1
|
Gu C, Chen Y, Li H, Wang J, Liu S. Considerations when treating influenza infections with oseltamivir. Expert Opin Pharmacother 2024; 25:1301-1316. [PMID: 38995220 DOI: 10.1080/14656566.2024.2376660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 07/02/2024] [Indexed: 07/13/2024]
Abstract
INTRODUCTION Since the coronavirus disease 2019-mandated social distancing policy has been lifted worldwide, the circulation of influenza is expected to resume. Currently, oseltamivir is approved as the first-line agent for influenza prevention and treatment. AREAS COVERED This paper reviews the updated evidence in the pharmacology, resistance mechanisms, clinical pharmacy management, and real-world data on oseltamivir for influenza. EXPERT OPINION Oseltamivir is an oral prodrug of oseltamivir carboxylate, an influenza A and B neuraminidase inhibitor. Recently, the therapeutic efficacy of oseltamivir has been demonstrated in several trials. Oseltamivir is generally well-tolerated but may lead to neuropsychiatric events and bleeding. Oseltamivir-resistant influenza virus has been associated with the H275Y mutation in the influenza A(H1N1)pdm09 virus, while most strains are still sensitive to oseltamivir. Dose adjustment for oseltamivir should be based on creatinine clearance and body weight in pediatric patients with renal failure. According to real-world data from Nanfang Hospital, the annual number of patients prescribed oseltamivir declined from 35,711 in 2019 to 8,971 in 2020, with marked increases in 2022 (20,213) and 2023 (18,071). Among the 206 inpatients, children aged < 6 years who were treated with oseltamivir had the shortest duration to defervescence.
Collapse
Affiliation(s)
- Chunping Gu
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yi Chen
- Department of Pharmacy, The Seventh Affiliated Hospital, Southern Medical University, Foshan, China
| | - Haobin Li
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory of Drug Metabolism Research and Evaluation, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Jinshen Wang
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory of Drug Metabolism Research and Evaluation, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Shuwen Liu
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory of Drug Metabolism Research and Evaluation, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Southern Medical University, Guangzhou, China
- MOE Innovation Center for Medical Basic Research on Inflammation and Immune Related Diseases, Southern Medical University, Guangzhou, China
| |
Collapse
|
2
|
Batool S, Chokkakula S, Song MS. Influenza Treatment: Limitations of Antiviral Therapy and Advantages of Drug Combination Therapy. Microorganisms 2023; 11:183. [PMID: 36677475 PMCID: PMC9865513 DOI: 10.3390/microorganisms11010183] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 12/23/2022] [Accepted: 01/09/2023] [Indexed: 01/15/2023] Open
Abstract
Influenza infection is serious and debilitating for humans and animals. The influenza virus undergoes incessant mutation, segment recombination, and genome reassortment. As a result, new epidemics and pandemics are expected to emerge, making the elimination challenging of the disease. Antiviral therapy has been used for the treatment of influenza since the development of amantadine in the 1960s; however, its use is hampered by the emergence of novel strains and the development of drug resistance. Thus, combinational therapy with two or more antivirals or immunomodulators with different modes of action is the optimal strategy for the effective treatment of influenza infection. In this review, we describe current options for combination therapy, their performance, and constraints imposed by resistance, calling attention to the advantages of combination therapy against severe influenza infections. We also discuss the challenges of influenza therapy and the limitations of approved antiviral drugs.
Collapse
Affiliation(s)
| | | | - Min-Suk Song
- Department of Microbiology, Chungbuk National University, College of Medicine and Medical Research Institute, Cheongju 28644, Chungbuk, Republic of Korea
| |
Collapse
|
3
|
Farah EM, Amine S, Ahmad S, Nonlaopon K, Allali K. Theoretical and numerical results of a stochastic model describing resistance and non-resistance strains of influenza. EUROPEAN PHYSICAL JOURNAL PLUS 2022; 137:1169. [PMID: 36310610 PMCID: PMC9589591 DOI: 10.1140/epjp/s13360-022-03302-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 09/18/2022] [Indexed: 06/16/2023]
Abstract
In this world, there are several acute viral infections. One of them is influenza, a respiratory disease caused by the influenza virus. Stochastic modelling of infectious diseases is now a popular topic in the current century. Several stochastic epidemiological models have been constructed in the research papers. In the present article, we offer a stochastic two-strain influenza epidemic model that includes both resistant and non-resistance strains. We demonstrate both the existence and uniqueness of the global positive solution using the stochastic Lyapunov function theory. The extinction of our research sickness results from favourable circumstances. Additionally, the infection's persistence in the mean is demonstrated. Finally, to demonstrate how well our theoretical analysis performs, various noise disturbances are simulated numerically.
Collapse
Affiliation(s)
- El Mehdi Farah
- Laboratory of Mathematics , Computer Science and Applications, Faculty of Sciences and Technologies, Hassan II University of Casablanca, PO Box 146, 20650 Mohammedia, Morocco
| | - Saida Amine
- Laboratory of Mathematics , Computer Science and Applications, Faculty of Sciences and Technologies, Hassan II University of Casablanca, PO Box 146, 20650 Mohammedia, Morocco
| | - Shabir Ahmad
- Department of Mathematics, University of Malakand, Chakdara, Dir Lower, Khyber Pakhtunkhwa Pakistan
| | - Kamsing Nonlaopon
- Present Address: Department of Mathematics, Faculty of Science, Khon Kaen University, Khon Kaen, 40002 Thailand
| | - Karam Allali
- Laboratory of Mathematics , Computer Science and Applications, Faculty of Sciences and Technologies, Hassan II University of Casablanca, PO Box 146, 20650 Mohammedia, Morocco
| |
Collapse
|
4
|
Saim-Mamoun A, Abed Y, Carbonneau J, Boivin G. Generation and Characterization of Drug-Resistant Influenza B Viruses Selected In Vitro with Baloxavir Acid. Pathogens 2022; 11:pathogens11091048. [PMID: 36145480 PMCID: PMC9505253 DOI: 10.3390/pathogens11091048] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 08/04/2022] [Accepted: 09/08/2022] [Indexed: 11/16/2022] Open
Abstract
Baloxavir marboxil (BXM) is an antiviral drug that targets the endonuclease of the influenza polymerase acidic (PA) protein. Antiviral resistance, mainly mediated by the I38T PA substitution, readily occurs in both A(H1N1) and A(H3N2) viruses following a single dose of BXM. Influenza B resistance to BXM remains poorly documented. We aimed to generate baloxavir-resistant contemporary influenza B/Yamagata/16/1988- and B/Victoria/2/1987-like viruses by in vitro passages under baloxavir acid (BXA) pressure to identify resistance mutations and to characterize the fitness of drug-resistant variants. Influenza B/Phuket/3073/2013 recombinant virus (rg-PKT13, a B/Yamagata/16/1988-like virus) and B/Quebec/MCV-11/2019 (MCV19, a B/Victoria/2/1987-like isolate) were passaged in ST6GalI-MDCK cells in the presence of increasing concentrations of BXA. At defined passages, viral RNA was extracted for sequencing the PA gene. The I38T PA substitution was selected in MCV19 after six passages in presence of BXA whereas no PA change was detected in rg-PKT13. The I38T substitution increased the BXA IC50 value by 13.7-fold in the MCV19 background and resulted in reduced viral titers compared to the wild type (WT) at early time points in ST6GalI-MDCK and at all time-points in human epithelial cells. By contrast, the I38T substitution had no impact on MCV19 polymerase activity, and this mutation was genetically stable over four passages. In conclusion, our results show a similar pathway of resistance to BXA in influenza B viruses highlighting the major role of the I38T PA substitution and suggest that I38T may differently impact the fitness of influenza variants depending on the viral type, subtype, or lineage.
Collapse
|
5
|
Alzabut J, Gao Z, Hussain S, Madi EN, Khan H. Stochastic dynamics of influenza infection: Qualitative analysis and numerical results. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2022; 19:10316-10331. [PMID: 36031995 DOI: 10.3934/mbe.2022482] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
In this paper, a novel influenza $ \mathcal{S}\mathcal{I}_N\mathcal{I}_R\mathcal{R} $ model with white noise is investigated. According to the research, white noise has a significant impact on the disease. First, we explain that there is global existence and positivity to the solution. Then we show that the stochastic basic reproduction $ {{\underset{\scriptscriptstyle\centerdot}{\text{R}}}} {_r} $ is a threshold that determines whether the disease is cured or persists. When the noise intensity is high, we get $ {{\underset{\scriptscriptstyle\centerdot}{\text{R}}}}{_r} < 1 $ and the disease goes away; when the white noise intensity is low, we get $ {{\underset{\scriptscriptstyle\centerdot}{\text{R}}}}{_r} > 1 $, and a sufficient condition for the existence of a stationary distribution is obtained, which suggests that the disease is still there. However, the main objective of the study is to produce a stochastic analogue of the deterministic model that we analyze using numerical simulations to get views on the infection dynamics in a stochastic environment that we can relate to the deterministic context.
Collapse
Affiliation(s)
- Jehad Alzabut
- Department of Mathematics and Sciences, Prince Sultan University, Riyadh 11586, Saudi Arabia
- Department of Industrial Engineering, OSTİM Technical University, Ankara 06374, Türkiye
| | - Zhengming Gao
- School of computer engineering, Jingchu University of Technology, Jingmen 448000, China
| | - Shah Hussain
- Faculty of Informatics and Computing, Universiti Sultan Zainal Abidin (UniSZA), Besut Campus, Terengganu, Malaysia
| | - Elissa Nadia Madi
- Faculty of Informatics and Computing, Universiti Sultan Zainal Abidin (UniSZA), Besut Campus, Terengganu, Malaysia
| | - Hasib Khan
- Department of Mathematics, Shaheed Benazir Bhutto University, Sheringal, Dir Upper, Khyber Pakhtunkhwa, Pakistan
| |
Collapse
|
6
|
Du W, de Vries E, van Kuppeveld FJM, Matrosovich M, de Haan CAM. Second sialic acid-binding site of influenza A virus neuraminidase: binding receptors for efficient release. FEBS J 2021; 288:5598-5612. [PMID: 33314755 PMCID: PMC8518505 DOI: 10.1111/febs.15668] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 12/02/2020] [Accepted: 12/08/2020] [Indexed: 12/11/2022]
Abstract
Influenza A viruses (IAVs) are a major cause of human respiratory tract infections and cause significant disease and mortality. Human IAVs originate from animal viruses that breached the host species barrier. IAV particles contain sialoglycan receptor-binding hemagglutinin (HA) and receptor-destroying neuraminidase (NA) in their envelope. When IAV crosses the species barrier, the functional balance between HA and NA needs to be adjusted to the sialoglycan repertoire of the novel host species. Relatively little is known about the role of NA in host adaptation in contrast to the extensively studied HA. NA prevents virion aggregation and facilitates release of (newly assembled) virions from cell surfaces and from decoy receptors abundantly present in mucus and cell glycocalyx. In addition to a highly conserved catalytic site, NA carries a second sialic acid-binding site (2SBS). The 2SBS preferentially binds α2,3-linked sialic acids and enhances activity of the neighboring catalytic site by bringing/keeping multivalent substrates in close contact with this site. In this way, the 2SBS contributes to the HA-NA balance of virus particles and affects virus replication. The 2SBS is highly conserved in all NA subtypes of avian IAVs, with some notable exceptions associated with changes in the receptor-binding specificity of HA and host tropism. Conservation of the 2SBS is invariably lost in human (pandemic) viruses and in several other viruses adapted to mammalian host species. Preservation or loss of the 2SBS is likely to be an important factor of the viral host range.
