1
|
Cho WK, Choi HJ, Ma JY. Antiviral Activity of Berbamine Against Influenza A Virus Infection. Int J Mol Sci 2025; 26:2819. [PMID: 40141460 PMCID: PMC11942913 DOI: 10.3390/ijms26062819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 03/17/2025] [Accepted: 03/19/2025] [Indexed: 03/28/2025] Open
Abstract
Berbamine (BBM) is a bibenzyl isoquinoline present in the traditional Chinese herbal medicine Berberis amurensisis Rupr. The present study demonstrates that BBM exerts strong antiviral efficacy against influenza A virus (IAV) infection. We examined the anti-IAV effect of BBM using green fluorescent protein (GFP)-expressing influenza A and H1N1 IAV. The fluorescence microscopy, fluorescence-activated cell sorting analysis, and plaque assay showed that BBM significantly hinders IAV infection. The immunofluorescence analysis confirmed the anti-influenza activity of BBM. From the time-of-addition and hemagglutination inhibition results, it is elucidated that the antiviral effect of BBM is closely related to its inhibitory effect against viral binding and entry at an early infection stage. Our findings imply that BBM has the potential to be developed as a potent antiviral drug against influenza viral infection.
Collapse
Affiliation(s)
- Won-Kyung Cho
- Korean Medicine (KM) Application Center, Korea Institute of Oriental Medicine, 70 Chemdanro, Dong-gu, Daegu 41062, Republic of Korea;
| | | | - Jin Yeul Ma
- Korean Medicine (KM) Application Center, Korea Institute of Oriental Medicine, 70 Chemdanro, Dong-gu, Daegu 41062, Republic of Korea;
| |
Collapse
|
2
|
Wang S, Zhang TH, Hu M, Tang K, Sheng L, Hong M, Chen D, Chen L, Shi Y, Feng J, Qian J, Sun L, Ding K, Sun R, Du Y. Deep mutational scanning of influenza A virus neuraminidase facilitates the identification of drug resistance mutations in vivo. mSystems 2023; 8:e0067023. [PMID: 37772870 PMCID: PMC10654105 DOI: 10.1128/msystems.00670-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 08/09/2023] [Indexed: 09/30/2023] Open
Abstract
IMPORTANCE NA is a crucial surface antigen and drug target of influenza A virus. A comprehensive understanding of NA's mutational effect and drug resistance profiles in vivo is essential for comprehending the evolutionary constraints and making informed choices regarding drug selection to combat resistance in clinical settings. In the current study, we established an efficient deep mutational screening system in mouse lung tissues and systematically evaluated the fitness effect and drug resistance to three neuraminidase inhibitors of NA single-nucleotide mutations. The fitness of NA mutants is generally correlated with a natural mutation in the database. The fitness of NA mutants is influenced by biophysical factors such as protein stability, complex formation, and the immune response triggered by viral infection. In addition to confirming previously reported drug-resistant mutations, novel mutations were identified. Interestingly, we identified an allosteric drug-resistance mutation that is not located within the drug-binding pocket but potentially affects drug binding by interfering with NA tetramerization. The dual assessments performed in this study provide a more accurate assessment of the evolutionary potential of drug-resistant mutations and offer guidance for the rational selection of antiviral drugs.
Collapse
Affiliation(s)
- Sihan Wang
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Tian-hao Zhang
- Molecular Biology Institute, University of California, Los Angeles, California, USA
| | - Menglong Hu
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Kejun Tang
- Department of Surgery, Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Li Sheng
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, California, USA
- School of Biomedical Sciences, LKS Faculty of Medicine, The Hong Kong University, Hong Kong, China
| | - Mengying Hong
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Dongdong Chen
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Liubo Chen
- Department of Medical Oncology, The Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China
| | - Yuan Shi
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, California, USA
| | - Jun Feng
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, California, USA
| | - Jing Qian
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Lifeng Sun
- Department of Colorectal Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Kefeng Ding
- Department of Colorectal Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Ren Sun
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, California, USA
- Center for Infectious Disease Research, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
| | - Yushen Du
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
3
|
Flynn JM, Huang QYJ, Zvornicanin SN, Schneider-Nachum G, Shaqra AM, Yilmaz NK, Moquin SA, Dovala D, Schiffer CA, Bolon DN. Systematic Analyses of the Resistance Potential of Drugs Targeting SARS-CoV-2 Main Protease. ACS Infect Dis 2023; 9:1372-1386. [PMID: 37390404 PMCID: PMC11161032 DOI: 10.1021/acsinfecdis.3c00125] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/02/2023]
Abstract
Drugs that target the main protease (Mpro) of SARS-CoV-2 are effective therapeutics that have entered clinical use. Wide-scale use of these drugs will apply selection pressure for the evolution of resistance mutations. To understand resistance potential in Mpro, we performed comprehensive surveys of amino acid changes that can cause resistance to nirmatrelvir (Pfizer), and ensitrelvir (Xocova) in a yeast screen. We identified 142 resistance mutations for nirmatrelvir and 177 for ensitrelvir, many of which have not been previously reported. Ninety-nine mutations caused apparent resistance to both inhibitors, suggesting likelihood for the evolution of cross-resistance. The mutation with the strongest drug resistance score against nirmatrelvir in our study (E166V) was the most impactful resistance mutation recently reported in multiple viral passaging studies. Many mutations that exhibited inhibitor-specific resistance were consistent with the distinct interactions of each inhibitor in the substrate binding site. In addition, mutants with strong drug resistance scores tended to have reduced function. Our results indicate that strong pressure from nirmatrelvir or ensitrelvir will select for multiple distinct-resistant lineages that will include both primary resistance mutations that weaken interactions with drug while decreasing enzyme function and compensatory mutations that increase enzyme activity. The comprehensive identification of resistance mutations enables the design of inhibitors with reduced potential of developing resistance and aids in the surveillance of drug resistance in circulating viral populations.
Collapse
Affiliation(s)
- Julia M. Flynn
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Qiu Yu J. Huang
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Sarah N. Zvornicanin
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Gila Schneider-Nachum
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Ala M. Shaqra
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Nese Kurt Yilmaz
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | | | - Dustin Dovala
- Novartis Institute for Biomedical Research, Emeryville, CA 94608, USA
| | - Celia A. Schiffer
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Daniel N.A. Bolon
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| |
Collapse
|
4
|
Prevalence and mechanisms of evolutionary contingency in human influenza H3N2 neuraminidase. Nat Commun 2022; 13:6443. [PMID: 36307418 PMCID: PMC9616408 DOI: 10.1038/s41467-022-34060-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 10/12/2022] [Indexed: 12/25/2022] Open
Abstract
Neuraminidase (NA) of human influenza H3N2 virus has evolved rapidly and been accumulating mutations for more than half-century. However, biophysical constraints that govern the evolutionary trajectories of NA remain largely elusive. Here, we show that among 70 natural mutations that are present in the NA of a recent human H3N2 strain, >10% are deleterious for an ancestral strain. By mapping the permissive mutations using combinatorial mutagenesis and next-generation sequencing, an extensive epistatic network is revealed. Biophysical and structural analyses further demonstrate that certain epistatic interactions can be explained by non-additive stability effect, which in turn modulates membrane trafficking and enzymatic activity of NA. Additionally, our results suggest that other biophysical mechanisms also contribute to epistasis in NA evolution. Overall, these findings not only provide mechanistic insights into the evolution of human influenza NA and elucidate its sequence-structure-function relationship, but also have important implications for the development of next-generation influenza vaccines.
Collapse
|
5
|
Cho WK, Lee MM, Ma JY. Antiviral Effect of Isoquercitrin against Influenza A Viral Infection via Modulating Hemagglutinin and Neuraminidase. Int J Mol Sci 2022; 23:13112. [PMID: 36361900 PMCID: PMC9653704 DOI: 10.3390/ijms232113112] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 10/13/2022] [Accepted: 10/27/2022] [Indexed: 07/30/2023] Open
Abstract
Isoquercitrin (IQC) is a component abundantly present in many plants and is known to have an anti-viral effect against various viruses. In this study, we demonstrate that IQC exhibits strong anti-influenza A virus infection, and its effect is closely related to the suppression of hemagglutinin (HA) and neuraminidase (NA) activities. We used green fluorescent protein-tagged Influenza A/PR/8/34 (H1N1), A/PR/8/34 (H1N1), and HBPV-VR-32 (H3N2) to evaluate the anti-IAV effect of IQC. The fluorescence microscopy and fluorescence-activated cell sorting analysis showed that IQC significantly decreases the levels of GFP expressed by IAV infection, dose-dependently. Consistent with that, IQC inhibited cytopathic effects by H1N1 or H3N2 IAV infection. Immunofluorescence analysis confirmed that IQC represses the IAV protein expression. Time-of-addition assay showed that IQC inhibits viral attachment and entry and exerts a strong virucidal effect during IAV infection. Hemagglutination assay confirmed that IQC affects IAV HA. Further, IQC potently reduced the NA activities of H1N1 and H3N2 IAV. Collectively, IQC prevents IAV infection at multi-stages via virucidal effects, inhibiting attachment, entry and viral release. Our results indicate that IQC could be developed as a potent antiviral drug to protect against influenza viral infection.
Collapse
Affiliation(s)
- Won-Kyung Cho
- Correspondence: (W.-K.C.); (J.Y.M.); Tel.: +82-53-940-3870 (W.-K.C.); +82-53-940-3812 (J.Y.M.)
| | | | - Jin Yeul Ma
- Correspondence: (W.-K.C.); (J.Y.M.); Tel.: +82-53-940-3870 (W.-K.C.); +82-53-940-3812 (J.Y.M.)
| |
Collapse
|
6
|
Predicting Permissive Mutations That Improve the Fitness of A(H1N1)pdm09 Viruses Bearing the H275Y Neuraminidase Substitution. J Virol 2022; 96:e0091822. [PMID: 35867563 PMCID: PMC9364793 DOI: 10.1128/jvi.00918-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Oseltamivir-resistant influenza viruses arise due to amino acid mutations in key residues of the viral neuraminidase (NA). These changes often come at a fitness cost; however, it is known that permissive mutations in the viral NA can overcome this cost. This result was observed in former seasonal A(H1N1) viruses in 2007 which expressed the H275Y substitution (N1 numbering) with no apparent fitness cost and lead to widespread oseltamivir resistance. Therefore, this study aims to predict permissive mutations that may similarly enable fit H275Y variants to arise in currently circulating A(H1N1)pdm09 viruses. The first approach in this study utilized in silico analyses to predict potentially permissive mutations. The second approach involved the generation of a virus library which encompassed all possible NA mutations while keeping H275Y fixed. Fit variants were then selected by serially passaging the virus library either through ferrets by transmission or passaging once in vitro. The fitness impact of selected substitutions was further evaluated experimentally. The computational approach predicted three candidate permissive NA mutations which, in combination with each other, restored the replicative fitness of an H275Y variant. The second approach identified a stringent bottleneck during transmission between ferrets; however, three further substitutions were identified which may improve transmissibility. A comparison of fit H275Y variants in vitro and in experimentally infected animals showed a statistically significant correlation in the variants that were positively selected. Overall, this study provides valuable tools and insights into potential permissive mutations that may facilitate the emergence of a fit H275Y A(H1N1)pdm09 variant. IMPORTANCE Oseltamivir (Tamiflu) is the most widely used antiviral for the treatment of influenza infections. Therefore, resistance to oseltamivir is a public health concern. This study is important as it explores the different evolutionary pathways available to current circulating influenza viruses that may lead to widespread oseltamivir resistance. Specifically, this study develops valuable experimental and computational tools to evaluate the fitness landscape of circulating A(H1N1)pmd09 influenza viruses bearing the H275Y mutation. The H275Y substitution is most commonly reported to confer oseltamivir resistance but also leads to loss of virus replication and transmission fitness, which limits its spread. However, it is known from previous influenza seasons that influenza viruses can evolve to overcome this loss of fitness. Therefore, this study aims to prospectively predict how contemporary A(H1N1)pmd09 influenza viruses may evolve to overcome the fitness cost of bearing the H275Y NA substitution, which could result in widespread oseltamivir resistance.
Collapse
|
7
|
Identification of a permissive secondary mutation that restores the enzymatic activity of oseltamivir resistance mutation H275Y. J Virol 2022; 96:e0198221. [PMID: 35045267 DOI: 10.1128/jvi.01982-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Many oseltamivir resistance mutations exhibit fitness defects in the absence of drug pressure that hinders their propagation in hosts. Secondary permissive mutations can rescue fitness defects and facilitate the segregation of resistance mutations in viral populations. Previous studies have identified a panel of permissive or compensatory mutations in neuraminidase (NA) that restore the growth defect of the predominant oseltamivir resistance mutation (H275Y) in H1N1 influenza A. In prior work, we identified a hyperactive mutation (Y276F) that increased NA activity by approximately 70%. While Y276F had not been previously identified as a permissive mutation, we hypothesized that Y276F may counteract the defects caused by H275Y by buffering its reduced NA expression and enzyme activity. In this study we measured the relative fitness, NA activity, and surface expression, as well as sensitivity to oseltamivir, for several oseltamivir resistance mutations including H275Y in the wildtype or Y276F genetic background. Our results demonstrate that Y276F selectively rescues the fitness defect of H275Y by restoring its NA surface expression and enzymatic activity, elucidating the local compensatory structural impacts of Y276F on the adjacent H275Y. Importance The potential for influenza A virus (IAV) to cause pandemics makes understanding evolutionary mechanisms that impact drug resistance critical for developing surveillance and treatment strategies. Oseltamivir is the most widely used therapeutic strategy to treat IAV infections, but mutations in IAV can lead to drug resistance. The main oseltamivir resistance mutation, H275Y, occurs in the neuraminidase (NA) protein of IAV and reduces drug binding as well as NA function. Here, we identify a new helper mutation, Y276F that can rescue the functional defects of H275Y and contribute to the evolution of drug resistance in IAV.
Collapse
|
8
|
Wang Y, Lei R, Nourmohammad A, Wu NC. Antigenic evolution of human influenza H3N2 neuraminidase is constrained by charge balancing. eLife 2021; 10:e72516. [PMID: 34878407 PMCID: PMC8683081 DOI: 10.7554/elife.72516] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 12/07/2021] [Indexed: 11/13/2022] Open
Abstract
As one of the main influenza antigens, neuraminidase (NA) in H3N2 virus has evolved extensively for more than 50 years due to continuous immune pressure. While NA has recently emerged as an effective vaccine target, biophysical constraints on the antigenic evolution of NA remain largely elusive. Here, we apply combinatorial mutagenesis and next-generation sequencing to characterize the local fitness landscape in an antigenic region of NA in six different human H3N2 strains that were isolated around 10 years apart. The local fitness landscape correlates well among strains and the pairwise epistasis is highly conserved. Our analysis further demonstrates that local net charge governs the pairwise epistasis in this antigenic region. In addition, we show that residue coevolution in this antigenic region is correlated with the pairwise epistasis between charge states. Overall, this study demonstrates the importance of quantifying epistasis and the underlying biophysical constraint for building a model of influenza evolution.
