1
|
Tan H, Pollard B, Li K, Wang J. Discovery of A-967079 as an Enterovirus D68 Antiviral by Targeting the Viral 2C Protein. ACS Infect Dis 2024; 10:4327-4336. [PMID: 39578369 DOI: 10.1021/acsinfecdis.4c00678] [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: 11/24/2024]
Abstract
Enterovirus D68 (EV-D68) has had several outbreaks worldwide, yet no FDA-approved antiviral is available for treating this viral infection. EV-D68 infection typically leads to respiratory illnesses and, in severe cases, can cause neurological complications and even death, particularly in children. This study identified a small molecule, A-967079, as an EV-D68 antiviral through phenotypical screening. A-967079 has shown potent and broad-spectrum antiviral activity with a high selectivity index against multiple strains of EV-D68. Pharmacological characterization of the mechanism of action involving time-of-addition, resistance selection, and differential scanning fluorimetry assays suggests that viral 2C protein is the drug target. Overall, A-967079 represents a promising candidate for further development as an EV-D68 antiviral.
Collapse
Affiliation(s)
- Haozhou Tan
- Department of Medicinal Chemistry, Ernest Mario School of Pharmacy, Rutgers, the State University of New Jersey, Piscataway, New Jersey 08854, United States
| | - Brian Pollard
- Department of Medicinal Chemistry, Ernest Mario School of Pharmacy, Rutgers, the State University of New Jersey, Piscataway, New Jersey 08854, United States
| | - Kan Li
- Department of Medicinal Chemistry, Ernest Mario School of Pharmacy, Rutgers, the State University of New Jersey, Piscataway, New Jersey 08854, United States
| | - Jun Wang
- Department of Medicinal Chemistry, Ernest Mario School of Pharmacy, Rutgers, the State University of New Jersey, Piscataway, New Jersey 08854, United States
| |
Collapse
|
2
|
Wu M, Wan Q, Dan X, Wang Y, Chen P, Chen C, Li Y, Yao X, He ML. Targeting Ser78 phosphorylation of Hsp27 achieves potent antiviral effects against enterovirus A71 infection. Emerg Microbes Infect 2024; 13:2368221. [PMID: 38932432 PMCID: PMC11212574 DOI: 10.1080/22221751.2024.2368221] [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: 01/29/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024]
Abstract
A positive-sense (+) single-stranded RNA (ssRNA) virus (e.g. enterovirus A71, EV-A71) depends on viral polypeptide translation for initiation of virus replication after entry. We reported that EV-A71 hijacks Hsp27 to induce hnRNP A1 cytosol redistribution to initiate viral protein translation, but the underlying mechanism is still elusive. Here, we show that phosphorylation-deficient Hsp27-3A (Hsp27S15/78/82A) and Hsp27S78A fail to translocate into the nucleus and induce hnRNP A1 cytosol redistribution, while Hsp27S15A and Hsp27S82A display similar effects to the wild type Hsp27. Furthermore, we demonstrate that the viral 2A protease (2Apro) activity is a key factor in regulating Hsp27/hnRNP A1 relocalization. Hsp27S78A dramatically decreases the IRES activity and viral replication, which are partially reduced by Hsp27S82A. However, Hsp27S15A displays the same activity as the wild-type Hsp27. Peptide S78 potently suppresses EV-A71 protein translation and reproduction through blockage of EV-A71-induced Hsp27 phosphorylation and Hsp27/hnRNP A1 relocalization. A point mutation (S78A) on S78 impairs its inhibitory functions on Hsp27/hnRNP A1 relocalization and viral replication. Taken together, we demonstrate the importance of Ser78 phosphorylation of Hsp27 regulated by virus infection in nuclear translocation, hnRNP A1 cytosol relocation, and viral replication, suggesting a new path (such as peptide S78) for target-based antiviral strategy.
Collapse
Affiliation(s)
- Mandi Wu
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong Special Administrative Region, People’s Republic of China
| | - Qianya Wan
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong Special Administrative Region, People’s Republic of China
| | - Xuelian Dan
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong Special Administrative Region, People’s Republic of China
- Department of Infectious Diseases, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, People’s Republic of China
| | - Yiran Wang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong Special Administrative Region, People’s Republic of China
| | - Peiran Chen
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong Special Administrative Region, People’s Republic of China
| | - Cien Chen
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong Special Administrative Region, People’s Republic of China
| | - Yichen Li
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong Special Administrative Region, People’s Republic of China
| | - Xi Yao
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong Special Administrative Region, People’s Republic of China
| | - Ming-Liang He
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong Special Administrative Region, People’s Republic of China
- CityU Shenzhen Research Institute, Shenzhen, People’s Republic of China
| |
Collapse
|
3
|
Xie H, Rhoden EE, Liu HM, Ogunsemowo F, Mainou BA, Burke RM, Burns CC. Antiviral Development for the Polio Endgame: Current Progress and Future Directions. Pathogens 2024; 13:969. [PMID: 39599522 PMCID: PMC11597170 DOI: 10.3390/pathogens13110969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 10/31/2024] [Accepted: 11/04/2024] [Indexed: 11/29/2024] Open
Abstract
As the world is approaching the eradication of wild poliovirus serotype 1, the last of the three wild types, the question of how to maintain a polio-free world becomes imminent. To mitigate the risk of sporadic vaccine-associated paralytic polio (VAPP) caused by oral polio vaccines (OPVs) that are routinely used in global immunization programs, the Polio Antivirals Initiative (PAI) was established in 2006. The primary goal of the PAI is to facilitate the discovery and development of antiviral drugs to stop the excretion of immunodeficiency-associated vaccine-derived poliovirus (iVDPV) in B cell-deficient individuals. This review summarizes the major progress that has been made in the development of safe and effective poliovirus antivirals and highlights the candidates that have shown promising results in vitro, in vivo, and in clinical trials.
Collapse
Affiliation(s)
- Hang Xie
- Poliovirus & Picornavirus Branch, Division of Viral Diseases, National Center for Immunization and Other Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA; (E.E.R.); (H.-M.L.); (F.O.); (B.A.M.); (C.C.B.)
| | - Eric E. Rhoden
- Poliovirus & Picornavirus Branch, Division of Viral Diseases, National Center for Immunization and Other Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA; (E.E.R.); (H.-M.L.); (F.O.); (B.A.M.); (C.C.B.)
| | - Hong-Mei Liu
- Poliovirus & Picornavirus Branch, Division of Viral Diseases, National Center for Immunization and Other Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA; (E.E.R.); (H.-M.L.); (F.O.); (B.A.M.); (C.C.B.)
| | - Folake Ogunsemowo
- Poliovirus & Picornavirus Branch, Division of Viral Diseases, National Center for Immunization and Other Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA; (E.E.R.); (H.-M.L.); (F.O.); (B.A.M.); (C.C.B.)
| | - Bernardo A. Mainou
- Poliovirus & Picornavirus Branch, Division of Viral Diseases, National Center for Immunization and Other Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA; (E.E.R.); (H.-M.L.); (F.O.); (B.A.M.); (C.C.B.)
| | | | - Cara C. Burns
- Poliovirus & Picornavirus Branch, Division of Viral Diseases, National Center for Immunization and Other Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA; (E.E.R.); (H.-M.L.); (F.O.); (B.A.M.); (C.C.B.)
| |
Collapse
|
4
|
Richter M, Khrenova M, Kazakova E, Riabova O, Egorova A, Makarov V, Schmidtke M. Dynamic features of virus protein 1 and substitutions in the 3-phenyl ring determine the potency and broad-spectrum activity of capsid-binding pyrazolo[3,4-d]pyrimidines against rhinoviruses. Antiviral Res 2024; 231:105993. [PMID: 39233314 DOI: 10.1016/j.antiviral.2024.105993] [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: 07/12/2024] [Revised: 08/15/2024] [Accepted: 08/24/2024] [Indexed: 09/06/2024]
Abstract
Pyrazolo[3,4-d]pyrimidines represent one potent class of well tolerated and highly active rhinovirus (RV) inhibitors that act as capsid binders. The lead compound OBR-5-340 inhibits a broad-spectrum of RVs. Aiming to improve lead activity, we evaluated the impact of structural modifications in the 3-phenyl ring of OBR-5-340 on its potency and spectrum of anti-RV activity vitro. Our results demonstrate the crucial role of substitution at position 4 for strong, broad-spectrum anti-RV activity. The 4-methyl (RCB23137) and 4-chloro (RCB23138) derivatives outperformed OBR-5-340 in terms of potency and anti-RV activity spectrum. Based on these findings, the compounds were selected for computational binding studies. Molecular dynamic simulations with six RVs differing in OBR-5-340, RCB23137, and RCB23138 sensitivity proved the impact of dynamic features of two VP1 loops enveloping these inhibitors on antiviral potency.
Collapse
Affiliation(s)
- Martina Richter
- Jena University Hospital, Institute of Medical Microbiology, Section Experimental Virology, Hans-Knoell-Str. 2, 07743, Jena, Germany
| | - Maria Khrenova
- Federal Research Centre "Fundamentals of Biotechnology" of the Russian Academy of Sciences (Research Centre of Biotechnology RAS), 33-2 Leninsky Prospect, 119071, Moscow, Russia; Department of Chemistry, Lomonosov Moscow State University, 1/3 Leninskie Gory, 119991, Moscow, Russia
| | - Elena Kazakova
- Federal Research Centre "Fundamentals of Biotechnology" of the Russian Academy of Sciences (Research Centre of Biotechnology RAS), 33-2 Leninsky Prospect, 119071, Moscow, Russia
| | - Olga Riabova
- Federal Research Centre "Fundamentals of Biotechnology" of the Russian Academy of Sciences (Research Centre of Biotechnology RAS), 33-2 Leninsky Prospect, 119071, Moscow, Russia
| | - Anna Egorova
- Federal Research Centre "Fundamentals of Biotechnology" of the Russian Academy of Sciences (Research Centre of Biotechnology RAS), 33-2 Leninsky Prospect, 119071, Moscow, Russia
| | - Vadim Makarov
- Federal Research Centre "Fundamentals of Biotechnology" of the Russian Academy of Sciences (Research Centre of Biotechnology RAS), 33-2 Leninsky Prospect, 119071, Moscow, Russia.
| | - Michaela Schmidtke
- Jena University Hospital, Institute of Medical Microbiology, Section Experimental Virology, Hans-Knoell-Str. 2, 07743, Jena, Germany.
| |
Collapse
|
5
|
Lin X, Sun Q, Cao Y, Li Z, Xu C, Liu J, Song J, Qin K, Zhang Y, Zhou J. A Novel Peptide from VP1 of EV-D68 Exhibits Broad-Spectrum Antiviral Activity Against Human Enteroviruses. Biomolecules 2024; 14:1331. [PMID: 39456264 PMCID: PMC11506774 DOI: 10.3390/biom14101331] [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: 09/10/2024] [Revised: 10/12/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024] Open
Abstract
Enteroviruses have been a historical concern since the identification of polioviruses in humans. Wild polioviruses have almost been eliminated, while multiple species of non-polio enteroviruses and their variants co-circulate annually. To date, at least 116 types have been found in humans and are grouped into the species Enterovirus A-D and Rhinovirus A-C. However, there are few available antiviral drugs, especially with a universal pharmaceutical effect. Here, we demonstrate that peptide P25 from EV-D68 has broad antiviral activity against EV A-D enteroviruses in vitro. P25, derived from the HI loop and β-I sheet of VP1, operates through a conserved hydrophilic motif -R---K-K--K- and the hydrophobic F near the N-terminus. It could prevent viral infection of EV-A71 by competing for the heparan sulfate (HS) receptor, binding and stabilizing virions by suppressing the release of the viral genome. P25 also inhibited the generation of infectious viral particles by reducing viral protein synthesis. The molecular docking revealed that P25 might bind to the pocket opening area, a potential target for broad-spectrum antivirals. Our findings implicate the multiple antiviral effects of peptide P25, including blocking viral binding to the HS receptor, impeding viral genome release, and reducing progeny particles, which could be a novel universal anti-enterovirus drug candidate.
