CA 94158, USA
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | | | | | | | | | - Akin Akinc
- Alnylam Pharmaceuticals; Cambridge, MA 002142, USA
| | | |
Collapse
|
24
|
Yin P, Sobolik EB, May NA, Wang S, Fayed A, Vyshenska D, Drobish AM, Parks MG, Lello LS, Merits A, Morrison TE, Greninger AL, Kielian M. Mutations in chikungunya virus nsP4 decrease viral fitness and sensitivity to the broad-spectrum antiviral 4'-Fluorouridine. PLoS Pathog 2025; 21:e1012859. [PMID: 39804924 PMCID: PMC11759387 DOI: 10.1371/journal.ppat.1012859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 01/24/2025] [Accepted: 12/23/2024] [Indexed: 01/16/2025] Open
Abstract
Chikungunya virus (CHIKV) is an arthritogenic alphavirus that has re-emerged to cause large outbreaks of human infections worldwide. There are currently no approved antivirals for treatment of CHIKV infection. Recently, we reported that the ribonucleoside analog 4'-fluorouridine (4'-FlU) is a highly potent inhibitor of CHIKV replication, and targets the viral nsP4 RNA dependent RNA polymerase. In mouse models, oral therapy with 4'-FlU diminished viral tissue burdens and virus-induced disease signs. To provide critical evidence for the potential of 4'-FlU as a CHIKV antiviral, here we selected for CHIKV variants with decreased 4'-FlU sensitivity, identifying two pairs of mutations in nsP2 and nsP4. The nsP4 mutations Q192L and C483Y were predominantly responsible for reduced sensitivity. These variants were still inhibited by higher concentrations of 4'-FlU, and the mutations did not change nsP4 fidelity or provide a virus fitness advantage in vitro or in vivo. Pathogenesis studies in mice showed that the nsP4-C483Y variant caused similar disease and viral tissue burden as WT CHIKV, while the nsP4-Q192L variant was strongly attenuated. Together these results support the potential of 4'-FlU to be an important antiviral against CHIKV.
Collapse
Affiliation(s)
- Peiqi Yin
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Elizabeth B. Sobolik
- Virology Division, Department of Laboratory Medicine and Pathology, University of Washington Medical Center, Seattle, Washington, United States of America
| | - Nicholas A. May
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - Sainan Wang
- Institute of Bioengineering, University of Tartu, Tartu, Estonia
| | - Atef Fayed
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Dariia Vyshenska
- Virology Division, Department of Laboratory Medicine and Pathology, University of Washington Medical Center, Seattle, Washington, United States of America
| | - Adam M. Drobish
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - M. Guston Parks
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | | | - Andres Merits
- Institute of Bioengineering, University of Tartu, Tartu, Estonia
| | - Thomas E. Morrison
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - Alexander L. Greninger
- Virology Division, Department of Laboratory Medicine and Pathology, University of Washington Medical Center, Seattle, Washington, United States of America
| | - Margaret Kielian
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York, United States of America
| |
Collapse
|
25
|
Sinha A, Tony AMC, Roy S. How fingers affect folding of a thumb: Inter-subdomain cooperation in the folding of SARS-CoV-2 RdRp protein. Biophys Chem 2025; 316:107342. [PMID: 39490134 DOI: 10.1016/j.bpc.2024.107342] [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: 07/05/2024] [Revised: 09/29/2024] [Accepted: 10/15/2024] [Indexed: 11/05/2024]
Abstract
The RNA-dependent RNA polymerase (RdRp) of SARS-CoV-2 is a critical enzyme essential for the virus's replication and transcription, making it a key therapeutic target. The RdRp protein exhibits a characteristic cupped right-hand shaped structure with two vital subdomains: the fingers and the thumb. Despite being distinct, biophysical experiments suggest that these subdomains cooperate to facilitate RNA accommodation, ensuring RdRp functionality. To investigate the structure-based mechanisms underlying the fingers-thumb interaction in both apo and RNA-bound RdRp, we constructed a coarse-grained structure-based model based on recent cryo-electron microscopy data. The simulations reveal frequent open-to-closed conformational transitions in apo RdRp, akin to a breathing-like motion. These conformational changes are regulated by the fingers-thumb association and the folding dynamics of the thumb subdomain. The thumb adopts a stable fold only when tethered by the fingers-thumb interface; when these subdomains are disconnected, the thumb transitions into an open state. A significant number of open-to-closed transition events were analyzed to generate a transition contact probability map, which highlights a few specific residues at the thumb-fingers interface, distant from the RNA accommodation sites, as essential for inducing the thumb's folding process. Given that thumb subdomain folding is critical for RNA binding and viral replication, the study proposes that these interfacial residues may function as remote regulatory switches and could be targeted for the development of allosteric drugs against SARS-CoV-2 and similar RNA viruses.
Collapse
Affiliation(s)
- Anushree Sinha
- Department of Chemical Sciences, Indian Institute of Science Education and Research Kolkata, West Bengal 741246, India
| | - Angel Mary Chiramel Tony
- Department of Chemical Sciences, Indian Institute of Science Education and Research Kolkata, West Bengal 741246, India
| | - Susmita Roy
- Department of Chemical Sciences, Indian Institute of Science Education and Research Kolkata, West Bengal 741246, India.
| |
Collapse
|
26
|
Schrell L, Fuchs HL, Dickmanns A, Scheibner D, Olejnik J, Hume AJ, Reineking W, Störk T, Müller M, Graaf-Rau A, Diederich S, Finke S, Baumgärtner W, Mühlberger E, Balkema-Buschmann A, Dobbelstein M. Inhibitors of dihydroorotate dehydrogenase synergize with the broad antiviral activity of 4'-fluorouridine. Antiviral Res 2025; 233:106046. [PMID: 39638153 DOI: 10.1016/j.antiviral.2024.106046] [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: 10/05/2024] [Revised: 12/01/2024] [Accepted: 12/02/2024] [Indexed: 12/07/2024]
Abstract
RNA viruses present a constant threat to human health, often with limited options for vaccination or therapy. Notable examples include influenza viruses and coronaviruses, which have pandemic potential. Filo- and henipaviruses cause more limited outbreaks, but with high case fatality rates. All RNA viruses rely on the activity of a virus-encoded RNA-dependent RNA polymerase (RdRp). An antiviral nucleoside analogue, 4'-Fluorouridine (4'-FlU), targets RdRp and diminishes the replication of several RNA viruses, including influenza A virus and SARS-CoV-2, through incorporation into nascent viral RNA and delayed chain termination. However, the effective concentration of 4'-FlU varied among different viruses, raising the need to fortify its efficacy. Here we show that inhibitors of dihydroorotate dehydrogenase (DHODH), an enzyme essential for pyrimidine biosynthesis, can synergistically enhance the antiviral effect of 4'-FlU against influenza A viruses, SARS-CoV-2, henipaviruses, and Ebola virus. Even 4'-FlU-resistant mutant influenza A virus was re-sensitized towards 4'-FlU by DHODH inhibition. The addition of uridine rescued influenza A virus replication, strongly suggesting uridine depletion as a mechanism of this synergy. 4'-FlU was also highly effective against SARS-CoV-2 in a hamster model of COVID. We propose that the impairment of endogenous uridine synthesis by DHODH inhibition enhances the incorporation of 4'-FlU into viral RNAs. This strategy may be broadly applicable to enhance the efficacy of pyrimidine nucleoside analogues for antiviral therapy.
Collapse
Affiliation(s)
- Leon Schrell
- Department of Molecular Oncology, Göttingen Center of Molecular Biosciences (GZMB), University Medical Center Göttingen, Justus-von-Liebig-Weg 11, 37077, Göttingen, Germany
| | - Hannah L Fuchs
- Department of Molecular Oncology, Göttingen Center of Molecular Biosciences (GZMB), University Medical Center Göttingen, Justus-von-Liebig-Weg 11, 37077, Göttingen, Germany
| | - Antje Dickmanns
- Department of Molecular Oncology, Göttingen Center of Molecular Biosciences (GZMB), University Medical Center Göttingen, Justus-von-Liebig-Weg 11, 37077, Göttingen, Germany
| | - David Scheibner
- Institute of Novel and Emerging Infectious Diseases, Friedrich-Loeffler-Institut, Südufer 10, 17493, Greifswald, Insel Riems, Germany
| | - Judith Olejnik
- Department of Virology, Immunology & Microbiology, Chobanian and Avedisian School of Medicine, Boston University, Boston, MA, 02218, USA; National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, 02218, USA
| | - Adam J Hume
- Department of Virology, Immunology & Microbiology, Chobanian and Avedisian School of Medicine, Boston University, Boston, MA, 02218, USA; National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, 02218, USA
| | - Wencke Reineking
- Department of Pathology, University of Veterinary Medicine Hannover, Bünteweg 17, 30559, Hannover, Germany
| | - Theresa Störk
- Department of Pathology, University of Veterinary Medicine Hannover, Bünteweg 17, 30559, Hannover, Germany
| | - Martin Müller
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Südufer 10, 17493, Greifswald, Insel Riems, Germany
| | - Annika Graaf-Rau
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, Südufer 10, 17493, Greifswald, Insel Riems, Germany
| | - Sandra Diederich
- Institute of Novel and Emerging Infectious Diseases, Friedrich-Loeffler-Institut, Südufer 10, 17493, Greifswald, Insel Riems, Germany
| | - Stefan Finke
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Südufer 10, 17493, Greifswald, Insel Riems, Germany
| | - Wolfgang Baumgärtner
- Department of Pathology, University of Veterinary Medicine Hannover, Bünteweg 17, 30559, Hannover, Germany
| | - Elke Mühlberger
- Department of Virology, Immunology & Microbiology, Chobanian and Avedisian School of Medicine, Boston University, Boston, MA, 02218, USA; National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, 02218, USA
| | - Anne Balkema-Buschmann
- Institute of Novel and Emerging Infectious Diseases, Friedrich-Loeffler-Institut, Südufer 10, 17493, Greifswald, Insel Riems, Germany
| | - Matthias Dobbelstein
- Department of Molecular Oncology, Göttingen Center of Molecular Biosciences (GZMB), University Medical Center Göttingen, Justus-von-Liebig-Weg 11, 37077, Göttingen, Germany; Max Planck Institute for Multidisciplinary Sciences, Am Fassberg 11, 37077, Göttingen, Germany.