Collapse
Affiliation(s)
- Yiquan Wang
- Department of Biochemistry, University of Illinois at Urbana-ChampaignUrbanaUnited States
| | - Ruipeng Lei
- Department of Biochemistry, University of Illinois at Urbana-ChampaignUrbanaUnited States
| | - Armita Nourmohammad
- Department of Physics, University of WashingtonSeattleUnited States
- Max Planck Institute for Dynamics and Self-OrganizationGöttingenGermany
- Fred Hutchinson Cancer Research CenterSeattleUnited States
| | - Nicholas C Wu
- Department of Biochemistry, University of Illinois at Urbana-ChampaignUrbanaUnited States
- Center for Biophysics and Quantitative Biology, University of Illinois at Urbana-ChampaignUrbanaUnited States
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-ChampaignUrbanaUnited States
- Carle Illinois College of Medicine, University of Illinois at Urbana-ChampaignUrbanaUnited States
| |
Collapse
|
9
|
Hayden FG, Asher J, Cowling BJ, Hurt AC, Ikematsu H, Kuhlbusch K, Lemenuel-Diot A, Du Z, Meyers LA, Piedra PA, Takazono T, Yen HL, Monto AS. Reducing influenza virus transmission: the value of antiviral treatment. Clin Infect Dis 2021; 74:532-540. [PMID: 34245250 PMCID: PMC8834654 DOI: 10.1093/cid/ciab625] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Indexed: 11/16/2022] Open
Abstract
Prompt antiviral treatment has the potential to reduce influenza virus transmission to close contacts, but rigorous data on the magnitude of treatment effects on transmission are limited. Animal model data indicate that rapid reductions in viral replication after antiviral treatment reduce the risk of transmission. Observational and clinical trial data with oseltamivir and other neuraminidase inhibitors indicate that prompt treatment of household index patients seems to reduce the risk of illness in contacts, although the magnitude of the reported effects has varied widely across studies. In addition, the potential risk of transmitting drug-resistant variants exists with all approved classes of influenza antivirals. A controlled trial examining baloxavir treatment efficacy to reduce transmission, including the risk of transmitting virus with reduced baloxavir susceptibility, is currently in progress. If reduced transmission risk is confirmed, modeling studies indicate that early treatment could have major epidemiologic benefits in seasonal and pandemic influenza.
Collapse
Affiliation(s)
- Frederick G Hayden
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, USA
| | | | - Benjamin J Cowling
- School of Public Health, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | | | | | | | | | - Zhanwei Du
- School of Public Health, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Lauren Ancel Meyers
- Department of Integrative Biology and Statistics & Data Sciences, University of Texas, Austin, Texas, USA
| | - Pedro A Piedra
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, USA
| | - Takahiro Takazono
- Department of Infectious Diseases, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Hui-Ling Yen
- School of Public Health, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Arnold S Monto
- University of Michigan School of Public Health, Ann Arbor, Michigan, USA
| |
Collapse
|
10
|
Medaglia C, Zwygart ACA, Silva PJ, Constant S, Huang S, Stellacci F, Tapparel C. Interferon Lambda Delays the Emergence of Influenza Virus Resistance to Oseltamivir. Microorganisms 2021; 9:1196. [PMID: 34205874 PMCID: PMC8227012 DOI: 10.3390/microorganisms9061196] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 04/28/2021] [Accepted: 05/28/2021] [Indexed: 12/26/2022] Open
Abstract
Influenza viruses are a leading cause of morbidity and mortality worldwide. These air-borne pathogens are able to cross the species barrier, leading to regular seasonal epidemics and sporadic pandemics. Influenza viruses also possess a high genetic variability, which allows for the acquisition of resistance mutations to antivirals. Combination therapies with two or more drugs targeting different mechanisms of viral replication have been considered an advantageous option to not only enhance the effectiveness of the individual treatments, but also reduce the likelihood of resistance emergence. Using an in vitro infection model, we assessed the barrier to viral resistance of a combination therapy with the neuraminidase inhibitor oseltamivir and human interferon lambda against the pandemic H1N1 A/Netherlands/602/2009 (H1N1pdm09) virus. We serially passaged the virus in a cell line derived from human bronchial epithelial cells in the presence or absence of increasing concentrations of oseltamivir alone or oseltamivir plus interferon lambda. While the treatment with oseltamivir alone quickly induced the emergence of antiviral resistance through a single mutation in the neuraminidase gene, the co-administration of interferon lambda delayed the emergence of drug-resistant influenza virus variants. Our results suggest a possible clinical application of interferon lambda in combination with oseltamivir to treat influenza.
Collapse
Affiliation(s)
- Chiara Medaglia
- Department of Microbiology and Molecular Medicine, University of Geneva, 1206 Geneva, Switzerland; (C.M.); (A.C.-A.Z.)
| | | | - Paulo Jacob Silva
- Insitute of Materials, Ecole polytechnique fédérale de Lausanne, 1015 Lausanne, Switzerland; (P.J.S.); (F.S.)
| | | | - Song Huang
- Epithelix Sas, 1228 Geneva, Switzerland; (S.C.); (S.H.)
| | - Francesco Stellacci
- Insitute of Materials, Ecole polytechnique fédérale de Lausanne, 1015 Lausanne, Switzerland; (P.J.S.); (F.S.)
| | - Caroline Tapparel
- Department of Microbiology and Molecular Medicine, University of Geneva, 1206 Geneva, Switzerland; (C.M.); (A.C.-A.Z.)
| |
Collapse
|
11
|
Radovanov J, Ristic M, Medic S, Kovacevic G, Dopud N, Nikolic N, Patic A, Cvjetkovic IH, Petrovic V. Genetic variability of the neuraminidase gene of influenza A(H1N1)pdm09 viruses circulating from the 2012/2013 to 2017/2018 season in Vojvodina Province, Serbia. Mol Cell Probes 2020; 52:101557. [PMID: 32147497 DOI: 10.1016/j.mcp.2020.101557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 02/24/2020] [Accepted: 03/02/2020] [Indexed: 11/25/2022]
Affiliation(s)
- Jelena Radovanov
- Institute of Public Health of Vojvodina, Futoska 121, 21000, Novi Sad, Serbia.
| | - Mioljub Ristic
- Institute of Public Health of Vojvodina, Futoska 121, 21000, Novi Sad, Serbia; University of Novi Sad, Medical Faculty, Hajduk Veljkova 1-3, 21000, Novi Sad, Serbia
| | - Snezana Medic
- Institute of Public Health of Vojvodina, Futoska 121, 21000, Novi Sad, Serbia; University of Novi Sad, Medical Faculty, Hajduk Veljkova 1-3, 21000, Novi Sad, Serbia
| | - Gordana Kovacevic
- Institute of Public Health of Vojvodina, Futoska 121, 21000, Novi Sad, Serbia
| | - Nela Dopud
- Institute of Public Health of Vojvodina, Futoska 121, 21000, Novi Sad, Serbia
| | - Natasa Nikolic
- Institute of Public Health of Vojvodina, Futoska 121, 21000, Novi Sad, Serbia; University of Novi Sad, Medical Faculty, Hajduk Veljkova 1-3, 21000, Novi Sad, Serbia
| | - Aleksandra Patic
- Institute of Public Health of Vojvodina, Futoska 121, 21000, Novi Sad, Serbia; University of Novi Sad, Medical Faculty, Hajduk Veljkova 1-3, 21000, Novi Sad, Serbia
| | - Ivana Hrnjakovic Cvjetkovic
- Institute of Public Health of Vojvodina, Futoska 121, 21000, Novi Sad, Serbia; University of Novi Sad, Medical Faculty, Hajduk Veljkova 1-3, 21000, Novi Sad, Serbia
| | - Vladimir Petrovic
- Institute of Public Health of Vojvodina, Futoska 121, 21000, Novi Sad, Serbia; University of Novi Sad, Medical Faculty, Hajduk Veljkova 1-3, 21000, Novi Sad, Serbia
| |
Collapse
|
12
|
Shie JJ, Fang JM. Development of effective anti-influenza drugs: congeners and conjugates - a review. J Biomed Sci 2019; 26:84. [PMID: 31640786 PMCID: PMC6806523 DOI: 10.1186/s12929-019-0567-0] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 09/16/2019] [Indexed: 12/20/2022] Open
Abstract
Influenza is a long-standing health problem. For treatment of seasonal flu and possible pandemic infections, there is a need to develop new anti-influenza drugs that have good bioavailability against a broad spectrum of influenza viruses, including the resistant strains. Relenza™ (zanamivir), Tamiflu™ (the phosphate salt of oseltamivir), Inavir™ (laninamivir octanoate) and Rapivab™ (peramivir) are four anti-influenza drugs targeting the viral neuraminidases (NAs). However, some problems of these drugs should be resolved, such as oral availability, drug resistance and the induced cytokine storm. Two possible strategies have been applied to tackle these problems by devising congeners and conjugates. In this review, congeners are the related compounds having comparable chemical structures and biological functions, whereas conjugate refers to a compound having two bioactive entities joined by a covalent bond. The rational design of NA inhibitors is based on the mechanism of the enzymatic hydrolysis of the sialic acid (Neu5Ac)-terminated glycoprotein. To improve binding affinity and lipophilicity of the existing NA inhibitors, several methods are utilized, including conversion of carboxylic acid to ester prodrug, conversion of guanidine to acylguanidine, substitution of carboxylic acid with bioisostere, and modification of glycerol side chain. Alternatively, conjugating NA inhibitors with other therapeutic entity provides a synergistic anti-influenza activity; for example, to kill the existing viruses and suppress the cytokines caused by cross-species infection.
Collapse
Affiliation(s)
- Jiun-Jie Shie
- Institute of Chemistry, Academia Sinica, Taipei, 115, Taiwan
| | - Jim-Min Fang
- Department of Chemistry, National Taiwan University, Taipei, 106, Taiwan. .,The Genomics Research Center, Academia Sinica, Taipei, 115, Taiwan.
| |
Collapse
|
13
|
Characterization of substitutions in the neuraminidase of A(H7N9) influenza viruses selected following serial passage in the presence of different neuraminidase inhibitors. Antiviral Res 2019; 168:68-75. [PMID: 31132385 DOI: 10.1016/j.antiviral.2019.05.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 05/23/2019] [Accepted: 05/24/2019] [Indexed: 02/06/2023]
Abstract
Avian A(H7N9) infections in humans have been reported in China since 2013 and are of public health concern due to their severity and pandemic potential. Oseltamivir and peramivir are neuraminidase inhibitors (NAIs) routinely used for the treatment of A(H7N9) infections, but variants with reduced sensitivity to these drugs can emerge in patients during treatment. Zanamivir and laninamivir are NAIs that are used less frequently. Herein, we performed in vitro serial passaging experiments with recombinant viruses, containing the neuraminidase (NA) from influenza A/Anhui/1/13 (H7N9) virus, in the presence of each NAI, to determine whether variants with reduced sensitivity would emerge. NA substitutions were characterized for their effect on the NA enzymatic activity and surface expression of the A/Anhui/1/13 (Anhui/1) NA, as well as NAs originating from contemporary A(H7N9) viruses of the Yangtze River Delta and Pearl River Delta lineages. In vitro passage in the presence of oseltamivir, peramivir and laninamivir selected for substitutions associated with reduced sensitivity (E119D, R292K and R152K), whereas passage in the presence of zanamivir did not select for any viruses with reduced sensitivity. All the NA substitutions significantly reduced activity, but not the expression of the Anhui/1 NA. In contemporary N9 NAs, all substitutions tested significantly reduced NA enzyme function in the Yangtze River lineage background, but not in the Pearl River Delta lineage background. Overall, these findings suggest that zanamivir may be less likely than the other NAIs to select for resistance in A(H7N9) viruses and that the impact of substitutions that reduce NAI susceptibility or enzyme function may be less in A(H7N9) viruses from the Pearl River lineage.
Collapse
|
14
|
Takashita E, Morita H, Ogawa R, Nakamura K, Fujisaki S, Shirakura M, Kuwahara T, Kishida N, Watanabe S, Odagiri T. Susceptibility of Influenza Viruses to the Novel Cap-Dependent Endonuclease Inhibitor Baloxavir Marboxil. Front Microbiol 2018; 9:3026. [PMID: 30574137 PMCID: PMC6291754 DOI: 10.3389/fmicb.2018.03026] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 11/22/2018] [Indexed: 01/17/2023] Open
Abstract
The novel cap-dependent endonuclease inhibitor baloxavir marboxil was approved for the treatment of influenza A and B virus infections in February 2018 in Japan. Because of the need to monitor influenza viruses for reduced susceptibility to this drug, we used two cell-based screening systems - a conventional plaque reduction assay and a focus reduction assay - to evaluate the susceptibility of influenza viruses to baloxavir. First, we generated a reference virus possessing an I38T substitution in the polymerase acidic subunit (PA), which is known to be associated with reduced susceptibility to baloxavir, and demonstrated the validity of our systems using this reference virus. We then determined the susceptibility of a panel of neuraminidase (NA) inhibitor-resistant viruses and their sensitive counterparts to baloxavir. No significant differences in baloxavir susceptibilities were found between the NA inhibitor-resistant and -sensitive viruses. We also examined seasonal influenza viruses isolated during the 2017-2018 influenza season in Japan and found that no currently circulating A(H1N1)pdm09, A(H3N2), or B viruses had significantly reduced susceptibility to baloxavir and none of the viruses possessed an amino acid substitution at PA residue 38. Use of a combination of methods to analyze antiviral susceptibility and detect amino acid substitutions is valuable for monitoring the emergence of baloxavir-resistant viruses.