Collapse
Affiliation(s)
- Xiaojing Lin
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China; (X.L.); (Q.S.); (Z.L.); (C.X.); (J.L.); (J.S.); (K.Q.)
| | - Qiang Sun
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China; (X.L.); (Q.S.); (Z.L.); (C.X.); (J.L.); (J.S.); (K.Q.)
| | - Yang Cao
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610064, China;
| | - Zi Li
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China; (X.L.); (Q.S.); (Z.L.); (C.X.); (J.L.); (J.S.); (K.Q.)
| | - Cuiling Xu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China; (X.L.); (Q.S.); (Z.L.); (C.X.); (J.L.); (J.S.); (K.Q.)
| | - Jun Liu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China; (X.L.); (Q.S.); (Z.L.); (C.X.); (J.L.); (J.S.); (K.Q.)
| | - Jingdong Song
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China; (X.L.); (Q.S.); (Z.L.); (C.X.); (J.L.); (J.S.); (K.Q.)
| | - Kun Qin
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China; (X.L.); (Q.S.); (Z.L.); (C.X.); (J.L.); (J.S.); (K.Q.)
| | - Yong Zhang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China; (X.L.); (Q.S.); (Z.L.); (C.X.); (J.L.); (J.S.); (K.Q.)
| | - Jianfang Zhou
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China; (X.L.); (Q.S.); (Z.L.); (C.X.); (J.L.); (J.S.); (K.Q.)
| |
Collapse
|
6
|
Roux H, Touret F, Rathelot P, Vanelle P, Roche M. From the "One-Molecule, One-Target, One-Disease" Concept towards Looking for Multi-Target Therapeutics for Treating Non-Polio Enterovirus (NPEV) Infections. Pharmaceuticals (Basel) 2024; 17:1218. [PMID: 39338380 PMCID: PMC11434921 DOI: 10.3390/ph17091218] [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: 07/11/2024] [Revised: 09/09/2024] [Accepted: 09/11/2024] [Indexed: 09/30/2024] Open
Abstract
Non-polio enteroviruses (NPEVs), namely coxsackieviruses (CV), echoviruses (E), enteroviruses (EV), and rhinoviruses (RV), are responsible for a wide variety of illnesses. Some infections can progress to life-threatening conditions in children or immunocompromised patients. To date, no treatments have been approved. Several molecules have been evaluated through clinical trials without success. To overcome these failures, the multi-target directed ligand (MTDL) strategy could be applied to tackle enterovirus infections. This work analyzes registered clinical trials involving antiviral drugs to highlight the best candidates and develops filters to apply to a selection for MTDL synthesis. We explicitly stated the methods used to answer the question: which solution can fight NPEVs effectively? We note the originality and relevance of this proposal in relation to the state of the art in the enterovirus-inhibitors field. Several combinations are possible to broaden the antiviral spectrum and potency. We discuss data related to the virus and data related to each LEAD compound identified so far. Overall, this study proposes a perspective on different strategies to overcome issues identified in clinical trials and evaluate the "MTDL" potential to improve the efficacy of drugs, broaden the antiviral targets, possibly reduce the adverse effects, drug design costs and limit the selection of drug-resistant virus variants.
Collapse
Affiliation(s)
- Hugo Roux
- Aix-Marseille Université, CNRS, ICR UMR_7273, LPCR, Faculté de Pharmacie, 13005 Marseille, France; (H.R.); (P.R.)
| | - Franck Touret
- Unité des Virus Émergents (UVE: Aix-Marseille Université, Università di Corsica, IRD 190, Inserm 1207, IRBA), 13005 Marseille, France;
| | - Pascal Rathelot
- Aix-Marseille Université, CNRS, ICR UMR_7273, LPCR, Faculté de Pharmacie, 13005 Marseille, France; (H.R.); (P.R.)
| | - Patrice Vanelle
- Aix-Marseille Université, CNRS, ICR UMR_7273, LPCR, Faculté de Pharmacie, 13005 Marseille, France; (H.R.); (P.R.)
| | - Manon Roche
- Aix-Marseille Université, CNRS, ICR UMR_7273, LPCR, Faculté de Pharmacie, 13005 Marseille, France; (H.R.); (P.R.)
| |
Collapse
|
7
|
Lin X, Gan J, Sun Q, Li Z, Qin K, Zhang Y, Cao Y, Zhou J. The Structural Framework and Opening Appearance of the VP1-Pocket of Enteroviruses Correlated with Viral Thermostability. Pathogens 2024; 13:711. [PMID: 39204311 PMCID: PMC11357065 DOI: 10.3390/pathogens13080711] [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: 07/09/2024] [Revised: 08/08/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024] Open
Abstract
Enteroviruses (EVs and RVs) are prevalent worldwide and cause various diseases in humans, of which the VP1-pocket is a target of antivirals, with a lipid molecule as a pocket factor to stabilize the virion. However, the characterization of the structure of the VP1-pocket in EVs is poor. Here, we compared the published capsid crystals of EVs and RVs and proposed a structural framework for the VP1-pocket: Frame 1-4, which is located at the CD loop, GH loop, and C-terminus, presenting with an outward opening appearance or not. The non-outward viral strains-CVB3, Echo 11, RV-A81, and RV-B70-are more thermally stable, with a breakpoint temperature (B.T.) of 51~62 °C for genome releasing, which is 4~10 °C higher than its outward temperature of 41~47 °C, and infectivity preservation when treated at 50 °C for 3 min. Its outward versus non-outward opening is correlated significantly with the B.T. for genome release (r = -0.90; p = 0.0004) and infectivity (r = -0.82, p = 0.0039). The energy of Frames 1, 2, and 4, including Van der Waals attractive and repulsive interactions and hydrogen bonds, showed significant correlations with the B.T. (r = -0.67, 0.75, and -0.8; p = 0.034, 0.013, and 0.006, respectively). These characters of the VP1-pocket could be predictors for virion thermostability and aid in the development of vaccines or antivirals.
Collapse
Affiliation(s)
- Xiaojing Lin
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China; (X.L.); (Q.S.); (Z.L.); (K.Q.); (Y.Z.)
| | - Jianhong Gan
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610064, China; (J.G.); (Y.C.)
| | - Qiang Sun
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China; (X.L.); (Q.S.); (Z.L.); (K.Q.); (Y.Z.)
| | - Zi Li
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China; (X.L.); (Q.S.); (Z.L.); (K.Q.); (Y.Z.)
| | - Kun Qin
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China; (X.L.); (Q.S.); (Z.L.); (K.Q.); (Y.Z.)
| | - Yong Zhang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China; (X.L.); (Q.S.); (Z.L.); (K.Q.); (Y.Z.)
| | - Yang Cao
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610064, China; (J.G.); (Y.C.)
| | - Jianfang Zhou
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China; (X.L.); (Q.S.); (Z.L.); (K.Q.); (Y.Z.)
| |
Collapse
|
8
|
Trujillo-Gómez J, Navarro CE, Atehortúa-Muñoz S, Florez ID. Acute infections of the central nervous system in children and adults: diagnosis and management. Minerva Med 2024; 115:476-502. [PMID: 39376101 DOI: 10.23736/s0026-4806.24.09097-9] [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: 10/09/2024]
Abstract
Central nervous system infections are due to different microorganisms such as viruses, bacteria, mycobacteria, fungi, amoebas, and other parasites. The etiology depends on multiple risk factors, and it defines the infection location because some microorganisms prefer meninges, brain tissue, cerebellum, brain stem or spinal cord. The microorganisms induce diseases in the nervous system through direct invasion, neurotoxin production, and the triggered immune response. To determine the infection etiology, there are several diagnostic tests which may be conducted with cerebrospinal fluid, blood, respiratory and stool samples. These tests include but are not limited to direct microscopic examination of the sample, stains, cultures, antigenic tests, nucleic acid amplification tests, metagenomic next-generation sequencing, immunologic biomarker and neuroimaging, especially contrast-enhanced magnetic resonance imaging. The treatment may consist of specific antimicrobial treatment and supportive standard care. Since viruses have no specific antiviral treatment, antimicrobial treatment is mainly targeted at non-viral infections. This article will focus on diagnosis and treatment of acute acquired infections of the central nervous system beyond the neonatal period. The discussion defines the disease, provides the clinical presentation, explains the etiology and risk factors, and briefly mentions potential complications. This updated review aims to provide the reader with all the elements needed to adequately approach a patient with a central nervous system infection. Mycobacterium tuberculosis infection, Cryptococcus spp. infection and vaccines are not within the scope of this article.
Collapse
Affiliation(s)
- Juliana Trujillo-Gómez
- Hospital General de Medellín, Medellín, Colombia
- School of Medicine, University of Antioquia, Medellín, Colombia
| | - Cristian E Navarro
- School of Medicine, University of Antioquia, Medellín, Colombia
- Grupo de Investigación, ESE Hospital Emiro Quintero Cañizares, Ocaña, Colombia
| | - Santiago Atehortúa-Muñoz
- Hospital Pablo Tobón Uribe, Medellín, Colombia
- Clínica Universitaria Bolivariana, Medellín, Colombia
| | - Ivan D Florez
- Department of Pediatrics, University of Antioquia, Medellín, Colombia -
- School of Rehabilitation Science, McMaster University, Hamilton, ON, Canada
- Pediatric Intensive Care Unit, Clínica Las Américas AUNA, Medellín, Colombia
| |
Collapse
|
9
|
Lü Z, Dai X, Xu J, Liu Z, Guo Y, Gao Z, Meng F. Medicinal chemistry strategies toward broad-spectrum antiviral agents to prevent next pandemics. Eur J Med Chem 2024; 271:116442. [PMID: 38685143 DOI: 10.1016/j.ejmech.2024.116442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 04/02/2024] [Accepted: 04/19/2024] [Indexed: 05/02/2024]
Abstract
The pandemic and tremendous impact of severe acute respiratory syndrome coronavirus 2 alert us, despite great achievements in prevention and control of infectious diseases, we still lack universal and powerful antiviral strategies to rapidly respond to the potential threat of serious infectious disease. Various highly contagious and pathogenic viruses, as well as other unknown viruses may appear or reappear in human society at any time, causing a catastrophic epidemic. Developing broad-spectrum antiviral drugs with high security and efficiency is of great significance for timely meeting public health emergency and protecting the lives and health of the people. Hence, in this review, we summarized diverse broad-spectrum antiviral targets and corresponding agents from a medicinal chemistry prospective, compared the pharmacological advantages and disadvantages of different targets, listed representative agents, showed their structures, pharmacodynamics and pharmacokinetics characteristics, and conducted a critical discussion on their development potential, in the hope of providing up-to-date guidance for the development of broad-spectrum antivirals and perspectives for applications of antiviral therapy.