| |
Collapse
|
27
|
Angius F, Puxeddu S, Zaimi S, Canton S, Nematollahzadeh S, Pibiri A, Delogu I, Alvisi G, Moi ML, Manzin A. SARS-CoV-2 Evolution: Implications for Diagnosis, Treatment, Vaccine Effectiveness and Development. Vaccines (Basel) 2024; 13:17. [PMID: 39852796 PMCID: PMC11769326 DOI: 10.3390/vaccines13010017] [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: 11/13/2024] [Revised: 12/13/2024] [Accepted: 12/23/2024] [Indexed: 01/26/2025] Open
Abstract
The COVID-19 pandemic, driven by the rapid evolution of the SARS-CoV-2 virus, presents ongoing challenges to global public health. SARS-CoV-2 is characterized by rapidly evolving mutations, especially in (but not limited to) the spike protein, complicating predictions about its evolutionary trajectory. These mutations have significantly affected transmissibility, immune evasion, and vaccine efficacy, leading to multiple pandemic waves with over half a billion cases and seven million deaths globally. Despite several strategies, from rapid vaccine development and administration to the design and availability of antivirals, including monoclonal antibodies, already having been employed, the persistent circulation of the virus and the emergence of new variants continue to result in high case numbers and fatalities. In the past four years, immense research efforts have contributed much to our understanding of the viral pathogenesis mechanism, the COVID-19 syndrome, and the host-microbe interactions, leading to the development of effective vaccines, diagnostic tools, and treatments. The focus of this review is to provide a comprehensive analysis of the functional impact of mutations on diagnosis, treatments, and vaccine effectiveness. We further discuss vaccine safety in pregnancy and the implications of hybrid immunity on long-term protection against infection, as well as the latest developments on a pan-coronavirus vaccine and nasal formulations, emphasizing the need for continued surveillance, research, and adaptive public health strategies in response to the ongoing SARS-CoV-2 evolution race.
Collapse
Affiliation(s)
- Fabrizio Angius
- Microbiology and Virology Unit, Department of Biomedical Sciences, University of Cagliari, University Campus, 09042 Monserrato, Italy (A.P.); (I.D.); (A.M.)
| | - Silvia Puxeddu
- Microbiology and Virology Unit, Department of Biomedical Sciences, University of Cagliari, University Campus, 09042 Monserrato, Italy (A.P.); (I.D.); (A.M.)
| | - Silvio Zaimi
- Microbiology and Virology Unit, Department of Biomedical Sciences, University of Cagliari, University Campus, 09042 Monserrato, Italy (A.P.); (I.D.); (A.M.)
| | - Serena Canton
- Microbiology and Virology Unit, Department of Biomedical Sciences, University of Cagliari, University Campus, 09042 Monserrato, Italy (A.P.); (I.D.); (A.M.)
| | - Sepehr Nematollahzadeh
- Department of Molecular Medicine, University of Padova, 35121 Padova, Italy; (S.N.); (G.A.)
| | - Andrea Pibiri
- Microbiology and Virology Unit, Department of Biomedical Sciences, University of Cagliari, University Campus, 09042 Monserrato, Italy (A.P.); (I.D.); (A.M.)
| | - Ilenia Delogu
- Microbiology and Virology Unit, Department of Biomedical Sciences, University of Cagliari, University Campus, 09042 Monserrato, Italy (A.P.); (I.D.); (A.M.)
| | - Gualtiero Alvisi
- Department of Molecular Medicine, University of Padova, 35121 Padova, Italy; (S.N.); (G.A.)
| | - Meng Ling Moi
- School of International Health, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan
| | - Aldo Manzin
- Microbiology and Virology Unit, Department of Biomedical Sciences, University of Cagliari, University Campus, 09042 Monserrato, Italy (A.P.); (I.D.); (A.M.)
| |
Collapse
|
28
|
Zhou S, Long N, Rosenke K, Jarvis MA, Feldmann H, Swanstrom R. Combined Treatment of Severe Acute Respiratory Syndrome Coronavirus 2 Reduces Molnupiravir-Induced Mutagenicity and Prevents Selection for Nirmatrelvir/Ritonavir Resistance Mutations. J Infect Dis 2024; 230:1380-1383. [PMID: 38973065 PMCID: PMC11646606 DOI: 10.1093/infdis/jiae213] [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: 01/05/2024] [Accepted: 03/22/2024] [Indexed: 07/09/2024] Open
Abstract
We investigated the mutation profiles of severe acute respiratory syndrome coronavirus 2 in samples collected from a molnupiravir and nirmatrelvir/ritonavir combination therapy in macaques. We found that molnupiravir induced several nirmatrelvir resistance mutations at low abundance that were not further selected in combination therapy. Coadministration of nirmatrelvir/ritonavir lowered the magnitude of the mutagenetic effect of molnupiravir.
Collapse
Affiliation(s)
- Shuntai Zhou
- Lineberger Comprehensive Cancer Center
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill
| | | | - Kyle Rosenke
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana
| | - Michael A Jarvis
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana
- School of Biomedical Sciences, University of Plymouth
- The Vaccine Group Ltd, Plymouth, Devon, United Kingdom
| | - Heinz Feldmann
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana
| | - Ronald Swanstrom
- Lineberger Comprehensive Cancer Center
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill
| |
Collapse
|
29
|
Boulon R, Mazeaud C, Farahani MD, Broquière M, Iddir M, Charpentier T, Anton A, Ayotte Y, Woo S, Lamarre A, Chatel-Chaix L, LaPlante SR. Repurposing Drugs and Synergistic Combinations as Potential Therapies for Inhibiting SARS-CoV-2 and Coronavirus Replication. ACS Pharmacol Transl Sci 2024; 7:4043-4055. [PMID: 39698276 PMCID: PMC11650740 DOI: 10.1021/acsptsci.4c00512] [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: 08/23/2024] [Revised: 10/21/2024] [Accepted: 10/25/2024] [Indexed: 12/20/2024]
Abstract
Drug repurposing can serve an important role in rapidly discovering medicament options for emerging microbial pandemics. In this study, a pragmatic approach is demonstrated for screening and testing drug combinations as potential broad-spectrum therapies against SARS-CoV-2 and other betacoronaviruses. Rapid cell-based phenotypic small molecule screens were executed using related common-cold-causing HCoV-OC43 betacoronavirus to identify replication inhibitors from a library of drugs approved by regulatory agencies for other indications. Given the best inhibitors, an expedient checkerboard strategy then served to identify synergistic drug combinations. These combinations were then validated using more challenging assays involving SARS-CoV-2 and variants. Promising drug combinations against multiple viral variants were discovered and involved Tilorone with Nelfinavir or Molnupiravir.
Collapse
Affiliation(s)
- Richard Boulon
- Institut
National de la Recherche Scientifique−Centre Armand-Frappier
Santé Biotechnologie, 531 boulevard des Prairies, Laval, Québec H7V
1B7, Canada
| | - Clément Mazeaud
- Institut
National de la Recherche Scientifique−Centre Armand-Frappier
Santé Biotechnologie, 531 boulevard des Prairies, Laval, Québec H7V
1B7, Canada
| | - Majid D. Farahani
- Institut
National de la Recherche Scientifique−Centre Armand-Frappier
Santé Biotechnologie, 531 boulevard des Prairies, Laval, Québec H7V
1B7, Canada
| | - Mathilde Broquière
- Institut
National de la Recherche Scientifique−Centre Armand-Frappier
Santé Biotechnologie, 531 boulevard des Prairies, Laval, Québec H7V
1B7, Canada
| | - Mustapha Iddir
- Institut
National de la Recherche Scientifique−Centre Armand-Frappier
Santé Biotechnologie, 531 boulevard des Prairies, Laval, Québec H7V
1B7, Canada
| | - Tania Charpentier
- Institut
National de la Recherche Scientifique−Centre Armand-Frappier
Santé Biotechnologie, 531 boulevard des Prairies, Laval, Québec H7V
1B7, Canada
| | - Anaïs Anton
- Institut
National de la Recherche Scientifique−Centre Armand-Frappier
Santé Biotechnologie, 531 boulevard des Prairies, Laval, Québec H7V
1B7, Canada
| | - Yann Ayotte
- NMX
Research and Solutions|Accelerating drug discovery, 500 boulevard Cartier Ouest, Laval, Quebec H7V 5B7, Canada
| | - Simon Woo
- Institut
National de la Recherche Scientifique−Centre Armand-Frappier
Santé Biotechnologie, 531 boulevard des Prairies, Laval, Québec H7V
1B7, Canada
- NMX
Research and Solutions|Accelerating drug discovery, 500 boulevard Cartier Ouest, Laval, Quebec H7V 5B7, Canada
| | - Alain Lamarre
- Institut
National de la Recherche Scientifique−Centre Armand-Frappier
Santé Biotechnologie, 531 boulevard des Prairies, Laval, Québec H7V
1B7, Canada
| | - Laurent Chatel-Chaix
- Institut
National de la Recherche Scientifique−Centre Armand-Frappier
Santé Biotechnologie, 531 boulevard des Prairies, Laval, Québec H7V
1B7, Canada
| | - Steven R. LaPlante
- Institut
National de la Recherche Scientifique−Centre Armand-Frappier
Santé Biotechnologie, 531 boulevard des Prairies, Laval, Québec H7V
1B7, Canada
| |
Collapse
|
30
|
Sousa BP, Mottin M, Seanego D, Jurisch CD, Rodrigues BSA, da Silva VLS, Andrade MA, Morais GS, Boerin DF, Froes TQ, Motta FN, Nonato MC, Bastos IDM, Chibale K, Gessner RK, Andrade CH. Discovery of Non-Covalent Inhibitors for SARS-CoV-2 PLpro: Integrating Virtual Screening, Synthesis, and Experimental Validation. ACS Med Chem Lett 2024; 15:2140-2149. [PMID: 39691531 PMCID: PMC11647681 DOI: 10.1021/acsmedchemlett.4c00420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 10/31/2024] [Accepted: 11/26/2024] [Indexed: 12/19/2024] Open
Abstract
The SARS-CoV-2 pandemic has significantly challenged global public health, highlighting the need for effective therapeutic options. This study focuses on the papain-like protease (PLpro) of SARS-CoV-2, which is a critical enzyme for viral polyprotein processing, maturation, and immune evasion. We employed a combined approach that began with computational models in a virtual screening campaign, prioritizing compounds from our in-house chemical library against PLpro. Out of 81 virtual hits evaluated through enzymatic and biophysical assays, we identified a modest inhibitor featuring a naphthyridine core with an IC50 of 73.61 μM and a K i of 22 μM. Expanding our exploration, we synthesized and assessed 30 naphthyridine analogues, three of which emerged as promising noncovalent, nonpeptidomimetic inhibitors with IC50 values between 15.06 and 51.81 μM. Furthermore, in vitro ADMET assays revealed these compounds to possess moderate aqueous solubility, low cytotoxicity, and high microsomal stability, making them excellent candidates for further development targeting SARS-CoV-2 PLpro.