Collapse
Affiliation(s)
- Emi Takashita
- Influenza Virus Research Center, National Institute of Infectious Diseases, Tokyo, Japan
| | - Hiroko Morita
- Influenza Virus Research Center, National Institute of Infectious Diseases, Tokyo, Japan
| | - Rie Ogawa
- Influenza Virus Research Center, National Institute of Infectious Diseases, Tokyo, Japan
| | - Kazuya Nakamura
- Influenza Virus Research Center, National Institute of Infectious Diseases, Tokyo, Japan
| | - Seiichiro Fujisaki
- Influenza Virus Research Center, National Institute of Infectious Diseases, Tokyo, Japan
| | - Masayuki Shirakura
- Influenza Virus Research Center, National Institute of Infectious Diseases, Tokyo, Japan
| | - Tomoko Kuwahara
- Influenza Virus Research Center, National Institute of Infectious Diseases, Tokyo, Japan
| | - Noriko Kishida
- Influenza Virus Research Center, National Institute of Infectious Diseases, Tokyo, Japan
| | - Shinji Watanabe
- Influenza Virus Research Center, National Institute of Infectious Diseases, Tokyo, Japan
| | - Takato Odagiri
- Influenza Virus Research Center, National Institute of Infectious Diseases, Tokyo, Japan
| |
Collapse
|
15
|
Abed Y, Tu V, Carbonneau J, Checkmahomed L, Venable MC, Fage C, Marie-Ève-Hamelin, Dufresne SF, Kobinger G, Boivin G. Comparison of early and recent influenza A(H1N1)pdm09 isolates harboring or not the H275Y neuraminidase mutation, in vitro and in animal models. Antiviral Res 2018; 159:26-34. [PMID: 30219318 DOI: 10.1016/j.antiviral.2018.09.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 09/10/2018] [Accepted: 09/12/2018] [Indexed: 11/29/2022]
Abstract
After 6 years of circulation in humans, a novel antigenic variant of influenza A(H1N1)pdm09 (i.e., A/Michigan/45/2015) emerged in 2015-16 and has predominated thereafter worldwide. Herein, we compared in vitro and in vivo properties of 2016 wild-type (WT) A/Michigan/45/15-like isolate and its H275Y neuraminidase (NA) variant to the original A/California/07/09-like counterparts. The H275Y mutation induced comparable levels of resistance to oseltamivir and peramivir without altering zanamivir susceptibility in both 2009 and 2016 isolates. In vitro, the two WT isolates had comparable replicative properties. The 2016-H275Y isolate had lower titers at 36 h post-inoculation (PI) (P < 0.05) while the 2009-H275Y titers were lower at both 24 h (P < 0.01) and 36 h PI (P < 0.001) vs the respective WTs. In mice, the 2016-WT isolate caused less weight losses (P < 0.001) and lower lung viral titers (LVTs) (P < 0.01) vs the 2009-WT. The LVTs of 2016-WT and 2016-H275Y groups were comparable whereas the 2009-H275Y LVTs were lower vs the respective WT (P < 0.01). Ferrets infected with the 2016-WT isolate and their contacts had higher nasal viral titers (NVTs) at early time points vs the 2009-WT group (P < 0.01). Also, NVTs of 2016-H275Y animals were lower vs the 2016-WT group at early time points in both infected (P < 0.01) and contact animals (P < 0.001). In conclusion, while the H275Y mutation similarly impacts the A/California/07/2009- and A/Michigan/45/2015-like A(H1N1)pdm09 NAs, the fitness of these isolates differs according to animal models with the 2016 virus being less virulent in mice but slightly more virulent in ferrets, potentially reflecting a period of cumulative changes in surface and internal genes.
Collapse
Affiliation(s)
- Yacine Abed
- CHUQ-CHUL and Laval University, Québec City, QC, Canada
| | - Véronique Tu
- CHUQ-CHUL and Laval University, Québec City, QC, Canada
| | | | | | | | - Clément Fage
- CHUQ-CHUL and Laval University, Québec City, QC, Canada
| | | | | | - Gary Kobinger
- CHUQ-CHUL and Laval University, Québec City, QC, Canada
| | - Guy Boivin
- CHUQ-CHUL and Laval University, Québec City, QC, Canada.
| |
Collapse
|
16
|
Lyons DM, Lauring AS. Mutation and Epistasis in Influenza Virus Evolution. Viruses 2018; 10:E407. [PMID: 30081492 PMCID: PMC6115771 DOI: 10.3390/v10080407] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 07/30/2018] [Accepted: 07/30/2018] [Indexed: 12/25/2022] Open
Abstract
Influenza remains a persistent public health challenge, because the rapid evolution of influenza viruses has led to marginal vaccine efficacy, antiviral resistance, and the annual emergence of novel strains. This evolvability is driven, in part, by the virus's capacity to generate diversity through mutation and reassortment. Because many new traits require multiple mutations and mutations are frequently combined by reassortment, epistatic interactions between mutations play an important role in influenza virus evolution. While mutation and epistasis are fundamental to the adaptability of influenza viruses, they also constrain the evolutionary process in important ways. Here, we review recent work on mutational effects and epistasis in influenza viruses.
Collapse
Affiliation(s)
- Daniel M Lyons
- Department of Ecology and Evolutionary Biology, University of Michigan, Ann Arbor, MI 48109, USA.
| | - Adam S Lauring
- Department of Ecology and Evolutionary Biology, University of Michigan, Ann Arbor, MI 48109, USA.
- Division of Infectious Diseases, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA.
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
17
|
Huang W, Cheng Y, Li X, Tan M, Wei H, Zhao X, Xiao N, Dong J, Wang D. Neuraminidase inhibitor susceptibility profile of human influenza viruses during the 2016-2017 influenza season in Mainland China. J Infect Chemother 2018; 24:729-733. [PMID: 29866491 DOI: 10.1016/j.jiac.2018.05.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2018] [Revised: 04/13/2018] [Accepted: 05/08/2018] [Indexed: 01/27/2023]
Abstract
To understand the current situation of antiviral-resistance of influenza viruses to neuraminidase inhibitors (NAIs) in Mainland China, The antiviral-resistant surveillance data of the circulating influenza viruses in Mainland China during the 2016-2017 influenza season were analyzed. The total 3215 influenza viruses were studied to determine 50% inhibitory concentration (IC50) for oseltamivir and zanamivir using a fluorescence-based assay. Approximately 0.3% (n = 10) of viruses showed either highly reduced inhibition (HRI) or reduced inhibition (RI) against at least one NAI. The most common neuraminidase (NA) amino acid substitution was H275Y in A (H1N1)pdm09 virus, which confers HRI by oseltamivir. Two A (H1N1)pdm09 viruses contained a new NA amino acid substitution respectively, S110F and D151E, which confers RI by oseltamivir or/and zanamivir. Two B/Victoria-lineage viruses harbored a new NA amino acid substitution respectively, H134Q and S246P, which confers RI by zanamivir. One B/Victoria-lineage virus contained dual amino acid substitution NA P124T and V422I, which confers HRI by zanamivir. One B/Yamagata-lineage virus was a reassortant virus that haemagglutinin (HA) from B/Yamagata-lineage virus and NA from B/Victoria-lineage virus, defined as B/Yamagata-lineage virus confers RI by oseltamivir, but as B/Victoria-lineage virus confers normal inhibition by oseltamivir. All new substitutions that have not been reported before, the correlation of these substitutions and observed changes in IC50 should be further assessed. During the 2016-2017 influenza season in Mainland China the majority tested viruses were susceptible to oseltamivir and zanamivir. Hence, NAIs remain the recommended antiviral for treatment and prophylaxis of influenza virus infections.
Collapse
Affiliation(s)
- Weijuan Huang
- Chinese National Influenza Center, National Institute for Viral Disease Control and Prevention, Collaboration Innovation Center for Diagnosis and Treatment of Infectious Diseases, Chinese Center for Disease Control and Prevention, Key Laboratory for Medical Virology, National Health Commission, Beijing, 102206, PR China
| | - Yanhui Cheng
- Chinese National Influenza Center, National Institute for Viral Disease Control and Prevention, Collaboration Innovation Center for Diagnosis and Treatment of Infectious Diseases, Chinese Center for Disease Control and Prevention, Key Laboratory for Medical Virology, National Health Commission, Beijing, 102206, PR China
| | - Xiyan Li
- Chinese National Influenza Center, National Institute for Viral Disease Control and Prevention, Collaboration Innovation Center for Diagnosis and Treatment of Infectious Diseases, Chinese Center for Disease Control and Prevention, Key Laboratory for Medical Virology, National Health Commission, Beijing, 102206, PR China
| | - Minju Tan
- Chinese National Influenza Center, National Institute for Viral Disease Control and Prevention, Collaboration Innovation Center for Diagnosis and Treatment of Infectious Diseases, Chinese Center for Disease Control and Prevention, Key Laboratory for Medical Virology, National Health Commission, Beijing, 102206, PR China
| | - Hejiang Wei
- Chinese National Influenza Center, National Institute for Viral Disease Control and Prevention, Collaboration Innovation Center for Diagnosis and Treatment of Infectious Diseases, Chinese Center for Disease Control and Prevention, Key Laboratory for Medical Virology, National Health Commission, Beijing, 102206, PR China
| | - Xiang Zhao
- Chinese National Influenza Center, National Institute for Viral Disease Control and Prevention, Collaboration Innovation Center for Diagnosis and Treatment of Infectious Diseases, Chinese Center for Disease Control and Prevention, Key Laboratory for Medical Virology, National Health Commission, Beijing, 102206, PR China
| | - Ning Xiao
- Chinese National Influenza Center, National Institute for Viral Disease Control and Prevention, Collaboration Innovation Center for Diagnosis and Treatment of Infectious Diseases, Chinese Center for Disease Control and Prevention, Key Laboratory for Medical Virology, National Health Commission, Beijing, 102206, PR China
| | - Jie Dong
- Chinese National Influenza Center, National Institute for Viral Disease Control and Prevention, Collaboration Innovation Center for Diagnosis and Treatment of Infectious Diseases, Chinese Center for Disease Control and Prevention, Key Laboratory for Medical Virology, National Health Commission, Beijing, 102206, PR China
| | - Dayan Wang
- Chinese National Influenza Center, National Institute for Viral Disease Control and Prevention, Collaboration Innovation Center for Diagnosis and Treatment of Infectious Diseases, Chinese Center for Disease Control and Prevention, Key Laboratory for Medical Virology, National Health Commission, Beijing, 102206, PR China.
| |
Collapse
|
18
|
Järhult JD. Environmental resistance development to influenza antivirals: a case exemplifying the need for a multidisciplinary One Health approach including physicians. Acta Vet Scand 2018; 60:6. [PMID: 29370857 PMCID: PMC5784702 DOI: 10.1186/s13028-018-0360-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 01/17/2018] [Indexed: 12/20/2022] Open
Abstract
A multidisciplinary approach is a prerequisite for One Health. Physicians are important players in the One Health team, yet they are often hard to convince of the benefits of the One Health approach. Here, the case for multidisciplinarity including physicians is made using the example of environmental resistance development to influenza antivirals. Neuraminidase inhibitors are the major class of anti-influenza pharmaceuticals, and extensively stockpiled globally as a cornerstone of pandemic preparedness, especially important in the first phase before vaccines can be mass-produced. The active metabolite of oseltamivir that is excreted from treated patients degrades poorly in conventional sewage treatment processes and has been found in river waters. Dabbling ducks constitute the natural influenza A virus reservoir and often reside near sewage treatment plant outlets, where they may be exposed to neuraminidase inhibitor residues. In vivo experiments using influenza-infected Mallards exposed to neuraminidase inhibitors present in their water have shown resistance development and persistence, demonstrating that resistance may be induced and become established in the influenza strains circulating in natural hosts. Neuraminidase inhibitor resistance genes may become part of a human-adapted influenza virus with pandemic potential through reassortment or direct transmission. A pandemic caused by a neuraminidase inhibitor-resistant influenza virus is a serious threat as the first line defense in pandemic preparedness would be disarmed. To assess the risk for environmental influenza resistance development, a broad multidisciplinary team containing chemists, social scientists, veterinarians, biologists, ecologists, virologists, epidemiologists, and physicians is needed. Information about One Health early in high school and undergraduate training, an active participation of One Health-engaged physicians in the debate, and more One Health-adapted funding and publication possibilities are suggested to increase the possibility to engage physicians.
Collapse
Affiliation(s)
- Josef D Järhult
- Zoonosis Science Center, Department of Medical Sciences, Uppsala University, 75185, Uppsala, Sweden.
| |
Collapse
|
19
|
Krammer F, Smith GJD, Fouchier RAM, Peiris M, Kedzierska K, Doherty PC, Palese P, Shaw ML, Treanor J, Webster RG, García-Sastre A. Influenza. Nat Rev Dis Primers 2018; 4:3. [PMID: 29955068 PMCID: PMC7097467 DOI: 10.1038/s41572-018-0002-y] [Citation(s) in RCA: 983] [Impact Index Per Article: 140.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Influenza is an infectious respiratory disease that, in humans, is caused by influenza A and influenza B viruses. Typically characterized by annual seasonal epidemics, sporadic pandemic outbreaks involve influenza A virus strains of zoonotic origin. The WHO estimates that annual epidemics of influenza result in ~1 billion infections, 3–5 million cases of severe illness and 300,000–500,000 deaths. The severity of pandemic influenza depends on multiple factors, including the virulence of the pandemic virus strain and the level of pre-existing immunity. The most severe influenza pandemic, in 1918, resulted in >40 million deaths worldwide. Influenza vaccines are formulated every year to match the circulating strains, as they evolve antigenically owing to antigenic drift. Nevertheless, vaccine efficacy is not optimal and is dramatically low in the case of an antigenic mismatch between the vaccine and the circulating virus strain. Antiviral agents that target the influenza virus enzyme neuraminidase have been developed for prophylaxis and therapy. However, the use of these antivirals is still limited. Emerging approaches to combat influenza include the development of universal influenza virus vaccines that provide protection against antigenically distant influenza viruses, but these vaccines need to be tested in clinical trials to ascertain their effectiveness.