Collapse
Affiliation(s)
- Wenjuan Du
- Section of VirologyDivision of Infectious Diseases & ImmunologyDepartment of Biomolecular Health SciencesFaculty of Veterinary MedicineUtrecht UniversityThe Netherlands
| | - Erik de Vries
- Section of VirologyDivision of Infectious Diseases & ImmunologyDepartment of Biomolecular Health SciencesFaculty of Veterinary MedicineUtrecht UniversityThe Netherlands
| | - Frank J. M. van Kuppeveld
- Section of VirologyDivision of Infectious Diseases & ImmunologyDepartment of Biomolecular Health SciencesFaculty of Veterinary MedicineUtrecht UniversityThe Netherlands
| | | | - Cornelis A. M. de Haan
- Section of VirologyDivision of Infectious Diseases & ImmunologyDepartment of Biomolecular Health SciencesFaculty of Veterinary MedicineUtrecht UniversityThe Netherlands
| |
Collapse
|
7
|
Roosenhoff R, Schutten M, Reed V, Clinch B, van der Linden A, Fouchier RAM, Fraaij PLA. Secondary substitutions in the hemagglutinin and neuraminidase genes associated with neuraminidase inhibitor resistance are rare in the Influenza Resistance Information Study (IRIS). Antiviral Res 2021; 189:105060. [PMID: 33713731 DOI: 10.1016/j.antiviral.2021.105060] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 02/16/2021] [Accepted: 03/05/2021] [Indexed: 10/21/2022]
Abstract
Amino acid substitutions in influenza virus neuraminidase (NA) that cause resistance to neuraminidase inhibitors (NAI) generally result in virus attenuation. However, influenza viruses may acquire secondary substitutions in the NA and hemagglutinin (HA) proteins that can restore viral fitness. To assess to which extent this happens, the emergence of NAI resistance substitutions and secondary - potentially compensatory - substitutions was quantified in influenza viruses of immunocompetent individuals included in the Influenza Resistance Information Study (IRIS; NCT00884117). Known resistance substitutions were detected by mutation specific RT-PCR in viruses of 57 of 1803 (3.2%) oseltamivir-treated individuals, including 39 individuals infected with A/H1N1pdm09 [H275Y] virus and 18 with A/H3N2 [R292K] virus. A total of fifteen and ten other amino acid substitutions were acquired in HA and NA respectively, of A/H1N1pdm09, A/H3N2 and influenza B viruses upon treatment with oseltamivir but none of these was associated with resistance to oseltamivir. All cultured viruses with the known resistance substitutions H275Y or R292K showed reduced susceptibility to oseltamivir in the NA-star assay. Upon next-generation sequencing, the vast majority of NAI resistant A/H1N1pdm09 and A/H3N2 viruses had no resistance-associated secondary substitutions at high frequency. Only in two A/H1N1pdm09 [H275Y] viruses, the potentially compensatory secondary substitutions HA-D52N and NA-R152K were detected. We conclude that the emergence of secondary substitutions that may restore viral fitness upon the emergence of known influenza virus NAI resistance substitutions was a rare event in this immunocompetent population.
Collapse
Affiliation(s)
- Rueshandra Roosenhoff
- Department of Viroscience, Erasmus Medical Center, Rotterdam, 3015GE, the Netherlands
| | - Martin Schutten
- Clinical Virology and Diagnostics, 1817HL, Alkmaar, the Netherlands
| | | | - Barry Clinch
- Roche Products Ltd, Welwyn Garden City, AL7 1TW, United Kingdom
| | - Anne van der Linden
- Department of Viroscience, Erasmus Medical Center, Rotterdam, 3015GE, the Netherlands
| | - Ron A M Fouchier
- Department of Viroscience, Erasmus Medical Center, Rotterdam, 3015GE, the Netherlands
| | - Pieter L A Fraaij
- Department of Viroscience, Erasmus Medical Center, Rotterdam, 3015GE, the Netherlands; Department of Pediatrics, Subdivision Infectious Diseases and Immunology, Erasmus Medical Center - Sophia, Rotterdam, the Netherlands.
| |
Collapse
|
8
|
Cox A, Schmierer J, D’Angelo J, Smith A, Levenson D, Treanor J, Kim B, Dewhurst S. A Mutated PB1 Residue 319 Synergizes with the PB2 N265S Mutation of the Live Attenuated Influenza Vaccine to Convey Temperature Sensitivity. Viruses 2020; 12:E1246. [PMID: 33142846 PMCID: PMC7693792 DOI: 10.3390/v12111246] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 10/20/2020] [Accepted: 10/24/2020] [Indexed: 01/07/2023] Open
Abstract
Current influenza vaccines have modest efficacy. This is especially true for current live attenuated influenza vaccines (LAIV), which have been inferior to the inactivated versions in recent years. Therefore, a new generation of live vaccines may be needed. We previously showed that a mutation at PB1 residue 319 confers enhanced temperature sensitivity and attenuation in an LAIV constructed in the genetic background of the mouse-adapted Influenza A Virus (IAV) strain A/PR/8/34 (PR8). Here, we describe the origin/discovery of this unique mutation and demonstrate that, when combined with the PB2 N265S mutation of LAIV, it conveys an even greater level of temperature sensitivity and attenuation on PR8 than the complete set of attenuating mutations from LAIV. Furthermore, we show that the combined PB1 L319Q and PB2 N265S mutations confer temperature sensitivity on IAV polymerase activity in two different genetic backgrounds, PR8 and A/Cal/04/09. Collectively, these findings show that the PB2 LAIV mutation synergizes with a mutation in PB1 and may have potential utility for improving LAIVs.
Collapse
Affiliation(s)
- Andrew Cox
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, New York, NY 14642, USA; (A.C.); (J.S.); (J.D.); (A.S.); (D.L.); (J.T.); (B.K.)
- Medical Scientist Training Program, University of Rochester School of Medicine and Dentistry, Rochester, New York, NY 14642, USA
- Department of Pediatrics, Pediatric Residency Program, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Jordana Schmierer
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, New York, NY 14642, USA; (A.C.); (J.S.); (J.D.); (A.S.); (D.L.); (J.T.); (B.K.)
| | - Josephine D’Angelo
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, New York, NY 14642, USA; (A.C.); (J.S.); (J.D.); (A.S.); (D.L.); (J.T.); (B.K.)
- Upstate Medical School, State University of New York, Syracuse, NY 13210, USA
| | - Andrew Smith
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, New York, NY 14642, USA; (A.C.); (J.S.); (J.D.); (A.S.); (D.L.); (J.T.); (B.K.)
- Medical Scientist Training Program, University of Rochester School of Medicine and Dentistry, Rochester, New York, NY 14642, USA
| | - Dustyn Levenson
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, New York, NY 14642, USA; (A.C.); (J.S.); (J.D.); (A.S.); (D.L.); (J.T.); (B.K.)
- M.D./Ph.D. Training Program, Wayne State University, Detroit, MI 48202, USA
| | - John Treanor
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, New York, NY 14642, USA; (A.C.); (J.S.); (J.D.); (A.S.); (D.L.); (J.T.); (B.K.)
- Division of Infectious Diseases, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York, NY 14642, USA
- Biomedical Advanced Research and Development Authority (BARDA)/HHS/ASPR, Influenza and Emerging Diseases Division 21J14, 200 C St SW, Washington, DC 20515, USA
| | - Baek Kim
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, New York, NY 14642, USA; (A.C.); (J.S.); (J.D.); (A.S.); (D.L.); (J.T.); (B.K.)
- Department of Pediatrics, Emory University, Atlanta, GA 30322, USA
- Center for Drug Discovery, Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA
| | - Stephen Dewhurst
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, New York, NY 14642, USA; (A.C.); (J.S.); (J.D.); (A.S.); (D.L.); (J.T.); (B.K.)
| |
Collapse
|
9
|
Kosik I, Yewdell JW. Influenza Hemagglutinin and Neuraminidase: Yin⁻Yang Proteins Coevolving to Thwart Immunity. Viruses 2019; 11:E346. [PMID: 31014029 PMCID: PMC6520700 DOI: 10.3390/v11040346] [Citation(s) in RCA: 116] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 04/11/2019] [Accepted: 04/13/2019] [Indexed: 01/04/2023] Open
Abstract
Influenza A virions possess two surface glycoproteins-the hemagglutinin (HA) and neuraminidase (NA)-which exert opposite functions. HA attaches virions to cells by binding to terminal sialic acid residues on glycoproteins/glycolipids to initiate the infectious cycle, while NA cleaves terminal sialic acids, releasing virions to complete the infectious cycle. Antibodies specific for HA or NA can protect experimental animals from IAV pathogenesis and drive antigenic variation in their target epitopes that impairs vaccine effectiveness in humans. Here, we review progress in understanding HA/NA co-evolution as each acquires epistatic mutations to restore viral fitness to mutants selected in the other protein by host innate or adaptive immune pressure. We also discuss recent exciting findings that antibodies to HA can function in vivo by blocking NA enzyme activity to prevent nascent virion release and enhance Fc receptor-based activation of innate immune cells.
Collapse
Affiliation(s)
- Ivan Kosik
- Laboratory of Viral Diseases, NIAID, NIH, Bethesda, MD 20892, USA.
| | | |
Collapse
|
10
|
In Vitro and In Vivo Characterization of Novel Neuraminidase Substitutions in Influenza A(H1N1)pdm09 Virus Identified Using Laninamivir-Mediated In Vitro Selection. J Virol 2019; 93:JVI.01825-18. [PMID: 30602610 DOI: 10.1128/jvi.01825-18] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 12/01/2018] [Indexed: 12/25/2022] Open
Abstract
Neuraminidase (NA) inhibitors (NAIs) are widely used antiviral drugs for the treatment of humans with influenza virus infections. There have been widespread reports of NAI resistance among seasonal A(H1N1) viruses, and most have been identified in oseltamivir-exposed patients or those treated with other NAIs. Thus, monitoring and identifying NA markers conferring resistance to NAIs-particularly newly introduced treatments-are critical to the management of viral infections. Therefore, we screened and identified substitutions conferring resistance to laninamivir by enriching random mutations in the NA gene of the 2009 pandemic influenza [A(H1N1)pdm09] virus followed by deep sequencing of the laninamivir-selected variants. After the generation of single mutants possessing each identified mutation, two A(H1N1)pdm09 recombinants possessing novel NA gene substitutions (i.e., D199E and P458T) were shown to exhibit resistance to more than one NAI. Of note, mutants possessing P458T-which is located outside of the catalytic or framework residue of the NA active site-exhibited highly reduced inhibition by all four approved NAIs. Using MDCK cells, we observed that the in vitro viral replication of the two recombinants was lower than that of the wild type (WT). Additionally, in infected mice, decreased mortality and/or mean lung viral titers were observed in mutants compared with the WT. Reverse mutations to the WT were observed in lung homogenate samples from D199E-infected mice after 3 serial passages. Overall, the novel NA substitutions identified could possibly emerge in influenza A(H1N1)pdm09 viruses during laninamivir therapy and the viruses could have altered NAI susceptibility, but the compromised in vitro/in vivo viral fitness may limit viral spreading.IMPORTANCE With the widespread emergence of NAI-resistant influenza virus strains, continuous monitoring of mutations that confer antiviral resistance is needed. Laninamivir is the most recently approved NAI in several countries; few data exist related to the in vitro selection of viral mutations conferring resistance to laninamivir. Thus, we screened and identified substitutions conferring resistance to laninamivir by random mutagenesis of the NA gene of the 2009 pandemic influenza [A(H1N1)pdm09] virus strain followed by deep sequencing of the laninamivir-selected variants. We found several novel substitutions in NA (D199E and P458T) in an A(H1N1)pdm09 background which conferred resistance to NAIs and which had an impact on viral fitness. Our study highlights the importance of continued surveillance for potential antiviral-resistant variants and the development of alternative therapeutics.