Collapse
Affiliation(s)
- Bruna
K. P. Sousa
- Center
for the Research and Advancement in Fragments and Molecular Targets
(CRAFT), Faculdade de Ciências Farmaceuticas de Ribeirão
Preto, Universidade de São Paulo, Ribeirão Preto, São
Paulo 05508-070, Brazil
- Laboratory
for Molecular Modeling and Drug Design (LabMol), Faculdade de Farmácia, Universidade Federal de Goiás, Goiânia, Goiás 74690-900, Brazil
| | - Melina Mottin
- Center
for the Research and Advancement in Fragments and Molecular Targets
(CRAFT), Faculdade de Ciências Farmaceuticas de Ribeirão
Preto, Universidade de São Paulo, Ribeirão Preto, São
Paulo 05508-070, Brazil
- Laboratory
for Molecular Modeling and Drug Design (LabMol), Faculdade de Farmácia, Universidade Federal de Goiás, Goiânia, Goiás 74690-900, Brazil
- Pathogen-Host
Interface Laboratory, Department of Cell Biology, University of Brasilia, Brasilia 73345-010, Brazil
| | - Donald Seanego
- Holistic
Drug Discovery and Development Centre (H3D), University of Cape Town, Cape Town 7701, South Africa
| | - Christopher D. Jurisch
- Holistic
Drug Discovery and Development Centre (H3D), University of Cape Town, Cape Town 7701, South Africa
| | - Beatriz S. A. Rodrigues
- Pathogen-Host
Interface Laboratory, Department of Cell Biology, University of Brasilia, Brasilia 73345-010, Brazil
| | - Verônica L. S. da Silva
- Pathogen-Host
Interface Laboratory, Department of Cell Biology, University of Brasilia, Brasilia 73345-010, Brazil
| | - Milene Aparecida Andrade
- Pathogen-Host
Interface Laboratory, Department of Cell Biology, University of Brasilia, Brasilia 73345-010, Brazil
| | - Gilberto S. Morais
- Pathogen-Host
Interface Laboratory, Department of Cell Biology, University of Brasilia, Brasilia 73345-010, Brazil
| | - Diogo F. Boerin
- Center
for the Research and Advancement in Fragments and Molecular Targets
(CRAFT), Faculdade de Ciências Farmaceuticas de Ribeirão
Preto, Universidade de São Paulo, Ribeirão Preto, São
Paulo 05508-070, Brazil
- Laboratório
de Cristalografia de Proteínas, Faculdade de Ciências
Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo 05508-070, Brazil
| | - Thamires Q. Froes
- Center
for the Research and Advancement in Fragments and Molecular Targets
(CRAFT), Faculdade de Ciências Farmaceuticas de Ribeirão
Preto, Universidade de São Paulo, Ribeirão Preto, São
Paulo 05508-070, Brazil
- Laboratório
de Cristalografia de Proteínas, Faculdade de Ciências
Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo 05508-070, Brazil
| | - Flávia Nader Motta
- Pathogen-Host
Interface Laboratory, Department of Cell Biology, University of Brasilia, Brasilia 73345-010, Brazil
- Faculdade
de Ceilândia, Universidade de Brasília, Brasília, Distrito
Federal 73345-010, Brazil
| | - M. Cristina Nonato
- Center
for the Research and Advancement in Fragments and Molecular Targets
(CRAFT), Faculdade de Ciências Farmaceuticas de Ribeirão
Preto, Universidade de São Paulo, Ribeirão Preto, São
Paulo 05508-070, Brazil
- Laboratório
de Cristalografia de Proteínas, Faculdade de Ciências
Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo 05508-070, Brazil
| | - Izabela D. M. Bastos
- Pathogen-Host
Interface Laboratory, Department of Cell Biology, University of Brasilia, Brasilia 73345-010, Brazil
| | - Kelly Chibale
- Holistic
Drug Discovery and Development Centre (H3D), University of Cape Town, Cape Town 7701, South Africa
- South
African Medical Research Council Drug Discovery and Development Research
Unit, University of Cape Town, Cape Town 7701, South Africa
- Institute
of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town 7701, South Africa
| | - Richard K. Gessner
- Holistic
Drug Discovery and Development Centre (H3D), University of Cape Town, Cape Town 7701, South Africa
| | - Carolina Horta Andrade
- Center
for the Research and Advancement in Fragments and Molecular Targets
(CRAFT), Faculdade de Ciências Farmaceuticas de Ribeirão
Preto, Universidade de São Paulo, Ribeirão Preto, São
Paulo 05508-070, Brazil
- Laboratory
for Molecular Modeling and Drug Design (LabMol), Faculdade de Farmácia, Universidade Federal de Goiás, Goiânia, Goiás 74690-900, Brazil
- Center
for Excellence in Artificial Intelligence (CEIA), Instituto de Informática, Universidade Federal de Goiás, Goiânia, Goiás 74690-900, Brazil
| |
Collapse
|
31
|
Duncan KE, Carstens RP, Butterfield KL, Jin Y, Inbody LR, Schaeffer AK, Matthews CZ, Zhao T, Patel S, Maas BM, Cheng MH, Stoch SA. Assessment of pharmacokinetics and tolerability following single-dose administration of molnupiravir in participants with hepatic or renal impairment. Clin Transl Sci 2024; 17:e70073. [PMID: 39601078 PMCID: PMC11599873 DOI: 10.1111/cts.70073] [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: 07/11/2024] [Revised: 10/22/2024] [Accepted: 10/31/2024] [Indexed: 11/29/2024] Open
Abstract
Individuals with chronic liver or kidney disease are at increased risk of severe COVID-19. Molnupiravir is an orally administered antiviral authorized for the treatment of mild-to-moderate COVID-19 in adults at risk of progression to severe disease. Two nonrandomized, open-label, single-dose, multicenter, phase 1 trials were conducted to investigate the effects of hepatic and renal impairment on the tolerability and pharmacokinetics of molnupiravir (800 mg) and its metabolite β-D-N4-hydroxycytidine (NHC; NCT05386589/NCT05386758). The impact of renal impairment on urinary excretion of NHC was also assessed. The 90% CI for the geometric mean ratio of the plasma NHC area under the concentration-time curve (AUC) from zero to infinity was <2.0 for participants with moderate hepatic or severe renal impairment versus healthy mean-matched controls. Comparable geometric mean values were observed for other pharmacokinetic parameters-including AUC from 0 to 12 h, AUC from zero to the last measurable concentration, and peak plasma concentration-in participants with moderate hepatic or severe renal impairment and in healthy mean-matched controls. Urinary excretion of NHC was low in healthy participants and participants with severe renal impairment; renal clearance was numerically lower in those with renal impairment. In both trials, all adverse events were of mild or moderate intensity and resolved by study completion. There were no clinically relevant treatment-related effects on other safety evaluations. Overall, molnupiravir was generally well-tolerated, with similar pharmacokinetic profiles in participants with hepatic or renal impairment and healthy participants, supporting its use for treating COVID-19 in these individuals without the need for dose adjustment.
Collapse
Affiliation(s)
| | | | | | - Yoon Jin
- Merck & Co., Inc.RahwayNew JerseyUSA
| | | | | | | | - Tian Zhao
- Merck & Co., Inc.RahwayNew JerseyUSA
| | | | | | | | | |
Collapse
|
32
|
Sasaki J, Sato A, Sasaki M, Okabe I, Kodama K, Otsuguro S, Yasuda K, Kojima H, Orba Y, Sawa H, Maenaka K, Yanagi Y, Hashiguchi T. X-206 exhibits broad-spectrum anti-β-coronavirus activity, covering SARS-CoV-2 variants and drug-resistant isolates. Antiviral Res 2024; 232:106039. [PMID: 39571911 DOI: 10.1016/j.antiviral.2024.106039] [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: 08/16/2024] [Revised: 11/12/2024] [Accepted: 11/18/2024] [Indexed: 11/26/2024]
Abstract
Coronaviruses such as the Middle East respiratory syndrome coronavirus (MERS-CoV), severe acute respiratory syndrome coronavirus (SARS-CoV), and SARS-CoV-2, causing MERS, SARS, and Coronavirus disease-19, respectively, are highly pathogenic to humans. Notably, several antiviral drugs against SARS-CoV-2, such as nirmatrelvir and remdesivir, have been approved. However, no approved vaccines or antiviral agents are available for other highly pathogenic β-coronaviruses. In this study, we identified two compounds, thapsigargin and X-206, that exhibit antiviral activities against SARS-CoV, MERS-CoV, and SARS-CoV-2. Notably, both compounds effectively inhibited the cell-to-cell fusion mediated by the Spike proteins of all three β-coronaviruses. X-206 exhibited antiviral activity against nirmatrelvir- and remdesivir-resistant SARS-CoV-2 isolates and SARS-CoV-2 variants, including Delta, BA.5, and XBB.1. Consequently, the mechanism of action of these compounds with anti-β-coronavirus activities may differ from that of the approved direct-acting drugs for SARS-CoV-2, thereby offering potential use as a cocktail with other antivirals, and serving as a chemical basis for developing therapeutic agents against β-coronaviruses in preparation for the next spillover and pandemic.