Collapse
Affiliation(s)
- Florian Krammer
- 0000 0001 0670 2351grid.59734.3cDepartment of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Gavin J. D. Smith
- 0000 0001 2180 6431grid.4280.eDuke–NUS Medical School, Singapore, Singapore ,0000 0004 1936 7961grid.26009.3dDuke Global Health Institute, Duke University, Durham, NC USA
| | - Ron A. M. Fouchier
- 000000040459992Xgrid.5645.2Department of Viroscience, Erasmus MC, Rotterdam, Netherlands
| | - Malik Peiris
- 0000000121742757grid.194645.bWHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, Hong Kong, China ,0000000121742757grid.194645.bCenter of Influenza Research, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, Hong Kong, China
| | - Katherine Kedzierska
- 0000 0001 2179 088Xgrid.1008.9Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria Australia
| | - Peter C. Doherty
- 0000 0001 2179 088Xgrid.1008.9Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria Australia ,0000 0001 0224 711Xgrid.240871.8Department of Immunology, St Jude Children’s Research Hospital, Memphis, TN USA
| | - Peter Palese
- 0000 0001 0670 2351grid.59734.3cDepartment of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY USA ,0000 0001 0670 2351grid.59734.3cDivision of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Megan L. Shaw
- 0000 0001 0670 2351grid.59734.3cDepartment of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - John Treanor
- 0000 0004 1936 9166grid.412750.5Division of Infectious Diseases, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY USA
| | - Robert G. Webster
- 0000 0001 0224 711Xgrid.240871.8Department of Infectious Diseases, St Jude Children’s Research Hospital, Memphis, TN USA
| | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA. .,Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA. .,Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
20
|
Farrukee R, Hurt AC. Antiviral Drugs for the Treatment and Prevention of Influenza. CURRENT TREATMENT OPTIONS IN INFECTIOUS DISEASES 2017. [DOI: 10.1007/s40506-017-0129-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
21
|
Hussain M, Galvin HD, Haw TY, Nutsford AN, Husain M. Drug resistance in influenza A virus: the epidemiology and management. Infect Drug Resist 2017; 10:121-134. [PMID: 28458567 PMCID: PMC5404498 DOI: 10.2147/idr.s105473] [Citation(s) in RCA: 321] [Impact Index Per Article: 40.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Influenza A virus (IAV) is the sole cause of the unpredictable influenza pandemics and deadly zoonotic outbreaks and constitutes at least half of the cause of regular annual influenza epidemics in humans. Two classes of anti-IAV drugs, adamantanes and neuraminidase (NA) inhibitors (NAIs) targeting the viral components M2 ion channel and NA, respectively, have been approved to treat IAV infections. However, IAV rapidly acquired resistance against both classes of drugs by mutating these viral components. The adamantane-resistant IAV has established itself in nature, and a majority of the IAV subtypes, especially the most common H1N1 and H3N2, circulating globally are resistant to adamantanes. Consequently, adamantanes have become practically obsolete as anti-IAV drugs. Similarly, up to 100% of the globally circulating IAV H1N1 subtypes were resistant to oseltamivir, the most commonly used NAI, until 2009. However, the 2009 pandemic IAV H1N1 subtype, which was sensitive to NAIs and has now become one of the dominant seasonal influenza virus strains, has replaced the pre-2009 oseltamivir-resistant H1N1 variants. This review traces the epidemiology of both adamantane- and NAI-resistant IAV subtypes since the approval of these drugs and highlights the susceptibility status of currently circulating IAV subtypes to NAIs. Further, it provides an overview of currently and soon to be available control measures to manage current and emerging drug-resistant IAV. Finally, this review outlines the research directions that should be undertaken to manage the circulation of IAV in intermediate hosts and develop effective and alternative anti-IAV therapies.
Collapse
Affiliation(s)
- Mazhar Hussain
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Henry D Galvin
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Tatt Y Haw
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Ashley N Nutsford
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Matloob Husain
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| |
Collapse
|
22
|
Gao Z, Niikura M, Withers SG. Ultrasensitive Fluorogenic Reagents for Neuraminidase Titration. Angew Chem Int Ed Engl 2017; 56:6112-6116. [PMID: 28191709 DOI: 10.1002/anie.201610544] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Indexed: 11/07/2022]
Abstract
Influenza viral neuraminidase plays a crucial role during infections. It is a major target for the development of anti-influenza drugs and is also attracting increasing attention as a vaccine target as evidence accumulates that neuraminidase-neutralizing antibodies contribute to protection. However, no method currently exists to accurately and efficiently measure concentrations of active neuraminidase in virus samples or other crude mixtures, which hampers development on both fronts. In this report, we describe the development of a selective and sensitive active-site titration reagent for neuraminidase that can quantify viral neuraminidases down to sub-nanomolar levels in crude samples, with no background from non-viral neuraminidases. By using this reagent, we determined accurate kcat values for six influenza A and two influenza B neuraminidases for the first time. We also quantified the neuraminidase content in a commercial influenza vaccine, thus demonstrating that this titration reagent opens the possibility for better vaccine analysis.
Collapse
Affiliation(s)
- Zhizeng Gao
- Department of Chemistry, University of British Columbia, Vancouver, British Columbia, V6T 1Z1, Canada
| | - Masahiro Niikura
- Faculty of Health Sciences, Simon Fraser University, Burnaby, British Columbia, V5A 1S6, Canada
| | - Stephen G Withers
- Department of Chemistry, University of British Columbia, Vancouver, British Columbia, V6T 1Z1, Canada
| |
Collapse
|
23
|
Gao Z, Niikura M, Withers SG. Ultrasensitive Fluorogenic Reagents for Neuraminidase Titration. Angew Chem Int Ed Engl 2017. [DOI: 10.1002/ange.201610544] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Zhizeng Gao
- Department of Chemistry; University of British Columbia; Vancouver British Columbia V6T 1Z1 Canada
| | - Masahiro Niikura
- Faculty of Health Sciences; Simon Fraser University; Burnaby British Columbia V5A 1S6 Canada
| | - Stephen G. Withers
- Department of Chemistry; University of British Columbia; Vancouver British Columbia V6T 1Z1 Canada
| |
Collapse
|
24
|
Antiviral Resistance in Influenza Viruses: Clinical and Epidemiological Aspects. ANTIMICROBIAL DRUG RESISTANCE 2017. [PMCID: PMC7122614 DOI: 10.1007/978-3-319-47266-9_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
There are three classes of antiviral drugs approved for the treatment of influenza: the M2 ion channel inhibitors (amantadine, rimantadine), neuraminidase (NA) inhibitors (laninamivir, oseltamivir, peramivir, zanamivir), and the protease inhibitor (favipiravir); some of the agents are only available in selected countries [1, 2]. These agents are effective at treating the signs and symptoms of influenza in patients infected with susceptible viruses. Clinical failure has been demonstrated in patients infected with viruses with primary resistance, i.e., antivirals can be present in the virus initially infecting the patient, or resistance may emerge during the course of therapy [3–5]. NA inhibitors are active against all nine NA subtypes recognized in nature [6], including highly pathogenic avian influenza A/H5N1 and recent low-pathogenic avian influenza A/H7N9 viruses [7]. Since seasonal influenza is usually an acute, self-limited illness in which viral clearance usually occurs rapidly due to innate and adaptive host immune responses, the emergence of drug-resistant variants would be anticipated to have limited effect on clinical recovery in otherwise healthy patients, as has been demonstrated clinically [3, 8, 9]. Unfortunately, immunocompromised or immunologically naïve hosts, such as young children and infants or those exposed to novel strains, are more likely to have mutations that confer resistance emergence during therapy; such resistant variants may also result in clinically significant adverse outcomes [10–13].
Collapse
|
25
|
Kossyvakis A, Mentis AFA, Tryfinopoulou K, Pogka V, Kalliaropoulos A, Antalis E, Lytras T, Meijer A, Tsiodras S, Karakitsos P, Mentis AF. Antiviral susceptibility profile of influenza A viruses; keep an eye on immunocompromised patients under prolonged treatment. Eur J Clin Microbiol Infect Dis 2016; 36:361-371. [PMID: 27848039 DOI: 10.1007/s10096-016-2809-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 10/03/2016] [Indexed: 11/24/2022]
Abstract
There was an increase in severe and fatal influenza cases in Greece during the 2011-2015 post-pandemic period. To investigate causality, we determined neuraminidase (NA) inhibitor susceptibility and resistance-conferring NA and hemagglutinin (HA) mutations in circulating influenza type A viruses during the pandemic (2009-2010) and post-pandemic periods in Greece. One hundred thirty-four influenza A(H1N1)pdm09 and 95 influenza A(H3N2) viruses submitted to the National Influenza Reference Laboratory of Southern Greece were tested for susceptibility to oseltamivir and zanamivir. Antiviral resistance was assessed by neuraminidase sequence analysis, as well as the fluorescence-based 50 % inhibitory concentration (IC50) method. Five influenza A(H1N1)pdm09 viruses (2.2 %) showed significantly reduced inhibition by oseltamivir (average IC50 300.60nM vs. 1.19nM) by Gaussian kernel density plot analysis. These viruses were isolated from immunocompromised patients and harbored the H275Y oseltamivir resistance-conferring NA substitution. All A(H1N1)pdm09 viruses were zanamivir-susceptible, and all A(H3N2) viruses were susceptible to both drugs. Oseltamivir-resistant viruses did not form a distinct cluster by phylogenetic analysis. Permissive mutations were detected in immunogenic and non immunogenic NA regions of both oseltamivir- resistant and susceptible viruses in the post-pandemic seasons. Several amino acid substitutions in the HA1 domain of the HA gene of post-pandemic viruses were identified. This study indicated low resistance to NAIs among tested influenza viruses. Antiviral resistance emerged only in immunocompromised patients under long-term oseltamivir treatment. Sequential sample testing in this vulnerable group of patients is recommended to characterise resistance or reinfection and viral evolution.
Collapse
Affiliation(s)
- A Kossyvakis
- National Influenza Reference Laboratory of Southern Greece, Hellenic Pasteur Institute, 127, Vas. Sofias Ave., 11521, Athens, Greece
| | - A-F A Mentis
- National Influenza Reference Laboratory of Southern Greece, Hellenic Pasteur Institute, 127, Vas. Sofias Ave., 11521, Athens, Greece.,Johns Hopkins University, AAP, Baltimore, MD, USA
| | - K Tryfinopoulou
- European Programme for Public Health Microbiology Training (EUPHEM), European Centre for Disease Prevention and Control, Stockholm, Sweden.,Antimicrobial Resistance and Healthcare-associated Infections Laboratory, National School of Public Health, Athens, Greece.,Hellenic Central Public Health Laboratory, Hellenic Centre for Disease Control and Prevention, Athens, Greece
| | - V Pogka
- National Influenza Reference Laboratory of Southern Greece, Hellenic Pasteur Institute, 127, Vas. Sofias Ave., 11521, Athens, Greece
| | - A Kalliaropoulos
- National Influenza Reference Laboratory of Southern Greece, Hellenic Pasteur Institute, 127, Vas. Sofias Ave., 11521, Athens, Greece
| | - E Antalis
- 4th Academic Department of Internal Medicine and Infectious Diseases, Attikon University Hospital, University of Athens Medical School, Athens, Greece
| | - T Lytras
- Department of Epidemiological Surveillance and Intervention, Hellenic Centre for Disease Control and Prevention, Athens, Greece.,Centre for Research in Environmental Epidemiology (CREAL), Barcelona, Spain.,Department of Experimental and Health Sciences, Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - A Meijer
- National Institute for Public Health and the Environment, Centre for Infectious Disease Control, Bilthoven, Netherlands
| | - S Tsiodras
- 4th Academic Department of Internal Medicine and Infectious Diseases, Attikon University Hospital, University of Athens Medical School, Athens, Greece
| | - P Karakitsos
- Department of Cytopathology, Attikon University Hospital, University of Athens Medical School, Athens, Greece
| | - A F Mentis
- National Influenza Reference Laboratory of Southern Greece, Hellenic Pasteur Institute, 127, Vas. Sofias Ave., 11521, Athens, Greece.
| |
Collapse
|
26
|
Tu V, Abed Y, Barbeau X, Carbonneau J, Fage C, Lagüe P, Boivin G. The I427T neuraminidase (NA) substitution, located outside the NA active site of an influenza A(H1N1)pdm09 variant with reduced susceptibility to NA inhibitors, alters NA properties and impairs viral fitness. Antiviral Res 2016; 137:6-13. [PMID: 27838351 DOI: 10.1016/j.antiviral.2016.11.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Revised: 10/28/2016] [Accepted: 11/09/2016] [Indexed: 01/16/2023]
Abstract
Emergence of pan neuraminidase inhibitor (NAI)-resistant variants constitutes a serious clinical concern. An influenza A(H1N1)pdm09 variant containing the I427T/Q313R neuraminidase (NA) substitutions was previously identified in a surveillance study. Although these changes are not part of the NA active site, the variant showed reduced susceptibility to many NAIs. In this study, we investigated the mechanism of resistance for the I427T/Q313R substitution and its impact on the NA enzyme and viral fitness. Recombinant wild-type (WT), I427T/Q313R and I427T A(H1N1)pdm09 viruses were generated by reverse genetics and tested for their drug susceptibilities, enzymatic properties and replication kinetics in vitro as well as their virulence in mice. Molecular dynamics (MD) simulations were performed for NA structural analysis. The I427T substitution, which was responsible for the resistance phenotype observed in the double (I427T/Q313R) mutant, induced 17-, 56-, 7-, and 14-fold increases in IC50 values against oseltamivir, zanamivir, peramivir and laninamivir, respectively. The I427T substitution alone or combined to Q313R significantly reduced NA affinity. The I427T/Q313R and to a lesser extent I427T recombinant viruses displayed reduced viral titers vs WT in vitro. In experimentally-infected mice, the mortality rates were 62.5%, 0% and 14.3% for the WT, I417T/Q313R and I427T viruses, respectively. There were about 2.5- and 2-Log reductions in mean lung viral titers on day 5 post-infection for the I427T/Q313R and I427T mutants, respectively, compared to WT. Results from simulations revealed that the I427T change indirectly altered the stability of the catalytic R368 residue of the NA enzyme causing its reduced binding to the substrate/inhibitor. This study demonstrates that the I427T/Q313R mutant, not only alters NAI susceptibility but also compromises NA properties and viral fitness, which could explain its infrequent detection in clinic.
Collapse
Affiliation(s)
- Véronique Tu
- Research Center in Infectious Diseases of the CHUQ-CHUL and Laval University, Québec City, QC, Canada
| | - Yacine Abed
- Research Center in Infectious Diseases of the CHUQ-CHUL and Laval University, Québec City, QC, Canada
| | - Xavier Barbeau
- Proteo and IBIS, Department of Chemistry, Faculty of Science and Engineering, Laval University, Québec City, QC, Canada
| | - Julie Carbonneau
- Research Center in Infectious Diseases of the CHUQ-CHUL and Laval University, Québec City, QC, Canada
| | - Clément Fage
- Research Center in Infectious Diseases of the CHUQ-CHUL and Laval University, Québec City, QC, Canada
| | - Patrick Lagüe
- Proteo and IBIS, Department of Biochemistry, Microbiology and Bioinformatics, Faculty of Science and Engineering, Laval University, Québec City, QC, Canada
| | - Guy Boivin
- Research Center in Infectious Diseases of the CHUQ-CHUL and Laval University, Québec City, QC, Canada.
| |
Collapse
|
27
|
Romero-Beltran L, Baker SF, Puerto-Solís M, González-Losa R, Conde-Ferraez L, Alvarez-Sánchez LC, Martínez-Sobrido L, Ayora-Talavera G. Mutations at highly conserved residues in influenza A(H1N1)pdm09 virus affect neuraminidase activity. Virus Res 2016; 225:1-9. [PMID: 27596738 DOI: 10.1016/j.virusres.2016.09.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 08/17/2016] [Accepted: 09/02/2016] [Indexed: 01/16/2023]
Abstract
Influenza virus neuraminidase (NA) plays a pivotal role during viral growth since its sialidase activity allows the efficient release of nascent virions from infected cells. Consequently, mutations in the NA catalytic site affecting sialic acid (SA) cleavage may influence the biological properties of influenza viruses. This study reports two amino acid substitutions (N386K and P431S) in the NA of the influenza A(H1N1)pdm09 virus that emerged in 2009 in Mexico. The NA sialidase activity to cleave SA-like substrates, and viral growth were examined and the mutant viruses had various deficiencies. NA mutations N386K and P431S together or separately, and in the presence or absence of H275Y were further evaluated using recombinant influenza A/California/04/2009 (pH1N1) viruses containing single, double, or triple mutations. Viral growth was reduced in the presence of mutation P431S alone or combined with N386K and/or H275Y. Substrates hydrolysis was reduced when recombinant pH1N1 viruses were analyzed by NA inhibitory assays. Moreover, elution assays with guinea pig red blood cells indicated an unbalanced hemagglutinin (HA):NA functionality. Altogether, our data underline the functional significance of mutations at highly conserved sites in influenza virus NA glycoprotein and the occurrence of permissive mutations to compensate virus viability in vitro.
