1
|
Rodríguez‐Negrete EA, Guevara‐Rivera EA, Arce‐Leal ÁP, Leyva‐López NE, Méndez‐Lozano J. A novel tomato spotted wilt virus isolate encoding a noncanonical NSm C118F substitution associated with Sw-5 tomato gene resistance breaking. MOLECULAR PLANT PATHOLOGY 2023; 24:1300-1311. [PMID: 37403515 PMCID: PMC10502823 DOI: 10.1111/mpp.13371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 06/07/2023] [Accepted: 06/08/2023] [Indexed: 07/06/2023]
Abstract
The nonstructural protein NSm of tomato spotted wilt virus (TSWV) has been identified as the avirulence determinant of the tomato single dominant Sw-5 resistance gene. Although Sw-5 effectiveness has been shown for most TSWV isolates, the emergence of resistance-breaking (RB) isolates has been observed. It is strongly associated with two point mutations (C118Y or T120N) in the NSm viral protein. TSWV-like symptoms were observed in tomato crop cultivars (+Sw-5) in the Baja California peninsula, Mexico, and molecular methods confirmed the presence of TSWV. Sequence analysis of the NSm 118-120 motif and three-dimensional protein modelling exhibited a noncanonical C118F substitution in seven isolates, suggesting that this substitution could emulate the C118Y-related RB phenotype. Furthermore, phylogenetic and molecular analysis of the full-length genome (TSWV-MX) revealed its reassortment-related evolution and confirmed that putative RB-related features are restricted to the NSm protein. Biological and mutational NSm 118 residue assays in tomato (+Sw-5) confirmed the RB nature of TSWV-MX isolate, and the F118 residue plays a critical role in the RB phenotype. The discovery of a novel TSWV-RB Mexican isolate with the presence of C118F substitution highlights a not previously described viral adaptation in the genus Orthotospovirus, and hence, the necessity of further crop monitoring to alert the establishment of novel RB isolates in cultivated tomatoes.
Collapse
|
2
|
Yang Z, Ye Z, Qiu J, Feng R, Li D, Hsieh C, Allcock J, Zhang S. A mutation-induced drug resistance database (MdrDB). Commun Chem 2023; 6:123. [PMID: 37316673 DOI: 10.1038/s42004-023-00920-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Accepted: 06/02/2023] [Indexed: 06/16/2023] Open
Abstract
Mutation-induced drug resistance is a significant challenge to the clinical treatment of many diseases, as structural changes in proteins can diminish drug efficacy. Understanding how mutations affect protein-ligand binding affinities is crucial for developing new drugs and therapies. However, the lack of a large-scale and high-quality database has hindered the research progresses in this area. To address this issue, we have developed MdrDB, a database that integrates data from seven publicly available datasets, which is the largest database of its kind. By integrating information on drug sensitivity and cell line mutations from Genomics of Drug Sensitivity in Cancer and DepMap, MdrDB has substantially expanded the existing drug resistance data. MdrDB is comprised of 100,537 samples of 240 proteins (which encompass 5119 total PDB structures), 2503 mutations, and 440 drugs. Each sample brings together 3D structures of wild type and mutant protein-ligand complexes, binding affinity changes upon mutation (ΔΔG), and biochemical features. Experimental results with MdrDB demonstrate its effectiveness in significantly enhancing the performance of commonly used machine learning models when predicting ΔΔG in three standard benchmarking scenarios. In conclusion, MdrDB is a comprehensive database that can advance the understanding of mutation-induced drug resistance, and accelerate the discovery of novel chemicals.