Collapse
Affiliation(s)
- Jiei Sasaki
- Laboratory of Medical Virology, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Akihiko Sato
- Division of Molecular Pathobiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan; Shionogi Pharmaceutical Research Center, Shionogi & Company, Limited, Toyonaka, Japan; Institute for Vaccine Research and Development, Hokkaido University, Sapporo, Japan
| | - Michihito Sasaki
- Division of Molecular Pathobiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan; Institute for Vaccine Research and Development, Hokkaido University, Sapporo, Japan
| | - Iori Okabe
- Department of Virology, Faculty of Medicine, Kyushu University, Fukuoka, Japan
| | - Kota Kodama
- Medical Data Science Lab., Hoshi University, Tokyo, Japan; Laboratory of Biomolecular Science, and Center for Research and Education on Drug Discovery, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Satoko Otsuguro
- Laboratory of Biomolecular Science, and Center for Research and Education on Drug Discovery, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Kosuke Yasuda
- Drug Discovery Initiative, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Hirotatsu Kojima
- Drug Discovery Initiative, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Yasuko Orba
- Division of Molecular Pathobiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan; Institute for Vaccine Research and Development, Hokkaido University, Sapporo, Japan
| | - Hirofumi Sawa
- Institute for Vaccine Research and Development, Hokkaido University, Sapporo, Japan
| | - Katsumi Maenaka
- Laboratory of Biomolecular Science, and Center for Research and Education on Drug Discovery, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan; Division of Pathogen Structure, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan; Global Station for Biosurfaces and Drug Discovery, Hokkaido University, Sapporo, Japan
| | - Yusuke Yanagi
- Department of Virology, Faculty of Medicine, Kyushu University, Fukuoka, Japan
| | - Takao Hashiguchi
- Laboratory of Medical Virology, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan; Kyoto University Immunomonitoring Center, Kyoto University, Kyoto, Japan.
| |
Collapse
|
33
|
Escobar PA, Sobol Z, Miller RR, Ferry-Martin S, Stermer A, Jacob B, Muniappa N, Sanchez RI, Blanchard KT, Galijatovic-Idrizbegovic A, Amin RP, Troth SP. Comprehensive genotoxicity and carcinogenicity assessment of molnupiravir. Toxicol Sci 2024; 202:278-290. [PMID: 39302733 PMCID: PMC11589102 DOI: 10.1093/toxsci/kfae112] [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] [Indexed: 09/22/2024] Open
Abstract
Molnupiravir is registered or authorized in several countries as a 5-d oral coronavirus disease 2019 treatment for adults. Molnupiravir is a prodrug of the antiviral ribonucleoside β-D-N4-hydroxycytidine (NHC) that distributes into cells, where it is phosphorylated to its pharmacologically active ribonucleoside triphosphate (NHC-TP) form. NHC-TP incorporates into severe acute respiratory syndrome coronavirus 2 RNA by the viral RNA-dependent RNA polymerase, resulting in an accumulation of errors in the viral genome, leading to inhibition of viral replication and loss of infectivity. The potential of molnupiravir to induce genomic mutations and DNA damage was comprehensively assessed in several in vitro and in vivo genotoxicity assays and a carcinogenicity study, in accordance with international guideline recommendations and expert opinion. Molnupiravir and NHC induced mutations in vitro in bacteria and mammalian cells but did not induce chromosome damage in in vitro or in vivo assays. The in vivo mutagenic and carcinogenic potential of molnupiravir was tested in a series of in vivo mutagenicity studies in somatic and germ cells (Pig-a Assay and Big Blue® TGR Mutation Assay) and in a carcinogenicity study (transgenic rasH2-Tg mouse), using durations of exposure and doses exceeding those used in clinical therapy. In vitro genotoxicity results are superseded by robustly conducted in vivo studies. Molnupiravir did not increase mutations in somatic or germ cells in the in vivo animal studies and was negative in the carcinogenicity study. The interpretation criteria for each study followed established regulatory guidelines. Taken together, these data indicate that molnupiravir use does not present a genotoxicity or carcinogenicity risk for patients.
Collapse
Affiliation(s)
- Patricia A Escobar
- Nonclinical Drug Safety and Pharmacokinetics Dynamics Metabolism and Bioanalysis, Preclinical Development, Merck & Co. Inc., Rahway, NJ 07065, United States
| | - Zhanna Sobol
- Nonclinical Drug Safety and Pharmacokinetics Dynamics Metabolism and Bioanalysis, Preclinical Development, Merck & Co. Inc., Rahway, NJ 07065, United States
| | - Randy R Miller
- Nonclinical Drug Safety and Pharmacokinetics Dynamics Metabolism and Bioanalysis, Preclinical Development, Merck & Co. Inc., Rahway, NJ 07065, United States
| | - Sandrine Ferry-Martin
- Nonclinical Drug Safety and Pharmacokinetics Dynamics Metabolism and Bioanalysis, Preclinical Development, Merck & Co. Inc., Rahway, NJ 07065, United States
| | - Angela Stermer
- Nonclinical Drug Safety and Pharmacokinetics Dynamics Metabolism and Bioanalysis, Preclinical Development, Merck & Co. Inc., Rahway, NJ 07065, United States
| | - Binod Jacob
- Nonclinical Drug Safety and Pharmacokinetics Dynamics Metabolism and Bioanalysis, Preclinical Development, Merck & Co. Inc., Rahway, NJ 07065, United States
| | - Nagaraja Muniappa
- Nonclinical Drug Safety and Pharmacokinetics Dynamics Metabolism and Bioanalysis, Preclinical Development, Merck & Co. Inc., Rahway, NJ 07065, United States
| | - Rosa I Sanchez
- Nonclinical Drug Safety and Pharmacokinetics Dynamics Metabolism and Bioanalysis, Preclinical Development, Merck & Co. Inc., Rahway, NJ 07065, United States
| | - Kerry T Blanchard
- Nonclinical Drug Safety and Pharmacokinetics Dynamics Metabolism and Bioanalysis, Preclinical Development, Merck & Co. Inc., Rahway, NJ 07065, United States
| | - Alema Galijatovic-Idrizbegovic
- Nonclinical Drug Safety and Pharmacokinetics Dynamics Metabolism and Bioanalysis, Preclinical Development, Merck & Co. Inc., Rahway, NJ 07065, United States
| | - Rupesh P Amin
- Nonclinical Drug Safety and Pharmacokinetics Dynamics Metabolism and Bioanalysis, Preclinical Development, Merck & Co. Inc., Rahway, NJ 07065, United States
| | - Sean P Troth
- Nonclinical Drug Safety and Pharmacokinetics Dynamics Metabolism and Bioanalysis, Preclinical Development, Merck & Co. Inc., Rahway, NJ 07065, United States
| |
Collapse
|
34
|
Barghash RF, Gemmati D, Awad AM, Elbakry MMM, Tisato V, Awad K, Singh AV. Navigating the COVID-19 Therapeutic Landscape: Unveiling Novel Perspectives on FDA-Approved Medications, Vaccination Targets, and Emerging Novel Strategies. Molecules 2024; 29:5564. [PMID: 39683724 PMCID: PMC11643501 DOI: 10.3390/molecules29235564] [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: 09/26/2024] [Revised: 11/21/2024] [Accepted: 11/22/2024] [Indexed: 12/18/2024] Open
Abstract
Amidst the ongoing global challenge of the SARS-CoV-2 pandemic, the quest for effective antiviral medications remains paramount. This comprehensive review delves into the dynamic landscape of FDA-approved medications repurposed for COVID-19, categorized as antiviral and non-antiviral agents. Our focus extends beyond conventional narratives, encompassing vaccination targets, repurposing efficacy, clinical studies, innovative treatment modalities, and future outlooks. Unveiling the genomic intricacies of SARS-CoV-2 variants, including the WHO-designated Omicron variant, we explore diverse antiviral categories such as fusion inhibitors, protease inhibitors, transcription inhibitors, neuraminidase inhibitors, nucleoside reverse transcriptase, and non-antiviral interventions like importin α/β1-mediated nuclear import inhibitors, neutralizing antibodies, and convalescent plasma. Notably, Molnupiravir emerges as a pivotal player, now licensed in the UK. This review offers a fresh perspective on the historical evolution of COVID-19 therapeutics, from repurposing endeavors to the latest developments in oral anti-SARS-CoV-2 treatments, ushering in a new era of hope in the battle against the pandemic.
Collapse
Affiliation(s)
- Reham F. Barghash
- Institute of Chemical Industries Research, National Research Centre, Dokki, Cairo 12622, Egypt
- Faculty of Biotechnology, October University for Modern Sciences and Arts (MSA), Cairo 12451, Egypt
| | - Donato Gemmati
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Ahmed M. Awad
- Department of Chemistry, California State University Channel Islands, Camarillo, CA 93012, USA
| | - Mustafa M. M. Elbakry
- Faculty of Biotechnology, October University for Modern Sciences and Arts (MSA), Cairo 12451, Egypt
- Biochemistry Department, Faculty of Science, Ain Shams University, Cairo 11566, Egypt
| | - Veronica Tisato
- Centre Hemostasis & Thrombosis, University of Ferrara, 44121 Ferrara, Italy
| | - Kareem Awad
- Institute of Pharmaceutical and Drug Industries Research, National Research Center, Dokki, Cairo 12622, Egypt;
| | - Ajay Vikram Singh
- Department of Chemical and Product Safety, German Federal Institute for Risk Assessment (BfR), Max-Dohrn-Strasse 8-10, 10589 Berlin, Germany
| |
Collapse
|
35
|
Focosi D, McNally D, Maggi F. The Fitness of Molnupiravir-Signed SARS-CoV-2 Variants: Imputation Analysis Based on Prescription Counts and Global Initiative on Sharing All Influenza Data Analyses by Country. Intervirology 2024; 67:114-118. [PMID: 39522507 PMCID: PMC11623959 DOI: 10.1159/000540282] [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: 10/07/2023] [Accepted: 07/04/2024] [Indexed: 11/16/2024] Open
Abstract
INTRODUCTION Molnupiravir is one of the oral direct-acting antivirals against SARS-CoV-2, largely deployed during the COVID-19 pandemic since the 2022 Omicron wave. While efficacy has been questioned in post-marketing clinical trials (leading to the EMA withdrawing its authorization), growing concerns have mounted regarding its possible mutagenic effects on the virus. While it has been assumed that either all the host viral load was cleared by the drug or drug-generated variants were not fit enough to survive, several lineages with a high transition/transversion ratio (a signature of molnupiravir action) have been recently reported from GISAID. METHODS We report here a systematic analysis of the GISAID database for sequences showing a molnupiravir signature, exposing a public web-based interface (https://ukcovid.xyz/molnupiravir/), and performing an imputation analysis based on per-country prescription (corrected by sequencing). RESULTS Our analysis confirms a direct correlation between the number of molnupiravir courses and the number of mutationally signed sequences deposited in GISAID in individual countries. CONCLUSIONS Molnupiravir can generate fit SARS-CoV-2 variants that transmit in the general population.