Collapse
Affiliation(s)
| | - Steven F Baker
- Department of Microbiology and Immunology, University of Rochester, Rochester, NY, USA
| | | | | | | | - Leidi C Alvarez-Sánchez
- Laboratorio de Virología, Universidad Autonoma de Yucatan, Mexico; Department of Microbiology and Immunology, University of Rochester, Rochester, NY, USA
| | - Luis Martínez-Sobrido
- Department of Microbiology and Immunology, University of Rochester, Rochester, NY, USA
| | | |
Collapse
|
28
|
Jones JC, Marathe BM, Lerner C, Kreis L, Gasser R, Pascua PNQ, Najera I, Govorkova EA. A Novel Endonuclease Inhibitor Exhibits Broad-Spectrum Anti-Influenza Virus Activity In Vitro. Antimicrob Agents Chemother 2016; 60:5504-14. [PMID: 27381402 PMCID: PMC4997863 DOI: 10.1128/aac.00888-16] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 06/27/2016] [Indexed: 11/20/2022] Open
Abstract
Antiviral drugs are important in preventing and controlling influenza, particularly when vaccines are ineffective or unavailable. A single class of antiviral drugs, the neuraminidase inhibitors (NAIs), is recommended for treating influenza. The limited therapeutic options and the potential risk of antiviral resistance are driving the search for additional small-molecule inhibitors that act on influenza virus proteins. The acid polymerase (PA) of influenza viruses is a promising target for new antivirals because of its essential role in initiating virus transcription. Here, we characterized a novel compound, RO-7, identified as a putative PA endonuclease inhibitor. RO-7 was effective when added before the cessation of genome replication, reduced polymerase activity in cell-free systems, and decreased relative amounts of viral mRNA and genomic RNA during influenza virus infection. RO-7 specifically inhibited the ability of the PA endonuclease domain to cleave a nucleic acid substrate. RO-7 also inhibited influenza A viruses (seasonal and 2009 pandemic H1N1 and seasonal H3N2) and B viruses (Yamagata and Victoria lineages), zoonotic viruses (H5N1, H7N9, and H9N2), and NAI-resistant variants in plaque reduction, yield reduction, and cell viability assays in Madin-Darby canine kidney (MDCK) cells with nanomolar to submicromolar 50% effective concentrations (EC50s), low toxicity, and favorable selective indices. RO-7 also inhibited influenza virus replication in primary normal human bronchial epithelial cells. Overall, RO-7 exhibits broad-spectrum activity against influenza A and B viruses in multiple in vitro assays, supporting its further characterization and development as a potential antiviral agent for treating influenza.
Collapse
Affiliation(s)
- Jeremy C Jones
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Bindumadhav M Marathe
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | | | | | | | - Philippe Noriel Q Pascua
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | | | - Elena A Govorkova
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| |
Collapse
|
29
|
Abstract
Antiviral drug resistance is a matter of great clinical importance that, historically, has been investigated mostly from a virological perspective. Although the proximate mechanisms of resistance can be readily uncovered using these methods, larger evolutionary trends often remain elusive. Recent interest by population geneticists in studies of antiviral resistance has spurred new metrics for evaluating mutation and recombination rates, demographic histories of transmission and compartmentalization, and selective forces incurred during viral adaptation to antiviral drug treatment. We present up-to-date summaries on antiviral resistance for a range of drugs and viral types, and review recent advances for studying their evolutionary histories. We conclude that information imparted by demographic and selective histories, as revealed through population genomic inference, is integral to assessing the evolution of antiviral resistance as it pertains to human health.
Collapse
Affiliation(s)
- Kristen K Irwin
- School of Life Sciences, École Polytechnique Fédéral de Lausanne (EPFL), Lausanne, Switzerland.,Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland
| | - Nicholas Renzette
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Timothy F Kowalik
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Jeffrey D Jensen
- School of Life Sciences, École Polytechnique Fédéral de Lausanne (EPFL), Lausanne, Switzerland.,Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland
| |
Collapse
|
30
|
Yen HL. Current and novel antiviral strategies for influenza infection. Curr Opin Virol 2016; 18:126-34. [DOI: 10.1016/j.coviro.2016.05.004] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 05/15/2016] [Accepted: 05/17/2016] [Indexed: 12/20/2022]
|
31
|
A Balance between Inhibitor Binding and Substrate Processing Confers Influenza Drug Resistance. J Mol Biol 2015; 428:538-553. [PMID: 26656922 DOI: 10.1016/j.jmb.2015.11.027] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Revised: 11/23/2015] [Accepted: 11/24/2015] [Indexed: 11/22/2022]
Abstract
The therapeutic benefits of the neuraminidase (NA) inhibitor oseltamivir are dampened by the emergence of drug resistance mutations in influenza A virus (IAV). To investigate the mechanistic features that underlie resistance, we developed an approach to quantify the effects of all possible single-nucleotide substitutions introduced into important regions of NA. We determined the experimental fitness effects of 450 nucleotide mutations encoding positions both surrounding the active site and at more distant sites in an N1 strain of IAV in the presence and absence of oseltamivir. NA mutations previously known to confer oseltamivir resistance in N1 strains, including H275Y and N295S, were adaptive in the presence of drug, indicating that our experimental system captured salient features of real-world selection pressures acting on NA. We identified mutations, including several at position 223, that reduce the apparent affinity for oseltamivir in vitro. Position 223 of NA is located adjacent to a hydrophobic portion of oseltamivir that is chemically distinct from the substrate, making it a hotspot for substitutions that preferentially impact drug binding relative to substrate processing. Furthermore, two NA mutations, K221N and Y276F, each reduce susceptibility to oseltamivir by increasing NA activity without altering drug binding. These results indicate that competitive expansion of IAV in the face of drug pressure is mediated by a balance between inhibitor binding and substrate processing.
Collapse
|
32
|
Tai SHS, Agafitei O, Gao Z, Liggins R, Petric M, Withers SG, Niikura M. Difluorosialic acids, potent novel influenza virus neuraminidase inhibitors, induce fewer drug resistance-associated neuraminidase mutations than does oseltamivir. Virus Res 2015; 210:126-32. [PMID: 26247418 DOI: 10.1016/j.virusres.2015.07.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Revised: 06/24/2015] [Accepted: 07/17/2015] [Indexed: 11/29/2022]
Abstract
Neuraminidase inhibitors (NAIs), including the most frequently prescribed oral therapeutic oseltamivir, play a critical role in the control of severe influenza virus (IFV) infections. However, recent reports of spread of an oseltamivir-resistant H1N1 pandemic strain in individuals who have never been exposed to oseltamivir highlight an urgent need for new antivirals against NAI-resistant IFVs. Difluorosialic acids (DFSAs) are a novel class of anti-IFV NAIs designed based on the mechanism of action of IFV NA, and distinguished by their covalent inhibition mode and their high structural similarity to the natural substrate, sialic acid. These characteristics should render the development of resistance a less rapid process. In this report, we evaluated the relative propensity of influenza A virus (IFV-A) NA to develop resistance against the DFSA class of inhibitor by passaging IFV-A strains in vitro in the presence of either oseltamivir or a representative DFSA (FeqGuDFSA). All the passage-selected lines gained mutations in hemagglutinin. Among the 12 oseltamivir-resistant passaged lines, five gained NA mutations and four of these were the well-defined H275Y mutation that causes oseltamivir resistance. In contrast, out of 15 DFSA-passaged lines, only 2 lines gained NA mutations. Further, NA inhibition assays indicated that these mutations did not change the sensitivity of NA to DFSA and thus the resistance to DFSA was not conferred by these NA mutations. These results strongly suggest that, compared to oseltamivir, IFV is less prone to development of resistance against DFSAs through NA mutations.
Collapse
Affiliation(s)
- S-H Sheldon Tai
- Faculty of Health Sciences, Simon Fraser University, 8888 University Drive, Burnaby, British Columbia V5A 1S6, Canada
| | - Olga Agafitei
- Faculty of Health Sciences, Simon Fraser University, 8888 University Drive, Burnaby, British Columbia V5A 1S6, Canada
| | - Zhizeng Gao
- Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, British Columbia V6T 1Z1, Canada
| | - Richard Liggins
- Centre for Drug Research and Development (CDRD), 2259 Lower Mall, Vancouver, British Columbia V6T 1Z4, Canada
| | - Martin Petric
- British Columbia Centre for Disease Control, 655 West 12th Avenue, Vancouver, British Columbia V5Z 4R4, Canada
| | - Stephen G Withers
- Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, British Columbia V6T 1Z1, Canada
| | - Masahiro Niikura
- Faculty of Health Sciences, Simon Fraser University, 8888 University Drive, Burnaby, British Columbia V5A 1S6, Canada.
| |
Collapse
|
33
|
Spanakis N, Pitiriga V, Gennimata V, Tsakris A. A review of neuraminidase inhibitor susceptibility in influenza strains. Expert Rev Anti Infect Ther 2015; 12:1325-36. [PMID: 25301229 DOI: 10.1586/14787210.2014.966083] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Influenza human infections are considered as a persistent global public health issue. Whereas vaccination is important for prevention, given its limitations, antiviral therapy is at the forefront of treatment, while it also plays a significant role in prevention. Currently, two classes of drugs, adamantanes (M2 blockers) and neuraminidase inhibitors (NAIs), are available for treatment and chemoprophylaxis of influenza infections. Given the resistance patterns of circulating influenza strains, adamantanes are not currently recommended. The current review mainly focuses on the development of resistance to NAIs among A and B subtypes of influenza virus strains over the last 5 years. 'Permissive' drift mutations and reassortment of viral gene segments have resulted in NAI oseltamivir-resistant A/(H1N1) variants that rapidly became predominant worldwide in the period 2007-2009. However, the prevalence of antiviral resistance to NAI zanamivir remains relatively low. In addition, the recently developed NAIs, peramivir and laninamivir, while licensed in certain countries, are still under evaluation and only a few reports have described resistance to peramivir. Although in 2014, the majority of circulating human influenza viruses remains susceptible to all NAIs, the emergence of oseltamivir-resistant influenza variants that could retain viral transmissibility, highlights the necessity for enhanced epidemiological and microbiological surveillance and clinical assessment of antiviral resistance.
Collapse
Affiliation(s)
- Nick Spanakis
- Department of Microbiology, Medical School, University of Athens, 11527 Athens, Greece
| | | | | | | |
Collapse
|
34
|
L'Huillier AG, Abed Y, Petty TJ, Cordey S, Thomas Y, Bouhy X, Schibler M, Simon A, Chalandon Y, van Delden C, Zdobnov E, Boquete-Suter P, Boivin G, Kaiser L. E119D Neuraminidase Mutation Conferring Pan-Resistance to Neuraminidase Inhibitors in an A(H1N1)pdm09 Isolate From a Stem-Cell Transplant Recipient. J Infect Dis 2015; 212:1726-34. [PMID: 25985905 DOI: 10.1093/infdis/jiv288] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Accepted: 05/08/2015] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND An influenza A(H1N1)pdm09 infection was diagnosed in a hematopoietic stem cell transplant recipient during conditioning regimen. He was treated with oral oseltamivir, later combined with intravenous zanamivir. The H275Y neuraminidase (NA) mutation was first detected, and an E119D NA mutation was identified during zanamivir therapy. METHODS Recombinant wild-type (WT) E119D and E119D/H275Y A(H1N1)pdm09 NA variants were generated by reverse genetics. Susceptibility to NA inhibitors (NAIs) was evaluated with a fluorometric assay using the 2'-(4-methylumbelliferyl)-α-D-N-acetylneuraminic acid (MUNANA) substrate. Susceptibility to favipiravir (T-705) was assessed using plaque reduction assays. The NA affinity and velocity values were determined with NA enzymatic studies. RESULTS We identified an influenza A(H1N1)pdm09 E119D mutant that exhibited a marked increase in the 50% inhibitory concentrations against all tested NAIs (827-, 25-, 286-, and 702-fold for zanamivir, oseltamivir, peramivir, and laninamivir, respectively). The double E119D/H275Y mutation further increased oseltamivir and peramivir 50% inhibitory concentrations by 790- and >5000-fold, respectively, compared with the WT. The mutant viruses remained susceptible to favipiravir. The NA affinity and velocity values of the E119D variant decreased by 8.1-fold and 4.5-fold, respectively, compared with the WT. CONCLUSIONS The actual emergence of a single NA mutation conferring pan-NAI resistance in the clinical setting reinforces the pressing need to develop new anti-influenza strategies.