Collapse
Affiliation(s)
- Ziyi Yang
- Tencent Quantum Laboratory, Shenzhen, 518057, Guangdong, China
| | - Zhaofeng Ye
- Tencent Quantum Laboratory, Shenzhen, 518057, Guangdong, China
| | - Jiezhong Qiu
- Tencent Quantum Laboratory, Shenzhen, 518057, Guangdong, China
| | - Rongjun Feng
- Tencent Quantum Laboratory, Shenzhen, 518057, Guangdong, China
| | - Danyu Li
- Tencent Quantum Laboratory, Shenzhen, 518057, Guangdong, China
| | - Changyu Hsieh
- Tencent Quantum Laboratory, Shenzhen, 518057, Guangdong, China
| | | | - Shengyu Zhang
- Tencent Quantum Laboratory, Shenzhen, 518057, Guangdong, China.
| |
Collapse
|
3
|
Wong KH, Lal SK. Alternative antiviral approaches to combat influenza A virus. Virus Genes 2023; 59:25-35. [PMID: 36260242 PMCID: PMC9832087 DOI: 10.1007/s11262-022-01935-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 09/06/2022] [Indexed: 01/14/2023]
Abstract
Influenza A (IAV) is a major human respiratory pathogen that contributes to a significant threat to health security, worldwide. Despite vaccinations and previous immunisations through infections, humans can still be infected with influenza several times throughout their lives. This phenomenon is attributed to the antigenic changes of hemagglutinin (HA) and neuraminidase (NA) proteins in IAV via genetic mutation and reassortment, conferring antigenic drift and antigenic shift, respectively. Numerous findings indicate that slow antigenic drift and reassortment-derived antigenic shift exhibited by IAV are key processes that allow IAVs to overcome the previously acquired host immunity, which eventually leads to the annual re-emergence of seasonal influenza and even pandemic influenza, in rare occasions. As a result, current therapeutic options hit a brick wall quickly. As IAV remains a constant threat for new outbreaks worldwide, the underlying processes of genetic changes and alternative antiviral approaches for IAV should be further explored to improve disease management. In the light of the above, this review discusses the characteristics and mechanisms of mutations and reassortments that contribute to IAV's evolution. We also discuss several alternative RNA-targeting antiviral approaches, namely the CRISPR/Cas13 systems, RNA interference (RNAi), and antisense oligonucleotides (ASO) as potential antiviral approaches against IAV.
Collapse
Affiliation(s)
- Ka Heng Wong
- School of Science, Monash University Malaysia, 47500, Bandar Sunway, Selangor DE, Malaysia
| | - Sunil K Lal
- School of Science, Monash University Malaysia, 47500, Bandar Sunway, Selangor DE, Malaysia.
- Tropical Medicine & Biology Multidisciplinary Platform, Monash University Malaysia, Bandar Sunway, 47500, Selangor, Malaysia.
| |
Collapse
|
4
|
Liu Y, Song X, Li C, Hu H, Li W, Wang L, Hu J, Liao C, Liang H, He Z, Ye L. Chrysin Ameliorates Influenza Virus Infection in the Upper Airways by Repressing Virus-Induced Cell Cycle Arrest and Mitochondria-Dependent Apoptosis. Front Immunol 2022; 13:872958. [PMID: 35432374 PMCID: PMC9009290 DOI: 10.3389/fimmu.2022.872958] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 03/14/2022] [Indexed: 11/13/2022] Open
Abstract
Chrysin has been proven to possess antiviral properties, but the precise underlying anti-influenza mechanism and its anti-influenza efficacy in vivo are largely unclear. In this study, we investigated the involvement of chrysin in the blockade of cell cycle and apoptosis in distinct cell lines subjected to two H1N1 influenza A virus (IAV) strains, as well as its anti-IAV activity in vivo. Here, we found an early unidentified finding that chrysin strongly impeded IAV replication through a mechanism that was autonomous of innate antiviral immune activation and viral protein interaction. Surprisingly, chrysin can suppress IAV-induced cell cycle arrest in the G0/G1 phase by downregulating the expression levels of P53 and P21 while promoting Cyclin D1/CDK4 and Cyclin E1/CDK2 activation. Furthermore, chrysin dramatically inhibited the IAV-triggered mitochondrial apoptotic pathway by altering the balance of Bax/Bcl-xl and reducing caspase-9 and caspase-3 activation. Accumulated reactive oxygen species (ROS) reduction may contribute to the inhibitory role of chrysin in cell cycle arrest and apoptosis following IAV infection. Notably, chrysin preferably inhibited IAV replication in the upper respiratory tract, indicating that it might be a promising drug for restraining the spread of respiratory viruses.