Collapse
Affiliation(s)
- Zirui Lü
- State Key Laboratory of NBC Protection for Civilian, Beijing, 102205, China
| | - Xiandong Dai
- State Key Laboratory of NBC Protection for Civilian, Beijing, 102205, China
| | - Jianjie Xu
- State Key Laboratory of NBC Protection for Civilian, Beijing, 102205, China
| | - Zhenming Liu
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, China
| | - Yongbiao Guo
- State Key Laboratory of NBC Protection for Civilian, Beijing, 102205, China
| | - Zhenhua Gao
- State Key Laboratory of NBC Protection for Civilian, Beijing, 102205, China
| | - Fanhua Meng
- State Key Laboratory of NBC Protection for Civilian, Beijing, 102205, China.
| |
Collapse
|
10
|
Wang S, Pang Z, Fan H, Tong Y. Advances in anti-EV-A71 drug development research. J Adv Res 2024; 56:137-156. [PMID: 37001813 PMCID: PMC10834817 DOI: 10.1016/j.jare.2023.03.007] [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: 01/17/2023] [Revised: 03/05/2023] [Accepted: 03/21/2023] [Indexed: 03/31/2023] Open
Abstract
BACKGROUND Enterovirus A71 (EV-A71) is capable of causing hand, foot and mouth disease (HFMD), which may lead to neurological sequelae and even death. As EV-A71 is resistant to environmental changes and mutates easily, there is still a lack of effective treatments or globally available vaccines. AIM OF REVIEW For more than 50 years since the HFMD epidemic, related drug research has been conducted. Progress in this area can promote the further application of existing potential drugs and develop more efficient and safe antiviral drugs, and provide useful reference for protecting the younger generation and maintaining public health security. KEY SCIENTIFIC CONCEPTS OF REVIEW At present, researchers have identified hundreds of EV-A71 inhibitors based on screening repurposed drugs, targeted structural design, and rational modification of previously effective drugs as the main development strategies. This review systematically introduces the current potential drugs to inhibit EV-A71 infection, including viral inhibitors targeting key sites such as the viral capsid, RNA-dependent RNA polymerase (RdRp), 2C protein, internal ribosome entry site (IRES), 3C proteinase (3Cpro), and 2A proteinase (2Apro), starting from each stage of the viral life cycle. Meanwhile, the progress of host-targeting antiviral drugs and their development are summarized in terms of regulating host immunity, inhibiting autophagy or apoptosis, and regulating the cellular redox environment. In addition, the current clinical methods for the prevention and treatment of HFMD are summarized and discussed with the aim of providing support and recommendations for the treatment of enterovirus infections including EV-A71.
Collapse
Affiliation(s)
- Shuqi Wang
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Zehan Pang
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Huahao Fan
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China.
| | - Yigang Tong
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China; Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing, China.
| |
Collapse
|
11
|
Richter M, Döring K, Blaas D, Riabova O, Khrenova M, Kazakova E, Egorova A, Makarov V, Schmidtke M. Molecular mechanism of rhinovirus escape from the Pyrazolo[3,4-d]pyrimidine capsid-binding inhibitor OBR-5-340 via mutations distant from the binding pocket: Derivatives that brake resistance. Antiviral Res 2024; 222:105810. [PMID: 38244889 DOI: 10.1016/j.antiviral.2024.105810] [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: 11/17/2023] [Revised: 01/04/2024] [Accepted: 01/14/2024] [Indexed: 01/22/2024]
Abstract
Rhinoviruses (RVs) cause the common cold. Attempts at discovering small molecule inhibitors have mainly concentrated on compounds supplanting the medium chain fatty acids residing in the sixty icosahedral symmetry-related hydrophobic pockets of the viral capsid of the Rhinovirus-A and -B species. High-affinity binding to these pockets stabilizes the capsid against structural changes necessary for the release of the ss(+) RNA genome into the cytosol of the host cell. However, single-point mutations may abolish this binding. RV-B5 is one of several RVs that are naturally resistant against the well-established antiviral agent pleconaril. However, RV-B5 is strongly inhibited by the pyrazolopyrimidine OBR-5-340. Here, we report on isolation and characterization of RV-B5 mutants escaping OBR-5-340 inhibition and show that substitution of amino acid residues not only within the binding pocket but also remote from the binding pocket hamper inhibition. Molecular dynamics network analysis revealed that strong inhibition occurs when an ensemble of several sequence stretches of the capsid proteins enveloping OBR-5-340 move together with OBR-5-340. Mutations abrogating this dynamic, regardless of whether being localized within the binding pocket or distant from it result in escape from inhibition. Pyrazolo [3,4-d]pyrimidine derivatives overcoming OBR-5-340 escape of various RV-B5 mutants were identified. Our work contributes to the understanding of the properties of capsid-binding inhibitors necessary for potent and broad-spectrum inhibition of RVs.
Collapse
Affiliation(s)
- Martina Richter
- Jena University Hospital, Department Medical Microbiology, Section Experimental Virology, Hans-Knoell-Str. 2, 07743 Jena, Germany
| | - Kristin Döring
- Jena University Hospital, Department Medical Microbiology, Section Experimental Virology, Hans-Knoell-Str. 2, 07743 Jena, Germany
| | - Dieter Blaas
- Medical University Vienna, Centre of Med. Biochem. Vienna Biocenter, Dr. Bohr Gasse 9/3, A-1030 Vienna, Austria
| | - Olga Riabova
- Federal Research Centre "Fundamentals of Biotechnology" of the Russian Academy of Sciences (Research Centre of Biotechnology RAS), 33-2 Leninsky Prospect, 119071 Moscow, Russia
| | - Maria Khrenova
- Federal Research Centre "Fundamentals of Biotechnology" of the Russian Academy of Sciences (Research Centre of Biotechnology RAS), 33-2 Leninsky Prospect, 119071 Moscow, Russia; Department of Chemistry, Lomonosov Moscow State University, 1/3 Leninskie Gory, 119991 Moscow, Russia
| | - Elena Kazakova
- Federal Research Centre "Fundamentals of Biotechnology" of the Russian Academy of Sciences (Research Centre of Biotechnology RAS), 33-2 Leninsky Prospect, 119071 Moscow, Russia
| | - Anna Egorova
- Federal Research Centre "Fundamentals of Biotechnology" of the Russian Academy of Sciences (Research Centre of Biotechnology RAS), 33-2 Leninsky Prospect, 119071 Moscow, Russia
| | - Vadim Makarov
- Federal Research Centre "Fundamentals of Biotechnology" of the Russian Academy of Sciences (Research Centre of Biotechnology RAS), 33-2 Leninsky Prospect, 119071 Moscow, Russia.
| | - Michaela Schmidtke
- Jena University Hospital, Department Medical Microbiology, Section Experimental Virology, Hans-Knoell-Str. 2, 07743 Jena, Germany.
| |
Collapse
|
12
|
Abd-Aziz N, Lee MF, Ong SK, Poh CL. Antiviral activity of SP81 peptide against Enterovirus A71 (EV-A71). Virology 2024; 589:109941. [PMID: 37984152 DOI: 10.1016/j.virol.2023.109941] [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: 07/05/2023] [Revised: 10/27/2023] [Accepted: 11/09/2023] [Indexed: 11/22/2023]
Abstract
The hand, food, and mouth disease (HFMD) is primarily caused by Enterovirus A71 (EV-A71). EV-A71 outbreaks in the Asia Pacific have been associated with severe neurological disease and high fatalities. Currently, there are no FDA-approved antivirals for the treatment of EV-A71 infections. In this study, the SP81 peptide, derived from the VP1 capsid protein of EV-A71 was shown to be a promising antiviral candidate for the treatment of EV-A71 infections. SP81 peptide was non-toxic to RD cells up to 45 μM, with a half-maximal cytotoxic concentration (CC50) of 90.32 μM. SP81 peptide exerted antiviral effects during the pre- and post-infection stages with 50% inhibitory concentrations (IC50) of 4.529 μM and 1.192 μM, respectively. Direct virus inactivation of EV-A71 by the SP81 peptide was also observed with an IC50 of 8.076 μM. Additionally, the SP81 peptide exhibited direct virus inactivation of EV-A71 at 95% upon the addition of the SP81 peptide within 5 min. This study showed that the SP81 peptide exhibited significant inhibition of EV-A71 and could serve as a promising antiviral agent for further clinical development against EV-A71 infections.
Collapse
Affiliation(s)
- Noraini Abd-Aziz
- Centre for Virus and Vaccine Research (CVVR), School of Medical and Life Sciences, Sunway University, 47500, Subang Jaya, Selangor, Malaysia
| | - Michelle Felicia Lee
- Centre for Virus and Vaccine Research (CVVR), School of Medical and Life Sciences, Sunway University, 47500, Subang Jaya, Selangor, Malaysia
| | - Seng-Kai Ong
- Department of Biological Sciences, School of Medical and Life Sciences, Sunway University, 47500, Subang Jaya, Selangor, Malaysia
| | - Chit Laa Poh
- Centre for Virus and Vaccine Research (CVVR), School of Medical and Life Sciences, Sunway University, 47500, Subang Jaya, Selangor, Malaysia.
| |
Collapse
|
13
|
Prapassornwattana P, Hannongbua S, Saparpakorn P. Elucidation of benzene sulfonamide derivative binding at a novel interprotomer pocket of wild type and mutants of coxsackievirus B3 viral capsid using molecular dynamics simulations and density functional theory. Biophys Chem 2023; 302:107109. [PMID: 37748430 DOI: 10.1016/j.bpc.2023.107109] [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: 05/01/2023] [Revised: 09/10/2023] [Accepted: 09/16/2023] [Indexed: 09/27/2023]
Abstract
Coxsackievirus B3 (CVB3), a serotype of enterovirus B, causes hand, foot, and mouth disease; pericarditis; and myocarditis. A benzene sulfonamide derivative is reported to have inhibitory activity against wild-type (WT) and eight mutants of the viral capsid of CVB3. Furthermore, the crystal structure of the complex formed between WT viral capsid of CVB3 and the derivative revealed binding at a novel druggable interprotomer pocket. We investigated how the compound could be a potent inhibitor of both WT and some mutants of CVB3 by determining binding to the viral capsid and the interaction energy with the binding pocket based on molecular dynamics simulations and density functional theory. We found that hydrogen bonds, pi-pi interactions, and electrostatic interactions are the key interactions with a protomer unit of CVB3 viral capsid. The residual interaction energy determined using density functional theory revealed key binding with VP1:Arg234 and a residue in the nearby VP1 unit (VP1':Arg219). These results explain why the compound is still a potent inhibitor against eight mutants. Moreover, the decreased inhibitory activity for some mutants could be explained by the calculated binding energy and the highest occupied molecular orbital and lowest unoccupied molecular orbital energy. The results will be helpful for the development of drugs resistant to CVB3.