Collapse
Affiliation(s)
- Arnaud G L'Huillier
- Laboratory of Virology, Divisions of Infectious Diseases and Laboratory Medicine
| | - Yacine Abed
- Centre Hospitalier Universitaire de Québec and Université Laval, Quebec City, Canada
| | - Tom J Petty
- Department of Genetic Medicine and Development, University of Geneva Medical School Swiss Institute of Bioinformatics, University of Geneva, Switzerland
| | - Samuel Cordey
- Laboratory of Virology, Divisions of Infectious Diseases and Laboratory Medicine
| | - Yves Thomas
- Laboratory of Virology, Divisions of Infectious Diseases and Laboratory Medicine
| | - Xavier Bouhy
- Centre Hospitalier Universitaire de Québec and Université Laval, Quebec City, Canada
| | - Manuel Schibler
- Laboratory of Virology, Divisions of Infectious Diseases and Laboratory Medicine
| | - Audrey Simon
- Division of Hematology, Department of Internal Medicine Specialties, University of Geneva Hospitals
| | - Yves Chalandon
- Division of Hematology, Department of Internal Medicine Specialties, University of Geneva Hospitals
| | - Christian van Delden
- Division of Infectious Diseases, Department of Internal Medicine Specialties, University of Geneva Hospitals
| | - Evgeny Zdobnov
- Department of Genetic Medicine and Development, University of Geneva Medical School Swiss Institute of Bioinformatics, University of Geneva, Switzerland
| | | | - Guy Boivin
- Centre Hospitalier Universitaire de Québec and Université Laval, Quebec City, Canada
| | - Laurent Kaiser
- Laboratory of Virology, Divisions of Infectious Diseases and Laboratory Medicine University of Geneva Medical School, University of Geneva, Switzerland
| |
Collapse
|
35
|
Takashita E, Meijer A, Lackenby A, Gubareva L, Rebelo-de-Andrade H, Besselaar T, Fry A, Gregory V, Leang SK, Huang W, Lo J, Pereyaslov D, Siqueira MM, Wang D, Mak GC, Zhang W, Daniels RS, Hurt AC, Tashiro M. Global update on the susceptibility of human influenza viruses to neuraminidase inhibitors, 2013–2014. Antiviral Res 2015; 117:27-38. [PMID: 25721488 PMCID: PMC9036627 DOI: 10.1016/j.antiviral.2015.02.003] [Citation(s) in RCA: 118] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Revised: 01/28/2015] [Accepted: 02/06/2015] [Indexed: 12/25/2022]
Abstract
Four World Health Organization (WHO) Collaborating Centres for Reference and Research on Influenza and one WHO Collaborating Centre for the Surveillance, Epidemiology and Control of Influenza (WHO CCs) tested 10,641 viruses collected by WHO-recognized National Influenza Centres between May 2013 and May 2014 to determine 50% inhibitory concentration (IC50) data for neuraminidase inhibitors (NAIs) oseltamivir, zanamivir, peramivir and laninamivir. In addition, neuraminidase (NA) sequence data, available from the WHO CCs and from sequence databases (n = 3206), were screened for amino acid substitutions associated with reduced NAI susceptibility. Ninety-five per cent of the viruses tested by the WHO CCs were from three WHO regions: Western Pacific, the Americas and Europe. Approximately 2% (n = 172) showed highly reduced inhibition (HRI) against at least one of the four NAIs, commonly oseltamivir, while 0.3% (n = 32) showed reduced inhibition (RI). Those showing HRI were A(H1N1)pdm09 with NA H275Y (n = 169), A(H3N2) with NA E119V (n = 1), B/Victoria-lineage with NA E117G (n = 1) and B/Yamagata-lineage with NA H273Y (n = 1); amino acid position numbering is A subtype and B type specific. Although approximately 98% of circulating viruses tested during the 2013–2014 period were sensitive to all four NAIs, a large community cluster of A(H1N1)pdm09 viruses with the NA H275Y substitution from patients with no previous exposure to antivirals was detected in Hokkaido, Japan. Significant numbers of A(H1N1)pdm09 NA H275Y viruses were also detected in China and the United States: phylogenetic analyses showed that the Chinese viruses were similar to those from Japan, while the United States viruses clustered separately from those of the Hokkaido outbreak, indicative of multiple resistance-emergence events. Consequently, global surveillance of influenza antiviral susceptibility should be continued from a public health perspective.
Collapse
Affiliation(s)
- Emi Takashita
- World Health Organization Collaborating Centre for Reference and Research on Influenza, National Institute of Infectious Diseases, Gakuen 4-7-1, Musashimurayama, Tokyo 208-0011, Japan.
| | - Adam Meijer
- National Institute for Public Health and the Environment, PO Box 1, 3720 BA Bilthoven, The Netherlands.
| | - Angie Lackenby
- Public Health England Colindale, 61 Colindale Avenue, London NW9 5EQ, United Kingdom.
| | - Larisa Gubareva
- World Health Organization Collaborating Centre for the Surveillance, Epidemiology and Control of Influenza, Centers for Disease Control and Prevention, 1600 Clifton RD NE, MS-G16 Atlanta, GA, United States.
| | - Helena Rebelo-de-Andrade
- Instituto Nacional de Saúde, Av. Padre Cruz, 1649-016 Lisboa, Portugal; Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal.
| | - Terry Besselaar
- Global Influenza Programme, World Health Organization, Avenue Appia 20, 1211 Geneva 27, Switzerland.
| | - Alicia Fry
- World Health Organization Collaborating Centre for the Surveillance, Epidemiology and Control of Influenza, Centers for Disease Control and Prevention, 1600 Clifton RD NE, MS-G16 Atlanta, GA, United States.
| | - Vicky Gregory
- World Health Organization Collaborating Centre for Reference and Research on Influenza, MRC-National Institute for Medical Research, The Ridgeway, Mill Hill, London NW7 1AA, United Kingdom.
| | - Sook-Kwan Leang
- World Health Organization Collaborating Centre for Reference and Research on Influenza, VIDRL, At the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia.
| | - Weijuan Huang
- World Health Organization Collaborating Centre for Reference and Research on Influenza, Chinese National Influenza Center, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, 155 Changbai Road, Changping District, Beijing 102206, China.
| | - Janice Lo
- Public Health Laboratory Centre, 382 Nam Cheong Street, Shek Kip Mei, Kowloon, Hong Kong, China.
| | - Dmitriy Pereyaslov
- Division of Communicable Diseases, Health Security, & Environment, World Health Organization Regional Office for Europe, UN City, Marmorvej 51, DK-2100 Copenhagen Ø, Denmark.
| | - Marilda M Siqueira
- Respiratory Viruses Laboratory/IOC, FIOCRUZ, Av Brasil, 4365 Rio de Janeiro, Brazil.
| | - Dayan Wang
- World Health Organization Collaborating Centre for Reference and Research on Influenza, Chinese National Influenza Center, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, 155 Changbai Road, Changping District, Beijing 102206, China.
| | - Gannon C Mak
- Public Health Laboratory Centre, 382 Nam Cheong Street, Shek Kip Mei, Kowloon, Hong Kong, China.
| | - Wenqing Zhang
- Global Influenza Programme, World Health Organization, Avenue Appia 20, 1211 Geneva 27, Switzerland.
| | - Rod S Daniels
- World Health Organization Collaborating Centre for Reference and Research on Influenza, MRC-National Institute for Medical Research, The Ridgeway, Mill Hill, London NW7 1AA, United Kingdom.
| | - Aeron C Hurt
- World Health Organization Collaborating Centre for Reference and Research on Influenza, VIDRL, At the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia; University of Melbourne, Melbourne School of Population and Global Health, Melbourne, VIC 3010, Australia.
| | - Masato Tashiro
- World Health Organization Collaborating Centre for Reference and Research on Influenza, National Institute of Infectious Diseases, Gakuen 4-7-1, Musashimurayama, Tokyo 208-0011, Japan.
| |
Collapse
|
36
|
Abstract
The ability of an influenza virus to transmit efficiently from human-to-human is a major factor in determining the epidemiological impact of that strain. The use of a relevant animal model to identify viral determinants of transmission, as well as host and environmental factors affecting transmission efficiency, is therefore critical for public health. The characterization of newly emerging influenza viruses in terms of their potential to transmit in a mammalian host is furthermore an important part of pandemic risk assessment. For these reasons, a guinea pig model of influenza virus transmission was developed in 2006. The guinea pig provides an important alternative to preexisting models for influenza. Most influenza viruses do not readily transmit among mice. Ferrets, while highly relevant, are expensive and can be difficult to obtain in high numbers. Moreover, it is generally accepted that efforts to accurately model human disease are strengthened by the use of multiple animal species. Herein, we provide an overview of influenza virus infectivity, growth, and transmission in the guinea pig and highlight knowledge gained on the topic of influenza virus transmission using the guinea pig model.
Collapse
Affiliation(s)
- Anice C Lowen
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
| | | | | |
Collapse
|
37
|
Characterization of a large cluster of influenza A(H1N1)pdm09 viruses cross-resistant to oseltamivir and peramivir during the 2013-2014 influenza season in Japan. Antimicrob Agents Chemother 2015; 59:2607-17. [PMID: 25691635 DOI: 10.1128/aac.04836-14] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 02/10/2015] [Indexed: 12/17/2022] Open
Abstract
Between September 2013 and July 2014, 2,482 influenza 2009 pandemic A(H1N1) [A(H1N1)pdm09] viruses were screened in Japan for the H275Y substitution in their neuraminidase (NA) protein, which confers cross-resistance to oseltamivir and peramivir. We found that a large cluster of the H275Y mutant virus was present prior to the main influenza season in Sapporo /: Hokkaido, with the detection rate for this mutant virus reaching 29% in this area. Phylogenetic analysis suggested the clonal expansion of a single mutant virus in Sapporo /: Hokkaido. To understand the reason for this large cluster, we examined the in vitro and in vivo properties of the mutant virus. We found that it grew well in cell culture, with growth comparable to that of the wild-type virus. The cluster virus also replicated well in the upper respiratory tract of ferrets and was transmitted efficiently between ferrets by way of respiratory droplets. Almost all recently circulating A(H1N1)pdm09 viruses, including the cluster virus, possessed two substitutions in NA, V241I and N369K, which are known to increase replication and transmission fitness. A structural analysis of NA predicted that a third substitution (N386K) in the NA of the cluster virus destabilized the mutant NA structure in the presence of the V241I and N369K substitutions. Our results suggest that the cluster virus retained viral fitness to spread among humans and, accordingly, caused the large cluster in Sapporo/Hokkaido. However, the mutant NA structure was less stable than that of the wild-type virus. Therefore, once the wild-type virus began to circulate in the community, the mutant virus could not compete and faded out.
Collapse
|
38
|
Permissive changes in the neuraminidase play a dominant role in improving the viral fitness of oseltamivir-resistant seasonal influenza A(H1N1) strains. Antiviral Res 2014; 114:57-61. [PMID: 25512229 DOI: 10.1016/j.antiviral.2014.12.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Revised: 11/26/2014] [Accepted: 12/03/2014] [Indexed: 11/24/2022]
Abstract
Permissive neuraminidase (NA) substitutions such as R222Q, V234M and D344N have facilitated the emergence and worldwide spread of oseltamivir-resistant influenza A/Brisbane/59/2007 (H1N1)-H275Y viruses. However, the potential contribution of genetic changes in other viral segments on viral fitness remains poorly investigated. A series of recombinant A(H1N1)pdm09 and A/WSN/33 7:1 reassortants containing the wild-type (WT) A/Brisbane/59/2007 NA gene or its single (H275Y) and double (H275Y/Q222R, H275Y/M234V and H275Y/N344D) variants were generated and their replicative properties were assessed in vitro. The Q222R reversion substitution significantly reduced viral titers when evaluated in both A(H1N1)pdm09 and A/WSN/33 backgrounds. The permissive role of the R222Q was further confirmed using A/WSN/33 7:1 reassortants containing the NA gene of the oseltamivir-susceptible or oseltamivir-resistant influenza A/Mississippi/03/2001 strains. Therefore, NA permissive substitutions play a dominant role for improving viral replication of oseltamivir-resistant A (H1N1)-H275Y viruses in vitro.
Collapse
|
39
|
Behera AK, Basu S, Cherian SS. Molecular mechanism of the enhanced viral fitness contributed by secondary mutations in the hemagglutinin protein of oseltamivir resistant H1N1 influenza viruses: modeling studies of antibody and receptor binding. Gene 2014; 557:19-27. [PMID: 25479009 DOI: 10.1016/j.gene.2014.12.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Revised: 11/26/2014] [Accepted: 12/01/2014] [Indexed: 11/25/2022]
Abstract
The envelope protein hemagglutinin (HA) of influenza viruses is primarily associated with host antibody and receptor interactions. The HA protein is known to maintain a functional balance with neuraminidase (NA), the other major envelope protein. Prior to 2007-2008, human seasonal H1N1 viruses possessing the NA H274Y mutation, which confers oseltamivir resistance, generally had low growth capability. Subsequently, secondary mutations that compensate for the deleterious effect of the NA H274Y mutation have been identified. The molecular mechanism of how the defect could be counteracted by these secondary mutations is not fully understood. We studied here the effect of three such mutations (T86K, K144E and R192K) in the HA protein, which are located at either the HA receptor binding site or in the H1N1 antigenic sites. Molecular docking and dynamics studies showed that, of the three mutations, the R192K mutation could have mediated neutralizing antibody escape and decreased receptor binding affinity, either or both of which may have contributed to increased viral fitness. The study suggests the molecular basis of enhanced viral fitness induced by secondary mutations in the evolution of oseltamivir-resistant influenza strains.
Collapse
Affiliation(s)
- Abhisek Kumar Behera
- Bioinformatics Group, National Institute of Virology, 20-A, Dr. Ambedkar Road, Post Box No. 11, Pune 411001, Maharashtra, India.
| | - Sushmita Basu
- Bioinformatics Group, National Institute of Virology, 20-A, Dr. Ambedkar Road, Post Box No. 11, Pune 411001, Maharashtra, India.
| | - Sarah S Cherian
- Bioinformatics Group, National Institute of Virology, 20-A, Dr. Ambedkar Road, Post Box No. 11, Pune 411001, Maharashtra, India.
| |
Collapse
|
40
|
Bank C, Hietpas RT, Jensen JD, Bolon DNA. A systematic survey of an intragenic epistatic landscape. Mol Biol Evol 2014; 32:229-38. [PMID: 25371431 DOI: 10.1093/molbev/msu301] [Citation(s) in RCA: 94] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Mutations are the source of evolutionary variation. The interactions of multiple mutations can have important effects on fitness and evolutionary trajectories. We have recently described the distribution of fitness effects of all single mutations for a nine-amino-acid region of yeast Hsp90 (Hsp82) implicated in substrate binding. Here, we report and discuss the distribution of intragenic epistatic effects within this region in seven Hsp90 point mutant backgrounds of neutral to slightly deleterious effect, resulting in an analysis of more than 1,000 double mutants. We find negative epistasis between substitutions to be common, and positive epistasis to be rare--resulting in a pattern that indicates a drastic change in the distribution of fitness effects one step away from the wild type. This can be well explained by a concave relationship between phenotype and genotype (i.e., a concave shape of the local fitness landscape), suggesting mutational robustness intrinsic to the local sequence space. Structural analyses indicate that, in this region, epistatic effects are most pronounced when a solvent-inaccessible position is involved in the interaction. In contrast, all 18 observations of positive epistasis involved at least one mutation at a solvent-exposed position. By combining the analysis of evolutionary and biophysical properties of an epistatic landscape, these results contribute to a more detailed understanding of the complexity of protein evolution.