Collapse
Affiliation(s)
- Ying Liu
- Department of Pharmacy, Department of Immunology, International Cancer Center, Shenzhen University Health Science Center, Shenzhen, China
| | - Xun Song
- Department of Pharmacy, Department of Immunology, International Cancer Center, Shenzhen University Health Science Center, Shenzhen, China
| | - Chenyang Li
- Department of Pharmacy, Department of Immunology, International Cancer Center, Shenzhen University Health Science Center, Shenzhen, China
| | - Hao Hu
- Department of Pharmacy, Department of Immunology, International Cancer Center, Shenzhen University Health Science Center, Shenzhen, China
| | - Wanlin Li
- Department of Pharmacy, Department of Immunology, International Cancer Center, Shenzhen University Health Science Center, Shenzhen, China
| | - Lu Wang
- Department of Respiratory Medicine, Shenzhen University General Hospital, Shenzhen University, Shenzhen, China
| | - Jing Hu
- Department of Pharmacy, Department of Immunology, International Cancer Center, Shenzhen University Health Science Center, Shenzhen, China
| | - Chenghui Liao
- Department of Pharmacy, Department of Immunology, International Cancer Center, Shenzhen University Health Science Center, Shenzhen, China
| | - Hanbai Liang
- Department of Pharmacy, Department of Immunology, International Cancer Center, Shenzhen University Health Science Center, Shenzhen, China
| | - Zhendan He
- Department of Pharmacy, Department of Immunology, International Cancer Center, Shenzhen University Health Science Center, Shenzhen, China
- College of Pharmacy, Shenzhen Technology University, Shenzhen, China
| | - Liang Ye
- Department of Pharmacy, Department of Immunology, International Cancer Center, Shenzhen University Health Science Center, Shenzhen, China
| |
Collapse
|
5
|
Sangtani R, Ghosh A, Jha HC, Parmar HS, Bala K. Potential of algal metabolites for the development of broad-spectrum antiviral therapeutics: Possible implications in COVID-19 therapy. Phytother Res 2021; 35:2296-2316. [PMID: 33210447 PMCID: PMC7753317 DOI: 10.1002/ptr.6948] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 10/26/2020] [Accepted: 10/30/2020] [Indexed: 01/25/2023]
Abstract
Covid-19 pandemic severely affected human health worldwide. Till October 19, 2020, total confirmed patients of COVID-19 are 39,944,882, whereas 1,111,998 people died across the globe. Till to date, we do not have any specific medicine and/or vaccine to treat COVID-19; however, research is still going on at war footing. So far vaccine development is concerned, here it is noteworthy that till now three major variants (named A, B, and C) of severe acute respiratory syndrome-coronavirus2 (SARS-CoV-2) have been recognized. Increased mutational rate and formation of new viral variants may increase the attrition rate of vaccines and/or candidate chemotherapies. Herbal remedies are chemical cocktails, thus open another avenue for effective antiviral therapeutics development. In fact, India is a large country, which is densely populated, but the overall severity of COVID-19 per million populations is lesser than any other country of the world. One of the major reasons for the aforesaid difference is the use of herbal remedies by the Government of India as a preventive measure for COVID-19. Therefore, the present review focuses on the epidemiology and molecular pathogenesis of COVID-19 and explores algal metabolites for their antiviral properties.