Collapse
Affiliation(s)
- Daniele Focosi
- North-Western Tuscany Blood Bank, Pisa University Hospital, Pisa, Italy
| | | | - Fabrizio Maggi
- Laboratory of Virology, National Institute for Infectious Diseases, Lazzaro Spallanzani IRCCS, Rome, Italy
| |
Collapse
|
36
|
Mao YQ, Jahanshahi S, Malty R, Van Ommen DAJ, Wan Y, Morey TM, Chuang SHW, Pavlova V, Ahmed C, Dahal S, Lin F, Mangos M, Nurtanto J, Song Y, Been T, Christie-Holmes N, Gray-Owen SD, Babu M, Wong AP, Batey RA, Attisano L, Cochrane A, Houry WA. Targeting protein homeostasis with small molecules as a strategy for the development of pan-coronavirus antiviral therapies. Commun Biol 2024; 7:1460. [PMID: 39511285 PMCID: PMC11543989 DOI: 10.1038/s42003-024-07143-z] [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: 07/17/2023] [Accepted: 10/24/2024] [Indexed: 11/15/2024] Open
Abstract
The COVID-19 pandemic has created a global health crisis, with challenges arising from the ongoing evolution of the SARS-CoV-2 virus, the emergence of new strains, and the long-term effects of COVID-19. Aiming to overcome the development of viral resistance, our study here focused on developing broad-spectrum pan-coronavirus antiviral therapies by targeting host protein quality control mechanisms essential for viral replication. Screening an in-house compound library led to the discovery of three candidate compounds targeting cellular proteostasis. The three compounds are (1) the nucleotide analog cordycepin, (2) a benzothiozole analog, and (3) an acyldepsipeptide analog initially developed as part of a campaign to target the mitochondrial ClpP protease. These compounds demonstrated dose-dependent efficacy against multiple coronaviruses, including SARS-CoV-2, effectively inhibiting viral replication in vitro as well as in lung organoids. Notably, the compounds also showed efficacy against SARS-CoV-2 delta and omicron strains. As part of this work, we developed a BSL2-level cell-integrated SARS-CoV-2 replicon, which could serve as a valuable tool for high-throughput screening and studying intracellular viral replication. Our study should aid in the advancement of antiviral drug development efforts.
Collapse
Affiliation(s)
- Yu-Qian Mao
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| | - Shahrzad Jahanshahi
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Ramy Malty
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
- Department of Chemistry and Biochemistry, University of Regina, Regina, SK, Canada
| | | | - Yimei Wan
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| | - Trevor M Morey
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| | | | - Veronika Pavlova
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| | - Choudhary Ahmed
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Subha Dahal
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Funing Lin
- Department of Chemistry, University of Toronto, Toronto, ON, Canada
| | - Maria Mangos
- Donnelly Centre, University of Toronto, Toronto, ON, Canada
| | | | - Yuetong Song
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children, Toronto, ON, Canada
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Terek Been
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Natasha Christie-Holmes
- Toronto High Containment Facility, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Scott D Gray-Owen
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- Toronto High Containment Facility, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Mohan Babu
- Department of Chemistry and Biochemistry, University of Regina, Regina, SK, Canada
| | - Amy P Wong
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children, Toronto, ON, Canada
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Robert A Batey
- Department of Chemistry, University of Toronto, Toronto, ON, Canada
| | - Liliana Attisano
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
- Donnelly Centre, University of Toronto, Toronto, ON, Canada
| | - Alan Cochrane
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Walid A Houry
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada.
- Department of Chemistry, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
37
|
Do TND, Abdelnabi R, Boda B, Constant S, Neyts J, Jochmans D. The triple combination of Remdesivir (GS-441524), Molnupiravir and Ribavirin is highly efficient in inhibiting coronavirus replication in human nasal airway epithelial cell cultures and in a hamster infection model. Antiviral Res 2024; 231:105994. [PMID: 39237005 PMCID: PMC11560660 DOI: 10.1016/j.antiviral.2024.105994] [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: 06/12/2024] [Revised: 08/20/2024] [Accepted: 09/01/2024] [Indexed: 09/07/2024]
Abstract
The use of fixed dose-combinations of antivirals with different mechanisms of action has proven key in the successful treatment of infections with HIV and HCV. For the treatment of infections with SARS-CoV-2 and possible future epi-/pandemic coronaviruses, it will be important to explore the efficacy of combinations of different drugs, in particular to avoid resistance development, such as in patients with immunodeficiencies. This work explores the effect of a combination of 3 broad-spectrum antiviral nucleosides on the replication of coronaviruses. To that end, we made use of primary human airway epithelial cell (HAEC) cultures grown at the air-liquid interface that were infected with the beta coronavirus OC43. We found that the triple combination of GS-441524 (the parent nucleoside of remdesivir), molnupiravir and ribavirin resulted in a more pronounced antiviral efficacy than what could be expected from a purely additive antiviral effect. The potency of this triple combination was next tested in SARS-CoV-2 infected hamsters in a prophylactic setup. To that end, for each of the drugs, intentionally suboptimal or even ineffective doses were selected. Yet, in the lungs of all hamsters that received triple prophylactic therapy (but not in those that received the respective double combinations) no infectious virus was detectable. Our findings indicate that co-administration of approved drugs for the treatment of coronavirus infections should be further explored but also against other families of viruses with epidemic and pandemic potential for which no effective antiviral treatment is available.
Collapse
Affiliation(s)
- Thuc Nguyen Dan Do
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute, Laboratory of Virology and Chemotherapy, 3000, Leuven, Belgium
| | - Rana Abdelnabi
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute, Laboratory of Virology and Chemotherapy, 3000, Leuven, Belgium; The VirusBank Platform, Gaston Geenslaan, B-3000, Leuven, Belgium
| | - Bernadett Boda
- Epithelix Sàrl, 18 Chemin des Aulx, Plan-les-Ouates, CH-1228, Geneva, Switzerland
| | - Samuel Constant
- Epithelix Sàrl, 18 Chemin des Aulx, Plan-les-Ouates, CH-1228, Geneva, Switzerland
| | - Johan Neyts
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute, Laboratory of Virology and Chemotherapy, 3000, Leuven, Belgium; The VirusBank Platform, Gaston Geenslaan, B-3000, Leuven, Belgium.
| | - Dirk Jochmans
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute, Laboratory of Virology and Chemotherapy, 3000, Leuven, Belgium.
| |
Collapse
|
38
|
Kumar P, Zhang X, Shaha R, Kschischo M, Dobbelstein M. Identification of antibody-resistant SARS-CoV-2 mutants via N4-Hydroxycytidine mutagenesis. Antiviral Res 2024; 231:106006. [PMID: 39293594 DOI: 10.1016/j.antiviral.2024.106006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 08/31/2024] [Accepted: 09/12/2024] [Indexed: 09/20/2024]
Abstract
Monoclonal antibodies targeting the Spike protein of SARS-CoV-2 are effective against COVID-19 and might mitigate future pandemics. However, their efficacy is challenged by the emergence of antibody-resistant virus variants. We developed a method to efficiently identify such resistant mutants based on selection from mutagenized virus pools. By inducing mutations with the active compound of Molnupiravir, N4-hydroxycytidine (NHC), and subsequently passaging the virus in the presence of antibodies, we identified specific Spike mutations linked to resistance. Validation of these mutations was conducted using pseudotypes and immunofluorescence analysis. From a Wuhan-like strain of SARS-CoV-2, we identified the following mutations conferring strong resistance towards the corresponding antibodies: Bamlanivimab - E484K, F490S and S494P; Sotrovimab - E340K; Cilgavimab - K444R/E and N450D. From the Omicron B.1.1.529 variant, the strongly selected mutations were: Bebtelovimab - V445A; Sotrovimab - E340K and K356M; Cilgavimab - K444R, V445A and N450D. We also identified escape mutations in the Wuhan-like Spike for the broadly neutralizing antibodies S2K146 - combined G485S and Q493R - and S2H97 - D428G, K462E and S514F. Structural analysis revealed that the selected mutations occurred at antibody-binding residues within the receptor-binding domains of the Spike protein. Most of the selected mutants largely maintained ACE2 binding and infectivity. Notably, many of the identified resistance-conferring mutations are prevalent in real-world SARS-CoV-2 variants, but some of them (G485S, D428G, and K462E) have not yet been observed in circulating strains. Our approach offers a strategy for predicting the therapeutic efficacy of antibodies against emerging virus variants.