Collapse
Affiliation(s)
- Pavinee Prapassornwattana
- Department of Chemistry, Faculty of Science, Kasetsart University, Chatuchak, Bangkok 10900, Thailand; Center for Advanced Studies in Nanotechnology for Chemical, Food and Agricultural Industries, KU Institute for Advanced Studies, Kasetsart University, Bangkok 10900, Thailand
| | - Supa Hannongbua
- Department of Chemistry, Faculty of Science, Kasetsart University, Chatuchak, Bangkok 10900, Thailand; Center for Advanced Studies in Nanotechnology for Chemical, Food and Agricultural Industries, KU Institute for Advanced Studies, Kasetsart University, Bangkok 10900, Thailand
| | - Patchreenart Saparpakorn
- Department of Chemistry, Faculty of Science, Kasetsart University, Chatuchak, Bangkok 10900, Thailand; Center for Advanced Studies in Nanotechnology for Chemical, Food and Agricultural Industries, KU Institute for Advanced Studies, Kasetsart University, Bangkok 10900, Thailand.
| |
Collapse
|
14
|
Lane TR, Fu J, Sherry B, Tarbet B, Hurst BL, Riabova O, Kazakova E, Egorova A, Clarke P, Leser JS, Frost J, Rudy M, Tyler KL, Klose T, Volobueva AS, Belyaevskaya SV, Zarubaev VV, Kuhn RJ, Makarov V, Ekins S. Efficacy of an isoxazole-3-carboxamide analog of pleconaril in mouse models of Enterovirus-D68 and Coxsackie B5. Antiviral Res 2023; 216:105654. [PMID: 37327878 PMCID: PMC10527014 DOI: 10.1016/j.antiviral.2023.105654] [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: 04/27/2023] [Revised: 06/05/2023] [Accepted: 06/13/2023] [Indexed: 06/18/2023]
Abstract
Enteroviruses (EV) cause a number of life-threatening infectious diseases. EV-D68 is known to cause respiratory illness in children that can lead to acute flaccid myelitis. Coxsackievirus B5 (CVB5) is commonly associated with hand-foot-mouth disease. There is no antiviral treatment available for either. We have developed an isoxazole-3-carboxamide analog of pleconaril (11526092) which displayed potent inhibition of EV-D68 (IC50 58 nM) as well as other enteroviruses including the pleconaril-resistant Coxsackievirus B3-Woodruff (IC50 6-20 nM) and CVB5 (EC50 1 nM). Cryo-electron microscopy structures of EV-D68 in complex with 11526092 and pleconaril demonstrate destabilization of the EV-D68 MO strain VP1 loop, and a strain-dependent effect. A mouse respiratory model of EV-D68 infection, showed 3-log decreased viremia, favorable cytokine response, as well as statistically significant 1-log reduction in lung titer reduction at day 5 after treatment with 11526092. An acute flaccid myelitis neurological infection model did not show efficacy. 11526092 was tested in a mouse model of CVB5 infection and showed a 4-log TCID50 reduction in the pancreas. In summary, 11526092 represents a potent in vitro inhibitor of EV with in vivo efficacy in EV-D68 and CVB5 animal models suggesting it is worthy of further evaluation as a potential broad-spectrum antiviral therapeutic against EV.
Collapse
Affiliation(s)
- Thomas R Lane
- Collaborations Pharmaceuticals Inc., Raleigh, NC, USA
| | - Jianing Fu
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA
| | - Barbara Sherry
- Department of Molecular Biomedical Sciences, North Carolina State University, College of Veterinary Medicine, Raleigh, NC, USA
| | - Bart Tarbet
- Institute for Antiviral Research, Utah State University, Logan, UT, USA; Department of Animal, Dairy and Veterinary Sciences, Utah State University, Logan, UT, USA
| | - Brett L Hurst
- Institute for Antiviral Research, Utah State University, Logan, UT, USA; Department of Animal, Dairy and Veterinary Sciences, Utah State University, Logan, UT, USA
| | - Olga Riabova
- Research Center of Biotechnology RAS, 33-1 Leninsky prospect, 119071, Moscow, Russia
| | - Elena Kazakova
- Research Center of Biotechnology RAS, 33-1 Leninsky prospect, 119071, Moscow, Russia
| | - Anna Egorova
- Research Center of Biotechnology RAS, 33-1 Leninsky prospect, 119071, Moscow, Russia
| | - Penny Clarke
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - J Smith Leser
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Joshua Frost
- Department of Immunology and Microbiology, Infectious Disease, Medicine and Neurology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | | | - Kenneth L Tyler
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Department of Veterans Affairs, Aurora, CO, USA
| | - Thomas Klose
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA
| | | | | | - Vladimir V Zarubaev
- Saint Petersburg Pasteur Institute, 14 Mira Street, 197101, Saint Petersburg, Russia
| | - Richard J Kuhn
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA
| | - Vadim Makarov
- Research Center of Biotechnology RAS, 33-1 Leninsky prospect, 119071, Moscow, Russia
| | - Sean Ekins
- Collaborations Pharmaceuticals Inc., Raleigh, NC, USA.
| |
Collapse
|
15
|
Real-Hohn A, Groznica M, Kontaxis G, Zhu R, Chaves OA, Vazquez L, Hinterdorfer P, Kowalski H, Blaas D. Stabilization of the Quadruplex-Forming G-Rich Sequences in the Rhinovirus Genome Inhibits Uncoating-Role of Na + and K . Viruses 2023; 15:1003. [PMID: 37112983 PMCID: PMC10141139 DOI: 10.3390/v15041003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 04/14/2023] [Accepted: 04/17/2023] [Indexed: 04/29/2023] Open
Abstract
Rhinoviruses (RVs) are the major cause of common cold, a respiratory disease that generally takes a mild course. However, occasionally, RV infection can lead to serious complications in patients debilitated by other ailments, e.g., asthma. Colds are a huge socioeconomic burden as neither vaccines nor other treatments are available. The many existing drug candidates either stabilize the capsid or inhibit the viral RNA polymerase, the viral proteinases, or the functions of other non-structural viral proteins; however, none has been approved by the FDA. Focusing on the genomic RNA as a possible target for antivirals, we asked whether stabilizing RNA secondary structures might inhibit the viral replication cycle. These secondary structures include G-quadruplexes (GQs), which are guanine-rich sequence stretches forming planar guanine tetrads via Hoogsteen base pairing with two or more of them stacking on top of each other; a number of small molecular drug candidates increase the energy required for their unfolding. The propensity of G-quadruplex formation can be predicted with bioinformatics tools and is expressed as a GQ score. Synthetic RNA oligonucleotides derived from the RV-A2 genome with sequences corresponding to the highest and lowest GQ scores indeed exhibited characteristics of GQs. In vivo, the GQ-stabilizing compounds, pyridostatin and PhenDC3, interfered with viral uncoating in Na+ but not in K+-containing phosphate buffers. The thermostability studies and ultrastructural imaging of protein-free viral RNA cores suggest that Na+ keeps the encapsulated genome more open, allowing PDS and PhenDC3 to diffuse into the quasi-crystalline RNA and promote the formation and/or stabilization of GQs; the resulting conformational changes impair RNA unraveling and release from the virion. Preliminary reports have been published.
Collapse
Affiliation(s)
- Antonio Real-Hohn
- Center of Medical Biochemistry, Vienna Biocenter, Max Perutz Laboratories, Medical University of Vienna, Dr. Bohr Gasse 9/3, 1030 Vienna, Austria; (M.G.)
| | - Martin Groznica
- Center of Medical Biochemistry, Vienna Biocenter, Max Perutz Laboratories, Medical University of Vienna, Dr. Bohr Gasse 9/3, 1030 Vienna, Austria; (M.G.)
- Institut Pasteur, CEDEX 15, 75724 Paris, France
| | - Georg Kontaxis
- Vienna Biocenter, Max Perutz Laboratories, Department of Structural and Computational Biology, University of Vienna, Campus Vienna BioCenter 5, 1030 Vienna, Austria;
| | - Rong Zhu
- Institute of Biophysics, Johannes Kepler University Linz, Gruberstr. 40, 4020 Linz, Austria; (R.Z.)
| | - Otávio Augusto Chaves
- Immunopharmacology Laboratory, Oswaldo Cruz Institute (IOC/Fiocruz), Av. Brasil, 4365, Rio de Janeiro 21040-360, Brazil
| | - Leonardo Vazquez
- Immunopharmacology Laboratory, Oswaldo Cruz Institute (IOC/Fiocruz), Av. Brasil, 4365, Rio de Janeiro 21040-360, Brazil
| | - Peter Hinterdorfer
- Institute of Biophysics, Johannes Kepler University Linz, Gruberstr. 40, 4020 Linz, Austria; (R.Z.)
| | - Heinrich Kowalski
- Center of Medical Biochemistry, Vienna Biocenter, Max Perutz Laboratories, Medical University of Vienna, Dr. Bohr Gasse 9/3, 1030 Vienna, Austria; (M.G.)
| | - Dieter Blaas
- Center of Medical Biochemistry, Vienna Biocenter, Max Perutz Laboratories, Medical University of Vienna, Dr. Bohr Gasse 9/3, 1030 Vienna, Austria; (M.G.)
| |
Collapse
|
16
|
Laajala M, Kalander K, Consalvi S, Amamuddy OS, Bishop ÖT, Biava M, Poce G, Marjomäki V. Antiviral Mechanisms of N-Phenyl Benzamides on Coxsackie Virus A9. Pharmaceutics 2023; 15:pharmaceutics15031028. [PMID: 36986888 PMCID: PMC10058015 DOI: 10.3390/pharmaceutics15031028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/14/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
Enteroviruses are one of the most abundant groups of viruses infecting humans, and yet there are no approved antivirals against them. To find effective antiviral compounds against enterovirus B group viruses, an in-house chemical library was screened. The most effective compounds against Coxsackieviruses B3 (CVB3) and A9 (CVA9) were CL212 and CL213, two N-phenyl benzamides. Both compounds were more effective against CVA9 and CL213 gave a better EC50 value of 1 µM with high a specificity index of 140. Both drugs were most effective when incubated directly with viruses suggesting that they mainly bound to the virions. A real-time uncoating assay showed that the compounds stabilized the virions and radioactive sucrose gradient as well as TEM confirmed that the viruses stayed intact. A docking assay, taking into account larger areas around the 2-and 3-fold axes of CVA9 and CVB3, suggested that the hydrophobic pocket gives the strongest binding to CVA9 but revealed another binding site around the 3-fold axis which could contribute to the binding of the compounds. Together, our data support a direct antiviral mechanism against the virus capsid and suggest that the compounds bind to the hydrophobic pocket and 3-fold axis area resulting in the stabilization of the virion.