Collapse
|
11
|
Lambkin-Williams R, Noulin N, Mann A, Catchpole A, Gilbert AS. The human viral challenge model: accelerating the evaluation of respiratory antivirals, vaccines and novel diagnostics. Respir Res 2018; 19:123. [PMID: 29929556 PMCID: PMC6013893 DOI: 10.1186/s12931-018-0784-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 04/19/2018] [Indexed: 12/15/2022] Open
Abstract
The Human Viral Challenge (HVC) model has, for many decades, helped in the understanding of respiratory viruses and their role in disease pathogenesis. In a controlled setting using small numbers of volunteers removed from community exposure to other infections, this experimental model enables proof of concept work to be undertaken on novel therapeutics, including vaccines, immunomodulators and antivirals, as well as new diagnostics.Crucially, unlike conventional phase 1 studies, challenge studies include evaluable efficacy endpoints that then guide decisions on how to optimise subsequent field studies, as recommended by the FDA and thus licensing studies that follow. Such a strategy optimises the benefit of the studies and identifies possible threats early on, minimising the risk to subsequent volunteers but also maximising the benefit of scarce resources available to the research group investing in the research. Inspired by the principles of the 3Rs (Replacement, Reduction and Refinement) now commonly applied in the preclinical phase, HVC studies allow refinement and reduction of the subsequent development phase, accelerating progress towards further statistically powered phase 2b studies. The breadth of data generated from challenge studies allows for exploration of a wide range of variables and endpoints that can then be taken through to pivotal phase 3 studies.We describe the disease burden for acute respiratory viral infections for which current conventional development strategies have failed to produce therapeutics that meet clinical need. The Authors describe the HVC model's utility in increasing scientific understanding and in progressing promising therapeutics through development.The contribution of the model to the elucidation of the virus-host interaction, both regarding viral pathogenicity and the body's immunological response is discussed, along with its utility to assist in the development of novel diagnostics.Future applications of the model are also explored.
Collapse
Affiliation(s)
- Rob Lambkin-Williams
- hVIVO Services Limited, Queen Mary BioEnterprises Innovation Centre, 42 New Road, London, England, E1 2AX, UK.
| | - Nicolas Noulin
- hVIVO Services Limited, Queen Mary BioEnterprises Innovation Centre, 42 New Road, London, England, E1 2AX, UK
| | - Alex Mann
- hVIVO Services Limited, Queen Mary BioEnterprises Innovation Centre, 42 New Road, London, England, E1 2AX, UK
| | - Andrew Catchpole
- hVIVO Services Limited, Queen Mary BioEnterprises Innovation Centre, 42 New Road, London, England, E1 2AX, UK
| | - Anthony S Gilbert
- hVIVO Services Limited, Queen Mary BioEnterprises Innovation Centre, 42 New Road, London, England, E1 2AX, UK
| |
Collapse
|
12
|
Eguchi N, Fujino K, Thanasut K, Taharaguchi M, Motoi M, Motoi A, Oonaka K, Taharaguchi S. In vitro Anti-Influenza Virus Activity of Agaricus brasiliensis KA21. Biocontrol Sci 2018; 22:171-174. [PMID: 28954960 DOI: 10.4265/bio.22.171] [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] [Indexed: 11/01/2022]
Abstract
Agaricus is known to have immunostimulatory and anti-tumor effects. However, the antiviral effects of Agaricus have not yet been examined. In the present study, the antiviral effects of an extract of Agaricus brasiliensis KA21 (AE) on the H1N1 influenza virus (PR8 strain) were investigated. The anti-influenza virus effects of AE were examined by using the plaque formation inhibition test. AE inhibited the plaque formation of PR8 in a dose-dependent manner: 98 and 50% (IC50) inhibition at 2.5 and 0.99 mg/mL, respectively. To elucidate the mechanisms of AE, the direct actions and adsorption and invasion inhibition of AE were examined, and were found to have no inhibitory effect on PR8 infection. Thus, in vitro antiviral effects may somehow inhibit PR8 after the viral invasion of cells. These results demonstrated that it is expected that AE can effectively prevent the spread of the influenza virus.
Collapse
Affiliation(s)
- Nao Eguchi
- Department of Veterinary Medicine, Laboratory of Microbiology II, School of Veterinary Medicine, Azabu University.,These authors contributed equally to this work
| | - Kan Fujino
- Department of Veterinary Medicine, Laboratory of Microbiology II, School of Veterinary Medicine, Azabu University.,These authors contributed equally to this work
| | - Khompakorn Thanasut
- Department of Veterinary Medicine, Laboratory of Microbiology II, School of Veterinary Medicine, Azabu University
| | - Motoko Taharaguchi
- Division of Experimental Animal Research, National Institute of Infectious Diseases
| | | | | | - Kenji Oonaka
- Department of Food and Life Science, Laboratory of Food and Hygiene, School of Life and Environmental Science, Azabu University
| | - Satoshi Taharaguchi
- Department of Veterinary Medicine, Laboratory of Microbiology II, School of Veterinary Medicine, Azabu University
| |
Collapse
|
13
|
Rath B, Chen X, Spies V, Muehlhans S, Obermeier P, Tief F, Seeber L, Karsch K, Milde J, Skopnik H, Schweiger B, Duwe SC. Prospective surveillance of antiviral resistance in hospitalized infants less than 12 months of age with A(H3N2) influenza infection and treated with oseltamivir. Antivir Ther 2017; 22:515-522. [PMID: 28205506 DOI: 10.3851/imp3141] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/14/2017] [Indexed: 10/20/2022]
Abstract
BACKGROUND Infants exhibit elevated influenza virus loads and prolonged viral shedding, which may increase the risk for resistance development, especially in cases of suboptimal exposure to antiviral therapy. METHODS We performed a prospective surveillance of hospitalized infants undergoing oseltamivir therapy during the 2008-2009 and 2011-2012 influenza seasons at two paediatric hospitals in Germany. A total of 37 infants less than 1 year of age with laboratory confirmed influenza A(H3N2) infection received oseltamivir as per physician's order for 5 days (2008-2009 season: 2 mg/kg twice daily; 2011-2012 season: 2.0 mg/kg; 2.5 mg/kg and 3.0 mg/kg twice daily for infants <1 month; 2-3 months and 4-12 months, respectively). Virus load, the susceptibility to neuraminidase inhibitors (NAIs), and the presence of molecular markers of resistance to NAIs was assessed for influenza viruses recovered from respiratory samples collected at baseline and during follow-up visits. RESULTS Overall, 73% of the infants continued to shed viral RNA detectable by reverse transcription (RT)-PCR after dose number 10 of oseltamivir; 12 infants shed viruses, 2 of them (both 9 months of age) shed resistant viruses. Resistance was characterized by ≥1,000-fold increase of 50% inhibitory concentration (IC50) for oseltamivir, up to 50-fold for zanamivir and elevated Km values when compared to susceptible A(H3N2) strains. Sanger sequencing revealed the selection of the NA-R292K substitution in both instances (after dose number 10 on day 6). CONCLUSIONS Our data suggest that it may be relevant to monitor antiviral resistance systematically in all infants, considering that the European Medicines Agency has recently extended the licensure for oseltamivir to include full-term infants.
Collapse
Affiliation(s)
- Barbara Rath
- Charité University Medical Center, Department of Pediatrics, Berlin, Germany.,Vienna Vaccine Safety Initiative, Berlin, Germany
| | - Xi Chen
- Charité University Medical Center, Department of Pediatrics, Berlin, Germany
| | - Vera Spies
- Klinikum Worms, Department of Pediatrics and Adolescent Medicine, Worms, Germany
| | - Susann Muehlhans
- Charité University Medical Center, Department of Pediatrics, Berlin, Germany
| | - Patrick Obermeier
- Charité University Medical Center, Department of Pediatrics, Berlin, Germany
| | - Franziska Tief
- Charité University Medical Center, Department of Pediatrics, Berlin, Germany
| | - Lea Seeber
- Charité University Medical Center, Department of Pediatrics, Berlin, Germany
| | - Katharina Karsch
- Charité University Medical Center, Department of Pediatrics, Berlin, Germany
| | - Jeanette Milde
- Robert Koch Institute, Division of Influenza Viruses and Other Respiratory Viruses, National Reference Centre for Influenza, Berlin, Germany
| | - Heino Skopnik
- Klinikum Worms, Department of Pediatrics and Adolescent Medicine, Worms, Germany
| | - Brunhilde Schweiger
- Robert Koch Institute, Division of Influenza Viruses and Other Respiratory Viruses, National Reference Centre for Influenza, Berlin, Germany
| | - Susanne C Duwe
- Robert Koch Institute, Division of Influenza Viruses and Other Respiratory Viruses, National Reference Centre for Influenza, Berlin, Germany
| |
Collapse
|
14
|
A new ion selective electrode method for determination of oseltamivir phosphate (Tamiflu) and its pharmaceutical applications. ARAB J CHEM 2017. [DOI: 10.1016/j.arabjc.2012.07.029] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
15
|
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
|
16
|
Weisshaar M, Cox R, Morehouse Z, Kyasa SK, Yan D, Oberacker P, Mao S, Golden JE, Lowen AC, Natchus MG, Plemper RK. Identification and Characterization of Influenza Virus Entry Inhibitors through Dual Myxovirus High-Throughput Screening. J Virol 2016; 90:7368-7387. [PMID: 27252534 PMCID: PMC4984618 DOI: 10.1128/jvi.00898-16] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2016] [Accepted: 05/26/2016] [Indexed: 12/31/2022] Open
Abstract
UNLABELLED Influenza A virus (IAV) infections cause major morbidity and mortality, generating an urgent need for novel antiviral therapeutics. We recently established a dual myxovirus high-throughput screening protocol that combines a fully replication-competent IAV-WSN strain and a respiratory syncytial virus reporter strain for the simultaneous identification of IAV-specific, paramyxovirus-specific, and broad-spectrum inhibitors. In the present study, this protocol was applied to a screening campaign to assess a diverse chemical library with over 142,000 entries. Focusing on IAV-specific hits, we obtained a hit rate of 0.03% after cytotoxicity testing and counterscreening. Three chemically distinct hit classes with nanomolar potency and favorable cytotoxicity profiles were selected. Time-of-addition, minigenome, and viral entry studies demonstrated that these classes block hemagglutinin (HA)-mediated membrane fusion. Antiviral activity extends to an isolate from the 2009 pandemic and, in one case, another group 1 subtype. Target identification through biolayer interferometry confirmed binding of all hit compounds to HA. Resistance profiling revealed two distinct escape mechanisms: primary resistance, associated with reduced compound binding, and secondary resistance, associated with unaltered binding. Secondary resistance was mediated, unusually, through two different pairs of cooperative mutations, each combining a mutation eliminating the membrane-proximal stalk N-glycan with a membrane-distal change in HA1 or HA2. Chemical synthesis of an analog library combined with in silico docking extracted a docking pose for the hit classes. Chemical interrogation spotlights IAV HA as a major druggable target for small-molecule inhibition. Our study identifies novel chemical scaffolds with high developmental potential, outlines diverse routes of IAV escape from entry inhibition, and establishes a path toward structure-aided lead development. IMPORTANCE This study is one of the first to apply a fully replication-competent third-generation IAV reporter strain to a large-scale high-throughput screen (HTS) drug discovery campaign, allowing multicycle infection and screening in physiologically relevant human respiratory cells. A large number of potential druggable targets was thus chemically interrogated, but mechanistic characterization, positive target identification, and resistance profiling demonstrated that three chemically promising and structurally distinct hit classes selected for further analysis all block HA-mediated membrane fusion. Viral escape from inhibition could be achieved through primary and secondary resistance mechanisms. In silico docking predicted compound binding to a microdomain located at the membrane-distal site of the prefusion HA stalk that was also previously suggested as a target site for chemically unrelated HA inhibitors. This study identifies an unexpected chemodominance of the HA stalk microdomain for small-molecule inhibitors in IAV inhibitor screening campaigns and highlights a novel mechanism of cooperative resistance to IAV entry blockers.