Collapse
MESH Headings
- SARS-CoV-2/genetics
- SARS-CoV-2/immunology
- SARS-CoV-2/drug effects
- Cytidine/analogs & derivatives
- Cytidine/pharmacology
- Cytidine/genetics
- Humans
- Spike Glycoprotein, Coronavirus/genetics
- Spike Glycoprotein, Coronavirus/immunology
- Drug Resistance, Viral/genetics
- Mutation
- Antibodies, Neutralizing/immunology
- Antibodies, Viral/immunology
- Mutagenesis
- COVID-19/virology
- COVID-19/immunology
- Antiviral Agents/pharmacology
- COVID-19 Drug Treatment
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal, Humanized/immunology
- Antibodies, Monoclonal, Humanized/pharmacology
- Hydroxylamines
Collapse
Affiliation(s)
- Priya Kumar
- Department of Molecular Oncology, Göttingen Center of Molecular Biosciences (GZMB), University Medical Center Göttingen, 37077, Göttingen, Germany
| | - Xiaoxiao Zhang
- Department of Mathematics and Technology, University of Applied Sciences Koblenz, 53424, Remagen, Germany; Department of Informatics, Technical University of Munich, 81675, Munich, Germany
| | - Rahul Shaha
- Department of Molecular Enzymology, Göttingen Center of Molecular Biosciences (GZMB), University of Göttingen, 37077, Göttingen, Germany
| | - Maik Kschischo
- Department of Mathematics and Technology, University of Applied Sciences Koblenz, 53424, Remagen, Germany
| | - Matthias Dobbelstein
- Department of Molecular Oncology, Göttingen Center of Molecular Biosciences (GZMB), University Medical Center Göttingen, 37077, Göttingen, Germany; Max Planck Institute for Multidisciplinary Sciences, Am Fassberg 11, 37077 Göttingen, Germany.
| |
Collapse
|
39
|
Alejandro B, Kim EJ, Hwang JY, Park JW, Smith M, Chung D. Genetic and phenotypic changes to Venezuelan equine encephalitis virus following treatment with β-D-N4-hydroxycytidine, an RNA mutagen. Sci Rep 2024; 14:25265. [PMID: 39448734 PMCID: PMC11502654 DOI: 10.1038/s41598-024-76788-x] [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: 08/27/2024] [Accepted: 10/16/2024] [Indexed: 10/26/2024] Open
Abstract
The high mutation rate of RNA viruses provides viral populations with the ability to adapt to new environments but also makes them vulnerable to extinction due to the deleterious effects of mutations, which is the conceptual basis for the antiviral activity of RNA mutagens. However, there are still gaps in the quantitative understanding of the dynamics between the mutations induced by an RNA mutagen and its effects on viral fitness. To address this, we used Venezuelan Equine Encephalitis Virus (VEEV) and the potent RNA mutagen β-d-N4-hydroxycytidine (NHC) as a model to analyze virus replication competency and mutation frequency following treatment in the total and replication-competent viral populations separately. We found that NHC induced transition mutations in a concentration dependent manner in the total population, while the replication-competent population maintained itself within an increased, yet narrow, mutation spectrum. The incorporation of NHC mainly happened during the positive sense RNA synthesis of VEEV. A growth kinetic analysis of VEEV population treated with NHC pointed to a lower but more diverse distribution in mutational fitness, demonstrating that NHC-induced mutations negatively and broadly affect the fitness of the virus. Together, our study provides mechanistic insight into how RNA mutagens affect viral population landscape and the potential of RNA mutagens as an antiviral strategy for alphaviruses.
Collapse
Affiliation(s)
- Brian Alejandro
- Center for Predictive Medicine, University of Louisville, Louisville, KY, USA
| | - Eun Jung Kim
- Center for Predictive Medicine, University of Louisville, Louisville, KY, USA
| | - Jae Yeon Hwang
- Department of Medicine, University of Louisville, Louisville, KY, USA
- Brown Cancer Center Bioinformatics Core, University of Louisville, Louisville, KY, USA
| | - Juw Won Park
- Department of Medicine, University of Louisville, Louisville, KY, USA
- Brown Cancer Center Bioinformatics Core, University of Louisville, Louisville, KY, USA
| | - Melissa Smith
- Department of Biochemistry and Molecular Genetics, School of Medicine, University of Louisville, Louisville, KY, USA
| | - Donghoon Chung
- Center for Predictive Medicine, University of Louisville, Louisville, KY, USA.
- Department of Microbiology and Immunology, School of Medicine, University of Louisville, Louisville, KY, USA.
| |
Collapse
|
40
|
Katiyar H, Arduini A, Li Y, Liang C. SARS-CoV-2 Assembly: Gaining Infectivity and Beyond. Viruses 2024; 16:1648. [PMID: 39599763 PMCID: PMC11598957 DOI: 10.3390/v16111648] [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: 10/02/2024] [Revised: 10/12/2024] [Accepted: 10/21/2024] [Indexed: 11/29/2024] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) was responsible for causing the COVID-19 pandemic. Intensive research has illuminated the complex biology of SARS-CoV-2 and its continuous evolution during and after the COVID-19 pandemic. While much attention has been paid to the structure and functions of the viral spike protein and the entry step of viral infection, partly because these are targets for neutralizing antibodies and COVID-19 vaccines, the later stages of SARS-CoV-2 replication, including the assembly and egress of viral progenies, remain poorly characterized. This includes insight into how the activities of the viral structural proteins are orchestrated spatially and temporally, which cellular proteins are assimilated by the virus to assist viral assembly, and how SARS-CoV-2 counters and evades the cellular mechanisms antagonizing virus assembly. In addition to becoming infectious, SARS-CoV-2 progenies also need to survive the hostile innate and adaptive immune mechanisms, such as recognition by neutralizing antibodies. This review offers an updated summary of the roles of SARS-CoV-2 structural proteins in viral assembly, the regulation of assembly by viral and cellular factors, and the cellular mechanisms that restrict this process. Knowledge of these key events often reveals the vulnerabilities of SARS-CoV-2 and aids in the development of effective antiviral therapeutics.
Collapse
Affiliation(s)
- Harshita Katiyar
- Lady Davis Institute, Jewish General Hospital, Montreal, QC H3T 1E2, Canada; (H.K.); (A.A.); (Y.L.)
- Department of Microbiology and Immunology, McGill University, Montreal, QC H3A 2B4, Canada
| | - Ariana Arduini
- Lady Davis Institute, Jewish General Hospital, Montreal, QC H3T 1E2, Canada; (H.K.); (A.A.); (Y.L.)
- Department of Medicine, McGill University, Montreal, QC H3G 2M1, Canada
| | - Yichen Li
- Lady Davis Institute, Jewish General Hospital, Montreal, QC H3T 1E2, Canada; (H.K.); (A.A.); (Y.L.)
- Department of Microbiology and Immunology, McGill University, Montreal, QC H3A 2B4, Canada
| | - Chen Liang
- Lady Davis Institute, Jewish General Hospital, Montreal, QC H3T 1E2, Canada; (H.K.); (A.A.); (Y.L.)
- Department of Microbiology and Immunology, McGill University, Montreal, QC H3A 2B4, Canada
- Department of Medicine, McGill University, Montreal, QC H3G 2M1, Canada
| |
Collapse
|
41
|
Jhanwar A, Sharma D, Das U. Unraveling the structural and functional dimensions of SARS-CoV2 proteins in the context of COVID-19 pathogenesis and therapeutics. Int J Biol Macromol 2024; 278:134850. [PMID: 39168210 DOI: 10.1016/j.ijbiomac.2024.134850] [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: 01/12/2024] [Revised: 08/14/2024] [Accepted: 08/16/2024] [Indexed: 08/23/2024]
Abstract
Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV2) has emerged as the causative agent behind the global pandemic of Coronavirus Disease 2019 (COVID-19). As the scientific community strives to comprehend the intricate workings of this virus, a fundamental aspect lies in deciphering the myriad proteins it expresses. This knowledge is pivotal in unraveling the complexities of the viral machinery and devising targeted therapeutic interventions. The proteomic landscape of SARS-CoV2 encompasses structural, non-structural, and open-reading frame proteins, each playing crucial roles in viral replication, host interactions, and the pathogenesis of COVID-19. This comprehensive review aims to provide an updated and detailed examination of the structural and functional attributes of SARS-CoV2 proteins. By exploring the intricate molecular architecture, we have highlighted the significance of these proteins in viral biology. Insights into their roles and interplay contribute to a deeper understanding of the virus's mechanisms, thereby paving the way for the development of effective therapeutic strategies. As the global scientific community strives to combat the ongoing pandemic, this synthesis of knowledge on SARS-CoV2 proteins serves as a valuable resource, fostering informed approaches toward mitigating the impact of COVID-19 and advancing the frontier of antiviral research.
Collapse
Affiliation(s)
- Aniruddh Jhanwar
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India
| | - Dipika Sharma
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India
| | - Uddipan Das
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India.
| |
Collapse
|
42
|
Wright NJ, Zhang F, Suo Y, Kong L, Yin Y, Fedor JG, Sharma K, Borgnia MJ, Im W, Lee SY. Antiviral drug recognition and elevator-type transport motions of CNT3. Nat Chem Biol 2024; 20:1144-1153. [PMID: 38418906 PMCID: PMC11625470 DOI: 10.1038/s41589-024-01559-8] [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: 09/01/2023] [Accepted: 01/22/2024] [Indexed: 03/02/2024]
Abstract
Nucleoside analogs have broad clinical utility as antiviral drugs. Key to their systemic distribution and cellular entry are human nucleoside transporters. Here, we establish that the human concentrative nucleoside transporter 3 (CNT3) interacts with antiviral drugs used in the treatment of coronavirus infections. We report high-resolution single-particle cryo-electron microscopy structures of bovine CNT3 complexed with antiviral nucleosides N4-hydroxycytidine, PSI-6206, GS-441524 and ribavirin, all in inward-facing states. Notably, we found that the orally bioavailable antiviral molnupiravir arrests CNT3 in four distinct conformations, allowing us to capture cryo-electron microscopy structures of drug-loaded outward-facing and drug-loaded intermediate states. Our studies uncover the conformational trajectory of CNT3 during membrane transport of a nucleoside analog antiviral drug, yield new insights into the role of interactions between the transport and the scaffold domains in elevator-like domain movements during drug translocation, and provide insights into the design of nucleoside analog antiviral prodrugs with improved oral bioavailability.