Collapse
Affiliation(s)
- Claudia Bank
- School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland
| | - Ryan T Hietpas
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA
| | - Jeffrey D Jensen
- School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland
| | - Daniel N A Bolon
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA
| |
Collapse
|
41
|
Duan S, Govorkova EA, Bahl J, Zaraket H, Baranovich T, Seiler P, Prevost K, Webster RG, Webby RJ. Epistatic interactions between neuraminidase mutations facilitated the emergence of the oseltamivir-resistant H1N1 influenza viruses. Nat Commun 2014; 5:5029. [PMID: 25297528 PMCID: PMC4197134 DOI: 10.1038/ncomms6029] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2014] [Accepted: 08/19/2014] [Indexed: 01/06/2023] Open
Abstract
Oseltamivir-resistant H1N1 influenza viruses carrying the H275Y neuraminidase mutation predominated worldwide during the 2007–2009 seasons. While several neuraminidase substitutions were found to be necessary to counteract the adverse effects of H275Y, the order and impact of evolutionary events involved remain elusive. Here, we reconstruct H1N1 neuraminidase phylogeny during 1999–2009, estimate the timing and order of crucial amino acid changes, and evaluate their impact on the biological outcome of the H275Y mutation. Of the twelve neuraminidase substitutions that occurred during 1999–2009, five (chronologically, V234M, R222Q, K329E, D344N, H275Y, and D354G) are necessary for maintaining full neuraminidase function in the presence of the H275Y mutation by altering protein accumulation or enzyme affinity/activity. The sequential emergence and cumulative effects of these mutations clearly illustrate a role for epistasis in shaping the emergence and subsequent evolution of a drug-resistant virus population, which can be useful in understanding emergence of novel viral phenotypes of influenza.
Collapse
Affiliation(s)
- Susu Duan
- Department of Infectious Diseases, St Jude Children's Research Hospital, 262 Danny Thomas Place, Mail Stop 330, Memphis, Tennessee 38105, USA
| | - Elena A Govorkova
- Department of Infectious Diseases, St Jude Children's Research Hospital, 262 Danny Thomas Place, Mail Stop 330, Memphis, Tennessee 38105, USA
| | - Justin Bahl
- 1] School of Public Health, The University of Texas Health Science Center at Houston, 1200 Pressler Street, Houston, Texas 77030, USA [2] Program in Emerging Infectious Diseases, Duke-National University of Singapore Graduate Medical School, 8 College Road, Singapore 169857, Singapore
| | - Hassan Zaraket
- Department of Infectious Diseases, St Jude Children's Research Hospital, 262 Danny Thomas Place, Mail Stop 330, Memphis, Tennessee 38105, USA
| | - Tatiana Baranovich
- Department of Infectious Diseases, St Jude Children's Research Hospital, 262 Danny Thomas Place, Mail Stop 330, Memphis, Tennessee 38105, USA
| | - Patrick Seiler
- Department of Infectious Diseases, St Jude Children's Research Hospital, 262 Danny Thomas Place, Mail Stop 330, Memphis, Tennessee 38105, USA
| | - Kristi Prevost
- Department of Infectious Diseases, St Jude Children's Research Hospital, 262 Danny Thomas Place, Mail Stop 330, Memphis, Tennessee 38105, USA
| | - Robert G Webster
- Department of Infectious Diseases, St Jude Children's Research Hospital, 262 Danny Thomas Place, Mail Stop 330, Memphis, Tennessee 38105, USA
| | - Richard J Webby
- Department of Infectious Diseases, St Jude Children's Research Hospital, 262 Danny Thomas Place, Mail Stop 330, Memphis, Tennessee 38105, USA
| |
Collapse
|
42
|
Evolution of oseltamivir resistance mutations in Influenza A(H1N1) and A(H3N2) viruses during selection in experimentally infected mice. Antimicrob Agents Chemother 2014; 58:6398-405. [PMID: 25114143 DOI: 10.1128/aac.02956-14] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The evolution of oseltamivir resistance mutations during selection through serial passages in animals is still poorly described. Herein, we assessed the evolution of neuraminidase (NA) and hemagglutinin (HA) genes of influenza A/WSN/33 (H1N1) and A/Victoria/3/75 (H3N2) viruses recovered from the lungs of experimentally infected BALB/c mice receiving suboptimal doses (0.05 and 1 mg/kg of body weight/day) of oseltamivir over two generations. The traditional phenotypic and genotypic methods as well as deep-sequencing analysis were used to characterize the potential selection of mutations and population dynamics of oseltamivir-resistant variants. No oseltamivir-resistant NA or HA changes were detected in the recovered A/WSN/33 viruses. However, we observed a positive selection of the I222T NA substitution in the recovered A/Victoria/3/75 viruses, with a frequency increasing over time and with an oseltamivir concentration from 4% in the initial pretherapy inoculum up to 28% after two lung passages. Although the presence of mixed I222T viral populations in mouse lungs only led to a minimal increase in oseltamivir 50% enzyme-inhibitory concentrations (IC50s) (by a mean of 5.7-fold) compared to that of the baseline virus, the expressed recombinant A/Victoria/3/75 I222T NA protein displayed a 16-fold increase in the oseltamivir IC50 level compared to that of the recombinant wild type (WT). In conclusion, the combination of serial in vivo passages under neuraminidase inhibitor (NAI) pressure and temporal deep-sequencing analysis enabled, for the first time, the identification and selection of the oseltamivir-resistant I222T NA mutation in an influenza H3N2 virus. Additional in vivo selection experiments with other antivirals and drug combinations might provide important information on the evolution of antiviral resistance in influenza viruses.
Collapse
|
43
|
Meijer A, Rebelo-de-Andrade H, Correia V, Besselaar T, Drager-Dayal R, Fry A, Gregory V, Gubareva L, Kageyama T, Lackenby A, Lo J, Odagiri T, Pereyaslov D, Siqueira MM, Takashita E, Tashiro M, Wang D, Wong S, Zhang W, Daniels RS, Hurt AC. Global update on the susceptibility of human influenza viruses to neuraminidase inhibitors, 2012-2013. Antiviral Res 2014; 110:31-41. [PMID: 25043638 PMCID: PMC8851378 DOI: 10.1016/j.antiviral.2014.07.001] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Revised: 06/30/2014] [Accepted: 07/01/2014] [Indexed: 01/14/2023]
Abstract
Emergence of influenza viruses with reduced susceptibility to neuraminidase inhibitors (NAIs) is sporadic, often follows exposure to NAIs, but occasionally occurs in the absence of NAI pressure. The emergence and global spread in 2007/2008 of A(H1N1) influenza viruses showing clinical resistance to oseltamivir due to neuraminidase (NA) H275Y substitution, in the absence of drug pressure, warrants continued vigilance and monitoring for similar viruses. Four World Health Organization (WHO) Collaborating Centres for Reference and Research on Influenza and one WHO Collaborating Centre for the Surveillance, Epidemiology and Control of Influenza (WHO CCs) tested 11,387 viruses collected by WHO-recognized National Influenza Centres (NIC) between May 2012 and May 2013 to determine 50% inhibitory concentration (IC50) data for oseltamivir, zanamivir, peramivir and laninamivir. The data were evaluated using normalized IC50 fold-changes rather than raw IC50 data. Nearly 90% of the 11,387 viruses were from three WHO regions: Western Pacific, the Americas and Europe. Only 0.2% (n = 27) showed highly reduced inhibition (HRI) against at least one of the four NAIs, usually oseltamivir, while 0.3% (n = 39) showed reduced inhibition (RI). NA sequence data, available from the WHO CCs and from sequence databases (n = 3661), were screened for amino acid substitutions associated with reduced NAI susceptibility. Those showing HRI were A(H1N1)pdm09 with NA H275Y (n = 18), A(H3N2) with NA E119V (n = 3) or NA R292K (n = 1) and B/Victoria-lineage with NA H273Y (n = 2); amino acid position numbering is A subtype and B type specific. Overall, approximately 99% of circulating viruses tested during the 2012–2013 period were sensitive to all four NAIs. Consequently, these drugs remain an appropriate choice for the treatment and prophylaxis of influenza virus infections.
Collapse
Affiliation(s)
- Adam Meijer
- National Institute for Public Health and the Environment, PO Box 1, 3720 BA Bilthoven, The Netherlands.
| | - Helena Rebelo-de-Andrade
- Instituto Nacional de Saúde, Av. Padre Cruz, 1649-016 Lisboa, Portugal; Faculdade de Farmácia, Universidade de Lisboa, Portugal.
| | - Vanessa Correia
- Instituto Nacional de Saúde, Av. Padre Cruz, 1649-016 Lisboa, Portugal; Faculdade de Farmácia, Universidade de Lisboa, Portugal.
| | - Terry Besselaar
- Global Influenza Programme, World Health Organization, Avenue Appia 20, 1211 Geneva 27, Switzerland.
| | - Renu Drager-Dayal
- Global Influenza Programme, World Health Organization, Avenue Appia 20, 1211 Geneva 27, Switzerland.
| | - Alicia Fry
- World Health Organization Collaborating Centre for the Surveillance, Epidemiology and Control of Influenza, Centers for Disease Control and Prevention, 1600 Clifton RD NE, MS-G16 Atlanta, GA, United States.
| | - Vicky Gregory
- World Health Organization Collaborating Centre for Reference and Research on Influenza, MRC-National Institute for Medical Research, The Ridgeway, Mill Hill, London NW7 1AA, United Kingdom.
| | - Larisa Gubareva
- World Health Organization Collaborating Centre for the Surveillance, Epidemiology and Control of Influenza, Centers for Disease Control and Prevention, 1600 Clifton RD NE, MS-G16 Atlanta, GA, United States.
| | - Tsutomu Kageyama
- World Health Organization Collaborating Centre for Reference and Research on Influenza, National Institute of Infectious Diseases, Gakuen 4-7-1, Musashimurayama, Tokyo 208-0011, Japan.
| | - Angie Lackenby
- Public Health England Colindale, 61 Colindale Avenue, London NW9 5EQ, United Kingdom.
| | - Janice Lo
- Public Health Laboratory Centre, 382 Nam Cheong Street, Shek Kip Mei, Kowloon, Hong Kong, China.
| | - Takato Odagiri
- World Health Organization Collaborating Centre for Reference and Research on Influenza, National Institute of Infectious Diseases, Gakuen 4-7-1, Musashimurayama, Tokyo 208-0011, Japan.
| | - Dmitriy Pereyaslov
- Division of Communicable Diseases, Health Security, & Environment, World Health Organization Regional Office for Europe, UN City, Marmorvej 51, DK-2100 Copenhagen Ø, Denmark.
| | - Marilda M Siqueira
- Respiratory Viruses Laboratory/IOC, FIOCRUZ Av Brasil, 4365 Rio de Janeiro, Brazil.
| | - Emi Takashita
- World Health Organization Collaborating Centre for Reference and Research on Influenza, National Institute of Infectious Diseases, Gakuen 4-7-1, Musashimurayama, Tokyo 208-0011, Japan.
| | - Masato Tashiro
- World Health Organization Collaborating Centre for Reference and Research on Influenza, National Institute of Infectious Diseases, Gakuen 4-7-1, Musashimurayama, Tokyo 208-0011, Japan.
| | - Dayan Wang
- World Health Organization Collaborating Centre for Reference and Research on Influenza, Chinese National Influenza Center, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, 155 Changbai Road, Changping District, Beijing 102206, China.
| | - Sun Wong
- Public Health Laboratory Centre, 382 Nam Cheong Street, Shek Kip Mei, Kowloon, Hong Kong, China.
| | - Wenqing Zhang
- Global Influenza Programme, World Health Organization, Avenue Appia 20, 1211 Geneva 27, Switzerland.
| | - Rod S Daniels
- World Health Organization Collaborating Centre for Reference and Research on Influenza, MRC-National Institute for Medical Research, The Ridgeway, Mill Hill, London NW7 1AA, United Kingdom.
| | - Aeron C Hurt
- World Health Organization Collaborating Centre for Reference and Research on Influenza, VIDRL, At the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia; University of Melbourne, Melbourne School of Population and Global Health, Melbourne, VIC 3010, Australia.
| |
Collapse
|
44
|
Park S, Kim JI, Lee I, Lee S, Hwang MW, Bae JY, Heo J, Kim D, Jang SI, Kim H, Cheong HJ, Song JW, Song KJ, Baek LJ, Park MS. Combination effects of peramivir and favipiravir against oseltamivir-resistant 2009 pandemic influenza A(H1N1) infection in mice. PLoS One 2014; 9:e101325. [PMID: 24992479 PMCID: PMC4081560 DOI: 10.1371/journal.pone.0101325] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Accepted: 06/04/2014] [Indexed: 12/18/2022] Open
Abstract
Antiviral drugs are being used for therapeutic purposes against influenza illness in humans. However, antiviral-resistant variants often nullify the effectiveness of antivirals. Combined medications, as seen in the treatment of cancers and other infectious diseases, have been suggested as an option for the control of antiviral-resistant influenza viruses. Here, we evaluated the therapeutic value of combination therapy against oseltamivir-resistant 2009 pandemic influenza H1N1 virus infection in DBA/2 mice. Mice were treated for five days with favipiravir and peramivir starting 4 hours after lethal challenge. Compared with either monotherapy, combination therapy saved more mice from viral lethality and resulted in increased antiviral efficacy in the lungs of infected mice. Furthermore, the synergism between the two antivirals, which was consistent with the survival outcomes of combination therapy, indicated that favipiravir could serve as a critical agent of combination therapy for the control of oseltamivir-resistant strains. Our results provide new insight into the feasibility of favipiravir in combination therapy against oseltamivir-resistant influenza virus infection.