Collapse
Affiliation(s)
- Mira Laajala
- Department of Biological and Environmental Science/Nanoscience Center, University of Jyväskylä, 40014 Jyväskylä, Finland
| | - Kerttu Kalander
- Department of Biological and Environmental Science/Nanoscience Center, University of Jyväskylä, 40014 Jyväskylä, Finland
| | - Sara Consalvi
- Department of Chemistry and Technologies of Drug, Sapienza University of Rome, Piazzale A. Moro 5, 00185 Rome, Italy
| | - Olivier Sheik Amamuddy
- Research Unit in Bioinformatics (RUBi), Department of Biochemistry and Microbiology, Rhodes University, Makhanda 6140, South Africa
| | - Özlem Tastan Bishop
- Research Unit in Bioinformatics (RUBi), Department of Biochemistry and Microbiology, Rhodes University, Makhanda 6140, South Africa
| | - Mariangela Biava
- Department of Chemistry and Technologies of Drug, Sapienza University of Rome, Piazzale A. Moro 5, 00185 Rome, Italy
| | - Giovanna Poce
- Department of Chemistry and Technologies of Drug, Sapienza University of Rome, Piazzale A. Moro 5, 00185 Rome, Italy
| | - Varpu Marjomäki
- Department of Biological and Environmental Science/Nanoscience Center, University of Jyväskylä, 40014 Jyväskylä, Finland
| |
Collapse
|
17
|
Ibba R, Corona P, Nonne F, Caria P, Serreli G, Palmas V, Riu F, Sestito S, Nieddu M, Loddo R, Sanna G, Piras S, Carta A. Design, Synthesis, and Antiviral Activities of New Benzotriazole-Based Derivatives. Pharmaceuticals (Basel) 2023; 16:ph16030429. [PMID: 36986528 PMCID: PMC10054465 DOI: 10.3390/ph16030429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 03/03/2023] [Accepted: 03/07/2023] [Indexed: 03/16/2023] Open
Abstract
Several human diseases are caused by enteroviruses and are currently clinically untreatable, pushing the research to identify new antivirals. A notable number of benzo[d][1,2,3]triazol-1(2)-yl derivatives were designed, synthesized, and in vitro evaluated for cytotoxicity and antiviral activity against a wide spectrum of RNA positive- and negative-sense viruses. Five of them (11b, 18e, 41a, 43a, 99b) emerged for their selective antiviral activity against Coxsackievirus B5, a human enteroviruses member among the Picornaviridae family. The EC50 values ranged between 6 and 18.5 μM. Among all derivatives, compounds 18e and 43a were interestingly active against CVB5 and were selected to better define the safety profile on cell monolayers by transepithelial resistance test (TEER). Results indicated compound 18e as the hit compound to investigate the potential mechanism of action by apoptosis assay, virucidal activity test, and the time of addition assay. CVB5 is known to be cytotoxic by inducing apoptosis in infected cells; in this study, compound 18e was proved to protect cells from viral infection. Notably, cells were mostly protected when pre-treated with derivative 18e, which had, however, no virucidal activity. From the performed biological assays, compound 18e turned out to be non-cytotoxic as well as cell protective against CVB5 infection, with a mechanism of action ascribable to an interaction on the early phase of infection, by hijacking the viral attachment process.
Collapse
Affiliation(s)
- Roberta Ibba
- Department of Medicine, Surgery and Pharmacy, University of Sassari, Via Muroni, 23/A, 07100 Sassari, Italy; (R.I.); (P.C.); (F.R.); (M.N.)
| | - Paola Corona
- Department of Medicine, Surgery and Pharmacy, University of Sassari, Via Muroni, 23/A, 07100 Sassari, Italy; (R.I.); (P.C.); (F.R.); (M.N.)
| | - Francesca Nonne
- GSK Vaccine Institute for Global Health GSK, Via Fiorentina, 1, 53100 Siena, Italy;
| | - Paola Caria
- Department of Biomedical Sciences, Section of Microbiology and Virology, University of Cagliari, Cittadella Universitaria, 09042 Monserrato, Italy; (P.C.); (G.S.); (V.P.); (R.L.)
| | - Gabriele Serreli
- Department of Biomedical Sciences, Section of Microbiology and Virology, University of Cagliari, Cittadella Universitaria, 09042 Monserrato, Italy; (P.C.); (G.S.); (V.P.); (R.L.)
| | - Vanessa Palmas
- Department of Biomedical Sciences, Section of Microbiology and Virology, University of Cagliari, Cittadella Universitaria, 09042 Monserrato, Italy; (P.C.); (G.S.); (V.P.); (R.L.)
| | - Federico Riu
- Department of Medicine, Surgery and Pharmacy, University of Sassari, Via Muroni, 23/A, 07100 Sassari, Italy; (R.I.); (P.C.); (F.R.); (M.N.)
- Department of Chemistry, Biomedicinskt Centrum, BMC, Uppsala University, Box 576, 75123 Uppsala, Sweden
| | - Simona Sestito
- Department of Chemical, Physical, Mathematical and Natural Sciences, University of Sassari, Via Vienna 2, 07100 Sassari, Italy;
| | - Maria Nieddu
- Department of Medicine, Surgery and Pharmacy, University of Sassari, Via Muroni, 23/A, 07100 Sassari, Italy; (R.I.); (P.C.); (F.R.); (M.N.)
| | - Roberta Loddo
- Department of Biomedical Sciences, Section of Microbiology and Virology, University of Cagliari, Cittadella Universitaria, 09042 Monserrato, Italy; (P.C.); (G.S.); (V.P.); (R.L.)
| | - Giuseppina Sanna
- Department of Biomedical Sciences, Section of Microbiology and Virology, University of Cagliari, Cittadella Universitaria, 09042 Monserrato, Italy; (P.C.); (G.S.); (V.P.); (R.L.)
- Correspondence: (G.S.); (S.P.)
| | - Sandra Piras
- Department of Medicine, Surgery and Pharmacy, University of Sassari, Via Muroni, 23/A, 07100 Sassari, Italy; (R.I.); (P.C.); (F.R.); (M.N.)
- Correspondence: (G.S.); (S.P.)
| | - Antonio Carta
- Department of Medicine, Surgery and Pharmacy, University of Sassari, Via Muroni, 23/A, 07100 Sassari, Italy; (R.I.); (P.C.); (F.R.); (M.N.)
| |
Collapse
|
18
|
Direct-Acting Antivirals and Host-Targeting Approaches against Enterovirus B Infections: Recent Advances. Pharmaceuticals (Basel) 2023. [DOI: 10.3390/ph16020203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Enterovirus B (EV-B)-related diseases, which can be life threatening in high-risk populations, have been recognized as a serious health problem, but their clinical treatment is largely supportive, and no selective antivirals are available on the market. As their clinical relevance has become more serious, efforts in the field of anti-EV-B inhibitors have greatly increased and many potential antivirals with very high selectivity indexes and promising in vitro activities have been discovered. The scope of this review encompasses recent advances in the discovery of new compounds with anti-viral activity against EV-B, as well as further progress in repurposing drugs to treat these infections. Current progress and future perspectives in drug discovery against EV-Bs are briefly discussed and existing gaps are spotlighted.
Collapse
|
19
|
Alhazmi A, Nekoua MP, Mercier A, Vergez I, Sane F, Alidjinou EK, Hober D. Combating coxsackievirus B infections. Rev Med Virol 2023; 33:e2406. [PMID: 36371612 DOI: 10.1002/rmv.2406] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 10/11/2022] [Accepted: 10/27/2022] [Indexed: 11/15/2022]
Abstract
Coxsackieviruses B (CVB) are small, non-enveloped, single-stranded RNA viruses belonging to the Enterovirus genus of the Picornaviridae family. They are common worldwide and cause a wide variety of human diseases ranging from those having relatively mild symptoms to severe acute and chronic pathologies such as cardiomyopathy and type 1 diabetes. The development of safe and effective strategies to combat these viruses remains a challenge. The present review outlines current approaches to control CVB infections and associated diseases. Various drugs targeting viral or host proteins involved in viral replication as well as vaccines have been developed and shown potential to prevent or combat CVB infections in vitro and in vivo in animal models. Repurposed drugs and alternative strategies targeting miRNAs or based on plant extracts and probiotics and their derivatives have also shown antiviral effects against CVB. In addition, clinical trials with vaccines and drugs are underway and offer hope for the prevention or treatment of CVB-induced diseases.
Collapse
Affiliation(s)
- Abdulaziz Alhazmi
- Laboratoire de Virologie ULR3610, Université de Lille et CHU de Lille, Lille, France.,Microbiology and Parasitology Department, Faculty of Medicine, Jazan University, Jazan, Saudi Arabia
| | | | - Ambroise Mercier
- Laboratoire de Virologie ULR3610, Université de Lille et CHU de Lille, Lille, France
| | - Ines Vergez
- Laboratoire de Virologie ULR3610, Université de Lille et CHU de Lille, Lille, France
| | - Famara Sane
- Laboratoire de Virologie ULR3610, Université de Lille et CHU de Lille, Lille, France
| | | | - Didier Hober
- Laboratoire de Virologie ULR3610, Université de Lille et CHU de Lille, Lille, France
| |
Collapse
|
20
|
On the Sensitivity of the Virion Envelope to Lipid Peroxidation. Microbiol Spectr 2022; 10:e0300922. [PMID: 36125312 PMCID: PMC9603946 DOI: 10.1128/spectrum.03009-22] [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] [Indexed: 01/04/2023] Open
Abstract
Emerging viruses are a public health threat best managed with broad spectrum antivirals. Common viral structures, like capsids or virion envelopes, have been proposed as targets for broadly active antiviral drugs. For example, a number of lipoperoxidators have been proposed to preferentially affect viral infectivity by targeting metabolically inactive enveloped virions while sparing metabolically active cells. However, this presumed preferential virion sensitivity to lipoperoxidation remains untested. To test whether virions are indeed more sensitive to lipoperoxidation than are cells, we analyzed the effects of two classic generic lipoperoxidators: lipophilic 2,2'-azobis(2,4-dimethylvaleronitrile) (AMVN) and hydrophilic 2,2'-azobis(2-methylpropionamidine) dihydrochloride (AAPH) on Vero and human foreskin fibroblasts (HFF) cell viability, HSV-1 plaquing efficiency, and virion and cell lipoperoxidation. Cells or virions were incubated with the lipoperoxidators at 37°C for 2 h or incubated in atmospheric O2, and dose responses (half maximal cytotoxic and effective concentration [CC50 and EC50]) were evaluated by three or four parameter regression. The HSV-1 virions were slightly more sensitive to lipoperoxidators than were the cells (selectivity index [SI], 3.3 to 7.4). The effects of the lipophilic AMVN on both cell and virion viability directly correlated with the extent of membrane lipoperoxidation as evaluated by two different probes, C11-Bodipy and liperfluo. Moreover, the hydrophilic AAPH-induced virion inactivation at lower concentrations than did lipoperoxidation. Known lipoperoxidators inhibit infectivity via lipoperoxidation-independent mechanisms. Antioxidants protected against a loss of viral infectivity by less than 5-fold. Carrier bovine serum albumin (BSA) protected against both peroxidators to a similar extent when present together with the lipoperoxidating agents, suggesting that BSA quenches them as expected. Virions incubated in atmospheric oxidative conditions suffered losses of infectivity that were similar to those of chemically peroxidated virions, and they were protected by water soluble vitamin C and BSA with no evident lipoperoxidation, indicating predominant peroxidative damage to nonlipid virion components. Thus, lipoperoxidation is not a mechanism by which to specifically inhibit the infectivity of enveloped viruses, and the effects of known lipoperoxidators on virion infectivity are not solely mediated by lipoperoxidation. IMPORTANCE Small molecules that induce lipoperoxidation have been proposed repeatedly as potential antiviral drugs based on a presumed unique sensitivity of virions to this type of damage. Several small molecules that inactivate virions without affecting cells have been proposed to act primarily by inducing lipoperoxidation. However, the preferential sensitivity of virions to lipoperoxidators had not been experimentally evaluated. Using two of the best characterized small molecule lipoperoxidators, which are widely considered to be the prototypical water soluble and liposoluble lipoperoxidators, we show that lipoperoxidators have no preference for virions over cells. Moreover, they also inactivate virions by mechanisms other than the induction of lipoperoxidation. Therefore, the general induction of lipoperoxidation is not a path by which to develop antivirals. Moreover, molecules with specific antiviral activity which are not cytotoxic and have no preference to localize to virions over cells are unlikely to act primarily by inducing lipoperoxidation.