Collapse
Affiliation(s)
- Marco Weisshaar
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Robert Cox
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Zachary Morehouse
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Shiva K Kyasa
- Emory Institute for Drug Development, Emory University, Atlanta, GA, USA
| | - Dan Yan
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Phil Oberacker
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Shuli Mao
- Emory Institute for Drug Development, Emory University, Atlanta, GA, USA
| | - Jennifer E Golden
- Department of Pharmacology, University of Wisconsin, Madison, WI, USA
| | - Anice C Lowen
- Department of Microbiology & Immunology, Emory University School of Medicine, Atlanta, GA, USA
| | - Michael G Natchus
- Emory Institute for Drug Development, Emory University, Atlanta, GA, USA
| | - Richard K Plemper
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| |
Collapse
|
17
|
Gaymard A, Charles-Dufant A, Sabatier M, Cortay JC, Frobert E, Picard C, Casalegno JS, Rosa-Calatrava M, Ferraris O, Valette M, Ottmann M, Lina B, Escuret V. Impact on antiviral resistance of E119V, I222L and R292K substitutions in influenza A viruses bearing a group 2 neuraminidase (N2, N3, N6, N7 and N9). J Antimicrob Chemother 2016; 71:3036-3045. [PMID: 27432605 DOI: 10.1093/jac/dkw275] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 06/01/2016] [Accepted: 06/02/2016] [Indexed: 01/02/2023] Open
Abstract
OBJECTIVES While subtype-specific substitutions linked to neuraminidase (NA) inhibitor resistance are well described in human N1 and N2 influenza NAs, little is known about other NA subtypes. The aim of this study was to determine whether the R292K and E119V ± I222L substitutions could be associated with oseltamivir resistance in all group 2 NAs and had an impact on virus fitness. METHODS Reassortant viruses with WT NA or variant N2, N3, N6, N7 or N9 NAs, bearing R292K or E119V ± I222L substitutions, were produced by reverse genetics. The antiviral susceptibility, activity, Km of the NA, mutation stability and in vitro virus fitness in MDCK cells were determined. RESULTS NA activities could be ranked as follows regardless of the substitution: N3 ≥ N6 > N2 ≥ N9 > N7. Using NA inhibitor resistance interpretation criteria used for human N1 or N2, the NA-R292K substitution conferred highly reduced inhibition by oseltamivir and the N6- or N9-R292K substitution conferred reduced inhibition by zanamivir and laninamivir. Viruses with the N3- or N6-E119V substitution showed normal inhibition by oseltamivir, while those with the N2-, N7- or N9-E119V substitution showed reduced inhibition by oseltamivir. Viruses with NA-E119V + I222L substitutions showed reduced inhibition (N3 and N6) or highly reduced inhibition (N2, N7 and N9) by oseltamivir. Viruses bearing the NA-R292K substitution had lower affinity and viruses bearing the NA-E119V substitution had higher affinity for the MUNANA substrate than viruses with corresponding WT NA. CONCLUSIONS NA-R292K and E119V + I222L substitutions conferred reduced inhibition by oseltamivir for all group 2 NAs. Surveillance of NA inhibitor resistance for zoonotic and human influenza viruses and the development of novel antiviral agents with different targets should be continued.
Collapse
Affiliation(s)
- Alexandre Gaymard
- Univ Lyon, Université Lyon 1, Faculté de Médecine Lyon Est, CIRI Inserm U1111, équipe Virpath, F-69008, Lyon, France.,Hospices Civils de Lyon, Centre National de Référence virus influenzae France Sud, Laboratoire de Virologie, Groupement Hospitalier Nord, F-69317, Lyon cedex 04, France
| | - Aymeric Charles-Dufant
- Univ Lyon, Université Lyon 1, Faculté de Médecine Lyon Est, CIRI Inserm U1111, équipe Virpath, F-69008, Lyon, France
| | - Murielle Sabatier
- Univ Lyon, Université Lyon 1, Faculté de Médecine Lyon Est, CIRI Inserm U1111, équipe Virpath, F-69008, Lyon, France
| | - Jean-Claude Cortay
- Univ Lyon, Université Lyon 1, Faculté de Médecine Lyon Est, CIRI Inserm U1111, équipe Virpath, F-69008, Lyon, France
| | - Emilie Frobert
- Univ Lyon, Université Lyon 1, Faculté de Médecine Lyon Est, CIRI Inserm U1111, équipe Virpath, F-69008, Lyon, France.,Hospices Civils de Lyon, Centre National de Référence virus influenzae France Sud, Laboratoire de Virologie, Groupement Hospitalier Nord, F-69317, Lyon cedex 04, France
| | - Caroline Picard
- Univ Lyon, Université Lyon 1, Faculté de Médecine Lyon Est, CIRI Inserm U1111, équipe Virpath, F-69008, Lyon, France
| | - Jean-Sébastien Casalegno
- Univ Lyon, Université Lyon 1, Faculté de Médecine Lyon Est, CIRI Inserm U1111, équipe Virpath, F-69008, Lyon, France.,Hospices Civils de Lyon, Centre National de Référence virus influenzae France Sud, Laboratoire de Virologie, Groupement Hospitalier Nord, F-69317, Lyon cedex 04, France
| | - Manuel Rosa-Calatrava
- Univ Lyon, Université Lyon 1, Faculté de Médecine Lyon Est, CIRI Inserm U1111, équipe Virpath, F-69008, Lyon, France
| | - Olivier Ferraris
- Univ Lyon, Université Lyon 1, Faculté de Médecine Lyon Est, CIRI Inserm U1111, équipe Virpath, F-69008, Lyon, France
| | - Martine Valette
- Univ Lyon, Université Lyon 1, Faculté de Médecine Lyon Est, CIRI Inserm U1111, équipe Virpath, F-69008, Lyon, France.,Hospices Civils de Lyon, Centre National de Référence virus influenzae France Sud, Laboratoire de Virologie, Groupement Hospitalier Nord, F-69317, Lyon cedex 04, France
| | - Michèle Ottmann
- Univ Lyon, Université Lyon 1, Faculté de Médecine Lyon Est, CIRI Inserm U1111, équipe Virpath, F-69008, Lyon, France
| | - Bruno Lina
- Univ Lyon, Université Lyon 1, Faculté de Médecine Lyon Est, CIRI Inserm U1111, équipe Virpath, F-69008, Lyon, France.,Hospices Civils de Lyon, Centre National de Référence virus influenzae France Sud, Laboratoire de Virologie, Groupement Hospitalier Nord, F-69317, Lyon cedex 04, France
| | - Vanessa Escuret
- Univ Lyon, Université Lyon 1, Faculté de Médecine Lyon Est, CIRI Inserm U1111, équipe Virpath, F-69008, Lyon, France .,Hospices Civils de Lyon, Centre National de Référence virus influenzae France Sud, Laboratoire de Virologie, Groupement Hospitalier Nord, F-69317, Lyon cedex 04, France
| |
Collapse
|
18
|
Oxford J, Lambkin-Williams R, Mann A. The Threat of Avian Influenza H5N1: ‘Do We Have the Tools for the Job?’. ACTA ACUST UNITED AC 2016; 18:71-4. [PMID: 17542151 DOI: 10.1177/095632020701800202] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
For the first time in human history virologists have the knowledge about the avian origin of pandemic influenza A viruses. Furthermore, in the last two decades a new class of anti influenza drugs, the neuraminidase inhibitors (NIs), has been developed from an academic discovery to a series of antiviral drugs to be used in the clinic. At present vaccinologists are producing influenza A (H5N1) vaccines to be stockpiled alongside the NIs to combat the first wave of an anticipated influenza pandemic. Studies from the 1918 infection calamity, the Spanish influenza, and the succeeding pandemics of 1957 and 1968, all caused by avian influenza A viruses, have shown how quickly such a virus can mutate to become less virulent (starting with 50% case fatality) and more infectious. Such a mutation cluster could lead to a rapid increase in world deaths, currently 170, to many millions. However there are optimistic analyses: judicious and swift application of NIs, vaccine and hygiene to an outbreak epicentre, most likely in South-East Asia, could break the chain of transmission.
Collapse
Affiliation(s)
- John Oxford
- Centre for Infectious Diseases, Retroscreen Virology Ltd, London, UK.
| | | | | |
Collapse
|
19
|
Yamada H, Nagase S, Takahashi K, Sakoda Y, Kida H, Okamoto S. Toll-like receptor 9 ligand D-type oligodeoxynucleotide D35 as a broad inhibitor for influenza A virus replication that is associated with suppression of neuraminidase activity. Antiviral Res 2016; 129:81-92. [PMID: 26923882 PMCID: PMC7113795 DOI: 10.1016/j.antiviral.2016.02.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 02/09/2016] [Accepted: 02/17/2016] [Indexed: 12/18/2022]
Abstract
The most effective drugs available to treat influenza are neuraminidase (NA) inhibitors, which provide important additional measures for the control of influenza virus infections. However, since the emergence of NA inhibitor-resistant viruses may compromise the clinical utility of this class of anti-influenza agents, it is very important to develop new anti-influenza agents which target a different region in NA responsible for its sensitivity from that for NA inhibitors and could be used to treat NA inhibitors-resistant isolates. The oligodeoxynucleotide D35, multimerized and aggregated, suppressed replication of influenza A viruses except A/WSN/33 (WSN). The suppressive viral replication by D35 depended on G-terad and multimer formation. The range of the suppressive viral replication at the late stage, including virus assembly and release from infected cells, was much larger than that at the initial stage, viral attachment and entry. D35 suppressed NA activity of influenza A viruses. Furthermore, replacing the NA gene of A/Puerto Rico/8/34 (PR8), in which viral replication was inhibited by D35 at the late stage, with the NA gene from WSN, in which viral replication was not inhibited, eliminated the D35-dependent suppression. D35 showed an additive anti-influenza effect with oseltamivir. It was also effective in vivo. These results suggest that the influenza virus NA mainly contributse to the D35-suppressible virus release from infected cells at the late stage. In addition, because administration of D35 into the virus-infected mice suppressed viral replication and weight loss, clinical application of D35 could be considered. The oligodeoxynucleotide D35 suppressed replication of some influenza A viruses. D35 inhibits viral replication at the late step which is dependent on NA activity. Antiviral mechanism by D35 is different from that by oseltamivir.
Collapse
Affiliation(s)
- Hiroshi Yamada
- Laboratory of Virology and Vaccinology, National Institute of Biomedical Innovation, Ibaraki, Osaka, Japan
| | - Satoshi Nagase
- Department of Laboratory Sciences, Division of Health Sciences, Kanazawa University Graduate School of Medical Science, Kanazawa, Japan
| | - Kazuo Takahashi
- Department of Infectious Diseases, Osaka Prefectural Institute of Public Health, Osaka, Japan
| | - Yoshihiro Sakoda
- Laboratory of Microbiology, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Hiroshi Kida
- Laboratory of Microbiology, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Japan; Research Center for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Shigefumi Okamoto
- Department of Laboratory Sciences, Division of Health Sciences, Kanazawa University Graduate School of Medical Science, Kanazawa, Japan.
| |
Collapse
|
20
|
Xu W, Li H, Jiang L. Human infection with a highly pathogenic avian influenza A (H5N6) virus in Yunnan province, China. Infect Dis (Lond) 2016; 48:477-82. [DOI: 10.3109/23744235.2015.1135253] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
21
|
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: 0.6] [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
|
22
|
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
|
23
|
Application of a seven-target pyrosequencing assay to improve the detection of neuraminidase inhibitor-resistant Influenza A(H3N2) viruses. Antimicrob Agents Chemother 2015; 59:2374-9. [PMID: 25645846 DOI: 10.1128/aac.04939-14] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
National U.S. influenza antiviral surveillance incorporates data generated by neuraminidase (NA) inhibition (NI) testing of isolates supplemented with NA sequence analysis and pyrosequencing analysis of clinical specimens. A lack of established correlates for clinically relevant resistance to NA inhibitors (NAIs) hinders interpretation of NI assay data. Nonetheless, A(H3N2) viruses are commonly monitored for moderately or highly reduced inhibition in the NI assay and/or for the presence of NA markers E119V, R292K, and N294S. In 2012 to 2013, three drug-resistant A(H3N2) viruses were detected by NI assay among isolates (n = 1,424); all showed highly reduced inhibition by oseltamivir and had E119V. In addition, one R292K variant was detected among clinical samples (n = 1,024) by a 3-target pyrosequencing assay. Overall, the frequency of NAI resistance was low (0.16% [4 of 2,448]). To screen for additional NA markers previously identified in viruses from NAI-treated patients, the pyrosequencing assay was modified to include Q136K, I222V, and deletions encompassing residues 245 to 248 (del245-248) and residues 247 to 250 (del247-250). The 7-target pyrosequencing assay detected NA variants carrying E119V, Q136, and del245-248 in an isolate from an oseltamivir-treated patient. Next, this assay was applied to clinical specimens collected from hospitalized patients and submitted for NI testing but failed cell culture propagation. Of the 27 clinical specimens tested, 4 (15%) contained NA changes: R292K (n = 2), E119V (n = 1), and del247-250 (n = 1). Recombinant NAs with del247-250 or del245-248 conferred highly reduced inhibition by oseltamivir, reduced inhibition by zanamivir, and normal inhibition by peramivir and laninamivir. Our results demonstrated the benefits of the 7-target pyrosequencing assay in conducting A(H3N2) antiviral surveillance and testing for clinical care.