Collapse
Affiliation(s)
- Nicholas J Wright
- Department of Biochemistry, Duke University School of Medicine, Durham, NC, USA
| | - Feng Zhang
- Department of Biochemistry, Duke University School of Medicine, Durham, NC, USA
| | - Yang Suo
- Department of Biochemistry, Duke University School of Medicine, Durham, NC, USA
| | - Lingyang Kong
- Departments of Biological Sciences, Chemistry, and Bioengineering, Lehigh University, Bethlehem, PA, USA
| | - Ying Yin
- Department of Biochemistry, Duke University School of Medicine, Durham, NC, USA
| | - Justin G Fedor
- Department of Biochemistry, Duke University School of Medicine, Durham, NC, USA
| | - Kedar Sharma
- Department of Health and Human Services, Genome Integrity and Structural Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, Durham, NC, USA
| | - Mario J Borgnia
- Department of Health and Human Services, Genome Integrity and Structural Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, Durham, NC, USA
| | - Wonpil Im
- Departments of Biological Sciences, Chemistry, and Bioengineering, Lehigh University, Bethlehem, PA, USA
| | - Seok-Yong Lee
- Department of Biochemistry, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|
43
|
Bolinger AA, Li J, Xie X, Li H, Zhou J. Lessons learnt from broad-spectrum coronavirus antiviral drug discovery. Expert Opin Drug Discov 2024; 19:1023-1041. [PMID: 39078037 PMCID: PMC11390334 DOI: 10.1080/17460441.2024.2385598] [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: 02/22/2024] [Accepted: 07/24/2024] [Indexed: 07/31/2024]
Abstract
INTRODUCTION Highly pathogenic coronaviruses (CoVs), such as severe acute respiratory syndrome CoV (SARS-CoV), Middle East respiratory syndrome CoV (MERS-CoV), and the most recent SARS-CoV-2 responsible for the COVID-19 pandemic, pose significant threats to human populations over the past two decades. These CoVs have caused a broad spectrum of clinical manifestations ranging from asymptomatic to severe distress syndromes (ARDS), resulting in high morbidity and mortality. AREAS COVERED The accelerated advancements in antiviral drug discovery, spurred by the COVID-19 pandemic, have shed new light on the imperative to develop treatments effective against a broad spectrum of CoVs. This perspective discusses strategies and lessons learnt in targeting viral non-structural proteins, structural proteins, drug repurposing, and combinational approaches for the development of antivirals against CoVs. EXPERT OPINION Drawing lessons from the pandemic, it becomes evident that the absence of efficient broad-spectrum antiviral drugs increases the vulnerability of public health systems to the potential onslaught by highly pathogenic CoVs. The rapid and sustained spread of novel CoVs can have devastating consequences without effective and specifically targeted treatments. Prioritizing the effective development of broad-spectrum antivirals is imperative for bolstering the resilience of public health systems and mitigating the potential impact of future highly pathogenic CoVs.
Collapse
Affiliation(s)
- Andrew A. Bolinger
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Jun Li
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Xuping Xie
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, USA
- Sealy Institute for Drug Discovery, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Hongmin Li
- Department of Pharmacology and Toxicology, College of Pharmacy, The BIO5 Institute, The University of Arizona, Tucson, AZ 85721, USA
| | - Jia Zhou
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, USA
- Sealy Institute for Drug Discovery, University of Texas Medical Branch, Galveston, TX 77555, USA
| |
Collapse
|
44
|
Istifli ES, Okumus N, Sarikurkcu C, Kuhn ER, Netz PA, Tepe AS. Comparative docking and molecular dynamics studies of molnupiravir (EIDD-2801): implications for novel mechanisms of action on influenza and SARS-CoV-2 protein targets. J Biomol Struct Dyn 2024; 42:8202-8214. [PMID: 37811782 DOI: 10.1080/07391102.2023.2267696] [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: 05/11/2023] [Accepted: 07/27/2023] [Indexed: 10/10/2023]
Abstract
Molnupiravir (EIDD-2801) (MLN) is an oral antiviral drug for COVID-19 treatment, being integrated into viral RNA through RNA-dependent RNA polymerase (RdRp). Upon ingestion, MLN is transformed into two active metabolites: β-d-N4-hydroxycytidine (NHC) (EIDD-1931) in the host plasma, and EIDD-1931-triphosphate (MTP) within the host cells. However, recent studies provide increasing evidence of MLN's interactions with off-target proteins beyond the viral genome, suggesting that the complete mechanisms of action of MLN remain unclear. The aim of this study was therefore to investigate the molecular interactions of MLN in the form of NHC and MTP with the non-RNA structural components of avian influenza (hemagglutinin, neuraminidase) and SARS-CoV-2 (spike glycoprotein, Mpro, and RdRp) viruses and to elucidate whether these two metabolites possess the ability to form stable complexes with these major viral components. Molecular docking of NHC and MTP was performed using AutoDock 4.2.6 and the obtained protein-drug complexes were submitted to 200-ns molecular dynamics simulations in triplicate with subsequent free energy calculations using GROMACS. Docking scores, molecular dynamics and MM/GBSA results showed that MTP was tightly bound within the active site of SARS-CoV-2 RdRp and remained highly stable throughout the 200-ns simulations. Besides, it was also shown that NHC and MTP formed moderately-to-highly stable molecular complexes with off-target receptors hemagglutinin, neuraminidase and Mpro, but rather weak interactions with spike glycoprotein. Our computational findings suggest that NHC and MTP may directly inhibit these receptors, and propose that additional studies on the off-target effects of MLN, i.e. real-time protein binding assays, should be performed.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Erman Salih Istifli
- Faculty of Science and Literature, Department of Biology, Cukurova University, Adana, Turkey
| | - Nurullah Okumus
- Faculty of Medicine, Department of Pediatrics, Afyonkarahisar Health Sciences University, Afyonkarahisar, Turkey
| | - Cengiz Sarikurkcu
- Faculty of Pharmacy, Department of Analytical Chemistry, Afyonkarahisar Health Sciences University, Afyonkarahisar, Turkey
| | - Eduardo Ramires Kuhn
- Theoretical Chemistry Group, Institute of Chemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Paulo A Netz
- Theoretical Chemistry Group, Institute of Chemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Arzuhan Sihoglu Tepe
- Department of Pharmacy Services, Kilis 7 Aralik University, Vocational High School of Health Services, Kilis, Turkey
| |
Collapse
|
45
|
Raczkiewicz I, Rivière C, Bouquet P, Desmarets L, Tarricone A, Camuzet C, François N, Lefèvre G, Silva Angulo F, Robil C, Trottein F, Sahpaz S, Dubuisson J, Belouzard S, Goffard A, Séron K. Hyperforin, the major metabolite of St. John's wort, exhibits pan-coronavirus antiviral activity. Front Microbiol 2024; 15:1443183. [PMID: 39176276 PMCID: PMC11339956 DOI: 10.3389/fmicb.2024.1443183] [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: 06/03/2024] [Accepted: 07/29/2024] [Indexed: 08/24/2024] Open
Abstract
Introduction The COVID-19 pandemic caused by the SARS-CoV-2 virus has underscored the urgent necessity for the development of antiviral compounds that can effectively target coronaviruses. In this study, we present the first evidence of the antiviral efficacy of hyperforin, a major metabolite of St. John's wort, for which safety and bioavailability in humans have already been established. Methods Antiviral assays were conducted in cell culture with four human coronaviruses: three of high virulence, SARS-CoV-2, SARS-CoV, and MERS-CoV, and one causing mild symptoms, HCoV-229E. The antiviral activity was also evaluated in human primary airway epithelial cells. To ascertain the viral step inhibited by hyperforin, time-of-addition assays were conducted. Subsequently, a combination assay of hyperforin with remdesivir was performed. Results The results demonstrated that hyperforin exhibited notable antiviral activity against the four tested human coronaviruses, with IC50 values spanning from 0.24 to 2.55 µM. Kinetic studies indicated that the observed activity occur at a post-entry step, potentially during replication. The antiviral efficacy of hyperforin was additionally corroborated in human primary airway epithelial cells. The results demonstrated a reduction in both intracellular and extracellular SARS-CoV-2 viral RNA, confirming that hyperforin targeted the replication step. Finally, an additive antiviral effect on SARS-CoV-2 was observed when hyperforin was combined with remdesivir. Discussion In conclusion, hyperforin has been identified as a novel pan-coronavirus inhibitor with activity in human primary airway epithelial cells, a preclinical model for coronaviruses. These findings collectively suggest that hyperforin has potential as a candidate antiviral agent against current and future human coronaviruses.
Collapse
Affiliation(s)
- Imelda Raczkiewicz
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 – UMR9017 – Center for Infection and Immunity of Lille (CIIL), Lille, France
| | - Céline Rivière
- BioEcoAgro, Joint Research Unit 1158, Univ. Lille, INRAE, Univ. Liège, UPJV, YNCREA, Univ. Artois, Univ. Littoral Côte d’Opale, ICV – Institut Charles Viollette, Lille, France
| | - Peggy Bouquet
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 – UMR9017 – Center for Infection and Immunity of Lille (CIIL), Lille, France
| | - Lowiese Desmarets
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 – UMR9017 – Center for Infection and Immunity of Lille (CIIL), Lille, France
| | - Audrey Tarricone
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 – UMR9017 – Center for Infection and Immunity of Lille (CIIL), Lille, France
| | - Charline Camuzet
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 – UMR9017 – Center for Infection and Immunity of Lille (CIIL), Lille, France
| | - Nathan François
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 – UMR9017 – Center for Infection and Immunity of Lille (CIIL), Lille, France
| | - Gabriel Lefèvre
- BioEcoAgro, Joint Research Unit 1158, Univ. Lille, INRAE, Univ. Liège, UPJV, YNCREA, Univ. Artois, Univ. Littoral Côte d’Opale, ICV – Institut Charles Viollette, Lille, France
| | - Fabiola Silva Angulo
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 – UMR9017 – Center for Infection and Immunity of Lille (CIIL), Lille, France
| | - Cyril Robil
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 – UMR9017 – Center for Infection and Immunity of Lille (CIIL), Lille, France
| | - François Trottein
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 – UMR9017 – Center for Infection and Immunity of Lille (CIIL), Lille, France
| | - Sevser Sahpaz
- BioEcoAgro, Joint Research Unit 1158, Univ. Lille, INRAE, Univ. Liège, UPJV, YNCREA, Univ. Artois, Univ. Littoral Côte d’Opale, ICV – Institut Charles Viollette, Lille, France
| | - Jean Dubuisson
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 – UMR9017 – Center for Infection and Immunity of Lille (CIIL), Lille, France
| | - Sandrine Belouzard
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 – UMR9017 – Center for Infection and Immunity of Lille (CIIL), Lille, France
| | - Anne Goffard
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 – UMR9017 – Center for Infection and Immunity of Lille (CIIL), Lille, France
| | - Karin Séron
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 – UMR9017 – Center for Infection and Immunity of Lille (CIIL), Lille, France
| |
Collapse
|
46
|
Du X, Yang X, Zhao J, Zhang J, Yu J, Ma L, Zhang W, Cen S, Ren X, He X. Design of novel broad-spectrum antiviral nucleoside analogues using natural bases ring-opening strategy. Drug Dev Res 2024; 85:e22237. [PMID: 39032059 DOI: 10.1002/ddr.22237] [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: 12/18/2023] [Revised: 06/19/2024] [Accepted: 07/07/2024] [Indexed: 07/22/2024]
Abstract
The global prevalence of RNA virus infections has presented significant challenges to public health in recent years, necessitating the expansion of its alternative therapeutic library. Due to its evolutional conservation, RNA-dependent RNA polymerase (RdRp) has emerged as a potential target for broad-spectrum antiviral nucleoside analogues. However, after over half a century of structural modification, exploring unclaimed chemical space using frequently-used structural substitution methods to design new nucleoside analogues is challenging. In this study, we explore the use of the "ring-opening" strategy to design new base mimics, thereby using these base mimics to design new nucleoside analogues with broad-spectrum antiviral activities. A total of 29 compounds were synthesized. Their activity against viral RdRp was initially screened using an influenza A virus RdRp high-throughput screening model. Then, the antiviral activity of 38a was verified against influenza virus strain A/PR/8/34 (H1N1), demonstrating a 50% inhibitory concentration (IC50) value of 9.95 μM, which was superior to that of ribavirin (the positive control, IC50 = 11.43 μM). Moreover, 38a also has inhibitory activity against coronavirus 229E with an IC50 of 30.82 μM. In addition, compounds 42 and 46f exhibit an 82% inhibition rate against vesicular stomatitis virus at a concentration of 20 μM and hardly induce cytotoxicity in host cells. This work demonstrates the feasibility of designing nucleoside analogues with "ring-opening" bases and suggests the "ring-opening" nucleosides may have greater polarity, and designing prodrugs is an important aspect of optimizing their antiviral activity. Future research should focus on enhancing the conformational restriction of open-loop bases to mimic Watson-Crick base pairing better and improve antiviral activity.