Collapse
|
21
|
Li X, Li Y, Fan S, Cao R, Li X, He X, Li W, Xu L, Cheng T, Li H, Zhong W. Discovery and Optimization of Quinoline Analogues as Novel Potent Antivirals against Enterovirus D68. J Med Chem 2022; 65:14792-14808. [PMID: 36254462 PMCID: PMC9661475 DOI: 10.1021/acs.jmedchem.2c01311] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
![]()
Enterovirus D68 (EV-D68)
is a nonpolio enterovirus that is mainly
transmitted through respiratory routes and poses a potential threat
for large-scale spread. EV-D68 infections mostly cause moderate to
severe respiratory diseases in children and potentially induce neurological
diseases. However, there are no specific antiviral drugs or vaccines
against EV-D68. Herein, through virtual screening and rational design,
a series of novel quinoline analogues as anti-EV-D68 agents targeting
VP1 were identified. Particularly, 19 exhibited potent
antiviral activity with an EC50 value ranging from 0.05
to 0.10 μM against various EV-D68 strains and showed inhibition
of viral replication verified by Western blot, immunofluorescence,
and plaque formation assay. Mechanistic studies indicated that the
anti-EV-D68 agents work mainly by interacting with VP1. The acceptable
bioavailability of 23.9% in rats and significant metabolic stability
in human liver microsome (Clint = 10.8 mL/min/kg, t1/2 = 148 min) indicated that compound 19 with a novel scaffold was worth further investigation.
Collapse
Affiliation(s)
- Xiaoyuan Li
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, P.R. China
| | - Yuexiang Li
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, P.R. China
| | - Shiyong Fan
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, P.R. China
| | - Ruiyuan Cao
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, P.R. China
| | - Xiaojia Li
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, P.R. China
| | - Xiaomeng He
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, P.R. China
| | - Wei Li
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, P.R. China
| | - Longfa Xu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiamen 361102, P.R. China
| | - Tong Cheng
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiamen 361102, P.R. China
| | - Honglin Li
- Shanghai Key Laboratory of New Drug Design, State Key Laboratory of Bioreactor Engineering, School of Pharmacy, East China University of Science & Technology, Shanghai 200237, P.R. China
| | - Wu Zhong
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, P.R. China
| |
Collapse
|
22
|
More-Adate P, Lokhande KB, Swamy KV, Nagar S, Baheti A. GC-MS profiling of Bauhinia variegata major phytoconstituents with computational identification of potential lead inhibitors of SARS-CoV-2 Mpro. Comput Biol Med 2022; 147:105679. [PMID: 35667152 PMCID: PMC9158327 DOI: 10.1016/j.compbiomed.2022.105679] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 05/26/2022] [Accepted: 05/30/2022] [Indexed: 01/18/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 was originally identified in Wuhan city of China in December 2019 and it spread rapidly throughout the globe, causing a threat to human life. Since targeted therapies are deficient, scientists all over the world have an opportunity to develop novel drug therapies to combat COVID-19. After the declaration of a global medical emergency, it was established that the Food and Drug Administration (FDA) could permit the use of emergency testing, treatments, and vaccines to decrease suffering, and loss of life, and restore the nation's health and security. The FDA has approved the use of remdesivir and its analogs as an antiviral medication, to treat COVID-19. The primary protease of SARS-CoV-2, which has the potential to regulate coronavirus proliferation, has been a viable target for the discovery of medicines against SARS-CoV-2. The present research deals with the in silico technique to screen phytocompounds from a traditional medicinal plant, Bauhinia variegata for potential inhibitors of the SARS-CoV-2 main protease. Dried leaves of the plant B. variegata were used to prepare aqueous and methanol extract and the constituents were analyzed using the GC-MS technique. A total of 57 compounds were retrieved from the aqueous and methanol extract analysis. Among these, three lead compounds (2,5 dimethyl 1-H Pyrrole, 2,3 diphenyl cyclopropyl methyl phenyl sulphoxide, and Benzonitrile m phenethyl) were shown to have the highest binding affinity (−5.719 to −5.580 kcal/mol) towards SARS-CoV-2 Mpro. The post MD simulation results also revealed the favorable confirmation and stability of the selected lead compounds with Mpro as per trajectory analysis. The Prime MM/GBSA binding free energy supports this finding, the top lead compound 2,3 diphenyl cyclopropyl methyl phenyl sulphoxide showed high binding free energy (−64.377 ± 5.24 kcal/mol) towards Mpro which reflects the binding stability of the molecule with Mpro. The binding free energy of the complexes was strongly influenced by His, Gln, and Glu residues. All of the molecules chosen are found to have strong pharmacokinetic characteristics and show drug-likeness properties. The lead compounds present acute toxicity (LD50) values ranging from 670 mg/kg to 2500 mg/kg; with toxicity classifications of 4 and 5 classes. Thus, these compounds could behave as probable lead candidates for treatment against SARS-CoV-2. However further in vitro and in vivo studies are required for the development of medication against SARS-CoV-2.
Collapse
|
23
|
Persistent Enterovirus Infection: Little Deletions, Long Infections. Vaccines (Basel) 2022; 10:vaccines10050770. [PMID: 35632526 PMCID: PMC9143164 DOI: 10.3390/vaccines10050770] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 05/04/2022] [Accepted: 05/10/2022] [Indexed: 01/27/2023] Open
Abstract
Enteroviruses have now been shown to persist in cell cultures and in vivo by a novel mechanism involving the deletion of varying amounts of the 5′ terminal genomic region termed domain I (also known as the cloverleaf). Molecular clones of coxsackievirus B3 (CVB3) genomes with 5′ terminal deletions (TD) of varying length allow the study of these mutant populations, which are able to replicate in the complete absence of wildtype virus genomes. The study of TD enteroviruses has revealed numerous significant differences from canonical enteroviral biology. The deletions appear and become the dominant population when an enterovirus replicates in quiescent cell populations, but can also occur if one of the cis-acting replication elements of the genome (CRE-2C) is artificially mutated in the element’s stem and loop structures. This review discusses how the TD genomes arise, how they interact with the host, and their effects on host biology.
Collapse
|
24
|
Wang J, Hu Y, Zheng M. Enterovirus A71 antivirals: Past, present, and future. Acta Pharm Sin B 2022; 12:1542-1566. [PMID: 35847514 PMCID: PMC9279511 DOI: 10.1016/j.apsb.2021.08.017] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 07/28/2021] [Accepted: 08/12/2021] [Indexed: 02/07/2023] Open
Abstract
Enterovirus A71 (EV-A71) is a significant human pathogen, especially in children. EV-A71 infection is one of the leading causes of hand, foot, and mouth diseases (HFMD), and can lead to neurological complications such as acute flaccid myelitis (AFM) in severe cases. Although three EV-A71 vaccines are available in China, they are not broadly protective and have reduced efficacy against emerging strains. There is currently no approved antiviral for EV-A71. Significant progress has been made in developing antivirals against EV-A71 by targeting both viral proteins and host factors. However, viral capsid inhibitors and protease inhibitors failed in clinical trials of human rhinovirus infection due to limited efficacy or side effects. This review discusses major discoveries in EV-A71 antiviral development, analyzes the advantages and limitations of each drug target, and highlights the knowledge gaps that need to be addressed to advance the field forward.
Collapse
Affiliation(s)
- Jun Wang
- Department of Pharmacology and Toxicology, College of Pharmacy, the University of Arizona, Tucson, AZ 85721, USA
| | - Yanmei Hu
- Department of Pharmacology and Toxicology, College of Pharmacy, the University of Arizona, Tucson, AZ 85721, USA
| | - Madeleine Zheng
- Department of Pharmacology and Toxicology, College of Pharmacy, the University of Arizona, Tucson, AZ 85721, USA
| |
Collapse
|
25
|
Antiviral effects of azithromycin: A narrative review. Biomed Pharmacother 2022; 147:112682. [PMID: 35131658 PMCID: PMC8813546 DOI: 10.1016/j.biopha.2022.112682] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/26/2022] [Accepted: 01/31/2022] [Indexed: 12/20/2022] Open
Abstract
Viral infections have a great impact on human health. The urgent need to find a cure against different viruses led us to investigations in a vast range of drugs. Azithromycin (AZT), classified as a macrolide, showed various effects on different known viruses such as severe acute respiratory syndrome coronavirus (SARS-CoV), Zika, Ebola, Enterovirus (EVs) and Rhinoviruses (RVs), and Influenza A previously; namely, these viruses, which caused global concerns, are considered as targets for AZT different actions. Due to AZT background in the treatment of known viral infections mentioned above (which is described in this study), in the early stages of COVID-19 (a new zoonotic disease caused by a novel coronavirus called severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)) development, AZT drew attention to itself due to its antiviral and immunomodulatory effects as a valuable candidate for COVID-19 treatment. AZT usage instructions for treating different viral infections have always been under observation, and COVID-19 is no exception. There are still debates about the use of AZT in COVID-19 treatment. However, eventually, novel researches convinced WHO to announce the discontinuation of AZT use (alone or in combination with hydroxychloroquine) in treating SARS-CoV-2 infection. This research aims to study the structure of all of the viruses mentioned above and the molecular and clinical effects of AZT against the virus.
Collapse
|
26
|
Targeting the Virus Capsid as a Tool to Fight RNA Viruses. Viruses 2022; 14:v14020174. [PMID: 35215767 PMCID: PMC8879806 DOI: 10.3390/v14020174] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/14/2022] [Accepted: 01/16/2022] [Indexed: 12/10/2022] Open
Abstract
Several strategies have been developed to fight viral infections, not only in humans but also in animals and plants. Some of them are based on the development of efficient vaccines, to target the virus by developed antibodies, others focus on finding antiviral compounds with activities that inhibit selected virus replication steps. Currently, there is an increasing number of antiviral drugs on the market; however, some have unpleasant side effects, are toxic to cells, or the viruses quickly develop resistance to them. As the current situation shows, the combination of multiple antiviral strategies or the combination of the use of various compounds within one strategy is very important. The most desirable are combinations of drugs that inhibit different steps in the virus life cycle. This is an important issue especially for RNA viruses, which replicate their genomes using error-prone RNA polymerases and rapidly develop mutants resistant to applied antiviral compounds. Here, we focus on compounds targeting viral structural capsid proteins, thereby inhibiting virus assembly or disassembly, virus binding to cellular receptors, or acting by inhibiting other virus replication mechanisms. This review is an update of existing papers on a similar topic, by focusing on the most recent advances in the rapidly evolving research of compounds targeting capsid proteins of RNA viruses.
Collapse
|
27
|
Compassionate-use pocapavir and immunoglobulin therapy for treatment of rituximab-associated enterovirus meningoencephalitis. J Neurovirol 2022; 28:329-334. [PMID: 34981437 DOI: 10.1007/s13365-021-01038-z] [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: 07/21/2021] [Revised: 11/12/2021] [Accepted: 12/09/2021] [Indexed: 10/19/2022]
Abstract
A 71-year-old woman previously on rituximab treatment for rheumatoid arthritis presented with 2 years of progressive neurologic symptoms. She was found to have persistent hypogammaglobulinemia and B cell depletion despite rituximab discontinuation a year prior. MRI revealed diffuse meningeal enhancement along the entire neuroaxis. LP showed a CSF lymphocytic pleocytosis, elevated protein, and presence of enterovirus by PCR. The patient was hospitalized several times for progressive clinical and radiologic decline, though she had transient improvements following treatment with immunoglobulin therapy. Her CSF remained positive for enterovirus PCR for at least 12 months. Though two brain biopsies were non-diagnostic, pan-Enterovirus was ultimately identified using a high-throughput next-generation sequencing technique. She was treated with compassionate-use pocapavir with clinical stabilization at 4-month follow-up; however, she expired 8 months later from a bacterial pneumonia.