Collapse
|
24
|
Antiviral Drugs for Influenza and Other Respiratory Virus Infections. MANDELL, DOUGLAS, AND BENNETT'S PRINCIPLES AND PRACTICE OF INFECTIOUS DISEASES 2015. [PMCID: PMC7152365 DOI: 10.1016/b978-1-4557-4801-3.00044-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
25
|
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: 11] [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
|
26
|
Hai R, Schmolke M, Leyva-Grado VH, Thangavel RR, Margine I, Jaffe EL, Krammer F, Solórzano A, García-Sastre A, Palese P, Bouvier NM. Influenza A(H7N9) virus gains neuraminidase inhibitor resistance without loss of in vivo virulence or transmissibility. Nat Commun 2014; 4:2854. [PMID: 24326875 PMCID: PMC3863970 DOI: 10.1038/ncomms3854] [Citation(s) in RCA: 136] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2013] [Accepted: 11/01/2013] [Indexed: 12/17/2022] Open
Abstract
Without baseline human immunity to the emergent avian influenza A(H7N9) virus, neuraminidase inhibitors are vital for controlling viral replication in severe infections. An amino acid change in the viral neuraminidase associated with drug resistance, NA-R292K (N2 numbering), has been found in some H7N9 clinical isolates. Here we assess the impact of the NA-R292K substitution on antiviral sensitivity and viral replication, pathogenicity and transmissibility of H7N9 viruses. Our data indicate that an H7N9 isolate encoding the NA-R292K substitution is highly resistant to oseltamivir and peramivir and partially resistant to zanamivir. Furthermore, H7N9 reassortants with and without the resistance mutation demonstrate comparable viral replication in primary human respiratory cells, virulence in mice and transmissibility in guinea pigs. Thus, in stark contrast to oseltamivir-resistant seasonal influenza A(H3N2) viruses, H7N9 virus replication and pathogenicity in these models are not substantially altered by the acquisition of high-level oseltamivir resistance due to the NA-R292K mutation. Some clinical isolates of influenza A(H7N9) virus encode a mutation within neuraminidase that could confer resistance to the only class of drugs active against H7N9. Here, the authors show that this mutation does not affect viral replication and pathogenicity while mediating resistance to antivirals in vivo.
Collapse
Affiliation(s)
- Rong Hai
- 1] Department of Microbiology, Icahn School of Medicine at Mount Sinai, One Gustave L Levy Place, Box 1124, New York, New York 10029, USA [2]
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Yamada H, Nagao C, Haredy AM, Mori Y, Mizuguchi K, Yamanishi K, Okamoto S. Dextran sulfate-resistant A/Puerto Rico/8/34 influenza virus is associated with the emergence of specific mutations in the neuraminidase glycoprotein. Antiviral Res 2014; 111:69-77. [PMID: 25234090 DOI: 10.1016/j.antiviral.2014.09.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Revised: 09/02/2014] [Accepted: 09/07/2014] [Indexed: 11/28/2022]
Abstract
Dextran sulfate (DS) is a negatively charged sulfated polysaccharide that suppresses the replication of influenza A viruses. The suppression was thought to be associated with inhibition of the hemagglutinin-dependent fusion activity. However, we previously showed that suppression by DS was observed not only at the initial stage of viral infection, but also later when virus is released from infected cells due to inhibition of neuraminidase (NA) activity. In the present study, we isolated DS-resistant A/Puerto Rico/8/34 (PR8) influenza viruses and analyzed the inhibition by DS. We found six mutations in NA genes of five independent resistant PR8 viruses and each resistant NA gene had two mutations. All mutations were from basic to acidic or neutral amino acids. In addition, R430L, K432E or K435E in the 430-435 region was a common mutation in all resistant NA genes. To determine which amino acid(s) are responsible for this resistance, a panel of recombinant viruses containing a PR8 and A/WSN/33(WSN) chimeric NA gene or an NA gene with different mutation(s) was generated using reverse genetics. Using recombinant viruses containing a PR8/WSN chimeric NA, we showed that one third of the C-terminal region of PR8 NA was responsible for DS-sensitivity. Recombinant viruses with a single mutation in NA replicated better than wild-type PR8 in the presence of DS, but were still DS-sensitive. However, replication of recombinant viruses with double mutations from the resistant viruses was not affected by the presence or absence of DS. In addition, resistant recombinant viruses were found to be sensitive to the NA inhibitor, oseltamivir and the oseltamivir-resistant recombinant virus was sensitive to DS. These results suggested that DS is an NA inhibitor with a different mechanism of action from the currently used NA inhibitors and that DS could be used in combination with these inhibitors to treat influenza virus infections.
Collapse
Affiliation(s)
- Hiroshi Yamada
- Laboratory of Virology and Vaccinology, Division of Biomedical Research, National Institute of Biomedical Innovation, Ibaraki, Osaka, Japan.
| | - Chioko Nagao
- National Institute of Biomedical Innovation, Ibaraki, Osaka, Japan
| | - Ahmad M Haredy
- Laboratory of Virology and Vaccinology, Division of Biomedical Research, National Institute of Biomedical Innovation, Ibaraki, Osaka, Japan
| | - Yasuko Mori
- Division of Clinical Virology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Kenji Mizuguchi
- National Institute of Biomedical Innovation, Ibaraki, Osaka, Japan
| | - Koichi Yamanishi
- Laboratory of Virology and Vaccinology, Division of Biomedical Research, National Institute of Biomedical Innovation, Ibaraki, Osaka, Japan
| | - Shigefumi Okamoto
- Laboratory of Virology and Vaccinology, Division of Biomedical Research, National Institute of Biomedical Innovation, Ibaraki, Osaka, Japan
| |
Collapse
|
28
|
Hossain MK, Choi HY, Hwang JS, Dayem AA, Kim JH, Kim YB, Poo H, Cho SG. Antiviral activity of 3,4'-dihydroxyflavone on influenza a virus. J Microbiol 2014; 52:521-6. [PMID: 24871979 DOI: 10.1007/s12275-014-4212-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Revised: 04/21/2014] [Accepted: 04/21/2014] [Indexed: 11/28/2022]
Abstract
Influenza virus infection causes thousands of deaths and millions of hospitalizations worldwide every year and the emergence of resistance to anti-influenza drugs has prompted scientists to seek new natural antiviral materials. In this study, we screened 13 different flavonoids from various flavonoid groups to identify the most potent antiviral flavonoid against human influenza A/PR/8/34 (H1N1). The 3-hydroxyl group flavonoids, including 3,2᾿dihydroxyflavone (3,2᾿DHF) and 3,4᾿dihydroxyflavone (3,4᾿DHF), showed potent anti-influenza activity. They inhibited viral neuraminidase activity and viral adsorption onto cells. To confirm the anti-influenza activity of these flavonoids, we used an in vivo mouse model. In mice infected with human influenza, oral administration of 3,4᾿DHF significantly decreased virus titers and pathological changes in the lung and reduced body weight loss and death. Our data suggest that 3-hydroxyl group flavonoids, particularly 3,4᾿DHF, have potent antiviral activity against human influenza A/PR/8/34 (H1N1) in vitro and in vivo. Further clinical studies are needed to investigate the therapeutic and prophylactic potential of the 3-hydroxyl group flavonoids in treating influenza pandemics.
Collapse
Affiliation(s)
- Mohammed Kawser Hossain
- Department of Animal Biotechnology, Animal Resources Research Center, and Incurable Disease Animal model and Stem cell Institute (IDASI), Konkuk University, Seoul, 143-701, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Oh DY, Hurt AC. A Review of the Antiviral Susceptibility of Human and Avian Influenza Viruses over the Last Decade. SCIENTIFICA 2014; 2014:430629. [PMID: 24800107 PMCID: PMC3995103 DOI: 10.1155/2014/430629] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Accepted: 03/06/2014] [Indexed: 06/03/2023]
Abstract
Antivirals play an important role in the prevention and treatment of influenza infections, particularly in high-risk or severely ill patients. Two classes of influenza antivirals have been available in many countries over the last decade (2004-2013), the adamantanes and the neuraminidase inhibitors (NAIs). During this period, widespread adamantane resistance has developed in circulating influenza viruses rendering these drugs useless, resulting in the reliance on the most widely available NAI, oseltamivir. However, the emergence of oseltamivir-resistant seasonal A(H1N1) viruses in 2008 demonstrated that NAI-resistant viruses could also emerge and spread globally in a similar manner to that seen for adamantane-resistant viruses. Previously, it was believed that NAI-resistant viruses had compromised replication and/or transmission. Fortunately, in 2013, the majority of circulating human influenza viruses remain sensitive to all of the NAIs, but significant work by our laboratory and others is now underway to understand what enables NAI-resistant viruses to retain the capacity to replicate and transmit. In this review, we describe how the susceptibility of circulating human and avian influenza viruses has changed over the last ten years and describe some research studies that aim to understand how NAI-resistant human and avian influenza viruses may emerge in the future.
Collapse
Affiliation(s)
- Ding Yuan Oh
- WHO Collaborating Centre for Reference and Research on Influenza, 10 Wreckyn Street, North Melbourne, VIC 3051, Australia
| | - Aeron C. Hurt
- WHO Collaborating Centre for Reference and Research on Influenza, 10 Wreckyn Street, North Melbourne, VIC 3051, Australia
- School of Applied Sciences and Engineering, Monash University, Churchill, VIC 3842, Australia
| |
Collapse
|
30
|
Sleeman K, Guo Z, Barnes J, Shaw M, Stevens J, Gubareva LV. R292K substitution and drug susceptibility of influenza A(H7N9) viruses. Emerg Infect Dis 2014; 19:1521-4. [PMID: 23965618 PMCID: PMC3810934 DOI: 10.3201/eid1909.130724] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Neuraminidase inhibitors are the only licensed antiviral medications available to treat avian influenza A(H7N9) virus infections in humans. According to a neuraminidase inhibition assay, an R292K substitution reduced antiviral efficacy of inhibitors, especially oseltamivir, and decreased viral fitness in cell culture. Monitoring emergence of R292K-carrying viruses using a pH-modified neuraminidase inhibition assay should be considered.