Collapse
Affiliation(s)
- Xingyi Du
- Key Laboratory of Structure-Based Drug Design and Discovery, Shenyang Pharmaceutical University, Shenyang, China
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
| | - Xingxing Yang
- Key Laboratory of Structure-Based Drug Design and Discovery, Shenyang Pharmaceutical University, Shenyang, China
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Jianyuan Zhao
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science, Beijing, China
| | - Jinyan Zhang
- Key Laboratory of Structure-Based Drug Design and Discovery, Shenyang Pharmaceutical University, Shenyang, China
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Jiahui Yu
- Key Laboratory of Structure-Based Drug Design and Discovery, Shenyang Pharmaceutical University, Shenyang, China
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Ling Ma
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science, Beijing, China
| | - Weina Zhang
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, National Center of Biomedical Analysis, Beijing, China
| | - Shan Cen
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science, Beijing, China
| | - Xuhong Ren
- Key Laboratory of Structure-Based Drug Design and Discovery, Shenyang Pharmaceutical University, Shenyang, China
| | - Xinhua He
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| |
Collapse
|
47
|
Jena NR, Pant S. Peptide inhibitors derived from the nsp7 and nsp8 cofactors of nsp12 targeting different substrate binding sites of nsp12 of the SARS-CoV-2. J Biomol Struct Dyn 2024; 42:7077-7089. [PMID: 37434315 DOI: 10.1080/07391102.2023.2235012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 07/05/2023] [Indexed: 07/13/2023]
Abstract
SARS-COV-2 is responsible for the COVID-19 pandemic, which has infected more than 767 million people worldwide including about 7 million deaths till 5 June 2023. Despite the emergency use of certain vaccines, deaths due to COVID-19 have not yet stopped completed. Therefore, it is imperative to design and develop drugs that can be used to treat patients suffering from COVID-19. Here, two peptide inhibitors derived from nsp7 and nsp8 cofactors of nsp12 have been shown to block different substrate binding sites of nsp12 that are mainly responsible for the replication of the viral genome of SARS-CoV-2. By using the docking, molecular dynamics (MD), and MM/GBSA techniques, it is shown that these inhibitors can bind to multiple binding sites of nsp12, such as the interface of nsp7 and nsp12, interface of nsp8 and nsp12, RNA primer entry site, and nucleoside triphosphate (NTP) entry site. The relative binding free energies of the most stable protein-peptide complexes are found to lie between ∼-34.20 ± 10.07 to -59.54 ± 9.96 kcal/mol. Hence, it is likely that these inhibitors may bind to different sites of nsp12 to block the access of its cofactors and the viral genome, thereby affecting the replication. It is thus proposed that these peptide inhibitors may be further developed as potential drug candidates to suppress the viral loads in COVID-19 patients.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- N R Jena
- Discipline of Natural Sciences, Indian Institute of Information Technology, Design, and Manufacturing, Jabalpur, India
| | - Suyash Pant
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research, Kolkata, India
| |
Collapse
|
48
|
Chan JFW, Yuan S, Chu H, Sridhar S, Yuen KY. COVID-19 drug discovery and treatment options. Nat Rev Microbiol 2024; 22:391-407. [PMID: 38622352 DOI: 10.1038/s41579-024-01036-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/28/2024] [Indexed: 04/17/2024]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused substantial morbidity and mortality, and serious social and economic disruptions worldwide. Unvaccinated or incompletely vaccinated older individuals with underlying diseases are especially prone to severe disease. In patients with non-fatal disease, long COVID affecting multiple body systems may persist for months. Unlike SARS-CoV and Middle East respiratory syndrome coronavirus, which have either been mitigated or remained geographically restricted, SARS-CoV-2 has disseminated globally and is likely to continue circulating in humans with possible emergence of new variants that may render vaccines less effective. Thus, safe, effective and readily available COVID-19 therapeutics are urgently needed. In this Review, we summarize the major drug discovery approaches, preclinical antiviral evaluation models, representative virus-targeting and host-targeting therapeutic options, and key therapeutics currently in clinical use for COVID-19. Preparedness against future coronavirus pandemics relies not only on effective vaccines but also on broad-spectrum antivirals targeting conserved viral components or universal host targets, and new therapeutics that can precisely modulate the immune response during infection.
Collapse
Affiliation(s)
- Jasper Fuk-Woo Chan
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Department of Infectious Diseases and Microbiology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, Guangdong Province, China
- Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Shatin, Hong Kong Special Administrative Region, China
| | - Shuofeng Yuan
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Department of Infectious Diseases and Microbiology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, Guangdong Province, China
- Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Shatin, Hong Kong Special Administrative Region, China
| | - Hin Chu
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Department of Infectious Diseases and Microbiology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, Guangdong Province, China
- Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Shatin, Hong Kong Special Administrative Region, China
| | - Siddharth Sridhar
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Department of Infectious Diseases and Microbiology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, Guangdong Province, China
| | - Kwok-Yung Yuen
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China.
- Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China.
- Department of Infectious Diseases and Microbiology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, Guangdong Province, China.
- Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Shatin, Hong Kong Special Administrative Region, China.
| |
Collapse
|
49
|
Hershan AA. Pathogenesis of COVID19 and the applications of US FDA-approved repurposed antiviral drugs to combat SARS-CoV-2 in Saudi Arabia: A recent update by review of literature. Saudi J Biol Sci 2024; 31:104023. [PMID: 38799719 PMCID: PMC11127266 DOI: 10.1016/j.sjbs.2024.104023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 05/05/2024] [Accepted: 05/10/2024] [Indexed: 05/29/2024] Open
Abstract
Still, there is no cure for the highly contagious severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-caused coronavirus disease 2019 (COVID19). The COVID19 pandemic caused health emergencies which resulted in enormous medical and financial consequences worldwide including Saudi Arabia. Saudi Arabia is the largest Arab country of the Middle East. The urban setting of Saudi Arabia makes it vulnerable towards SARS-CoV-2 (SCV-2). Religious areas of this country are visited by millions of pilgrims every year for the Umrah and Hajj pilgrimage, which contributes to the potential COVID19 epidemic risk. COVID19 throws various challenges to healthcare professionals to choose the right drugs or therapy in clinical settings because of the lack of availability of newer drugs. Current drug development and discovery is an expensive, complex, and long process, which involves a high failure rate in clinical trials. While repurposing of United States Food and Drug Administration (US FDA)-approved antiviral drugs offers numerous benefits including complete pharmacokinetic and safety profiles, which significantly shorten drug development cycles and reduce costs. A range of repurposed US FDA-approved antiviral drugs including ribavirin, lopinavir/ritonavir combination, oseltamivir, darunavir, remdesivir, nirmatrelvir/ritonavir combination, and molnupiravir showed encouraging results in clinical trials in COVID19 treatment. In this article, several COVID19-related discussions have been provided including emerging variants of concern of, COVID19 pathogenesis, COVID19 pandemic scenario in Saudi Arabia, drug repurposing strategies against SCV-2, as well as repurposing of US FDA-approved antiviral drugs that might be considered to combat SCV-2 in Saudi Arabia. Moreover, drug repurposing in the context of COVID19 management along with its limitations and future perspectives have been summarized.
Collapse
Affiliation(s)
- Almonther Abdullah Hershan
- The University of Jeddah, College of Medicine, Department of Medical microbiology and parasitology, Jeddah, Saudi Arabia
| |
Collapse
|
50
|
Focosi D, Franchini M, Maggi F, Shoham S. COVID-19 therapeutics. Clin Microbiol Rev 2024; 37:e0011923. [PMID: 38771027 PMCID: PMC11237566 DOI: 10.1128/cmr.00119-23] [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: 05/22/2024] Open
Abstract
SUMMARYSince the emergence of COVID-19 in 2020, an unprecedented range of therapeutic options has been studied and deployed. Healthcare providers have multiple treatment approaches to choose from, but efficacy of those approaches often remains controversial or compromised by viral evolution. Uncertainties still persist regarding the best therapies for high-risk patients, and the drug pipeline is suffering fatigue and shortage of funding. In this article, we review the antiviral activity, mechanism of action, pharmacokinetics, and safety of COVID-19 antiviral therapies. Additionally, we summarize the evidence from randomized controlled trials on efficacy and safety of the various COVID-19 antivirals and discuss unmet needs which should be addressed.
Collapse
Affiliation(s)
- Daniele Focosi
- North-Western Tuscany Blood Bank, Pisa University Hospital, Pisa, Italy
| | - Massimo Franchini
- Division of Hematology and Transfusion Medicine, Carlo Poma Hospital, Mantua, Italy
| | - Fabrizio Maggi
- National Institute for Infectious Diseases "Lazzaro Spallanzani" IRCCS, Rome, Italy
| | - Shmuel Shoham
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|