Collapse
|
28
|
Nagafuchi S. Regulation of Viral Infection in Diabetes. BIOLOGY 2021; 10:biology10060529. [PMID: 34199246 PMCID: PMC8231782 DOI: 10.3390/biology10060529] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 06/11/2021] [Indexed: 11/16/2022]
Abstract
Though there is no 'Diabetes Virus', multiple agents such as mumps virus, rubella virus, influenza virus, type A hepatitis virus, enterovirus, rotavirus, cytomegalovirus, varicella-zoster virus, human herpesvirus 6, Epstein-Barr virus, and also SARS-CoV-2 have been reported to be associated to diabetes [...].
Collapse
Affiliation(s)
- Seiho Nagafuchi
- Department of Internal Medicine, Division of Metabolism and Endocrinology, Faculty of Medicine, Saga University, Saga 849-8501, Japan
| |
Collapse
|
29
|
Licochalcone A inhibits enterovirus A71 replication in vitro and in vivo. Antiviral Res 2021; 195:105091. [PMID: 34044060 DOI: 10.1016/j.antiviral.2021.105091] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 05/17/2021] [Accepted: 05/19/2021] [Indexed: 01/17/2023]
Abstract
Enterovirus A71 (EV-A71) is one of the main causative agents of hand-foot-mouth disease (HFMD) and causes serious neurological complications. However, no effective therapy is currently available for treating these infections. Therefore, effective drugs to prevent and treat EV-A71 infections are urgently needed. Here, we demonstrated that treatment with Licochalcone A (LCA) significantly inhibited EV-A71 replication in a dose-dependent manner, with an EC50 of 9.30 μM in RD cells and 5.73 μM in Vero cells. The preliminary results on the inhibition mechanism showed that LCA exerted antiviral effects by interfering with the early step of viral replication. We further demonstrated that LCA showed potent antiviral activity against many enteroviruses, including EV-A71 (strain C4), EV-A71 (strain H), and coxsackievirus A16 (CV-A16). Furthermore, LCA could effectively prevent the clinical symptoms and death of virus infected mice and decreased viral load in EV-A71-infected mice. Taken together, our studies showed for the first time, that LCA is a promising EV-A71 inhibitor and provide important information for the clinical development of LCA as a potential new anti-EV-A71 agent.
Collapse
|
30
|
Anasir MI, Zarif F, Poh CL. Antivirals blocking entry of enteroviruses and therapeutic potential. J Biomed Sci 2021; 28:10. [PMID: 33451326 PMCID: PMC7811253 DOI: 10.1186/s12929-021-00708-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Accepted: 01/08/2021] [Indexed: 01/26/2023] Open
Abstract
Viruses from the genus Enterovirus (EV) of the Picornaviridae family are known to cause diseases such as hand foot and mouth disease (HFMD), respiratory diseases, encephalitis and myocarditis. The capsid of EV is an attractive target for the development of direct-acting small molecules that can interfere with viral entry. Some of the capsid binders have been evaluated in clinical trials but the majority have failed due to insufficient efficacy or unacceptable off-target effects. Furthermore, most of the capsid binders exhibited a low barrier to resistance. Alternatively, host-targeting inhibitors such as peptides derived from the capsid of EV that can recognize cellular receptors have been identified. However, the majority of these peptides displayed low anti-EV potency (µM range) as compared to the potency of small molecule compounds (nM range). Nonetheless, the development of anti-EV peptides is warranted as they may complement the small-molecules in a drug combination strategy to treat EVs. Lastly, structure-based approach to design antiviral peptides should be utilized to unearth potent anti-EV peptides.
Collapse
Affiliation(s)
- Mohd Ishtiaq Anasir
- Centre for Virus and Vaccine Research, Sunway University, 5, Jalan Universiti, 47500, Bandar Sunway, Selangor, Malaysia
| | - Faisal Zarif
- Centre for Virus and Vaccine Research, Sunway University, 5, Jalan Universiti, 47500, Bandar Sunway, Selangor, Malaysia
| | - Chit Laa Poh
- Centre for Virus and Vaccine Research, Sunway University, 5, Jalan Universiti, 47500, Bandar Sunway, Selangor, Malaysia.
| |
Collapse
|
31
|
Walther C, Döring K, Schmidtke M. Comparative in vitro analysis of inhibition of rhinovirus and influenza virus replication by mucoactive secretolytic agents and plant extracts. BMC Complement Med Ther 2020; 20:380. [PMID: 33357221 PMCID: PMC7757078 DOI: 10.1186/s12906-020-03173-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 12/02/2020] [Indexed: 01/23/2023] Open
Abstract
Background Rhinoviruses and influenza viruses cause millions of acute respiratory infections annually. Symptoms of mild acute respiratory infections are commonly treated with over-the-counter products like ambroxol, bromhexine, and N-acetyl cysteine, as well as of thyme and pelargonium extracts today. Because the direct antiviral activity of these over-the-counter products has not been studied in a systematic way, the current study aimed to compare their inhibitory effect against rhinovirus and influenza virus replication in an in vitro setting. Methods The cytotoxicity of ambroxol, bromhexine, and N-acetyl cysteine, as well as of thyme and pelargonium extracts was analyzed in Madin Darby canine kidney (MDCK) and HeLa Ohio cells. The antiviral effect of these over-the-counter products was compared by analyzing the dose-dependent inhibition (i) of rhinovirus A2- and B14-induced cytopathic effect in HeLa Ohio cells and (ii) of influenza virus A/Hong Kong/68 (subtype H3N2)- and A/Jena/8178/09 (subtype H1N1, pandemic)-induced cytopathic effect in MDCK cells at non-cytotoxic concentrations. To get insights into the mechanism of action of pelargonium extract against influenza virus, we performed time-of-addition assays as well as hemagglutination and neuraminidase inhibition assays. Results N-acetyl cysteine, thyme and pelargonium extract showed no or only marginal cytotoxicity in MDCK and HeLa Ohio cells in the tested concentration range. The 50% cytotoxic concentration of ambroxol and bromhexine was 51.85 and 61.24 μM, respectively. No anti-rhinoviral activity was detected at non-cytotoxic concentrations in this in vitro study setting. Ambroxol, bromhexine, and N-acetyl cysteine inhibited the influenza virus-induced cytopathic effect in MDCK cells no or less than 50%. In contrast, a dose-dependent anti-influenza virus activity of thyme and pelargonium extracts was demonstrated. The time-of addition assays revealed an inhibition of early and late steps of influenza virus replication by pelargonium extract whereas zanamivir acted on late steps only. The proven block of viral neuraminidase activity might explain the inhibition of influenza virus replication when added after viral adsorption. Conclusion The study results indicate a distinct inhibition of influenza A virus replication by thyme and pelargonium extract which might contribute to the beneficial effects of these plant extracts on acute respiratory infections symptoms.
Collapse
Affiliation(s)
- Christin Walther
- Department Medical Microbiology, Section Experimental Virology, Jena University Hospital, Hans-Knöll-Str. 2, D-07745, Jena, Germany
| | - Kristin Döring
- Department Medical Microbiology, Section Experimental Virology, Jena University Hospital, Hans-Knöll-Str. 2, D-07745, Jena, Germany
| | - Michaela Schmidtke
- Department Medical Microbiology, Section Experimental Virology, Jena University Hospital, Hans-Knöll-Str. 2, D-07745, Jena, Germany.
| |
Collapse
|
32
|
Hwu JR, Panja A, Jayakumar S, Tsay SC, Tan KT, Huang WC, Hu YC, Leyssen P, Neyts J. Enterovirus Inhibition by Hinged Aromatic Compounds with Polynuclei. Molecules 2020; 25:molecules25173821. [PMID: 32842645 PMCID: PMC7503712 DOI: 10.3390/molecules25173821] [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: 07/30/2020] [Revised: 08/17/2020] [Accepted: 08/19/2020] [Indexed: 11/16/2022] Open
Abstract
The modern world has no available drugs for the treatment of enteroviruses (EV), which affect millions of people worldwide each year. The EV71 is a major causative disease for hand, foot, and mouth disease; sometimes it is associated with severe central nervous system diseases. Treatment for enteroviral infection is mainly supportive; treatment for aseptic meningitis caused by enteroviruses is also generally symptomatic. Upon the urgent request of new anti-enterovirus drugs, a series of hinged aromatic compounds with polynulei were synthesized through two different chemical pathways. Among these morpholine–furan/thiophene/pyrrole–benzene–pyrazole conjugates, three new agents exhibited inhibitory activity with EC50 = 2.29–6.16 μM toward EV71 strain BrCr in RD cells. Their selectivity index values were reached as high as 33.4. Their structure–activity relationship was deduced that a thiophene derivative with morpholine and trifluorobenzene rings showed the greatest antiviral activity, with EC50 = 2.29 μM.
Collapse
Affiliation(s)
- Jih Ru Hwu
- Department of Chemistry, National Tsing Hua University, Hsinchu 300, Taiwan; (A.P.); (S.J.); (S.-C.T.); (K.-T.T.); (W.-C.H.)
- Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 300, Taiwan;
- Department of Chemistry, National Central University, Jhongli City, Taoyuan 320, Taiwan
- Correspondence: (J.R.H.); (J.N.)
| | - Avijit Panja
- Department of Chemistry, National Tsing Hua University, Hsinchu 300, Taiwan; (A.P.); (S.J.); (S.-C.T.); (K.-T.T.); (W.-C.H.)
| | - Srinivasan Jayakumar
- Department of Chemistry, National Tsing Hua University, Hsinchu 300, Taiwan; (A.P.); (S.J.); (S.-C.T.); (K.-T.T.); (W.-C.H.)
| | - Shwu-Chen Tsay
- Department of Chemistry, National Tsing Hua University, Hsinchu 300, Taiwan; (A.P.); (S.J.); (S.-C.T.); (K.-T.T.); (W.-C.H.)
- Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 300, Taiwan;
- Department of Chemistry, National Central University, Jhongli City, Taoyuan 320, Taiwan
| | - Kui-Thong Tan
- Department of Chemistry, National Tsing Hua University, Hsinchu 300, Taiwan; (A.P.); (S.J.); (S.-C.T.); (K.-T.T.); (W.-C.H.)
- Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 300, Taiwan;
| | - Wen-Chieh Huang
- Department of Chemistry, National Tsing Hua University, Hsinchu 300, Taiwan; (A.P.); (S.J.); (S.-C.T.); (K.-T.T.); (W.-C.H.)
- Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 300, Taiwan;
| | - Yu-Chen Hu
- Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 300, Taiwan;
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Pieter Leyssen
- Rega Institute for Medical Research, Katholieke Universiteit Leuven, Minderbroedersstraat 10, Leuven B-3000, Belgium;
| | - Johan Neyts
- Rega Institute for Medical Research, Katholieke Universiteit Leuven, Minderbroedersstraat 10, Leuven B-3000, Belgium;
- Correspondence: (J.R.H.); (J.N.)
| |
Collapse
|
33
|
Schlicksup CJ, Zlotnick A. Viral structural proteins as targets for antivirals. Curr Opin Virol 2020; 45:43-50. [PMID: 32777753 DOI: 10.1016/j.coviro.2020.07.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 06/30/2020] [Accepted: 07/01/2020] [Indexed: 12/29/2022]
Abstract
Viral structural proteins are emerging as effective targets for new antivirals. In a viral lifecycle, the capsid must assemble, disassemble, and respond to host proteins, all at the right time and place. These reactions work within a narrow range of conditions, making them susceptible to small molecule interference. In at least three specific viruses, this approach has had met with preliminary success. In rhinovirus and poliovirus, compounds like pleconaril bind capsid and block RNA release. Bevirimat binds to Gag protein in HIV, inhibiting maturation. In Hepatitis B virus, core protein allosteric modulators (CpAMs) promote spontaneous assembly of capsid protein leading to empty and aberrant particles. Despite the biological diversity between viruses and the chemical diversity between antiviral molecules, we observe common features in these antivirals' mechanisms of action. These approaches work by stabilizing protein-protein interactions.
Collapse
Affiliation(s)
- Christopher John Schlicksup
- Molecular and Cellular Biology Department, Indiana University-Bloomington, Bloomington, IN 47401, United States
| | - Adam Zlotnick
- Molecular and Cellular Biology Department, Indiana University-Bloomington, Bloomington, IN 47401, United States.
| |
Collapse
|
34
|
Abstract
Enterovirus D68 (EV-D68) is an RNA virus that causes respiratory illnesses mainly in children. In severe cases, it can lead to neurological complications such as acute flaccid myelitis (AFM). EV-D68 belongs to the enterovirus genera of the Picornaviridae family, which also includes many other significant human pathogens such as poliovirus, enterovirus A71, and rhinovirus. There are currently no vaccines or antivirals against EV-D68. In this review, we present the current understanding of the link between EV-D68 and AFM, the mechanism of viral replication, and recent progress in developing EV-D68 antivirals by targeting various viral proteins and host factors that are essential for viral replication. The future directions of EV-D68 antiviral drug discovery and the criteria for drugs to reach clinical trials are also discussed.
Collapse
Affiliation(s)
- Yanmei Hu
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, USA, 85721
| | - Rami Musharrafieh
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, USA, 85721
| | - Madeleine Zheng
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, USA, 85721
| | - Jun Wang
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, USA, 85721
| |
Collapse
|
35
|
Musharrafieh R, Kitamura N, Hu Y, Wang J. Development of broad-spectrum enterovirus antivirals based on quinoline scaffold. Bioorg Chem 2020; 101:103981. [PMID: 32559580 DOI: 10.1016/j.bioorg.2020.103981] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 04/09/2020] [Accepted: 05/29/2020] [Indexed: 12/11/2022]
Abstract
Non-polio enteroviruses such as enterovirus A71 (EV-A71), EV-D68, and coxsackievirus B3 (CVB3) are significant human pathogens with disease manifestations ranging from mild flu-like symptoms to more severe encephalitis, myocarditis, acute flaccid paralysis/myelitis, and even death. There is currently no effective antivirals to prevent or treat non-polio enterovirus infection. In this study, we report our progress in developing potent and broad-spectrum antivirals against these non-polio enteroviruses. Starting from our previously developed lead compounds that had potent antiviral activity against EV-D68, we synthesized 43 analogs and profiled their broad-spectrum antiviral activity against additional EV-D68, EV-A71, and CVB3 viruses. Promising candidates were also selected for mouse microsomal stability test to prioritize lead compounds for future in vivo mouse model studies. Collectively, this multi-parameter optimization process revealed a promising lead compound 6aw that showed single-digit to submicromolar EC50 values against two EV-D68 strains (US/KY and US/MO), two EV-A71 strains (Tainan and US/AK), and one CVB3 strain, with a high selectivity index. Encouragingly, 6aw was stable in mouse microsomes with a half-life of 114.7 min. Overall, 6aw represents one of the most potent broad-spectrum antiviral against non-polio enteroviruses, rendering it a promising lead candidate for non-polio enteroviruses with translational potential.
Collapse
Affiliation(s)
- Rami Musharrafieh
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, AZ 85721, United States; Department of Chemistry and Biochemistry, The University of Arizona, Tucson, AZ 85721, United States
| | - Naoya Kitamura
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, AZ 85721, United States
| | - Yanmei Hu
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, AZ 85721, United States
| | - Jun Wang
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, AZ 85721, United States.
| |
Collapse
|
36
|
Liu Z, Xia S, Wang X, Lan Q, Li P, Xu W, Wang Q, Lu L, Jiang S. Sodium Copper Chlorophyllin Is Highly Effective against Enterovirus (EV) A71 Infection by Blocking Its Entry into the Host Cell. ACS Infect Dis 2020; 6:882-890. [PMID: 32233455 DOI: 10.1021/acsinfecdis.0c00096] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Human enteroviruses (HEVs) pose an ongoing threat to global public health. Particularly, enterovirus-A71 (EV-A71), the main pathogen causing hand-foot-and-mouth disease (HFMD), has caused ongoing outbreaks globally in recent years associated with severe neurological manifestations and several deaths. Currently, no effective antivirals are available for the prevention or treatment of EV-A71 infection. In this study, we found that sodium copper chlorophyllin (CHL), a health food additive and an over-the-counter anticancer medicine or treatment to reduce the odor of urine or feces, exhibited potent inhibitory activity against infection by divergent EV-A71 and coxsackievirus-A16 (CV-A16) isolates at a low micromolar concentration with excellent safety. The antiviral activity of each was confirmed by colorimetric viral infection and qRT-PCR assays. A series of mechanistic studies showed that CHL did not target the host cell but blocked the entry of EV-A71 and CV-A16 into the host cell at the postattachment stage. In the mouse model, CHL could significantly reduce the viral titer in the lungs and muscles. Since CHL has been used in clinics for many years with excellent safety, it has the potential to be further developed into a prophylactic or therapeutic to prevent or treat HFMD caused by EV-A71 or CV-A16 infection.
Collapse
Affiliation(s)
- Zezhong Liu
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Shuai Xia
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Xinling Wang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Qiaoshuai Lan
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Peiyu Li
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Wei Xu
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Qian Wang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Lu Lu
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Shibo Jiang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, New York 10065, United States
| |
Collapse
|
37
|
Egorova A, Kazakova E, Jahn B, Ekins S, Makarov V, Schmidtke M. Novel pleconaril derivatives: Influence of substituents in the isoxazole and phenyl rings on the antiviral activity against enteroviruses. Eur J Med Chem 2019; 188:112007. [PMID: 31881489 DOI: 10.1016/j.ejmech.2019.112007] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 12/20/2019] [Accepted: 12/20/2019] [Indexed: 01/21/2023]
Abstract
Today, there are no medicines to treat enterovirus and rhinovirus infections. In the present study, a series of novel pleconaril derivatives with substitutions in the isoxazole and phenyl rings was synthesized and evaluated for their antiviral activity against a panel of pleconaril-sensitive and -resistant enteroviruses. Studies of the structure-activity relationship demonstrate the crucial role of the N,N-dimethylcarbamoyl group in the isoxazole ring for antiviral activity against pleconaril-resistant viruses. In addition, one or two substituents in the phenyl ring directly impact on the spectrum of antienteroviral activity. The 3-(3-methyl-4-(3-(3-N,N-dimethylcarbamoyl-isoxazol-5-yl)propoxy)phenyl)-5-trifluoromethyl-1,2,4-oxadiazole 10g was among the compounds exhibiting the strongest activity against pleconaril-resistant as well as pleconaril-susceptible enteroviruses with IC50 values from 0.02 to 5.25 μM in this series. Compound 10g demonstrated markedly less CYP3A4 induction than pleconaril, was non-mutagenic, and was bioavailable after intragastric administration in mice. These results highlight compound 10g as a promising potential candidate as a broad spectrum enterovirus and rhinovirus inhibitor for further preclinical investigations.
Collapse
Affiliation(s)
- Anna Egorova
- Bach Institute of Biochemistry, Research Center of Biotechnology of the Russian Academy of Sciences, 33-2 Leninsky Prospect, Moscow, 119071, Russia
| | - Elena Kazakova
- Bach Institute of Biochemistry, Research Center of Biotechnology of the Russian Academy of Sciences, 33-2 Leninsky Prospect, Moscow, 119071, Russia
| | - Birgit Jahn
- Jena University Hospital, Department of Medical Microbiology, Section Experimental Virology, Hans-Knöll-Strasse 2, Jena, 07745, Germany
| | - Sean Ekins
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, NC27606, USA
| | - Vadim Makarov
- Bach Institute of Biochemistry, Research Center of Biotechnology of the Russian Academy of Sciences, 33-2 Leninsky Prospect, Moscow, 119071, Russia.
| | - Michaela Schmidtke
- Jena University Hospital, Department of Medical Microbiology, Section Experimental Virology, Hans-Knöll-Strasse 2, Jena, 07745, Germany.
| |
Collapse
|
38
|
Ma C, Hu Y, Zhang J, Musharrafieh R, Wang J. A Novel Capsid Binding Inhibitor Displays Potent Antiviral Activity against Enterovirus D68. ACS Infect Dis 2019; 5:1952-1962. [PMID: 31532189 DOI: 10.1021/acsinfecdis.9b00284] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Enterovirus D68 (EV-D68) is a respiratory viral pathogen that primarily infects children under the age of 8. Although EV-D68 infection typically leads to moderate to severe respiratory illnesses, recent years have seen increasing cases of EV-D68 triggered neurological complications such as acute flaccid myelitis (AFM). There is currently no vaccine or antiviral available for EV-D68; we therefore aimed to develop potent and specific small molecule antivirals against EV-D68. In this study, we report our discovery of a viral capsid inhibitor R856932 that inhibits multiple contemporary EV-D68 strains with single-digit to submicromolar efficacy. Mechanistic studies have shown that the tetrazole compound R856932 binds to the hydrophobic pocket of viral capsid protein VP1, thereby preventing viral uncoating and release of viral genome in the infected cells. The mechanism of action of R856932 was confirmed by time-of-addition, Western blot, RT-qPCR, viral heat inactivation, serial viral passage, and reverse genetics experiments. A single mutation located at VP1, A129V, confers resistance against R856932. However, a recombination virus encoding VP1-A129V appeared to have compromised fitness of replication compared to the wild-type EV-D68 virus as shown by the competition growth assay. Overall, the hit compound identified in this study, R856932, represents a promising starting point with a confirmed mechanism of action that can be further developed into EV-D68 antivirals.
Collapse
Affiliation(s)
- Chunlong Ma
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, 1657 East Helen Street, Tucson, Arizona 85721, United States
| | - Yanmei Hu
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, 1657 East Helen Street, Tucson, Arizona 85721, United States
| | - Jiantao Zhang
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, 1657 East Helen Street, Tucson, Arizona 85721, United States
| | - Rami Musharrafieh
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, 1657 East Helen Street, Tucson, Arizona 85721, United States
| | - Jun Wang
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, 1657 East Helen Street, Tucson, Arizona 85721, United States
| |
Collapse
|