Collapse
|
31
|
A conformational restriction in the influenza A virus neuraminidase binding site by R152 results in a combinational effect of I222T and H274Y on oseltamivir resistance. Antimicrob Agents Chemother 2013; 58:1639-45. [PMID: 24366752 DOI: 10.1128/aac.01848-13] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The I222K, I222R, and I222T substitutions in neuraminidase (NA) have been found in clinically derived 2009 pandemic influenza A/H1N1 viruses with altered susceptibilities to NA inhibitors (NAIs). The effects of these substitutions, together with the most frequently observed resistance-related substitution, H274Y, on viral fitness and resistance mechanisms were further investigated in this study. Reduced sensitivities to oseltamivir were observed in all three mutants (I222K, I222R, and I222T). Furthermore, the I222K and I222T substitutions had a combinational effect of further increasing resistance in the presence of H274Y, which might result from a conformational restriction in the NA binding site. Of note, by using molecular dynamics simulations, R152, the neighbor of T222, was observed to translate to a position closer to T222, resulting in the narrowing of the binding pocket, which otherwise only subtends the residue substitution of H274Y. Moreover, significantly attenuated NA function and viral growth abilities were found in the I222K+H274Y double mutant, while the I222T+H274Y double mutant exhibited slightly delayed growth but had a peak viral titer similar to that of the wild-type virus in MDCK cells. The relative growth advantage of the I222T mutant versus the I222K mutant and the higher frequency of I222T emerging in N1 subtype influenza viruses raise concerns necessitating close monitoring of the dual substitutions I222T and H274Y.
Collapse
|
32
|
YANG ZHIWEI, WU FEI, LIU JUNXING, WANG SHUQIU, YUAN XIAOHUI. SUSCEPTIBILITY OF COMMERCIAL NEURAMINIDASE INHIBITORS AGAINST 2013 A/H7N9 INFLUENZA VIRUS: A DOCKING AND MOLECULAR DYNAMICS STUDY. JOURNAL OF THEORETICAL & COMPUTATIONAL CHEMISTRY 2013. [DOI: 10.1142/s0219633613500697] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The latest influenza A ( H 7 N 9) virus attracted a worldwide attention due to the first report of human infections and the continuing reported cases in China. In this work, homology modeling, docking and molecular dynamics simulations were combined to study the interactions between neuraminidase ( N 9_2013, from novel A/ H 7 N 9 virus) and agents zanamivir, oseltamivir, peramivir. It was found that N 9_2013 protein is structurally close to the template (PDB code: 1F8B), especially the active site. The binding properties of N 9_2013 protein were nearly identical to those of template. As a result, the three available drugs should be still efficacious for the new emerging A ( H 7 N 9) virus. However, the stabilities of docked complexes and binding affinities (Eint) were slightly reduced, in contrast to the corresponding inhibitor-template complexes, with the values of -82.27 (-84.30), -78.84 (-80.28) and -77.52 (-81.94) kcal mol-1, respectively. Besides, R292K mutation might induce the resistance of the novel virus to the commercial inhibitors. Thus, it arouses the need for continuous monitoring of antiviral drug susceptibilities.
Collapse
Affiliation(s)
- ZHIWEI YANG
- School of Basic Medical Sciences, Jiamusi University, Jiamusi 154007, P. R. China
| | - FEI WU
- School of Basic Medical Sciences, Jiamusi University, Jiamusi 154007, P. R. China
| | - JUNXING LIU
- School of Basic Medical Sciences, Jiamusi University, Jiamusi 154007, P. R. China
| | - SHUQIU WANG
- School of Basic Medical Sciences, Jiamusi University, Jiamusi 154007, P. R. China
| | - XIAOHUI YUAN
- Institute of Biomedicine, Jinan University, Guangzhou 510632, P. R. China
| |
Collapse
|
33
|
Smee DF, Tarbet EB, Furuta Y, Morrey JD, Barnard DL. Synergistic combinations of favipiravir and oseltamivir against wild-type pandemic and oseltamivir-resistant influenza A virus infections in mice. Future Virol 2013; 8:1085-1094. [PMID: 24563658 DOI: 10.2217/fvl.13.98] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
AIM Favipiravir and oseltamivir are antiviral compounds used for the treatment of influenza infections. We have aimed to investigate the efficacy of the compounds in combination to treat influenza H1N1 virus infections in mice. MATERIALS & METHODS Mice infected with pandemic influenza A/California/04/2009 (H1N1pdm) virus or an oseltamivir-resistant (H275Y neuraminidase mutation) influenza A/Mississippi/ 3/2001 (H1N1) virus were treated orally with inhibitors twice a day for 5 days starting 4 h after infection. RESULTS Complete protection from death was afforded by favipiravir treatments of 100 mg/kg/day, but lower doses were less effective. Combinations of oseltamivir (1 and 3 mg/kg/day) with favipiravir (3, 10 and 30 mg/kg/day) resulted in a synergistic improvement in survival rates against H1N1pdm infections. Significant reductions in lung virus titers also occurred. Against the H275Y virus infection, oseltamivir alone was only 30% protective from death at 100 mg/kg/day, but combinations of the two compounds produced a synergistic improvement in survival rate. CONCLUSION The utility of treating H1N1 influenza virus infections with oseltamivir and favipiravir in combination has been established.
Collapse
Affiliation(s)
- Donald F Smee
- Institute for Antiviral Research, Department of Animal, Dairy & Veterinary Sciences, Utah State University, Logan, UT, USA
| | - E Bart Tarbet
- Institute for Antiviral Research, Department of Animal, Dairy & Veterinary Sciences, Utah State University, Logan, UT, USA
| | | | - John D Morrey
- Institute for Antiviral Research, Department of Animal, Dairy & Veterinary Sciences, Utah State University, Logan, UT, USA
| | - Dale L Barnard
- Institute for Antiviral Research, Department of Animal, Dairy & Veterinary Sciences, Utah State University, Logan, UT, USA
| |
Collapse
|
34
|
Characterization of two distinct neuraminidases from avian-origin human-infecting H7N9 influenza viruses. Cell Res 2013; 23:1347-55. [PMID: 24165891 PMCID: PMC3847574 DOI: 10.1038/cr.2013.144] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Revised: 09/24/2013] [Accepted: 09/25/2013] [Indexed: 12/15/2022] Open
Abstract
An epidemic of an avian-origin H7N9 influenza virus has recently emerged in China, infecting 134 patients of which 45 have died. This is the first time that an influenza virus harboring an N9 serotype neuraminidase (NA) has been known to infect humans. H7N9 viruses are divergent and at least two distinct NAs and hemagglutinins (HAs) have been found, respectively, from clinical isolates. The prototypes of these viruses are A/Anhui/1/2013 and A/Shanghai/1/2013. NAs from these two viruses are distinct as the A/Shanghai/1/2013 NA has an R294K substitution that can confer NA inhibitor oseltamivir resistance. Oseltamivir is by far the most commonly used anti-influenza drug due to its potency and high bioavailability. In this study, we show that an R294K substitution results in multidrug resistance with extreme oseltamivir resistance (over 100 000-fold) using protein- and virus-based assays. To determine the molecular basis for the inhibitor resistance, we solved high-resolution crystal structures of NAs from A/Anhui/1/2013 N9 (R294-containing) and A/Shanghai/1/2013 N9 (K294-containing). R294K substitution results in an unfavorable E276 conformation for oseltamivir binding, and consequently loss of inhibitor carboxylate interactions, which compromises the binding of all classical NA ligands/inhibitors. Moreover, we found that R294K substitution results in reduced NA catalytic efficiency along with lower viral fitness. This helps to explain why K294 has predominantly been found in clinical cases of H7N9 infection under the selective pressure of oseltamivir treatment and not in the dominant human-infecting viruses. This implies that oseltamivir can still be efficiently used in the treatment of H7N9 infections.
Collapse
|
35
|
Baranovich T, Burnham AJ, Marathe BM, Armstrong J, Guan Y, Shu Y, Peiris JMS, Webby RJ, Webster RG, Govorkova EA. The neuraminidase inhibitor oseltamivir is effective against A/Anhui/1/2013 (H7N9) influenza virus in a mouse model of acute respiratory distress syndrome. J Infect Dis 2013; 209:1343-53. [PMID: 24133191 DOI: 10.1093/infdis/jit554] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND High mortality and uncertainty about the effectiveness of neuraminidase inhibitors (NAIs) in humans infected with influenza A(H7N9) viruses are public health concerns. METHODS Susceptibility of N9 viruses to NAIs was determined in a fluorescence-based assay. The NAI oseltamivir (5, 20, or 80 mg/kg/day) was administered to BALB/c mice twice daily starting 24, 48, or 72 hours after A/Anhui/1/2013 (H7N9) virus challenge. RESULTS All 12 avian N9 and 3 human H7N9 influenza viruses tested were susceptible to NAIs. Without prior adaptation, A/Anhui/1/2013 (H7N9) caused lethal infection in mice that was restricted to the respiratory tract and resulted in pulmonary edema and acute lung injury with hyaline membrane formation, leading to decreased oxygenation, all characteristics of human acute respiratory distress syndrome. Oseltamivir at 20 and 80 mg/kg protected 80% and 88% of mice when initiated after 24 hours, and the efficacy decreased to 70% and 60%, respectively, when treatment was delayed by 48 hours. Emergence of oseltamivir-resistant variants was not detected. CONCLUSIONS H7N9 viruses are comparable to currently circulating influenza A viruses in susceptibility to NAIs. Based on these animal studies, early treatment is associated with improved outcomes.
Collapse
Affiliation(s)
- Tatiana Baranovich
- Department of Infectious Diseases, St Jude Children's Research Hospital, Memphis, Tennessee
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Gillman A, Muradrasoli S, Söderström H, Nordh J, Bröjer C, Lindberg RH, Latorre-Margalef N, Waldenström J, Olsen B, Järhult JD. Resistance mutation R292K is induced in influenza A(H6N2) virus by exposure of infected mallards to low levels of oseltamivir. PLoS One 2013; 8:e71230. [PMID: 23951116 PMCID: PMC3741332 DOI: 10.1371/journal.pone.0071230] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2012] [Accepted: 06/29/2013] [Indexed: 01/24/2023] Open
Abstract
Resistance to neuraminidase inhibitors (NAIs) is problematic as these drugs constitute the major treatment option for severe influenza. Extensive use of the NAI oseltamivir (Tamiflu®) results in up to 865 ng/L of its active metabolite oseltamivir carboxylate (OC) in river water. There one of the natural reservoirs of influenza A, dabbling ducks, can be exposed. We previously demonstrated that an influenza A(H1N1) virus in mallards (Anas platyrhynchos) exposed to 1 µg/L of OC developed oseltamivir resistance through the mutation H274Y (N2-numbering). In this study, we assessed the resistance development in an A(H6N2) virus, which belongs to the phylogenetic N2 group of neuraminidases with distinct functional and resistance characteristics. Mallards were infected with A(H6N2) while exposed to 120 ng/L, 1.2 µg/L or 12 µg/L of OC in their sole water source. After 4 days with 12 µg/L of OC exposure, the resistance mutation R292K emerged and then persisted. Drug sensitivity was decreased ≈13,000-fold for OC and ≈7.8-fold for zanamivir. Viral shedding was similar when comparing R292K and wild-type virus indicating sustained replication and transmission. Reduced neuraminidase activity and decrease in recovered virus after propagation in embryonated hen eggs was observed in R292K viruses. The initial, but not the later R292K isolates reverted to wild-type during egg-propagation, suggesting a stabilization of the mutation, possibly through additional mutations in the neuraminidase (D113N or D141N) or hemagglutinin (E216K). Our results indicate a risk for OC resistance development also in a N2 group influenza virus and that exposure to one NAI can result in a decreased sensitivity to other NAIs as well. If established in influenza viruses circulating among wild birds, the resistance could spread to humans via re-assortment or direct transmission. This could potentially cause an oseltamivir-resistant pandemic; a serious health concern as preparedness plans rely heavily on oseltamivir before vaccines can be mass-produced.