Collapse
Affiliation(s)
- Sehee Park
- Department of Microbiology, College of Medicine, and the Institute for Viral Diseases, Korea University, Seoul, Republic of Korea; Department of Microbiology, College of Medicine, Hallym University, Chuncheon, Gangwon-do, Republic of Korea
| | - Jin Il Kim
- Department of Microbiology, College of Medicine, and the Institute for Viral Diseases, Korea University, Seoul, Republic of Korea; Department of Microbiology, College of Medicine, Hallym University, Chuncheon, Gangwon-do, Republic of Korea
| | - Ilseob Lee
- Department of Microbiology, College of Medicine, and the Institute for Viral Diseases, Korea University, Seoul, Republic of Korea; Department of Microbiology, College of Medicine, Hallym University, Chuncheon, Gangwon-do, Republic of Korea
| | - Sangmoo Lee
- Department of Microbiology, College of Medicine, and the Institute for Viral Diseases, Korea University, Seoul, Republic of Korea; Department of Microbiology, College of Medicine, Hallym University, Chuncheon, Gangwon-do, Republic of Korea
| | - Min-Woong Hwang
- Department of Microbiology, College of Medicine, and the Institute for Viral Diseases, Korea University, Seoul, Republic of Korea; Department of Microbiology, College of Medicine, Hallym University, Chuncheon, Gangwon-do, Republic of Korea
| | - Joon-Yong Bae
- Department of Microbiology, College of Medicine, and the Institute for Viral Diseases, Korea University, Seoul, Republic of Korea; Department of Microbiology, College of Medicine, Hallym University, Chuncheon, Gangwon-do, Republic of Korea
| | - Jun Heo
- Department of Microbiology, College of Medicine, and the Institute for Viral Diseases, Korea University, Seoul, Republic of Korea; Department of Microbiology, College of Medicine, Hallym University, Chuncheon, Gangwon-do, Republic of Korea
| | - Donghwan Kim
- Department of Microbiology, College of Medicine, Hallym University, Chuncheon, Gangwon-do, Republic of Korea
| | - Seok-Il Jang
- Department of Microbiology, College of Medicine, Hallym University, Chuncheon, Gangwon-do, Republic of Korea
| | - Hyejin Kim
- Department of Microbiology, College of Medicine, and the Institute for Viral Diseases, Korea University, Seoul, Republic of Korea
| | - Hee Jin Cheong
- Division of Infectious Diseases, Korea University Guro Hospital, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Jin-Won Song
- Department of Microbiology, College of Medicine, and the Institute for Viral Diseases, Korea University, Seoul, Republic of Korea
| | - Ki-Joon Song
- Department of Microbiology, College of Medicine, and the Institute for Viral Diseases, Korea University, Seoul, Republic of Korea
| | - Luck Ju Baek
- Department of Microbiology, College of Medicine, and the Institute for Viral Diseases, Korea University, Seoul, Republic of Korea
| | - Man-Seong Park
- Department of Microbiology, College of Medicine, and the Institute for Viral Diseases, Korea University, Seoul, Republic of Korea; Department of Microbiology, College of Medicine, Hallym University, Chuncheon, Gangwon-do, Republic of Korea
| |
Collapse
|
45
|
Boivin G. Detection and management of antiviral resistance for influenza viruses. Influenza Other Respir Viruses 2014; 7 Suppl 3:18-23. [PMID: 24215378 DOI: 10.1111/irv.12176] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/11/2013] [Indexed: 12/25/2022] Open
Abstract
Neuraminidase inhibitors (NAIs) are first-line agents for the treatment and prevention of influenza virus infections. As for other antivirals, the development of resistance to NAIs has become an important concern particularly in the case of A(H1N1) viruses and oseltamivir. The most frequently reported change conferring oseltamivir resistance in that viral context is the H275Y neuraminidase mutation (N1 numbering). Recent studies have shown that, in the presence of the appropriate permissive mutations, the H275Y variant can retain virulence and transmissibility in some viral backgrounds. Most oseltamivir-resistant influenza A virus infections can be managed with the use of inhaled or intravenous zanamivir, another NAI. New NAI compounds and non-neuraminidase agents as well as combination therapies are currently in clinical evaluation for the treatment for severe influenza infections.
Collapse
Affiliation(s)
- Guy Boivin
- CHUQ-CHUL and Laval University, Quebec, QC, Canada
| |
Collapse
|
46
|
Impact of different oseltamivir regimens on treating influenza A virus infection and resistance emergence: insights from a modelling study. PLoS Comput Biol 2014; 10:e1003568. [PMID: 24743564 PMCID: PMC3990489 DOI: 10.1371/journal.pcbi.1003568] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Accepted: 02/26/2014] [Indexed: 12/27/2022] Open
Abstract
Several studies have proven oseltamivir to be efficient in reducing influenza viral titer and symptom intensity. However, the usefulness of oseltamivir can be compromised by the emergence and spread of drug-resistant virus. The selective pressure exerted by different oseltamivir therapy regimens have received little attention. Combining models of drug pharmacokinetics, pharmacodynamics, viral kinetics and symptom dynamics, we explored the efficacy of oseltamivir in reducing both symptoms (symptom efficacy) and viral load (virological efficacy). We simulated samples of 1000 subjects using previously estimated between-subject variability in viral and symptom dynamic parameters to describe the observed heterogeneity in a patient population. We simulated random mutations conferring resistance to oseltamivir. We explored the effect of therapy initiation time, dose, intake frequency and therapy duration on influenza infection, illness dynamics, and emergence of viral resistance. Symptom and virological efficacies were strongly associated with therapy initiation time. The proportion of subjects shedding resistant virus was 27-fold higher when prophylaxis was initiated during the incubation period compared with no treatment. It fell to below 1% when treatment was initiated after symptom onset for twice-a-day intakes. Lower doses and prophylaxis regimens led to lower efficacies and increased risk of resistance emergence. We conclude that prophylaxis initiated during the incubation period is the main factor leading to resistance emergence. Oseltamivir is currently the most commonly used drug against influenza but the emergence and spread of oseltamivir-resistant virus is threatening its usefulness. A previously published study quantified the risk of drug-resistance emergence and spread. In this work we investigate under what conditions drug-resistance is likely to occur and how we can mitigate it. For this purpose, we simulated populations of influenza-infected subjects under different treatment conditions varying drug dose, intake frequency and duration of therapy. We used an approach that mimics the randomness of drug-resistance emergence and allowed for between-subject variability. We measured the effect of treatment on reducing infection and symptoms and on drug-resistance emergence. We found that for subjects starting oseltamivir during the influenza incubation period, the risk of resistance emergence is dramatically increased. Thus, our findings suggest that standard prophylaxis should only be used after exclusion of an influenza infection in the incubation period by use of a rapid test. If existing infection cannot be excluded, then prophylaxis should be done with increased dose, intake frequency and duration in order to avoid emergence of drug-resistant strains and to preserve oseltamivir efficacy.
Collapse
|
47
|
Thangavel RR, Bouvier NM. Animal models for influenza virus pathogenesis, transmission, and immunology. J Immunol Methods 2014; 410:60-79. [PMID: 24709389 PMCID: PMC4163064 DOI: 10.1016/j.jim.2014.03.023] [Citation(s) in RCA: 138] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Revised: 03/22/2014] [Accepted: 03/24/2014] [Indexed: 12/24/2022]
Abstract
In humans, infection with an influenza A or B virus manifests typically as an acute and self-limited upper respiratory tract illness characterized by fever, cough, sore throat, and malaise. However, influenza can present along a broad spectrum of disease, ranging from sub-clinical or even asymptomatic infection to a severe primary viral pneumonia requiring advanced medical supportive care. Disease severity depends upon the virulence of the influenza virus strain and the immune competence and previous influenza exposures of the patient. Animal models are used in influenza research not only to elucidate the viral and host factors that affect influenza disease outcomes in and spread among susceptible hosts, but also to evaluate interventions designed to prevent or reduce influenza morbidity and mortality in man. This review will focus on the three animal models currently used most frequently in influenza virus research - mice, ferrets, and guinea pigs - and discuss the advantages and disadvantages of each.
Collapse
Affiliation(s)
- Rajagowthamee R Thangavel
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
| | - Nicole M Bouvier
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA; Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA.
| |
Collapse
|
48
|
Butler J, Hooper KA, Petrie S, Lee R, Maurer-Stroh S, Reh L, Guarnaccia T, Baas C, Xue L, Vitesnik S, Leang SK, McVernon J, Kelso A, Barr IG, McCaw JM, Bloom JD, Hurt AC. Estimating the fitness advantage conferred by permissive neuraminidase mutations in recent oseltamivir-resistant A(H1N1)pdm09 influenza viruses. PLoS Pathog 2014; 10:e1004065. [PMID: 24699865 PMCID: PMC3974874 DOI: 10.1371/journal.ppat.1004065] [Citation(s) in RCA: 100] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Accepted: 02/27/2014] [Indexed: 01/06/2023] Open
Abstract
Oseltamivir is relied upon worldwide as the drug of choice for the treatment of human influenza infection. Surveillance for oseltamivir resistance is routinely performed to ensure the ongoing efficacy of oseltamivir against circulating viruses. Since the emergence of the pandemic 2009 A(H1N1) influenza virus (A(H1N1)pdm09), the proportion of A(H1N1)pdm09 viruses that are oseltamivir resistant (OR) has generally been low. However, a cluster of OR A(H1N1)pdm09 viruses, encoding the neuraminidase (NA) H275Y oseltamivir resistance mutation, was detected in Australia in 2011 amongst community patients that had not been treated with oseltamivir. Here we combine a competitive mixtures ferret model of influenza infection with a mathematical model to assess the fitness, both within and between hosts, of recent OR A(H1N1)pdm09 viruses. In conjunction with data from in vitro analyses of NA expression and activity we demonstrate that contemporary A(H1N1)pdm09 viruses are now more capable of acquiring H275Y without compromising their fitness, than earlier A(H1N1)pdm09 viruses circulating in 2009. Furthermore, using reverse engineered viruses we demonstrate that a pair of permissive secondary NA mutations, V241I and N369K, confers robust fitness on recent H275Y A(H1N1)pdm09 viruses, which correlated with enhanced surface expression and enzymatic activity of the A(H1N1)pdm09 NA protein. These permissive mutations first emerged in 2010 and are now present in almost all circulating A(H1N1)pdm09 viruses. Our findings suggest that recent A(H1N1)pdm09 viruses are now more permissive to the acquisition of H275Y than earlier A(H1N1)pdm09 viruses, increasing the risk that OR A(H1N1)pdm09 will emerge and spread worldwide.
Collapse
Affiliation(s)
- Jeff Butler
- World Health Organization Collaborating Centre for Reference and Research on Influenza, North Melbourne, Australia
| | - Kathryn A. Hooper
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- Molecular and Cellular Biology Program, University of Washington, Seattle, Washington, United States of America
| | - Stephen Petrie
- Melbourne School of Population and Global Health, The University of Melbourne, Parkville, Australia
| | - Raphael Lee
- Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Sebastian Maurer-Stroh
- Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), Singapore
- National Public Health Laboratory, Communicable Diseases Division Ministry of Health, Singapore
- School of Biological Sciences (SBS), Nanyang Technological University (NTU), Singapore
| | - Lucia Reh
- World Health Organization Collaborating Centre for Reference and Research on Influenza, North Melbourne, Australia
| | - Teagan Guarnaccia
- World Health Organization Collaborating Centre for Reference and Research on Influenza, North Melbourne, Australia
| | - Chantal Baas
- World Health Organization Collaborating Centre for Reference and Research on Influenza, North Melbourne, Australia
- Monash University, School of Applied Sciences, Churchill, Victoria, Australia
| | - Lumin Xue
- World Health Organization Collaborating Centre for Reference and Research on Influenza, North Melbourne, Australia
| | - Sophie Vitesnik
- World Health Organization Collaborating Centre for Reference and Research on Influenza, North Melbourne, Australia
| | - Sook-Kwan Leang
- World Health Organization Collaborating Centre for Reference and Research on Influenza, North Melbourne, Australia
| | - Jodie McVernon
- Melbourne School of Population and Global Health, The University of Melbourne, Parkville, Australia
- Murdoch Childrens Research Institute, The Royal Children's Hospital, Melbourne, Victoria, Australia
| | - Anne Kelso
- World Health Organization Collaborating Centre for Reference and Research on Influenza, North Melbourne, Australia
| | - Ian G. Barr
- World Health Organization Collaborating Centre for Reference and Research on Influenza, North Melbourne, Australia
- Monash University, School of Applied Sciences, Churchill, Victoria, Australia
| | - James M. McCaw
- Melbourne School of Population and Global Health, The University of Melbourne, Parkville, Australia
- Murdoch Childrens Research Institute, The Royal Children's Hospital, Melbourne, Victoria, Australia
| | - Jesse D. Bloom
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Aeron C. Hurt
- World Health Organization Collaborating Centre for Reference and Research on Influenza, North Melbourne, Australia
- Monash University, School of Applied Sciences, Churchill, Victoria, Australia
| |
Collapse
|
49
|
Impact of potential permissive neuraminidase mutations on viral fitness of the H275Y oseltamivir-resistant influenza A(H1N1)pdm09 virus in vitro, in mice and in ferrets. J Virol 2013; 88:1652-8. [PMID: 24257597 DOI: 10.1128/jvi.02681-13] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Neuraminidase (NA) mutations conferring resistance to NA inhibitors (NAIs) generally compromise the fitness of influenza viruses. The only NAI-resistant virus that widely spread in the population, the A/Brisbane/59/2007 (H1N1) strain, contained permissive mutations that restored the detrimental effect caused by the H275Y change. Computational analysis predicted other permissive NA mutations for A(H1N1)pdm09 viruses. Here, we investigated the effect of T289M and N369K mutations on the viral fitness of the A(H1N1)pdm09 H275Y variant. Recombinant wild-type (WT) A(H1N1)pdm09 and the H275Y, H275Y/T289M, H275Y/N369K, and H275Y/V241I/N369K (a natural variant) NA mutants were generated by reverse genetics. Replication kinetics were performed by using ST6GalI-MDCK cells. Virulence was assessed in C57BL/6 mice, and contact transmission was evaluated in ferrets. The H275Y mutation significantly reduced viral titers during the first 12 to 36 h postinfection (p.i.) in vitro. Nevertheless, the WT and H275Y viruses induced comparable mortality rates, weight loss, and lung titers in mice. The T289M mutation eliminated the detrimental effect caused by the H275Y change in vitro while causing greater weight loss and mortality in mice, with significantly higher lung viral titers on days 3 and 6 p.i. than with the H275Y mutant. In index ferrets, the WT, H275Y, H275Y/T289M, and H275Y/V241I/N369K recombinants induced comparable fever, weight loss, and nasal wash viral titers. All tested viruses were transmitted at comparable rates in contact ferrets, with the H275Y/V241I/N369K recombinant demonstrating higher nasal wash viral titers than the H275Y mutant. Permissive mutations may enhance the fitness of A(H1N1)pdm09 H275Y viruses in vitro and in vivo. The emergence of such variants should be carefully monitored.
Collapse
|
50
|
Emergence of an oseltamivir-resistant influenza A/H3N2 virus in an elderly patient receiving a suboptimal dose of antiviral prophylaxis. J Clin Microbiol 2013; 51:4234-6. [PMID: 24088848 DOI: 10.1128/jcm.02659-13] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
We report the emergence of an influenza virus A/H3N2-E119V neuraminidase variant from an elderly patient with renal dysfunction who received a suboptimal dose of oseltamivir prophylaxis. In neuraminidase inhibition assays, the E119V variant showed a 413-fold increase in the 50% inhibitory oseltamivir concentration and grew at titers comparable to those of the wild type in vitro.
Collapse
|