Collapse
Affiliation(s)
- Anna Gillman
- Section of Infectious Diseases, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Shaman Muradrasoli
- Section of Bacteriology and Food Safety, Department of Biomedical Sciences and Veterinary Public Health, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | | | - Johan Nordh
- Section of Infectious Diseases, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Caroline Bröjer
- Section of Pathology, Pharmacology and Toxicology, Department of Biomedical Sciences and Veterinary Public Health, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | | | - Neus Latorre-Margalef
- Section for Zoonotic Ecology and Epidemiology, School of Natural Sciences, Linnaeus University, Kalmar, Sweden
| | - Jonas Waldenström
- Section for Zoonotic Ecology and Epidemiology, School of Natural Sciences, Linnaeus University, Kalmar, Sweden
| | - Björn Olsen
- Section of Infectious Diseases, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Josef D. Järhult
- Section of Infectious Diseases, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
- * E-mail:
| |
Collapse
|
37
|
Abstract
Despite 75 years of research into prevention and treatment of influenza, the viruses that cause this disease continue to rank as some of the most important pathogens afflicting humans today. Progress in development of therapeutics for influenza has been slow for much of that time, but has accelerated in pace over the last two decades. Two classes of antiviral medications are used in humans at present, but each has limitations in scope and effectiveness of use. New strategies involving these licensed agents, including alternate forms of delivery and combination therapy with other drugs, are currently being explored. In addition, several novel antiviral compounds are in various clinical phases of development. Together with strategies designed to target the virus itself, new approaches to interrupt host–pathogen interactions or modulate detrimental aspects of the immune response have been proposed. Therapy for influenza will likely undergo substantial changes in the decades to come, evolving with our knowledge of pathogenesis as new approaches become viable and are validated clinically.
Collapse
Affiliation(s)
- Elena A Govorkova
- Department of Infectious Diseases, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105-3678, USA.
| | | |
Collapse
|
38
|
An Y, Rininger JA, Jarvis DL, Jing X, Ye Z, Aumiller JJ, Eichelberger M, Cipollo JF. Comparative glycomics analysis of influenza Hemagglutinin (H5N1) produced in vaccine relevant cell platforms. J Proteome Res 2013; 12:3707-20. [PMID: 23848607 DOI: 10.1021/pr400329k] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Hemagglutinin (HA) is the major antigen in influenza vaccines, and glycosylation is known to influence its antigenicity. Embryonated hen eggs are traditionally used for influenza vaccine production, but vaccines produced in mammalian and insect cells were recently licensed. This raises the concern that vaccines produced with different cell systems might not be equivalent due to differences in their glycosylation patterns. Thus, we developed an analytical method to monitor vaccine glycosylation through a combination of nanoLC/MS(E) and quantitative MALDI-TOF MS permethylation profiling. We then used this method to examine glycosylation of HAs from two different influenza H5N1 strains produced in five different platforms, including hen eggs, three different insect cell lines (High Five, expresSF+ and glycoengineered expresSF+), and a human cell line (HEK293). Our results demonstrated that (1) sequon utilization is not necessarily equivalent in different cell types, (2) there are quantitative and qualitative differences in the overall N-glycosylation patterns and structures produced by different cell types, (3) ∼20% of the N-glycans on the HAs produced by High Five cells are core α1,3-fucosylated structures, which may be allergenic in humans, and (4) our method can be used to monitor differences in glycosylation during the cellular glycoengineering stages of vaccine development.
Collapse
Affiliation(s)
- Yanming An
- Center for Biologics Evaluation and Research, Food and Drug Administration, Bethesda, Maryland 20892, USA
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Govorkova EA. Consequences of resistance: in vitro fitness, in vivo infectivity, and transmissibility of oseltamivir-resistant influenza A viruses. Influenza Other Respir Viruses 2013; 7 Suppl 1:50-7. [PMID: 23279897 DOI: 10.1111/irv.12044] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
The development of drug resistance is a major drawback to any antiviral therapy, and the specific anti-influenza drugs, the neuraminidase (NA) inhibitors (NAIs), are not excluded from this rule. The impact of drug resistance depends on the degree of reduction in fitness of the particular drug-resistant virus. If the resistance mutations lead to only a modest biological fitness cost and the virus remains highly transmissible, the effectiveness of antiviral use is likely to be reduced. This review focuses on the fitness of oseltamivir-resistant seasonal H1N1 and H3N2, 2009 pandemic H1N1 (H1N1pdm09), and highly pathogenic H5N1 influenza A viruses carrying clinically derived NAI resistance-associated NA mutations.
Collapse
Affiliation(s)
- Elena A Govorkova
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN 38105-2794, USA.
| |
Collapse
|
40
|
Chao DL. Modeling the global transmission of antiviral-resistant influenza viruses. Influenza Other Respir Viruses 2013; 7 Suppl 1:58-62. [PMID: 23279898 DOI: 10.1111/irv.12043] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
Abstract
BACKGROUND The mutations that confer resistance to antiviral agents are thought to be detrimental, or at best neutral, to influenza virus fitness. The fact that resistant influenza strains can circulate and sometimes replace sensitive strains is of great public health concern. OBJECTIVES We used mathematical modeling to test various hypotheses about the transmission of antiviral-resistant influenza viruses by comparing the model's output with the observed rise in antiviral resistance of seasonal A(H1N1) influenza viruses between 2006 and 2009. METHODS We developed a mathematical model of the transmission of influenza among 321 cities around the globe. In the model, influenza strains resistant to antiviral agents competed with sensitive strains. RESULTS AND CONCLUSIONS We found that a resistant strain of influenza could not displace the sensitive strain as rapidly as has been observed unless it was more transmissible than the sensitive strain in the general population. We believe that an antiviral-resistant strain displaced the antiviral-sensitive seasonal A(H1N1) virus by hitchhiking on an escape mutation. Because of the complex global patterns of influenza circulation, tracking the emergence and spread of antiviral resistance must be a coordinated global effort.
Collapse
Affiliation(s)
- Dennis L Chao
- Center for Statistics and Quantitative Infectious Diseases, Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA.
| |
Collapse
|
41
|
Abstract
ABSTRACTObjectives: The primary objective of this study was to determine the preparedness for pandemic influenza of hospitals, in terms of amount of antiviral drugs on hand and employee vaccination rates, in the Finger Lakes region (FLR) of western New York.Methods: A survey of the 17 FLR hospitals was conducted via e-mail during the period of June 2007 to August 2007.Results: A total of 13 of 17 hospitals responded for a response rate of 76.5%. Only 23.1% of responding hospitals stockpile antiviral drugs. Vaccination rates for personnel with patient contact ranged from 36.8% to 76.1%.Conclusions: Hospitals in the FLR have insufficient quantities of antiviral agents stockpiled to provide for the protection of health care workers, and influenza vaccination rates for health care workers are low. To ensure that a high level of care is maintained during a pandemic, health care workers need to be provided with appropriate protection. This can be accomplished if hospitals stockpile antiviral agents designated for the treatment and prophylaxis of health care workers with patient contact and their families.(Disaster Med Public Health Preparedness. 2010;4:55-61)
Collapse
|
42
|
Influenza virus resistance to neuraminidase inhibitors. Antiviral Res 2013; 98:174-85. [PMID: 23523943 DOI: 10.1016/j.antiviral.2013.03.014] [Citation(s) in RCA: 246] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Revised: 02/26/2013] [Accepted: 03/14/2013] [Indexed: 11/23/2022]
Abstract
In addition to immunization programs, antiviral agents can play a major role for the control of seasonal influenza epidemics and may also provide prophylactic and therapeutic benefits during an eventual pandemic. The purpose of this article is to review the mechanism of action, pharmacokinetics and clinical indications of neuraminidase inhibitors (NAIs) with an emphasis on the emergence of antiviral drug resistance. There are two approved NAIs compounds in US: inhaled zanamivir and oral oseltamivir, which have been commercially available since 1999-2000. In addition, two other NAIs, peramivir (an intravenous cyclopentane derivative) and laninamivir (a long-acting NAI administered by a single nasal inhalation) have been approved in certain countries and are under clinical evaluations in others. As for other antivirals, the development and dissemination of drug resistance is a significant threat to the clinical utility of NAIs. The emergence and worldwide spread of oseltamivir-resistant seasonal A(H1N1) viruses during the 2007-2009 seasons emphasize the need for continuous monitoring of antiviral drug susceptibilities. Further research priorities should include a better understanding of the mechanisms of resistance to existing antivirals, the development of novel compounds which target viral or host proteins and the evaluation of combination therapies for improved treatment of severe influenza infections, particularly in immunocompromised individuals. This article forms part of a symposium in Antiviral Research on "Treatment of influenza: targeting the virus or the host."
Collapse
|
43
|
Hsu KC, Hung HC, Horng JT, Fang MY, Chang CY, Li LT, Chen IJ, Chen YC, Chou DL, Chang CW, Hsieh HP, Yang JM, Hsu JTA. Parallel screening of wild-type and drug-resistant targets for anti-resistance neuraminidase inhibitors. PLoS One 2013; 8:e56704. [PMID: 23437217 PMCID: PMC3577712 DOI: 10.1371/journal.pone.0056704] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2012] [Accepted: 01/14/2013] [Indexed: 11/19/2022] Open
Abstract
Infection with influenza virus is a major public health problem, causing serious illness and death each year. Emergence of drug-resistant influenza virus strains limits the effectiveness of drug treatment. Importantly, a dual H275Y/I223R mutation detected in the pandemic influenza A 2009 virus strain results in multidrug resistance to current neuraminidase (NA) drugs. Therefore, discovery of new agents for treating multiple drug-resistant (MDR) influenza virus infections is important. Here, we propose a parallel screening strategy that simultaneously screens wild-type (WT) and MDR NAs, and identifies inhibitors matching the subsite characteristics of both NA-binding sites. These may maintain their potency when drug-resistant mutations arise. Initially, we analyzed the subsite of the dual H275Y/I223R NA mutant. Analysis of the site-moiety maps of NA protein structures show that the mutant subsite has a relatively small volume and is highly polar compared with the WT subsite. Moreover, the mutant subsite has a high preference for forming hydrogen-bonding interactions with polar moieties. These changes may drive multidrug resistance. Using this strategy, we identified a new inhibitor, Remazol Brilliant Blue R (RB19, an anthraquinone dye), which inhibited WT NA and MDR NA with IC50 values of 3.4 and 4.5 µM, respectively. RB19 comprises a rigid core scaffold and a flexible chain with a large polar moiety. The former interacts with highly conserved residues, decreasing the probability of resistance. The latter forms van der Waals contacts with the WT subsite and yields hydrogen bonds with the mutant subsite by switching the orientation of its flexible side chain. Both scaffolds of RB19 are good starting points for lead optimization. The results reveal a parallel screening strategy for identifying resistance mechanisms and discovering anti-resistance neuraminidase inhibitors. We believe that this strategy may be applied to other diseases with high mutation rates, such as cancer and human immunodeficiency virus type 1.
Collapse
Affiliation(s)
- Kai-Cheng Hsu
- Institute of Bioinformatics and Systems Biology, National Chiao Tung University, Hsinchu, Taiwan
| | - Hui-Chen Hung
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli, Taiwan
| | - Jim-Tong Horng
- Department of Biochemistry, Chang Gung University, Taoyuan, Taiwan
| | - Ming-Yu Fang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli, Taiwan
| | - Chun-Yu Chang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli, Taiwan
| | - Ling-Ting Li
- Institute of Bioinformatics and Systems Biology, National Chiao Tung University, Hsinchu, Taiwan
| | - I-Jung Chen
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli, Taiwan
| | - Yun-Chu Chen
- Department of Biochemistry, Chang Gung University, Taoyuan, Taiwan
| | - Ding-Li Chou
- Department of Biochemistry, Chang Gung University, Taoyuan, Taiwan
| | - Chun-Wei Chang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli, Taiwan
| | - Hsing-Pang Hsieh
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli, Taiwan
| | - Jinn-Moon Yang
- Institute of Bioinformatics and Systems Biology, National Chiao Tung University, Hsinchu, Taiwan
- Department of Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan
- Center for Bioinformatics Research, National Chiao Tung University, Hsinchu, Taiwan
- * E-mail: (JMY); (JTAH)
| | - John T.-A. Hsu
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli, Taiwan
- Department of Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan
- * E-mail: (JMY); (JTAH)
| |
Collapse
|
44
|
van der Vries E, Schutten M, Fraaij P, Boucher C, Osterhaus A. Influenza virus resistance to antiviral therapy. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2013; 67:217-46. [PMID: 23886002 DOI: 10.1016/b978-0-12-405880-4.00006-8] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Antiviral drugs for influenza therapy and prophylaxis are either of the adamantane or neuraminidase inhibitor (NAI) class. However, the NAIs are mainly prescribed nowadays, because of widespread adamantane resistance among influenza A viruses and ineffectiveness of adamantanes against influenza B. Emergence and spread of NAI resistance would further limit our therapeutic options. Taking into account the previous spread of oseltamivir-resistant viruses during the 2007/2008 season preceding the last pandemic, emergence of yet another naturally NAI-resistant influenza virus may not be an unlikely event. This previous incident also underlines the importance of resistance surveillance and asks for a better understanding of the mechanisms underlying primary resistance development. We provide an overview of the major influenza antiviral resistance mechanisms and future therapies for influenza. Here, we call for a better understanding of the effect of virus mutations upon antiviral treatment and for a tailored antiviral approach to severe influenza virus infections.
Collapse
|
45
|
Pandemic 2009 H1N1 influenza A virus carrying a Q136K mutation in the neuraminidase gene is resistant to zanamivir but exhibits reduced fitness in the guinea pig transmission model. J Virol 2012. [PMID: 23192869 DOI: 10.1128/jvi.02507-12] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Resistance of influenza A viruses to neuraminidase inhibitors can arise through mutations in the neuraminidase (NA) gene. We show here that a Q136K mutation in the NA of the 2009 pandemic H1N1 virus confers a high degree of resistance to zanamivir. Resistance is accompanied by reduced numbers of NA molecules in viral particles and reduced intrinsic enzymatic activity of mutant NA. Interestingly, the Q136K mutation strongly impairs viral fitness in the guinea pig transmission model.
Collapse
|
46
|
van der Vries E, Collins PJ, Vachieri SG, Xiong X, Liu J, Walker PA, Haire LF, Hay AJ, Schutten M, Osterhaus ADME, Martin SR, Boucher CAB, Skehel JJ, Gamblin SJ. H1N1 2009 pandemic influenza virus: resistance of the I223R neuraminidase mutant explained by kinetic and structural analysis. PLoS Pathog 2012; 8:e1002914. [PMID: 23028314 PMCID: PMC3447749 DOI: 10.1371/journal.ppat.1002914] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2012] [Accepted: 08/05/2012] [Indexed: 12/16/2022] Open
Abstract
Two classes of antiviral drugs, neuraminidase inhibitors and adamantanes, are approved for prophylaxis and therapy against influenza virus infections. A major concern is that antiviral resistant viruses emerge and spread in the human population. The 2009 pandemic H1N1 virus is already resistant to adamantanes. Recently, a novel neuraminidase inhibitor resistance mutation I223R was identified in the neuraminidase of this subtype. To understand the resistance mechanism of this mutation, the enzymatic properties of the I223R mutant, together with the most frequently observed resistance mutation, H275Y, and the double mutant I223R/H275Y were compared. Relative to wild type, K(M) values for MUNANA increased only 2-fold for the single I223R mutant and up to 8-fold for the double mutant. Oseltamivir inhibition constants (K(I)) increased 48-fold in the single I223R mutant and 7500-fold in the double mutant. In both cases the change was largely accounted for by an increased dissociation rate constant for oseltamivir, but the inhibition constants for zanamivir were less increased. We have used X-ray crystallography to better understand the effect of mutation I223R on drug binding. We find that there is shrinkage of a hydrophobic pocket in the active site as a result of the I223R change. Furthermore, R223 interacts with S247 which changes the rotamer it adopts and, consequently, binding of the pentoxyl substituent of oseltamivir is not as favorable as in the wild type. However, the polar glycerol substituent present in zanamivir, which mimics the natural substrate, is accommodated in the I223R mutant structure in a similar way to wild type, thus explaining the kinetic data. Our structural data also show that, in contrast to a recently reported structure, the active site of 2009 pandemic neuraminidase can adopt an open conformation.
Collapse
Affiliation(s)
| | - Patrick J. Collins
- Medical Research Council, National Institute for Medical Research, The Ridgeway, Mill Hill, London, United Kingdom
| | - Sebastien G. Vachieri
- Medical Research Council, National Institute for Medical Research, The Ridgeway, Mill Hill, London, United Kingdom
| | - Xiaoli Xiong
- Medical Research Council, National Institute for Medical Research, The Ridgeway, Mill Hill, London, United Kingdom
| | - Junfeng Liu
- Medical Research Council, National Institute for Medical Research, The Ridgeway, Mill Hill, London, United Kingdom
- MOA Key Laboratory of Plant Pathology, China Agricultural University, Beijing, People's Republic of China
| | - Philip A. Walker
- Medical Research Council, National Institute for Medical Research, The Ridgeway, Mill Hill, London, United Kingdom
| | - Lesley F. Haire
- Medical Research Council, National Institute for Medical Research, The Ridgeway, Mill Hill, London, United Kingdom
| | - Alan J. Hay
- Medical Research Council, National Institute for Medical Research, The Ridgeway, Mill Hill, London, United Kingdom
| | - Martin Schutten
- Erasmus Medical Centre, Department of Virology, Rotterdam, The Netherlands
| | | | - Steve R. Martin
- Medical Research Council, National Institute for Medical Research, The Ridgeway, Mill Hill, London, United Kingdom
| | | | - John J. Skehel
- Medical Research Council, National Institute for Medical Research, The Ridgeway, Mill Hill, London, United Kingdom
- * E-mail:
| | - Steve J. Gamblin
- Medical Research Council, National Institute for Medical Research, The Ridgeway, Mill Hill, London, United Kingdom
| |
Collapse
|
47
|
Smee DF, Julander JG, Tarbet EB, Gross M, Nguyen J. Treatment of oseltamivir-resistant influenza A (H1N1) virus infections in mice with antiviral agents. Antiviral Res 2012; 96:13-20. [PMID: 22809862 DOI: 10.1016/j.antiviral.2012.07.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2011] [Revised: 06/11/2012] [Accepted: 07/06/2012] [Indexed: 12/13/2022]
Abstract
Influenza A/Mississippi/03/2001 (H1N1) and A/Hong Kong/2369/2009 (H1N1) viruses containing the neuraminidase gene mutation H275Y (conferring resistance to oseltamivir) were adapted to mice and evaluated for suitability as models for lethal infection and antiviral treatment. The viral neuraminidases were resistant to peramivir and oseltamivir carboxylate but sensitive to zanamivir. Similar pattern of antiviral activity were seen in MDCK cell assays. Lethal infections were achieved in mice with the two viruses. Oral oseltamivir at 100 and 300mg/kg/day bid for 5day starting at -2h gave 30% and 60% protection from death, respectively, due to the A/Mississippi/03/2001 infection. Intraperitoneal treatments with zanamivir at 30 and 100mg/kg/day starting at -2h gave 60% and 90% protection, respectively. Neither compound at <300mg/kg/day protected mice when treatments began at +24h. Amantadine was effective at 10, 30, and 100mg/kg/day, rimantadine was protective at 10 and 30mg/kg/day (highest dose tested), and ribavirin was active at 30 and 75mg/kg/day, with survival ranging from 60-100% for oral treatments initiated at -2h. For treatments begun at +24h, amantadine was protective at 30 and 100mg/kg/day, rimantadine showed efficacy at 10 and 30mg/kg/day, and ribavirin was active at 75mg/kg/day, with 60-100% survival per group. In the A/Hong Kong/2369/2009 infection, oral oseltamivir at 100 and 300mg/kg/day starting at -2h gave 50% and 70% protection from death, respectively. These infection models will be useful to study newly discovered anti-influenza virus agents and to evaluate compounds in combination.
Collapse
Affiliation(s)
- Donald F Smee
- Institute for Antiviral Research, Utah State University, Logan, UT 84322, USA.
| | | | | | | | | |
Collapse
|
48
|
Hung HC, Liu CL, Hsu JTA, Horng JT, Fang MY, Wu SY, Ueng SH, Wang MY, Yaw CW, Hou MH. Development of an anti-influenza drug screening assay targeting nucleoproteins with tryptophan fluorescence quenching. Anal Chem 2012; 84:6391-9. [PMID: 22712523 DOI: 10.1021/ac2022426] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Recent studies have shown that NP (nucleoprotein), which possesses multiple functions in the viral life cycle, is a new potential anti-influenza drug target. NP inhibitors reliably induce conformational changes in NPs, and these changes may confer inhibition of the influenza virus. The six conserved tryptophan residues in NP can be used as an intrinsic probe to monitor the change in fluorescence of the tryptophan residues in the protein upon binding to an NP inhibitor. In the present study, we found that the fluorescence of recombinant NP proteins was quenched following the binding of available NP inhibitors (such as nucleozin) in a concentration- and time-dependent manner, which suggests that the inhibitor induced conformational changes in the NPs. The minimal fluorescence-quenching effect and weak binding constant of nucleozin to the swine-origin influenza virus H1N1pdm09 (SOIV) NP revealed that the SOIV is resistant to nucleozin. We have used the fluorescence-quenching property of tryptophans in NPs that were bound to ligands in a 96-well-plate-based drug screen to assess the ability of promising small molecules to interact with NPs and have identified one new anti-influenza drug, CSV0C001018, with a high SI value. This convenient method for drug screening may facilitate the development of antiviral drugs that target viruses other than the influenza virus, such as HIV and HBV.
Collapse
Affiliation(s)
- Hui-Chen Hung
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli, Taiwan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Johansson BE, Cox MMJ. Influenza viral neuraminidase: the forgotten antigen. Expert Rev Vaccines 2012; 10:1683-95. [PMID: 22085172 DOI: 10.1586/erv.11.130] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Influenza is the most common cause of vaccine-preventable morbidity and mortality despite the availability of the conventional trivalent inactivated vaccine and the live-attenuated influenza vaccine. These vaccines induce an immunity dominated by the response to hemagglutinin (HA) and are most effective when there is sufficient antigenic relatedness between the vaccine strain and the HA of the circulating wild-type virus. Vaccine strategies against influenza may benefit from inclusion of other viral antigens in addition to HA. Epidemiologic evidence and studies in animals and humans indicate that anti-neuraminidase (NA) immunity will provide protection against severe illness or death in the event of a significant antigenic change in the HA component of the vaccine. However, there is little NA immunity induced by trivalent inactivated vaccine and live-attenuated influenza vaccine. The quantity of NA in influenza vaccines is not standardized and varies significantly among manufacturers, production lots and tested strains. The activity and stability of the NA enzyme is influenced by concentration of divalent cations. If immunity against NA is desirable, a better understanding of how the enzymatic properties affect the immunogenicity is needed.
Collapse
Affiliation(s)
- Bert E Johansson
- Department of Pediatrics, Texas Tech University Health Sciences Center, Paul H Foster School of Medicine and El Paso Children?s Hospital, 4825 Alameda Avenue El Paso, TX 79905, USA.
| | | |
Collapse
|
50
|
Abstract
PURPOSE OF REVIEW The 2009 influenza pandemic introduced a new influenza A/H1N1 subtype in the human population. This pandemic 2009 influenza A/H1N1 virus has natural resistance to the adamantanes class and has a low threshold to become resistant to the neuraminidase class of antiviral drugs. This review describes recent findings on influenza antiviral resistance in pandemic 2009 influenza A/H1N1 virus. RECENT FINDINGS Pandemic 2009 viruses have emerged with novel resistance patterns to the neuraminidase inhibitors. In addition, the identification of mutations that facilitated oseltamivir resistance in prepandemic influenza emphasizes the ability of influenza to become resistant to antiviral drugs without significant loss of fitness. SUMMARY Novel initiatives are required to find and develop high genetic barrier influenza therapeutic regimens for effective treatment of severe influenza virus infections.
Collapse
|