Collapse
Affiliation(s)
- Rimjhim Sangtani
- Discipline of Biosciences and Biomedical EngineeringIndian Institute of TechnologyIndoreIndia
| | - Atreyee Ghosh
- Discipline of Biosciences and Biomedical EngineeringIndian Institute of TechnologyIndoreIndia
| | - Hem C. Jha
- Discipline of Biosciences and Biomedical EngineeringIndian Institute of TechnologyIndoreIndia
| | | | - Kiran Bala
- Discipline of Biosciences and Biomedical EngineeringIndian Institute of TechnologyIndoreIndia
| |
Collapse
|
6
|
Abstract
Influenza poses a significant burden on society and health care systems. Although antivirals are an integral tool in effective influenza management, the potential for the emergence of antiviral-resistant viruses can lead to uncertainty and hesitation among front-line prescribers and policy makers. Here, we provide an overview of influenza antiviral resistance in context, exploring the key concepts underlying its development and clinical impact. Due to the acute nature of influenza in immunocompetent patients, resistant viruses that develop during antiviral treatment of a single patient ("treatment-emergent resistance") are usually cleared in a relatively short time, with no impact on future antiviral efficacy. In addition, although available data are limited by small numbers of patients, they show that antiviral treatment still provides clinical benefit to the patient within whom resistance emerges. In contrast, the sustained community transmission of resistant variants in the absence of treatment ("acquired resistance") is of greater concern and can potentially render front-line antivirals ineffective. Importantly, however, resistant viruses are usually associated with reduced fitness such that their widespread transmission is relatively rare. Influenza antivirals are an essential part of effective influenza management due to their ability to reduce the risk of complications and death in infected patients. Although antiviral resistance should be taken seriously and requires continuous careful monitoring, it is not comparable to antibiotic resistance in bacteria, which can become permanent and widespread, with far-reaching medical consequences. The benefits of antiviral treatment far outweigh concerns of potential resistance, which in the vast majority of cases does not have a significant clinical impact.
Collapse
|
7
|
Prediction and characterization of influenza virus polymerase inhibitors through blind docking and ligand based virtual screening. J Mol Liq 2021. [DOI: 10.1016/j.molliq.2020.114784] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
8
|
Abstract
PURPOSE OF REVIEW Neuraminidase inhibitors (NAIs), including oseltamivir, zanamivir, and peramivir, is the main class of antiviral available for clinical use. As such, development of resistance toward these agents is of great clinical and public health concern. RECENT FINDINGS At present, NAI resistance remains uncommon among the circulating viruses (oseltamivir <3.5%, zanamivir <1%). Resistance risk is slightly higher in A(H1N1) than A(H3N2) and B viruses. Resistance may emerge during drug exposure, particularly among young children (<5 years), the immunocompromised, and individuals receiving prophylactic regimens. H275Y A(H1N1) variant, showing high-level oseltamivir resistance, is capable of causing outbreaks. R294K A(H7N9) variant shows reduced inhibition across NAIs. Multi-NAI resistance has been reported in the immunocompromised. SUMMARY These findings highlight the importance of continuous surveillance, and assessment of viral fitness and transmissibility of resistant virus strains. Detection can be challenging, especially in a mix of resistant and wild-type viruses. Recent advances in molecular techniques (e.g. targeted mutation PCR, iART, ddPCR, pyrosequencing, next-generation sequencing) have improved detection and our understanding of viral dynamics. Treatment options available for oseltamivir-resistant viruses are limited, and susceptibility testing of other NAIs may be required, but non-NAI antivirals (e.g. polymerase inhibitors) that are active against these resistant viruses are in late-stage clinical development.
Collapse
|
9
|
Bassetti M, Castaldo N, Carnelutti A. Neuraminidase inhibitors as a strategy for influenza treatment: pros, cons and future perspectives. Expert Opin Pharmacother 2019; 20:1711-1718. [PMID: 31169040 DOI: 10.1080/14656566.2019.1626824] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Introduction: Influenza represents a major public health threat worldwide. Implementation of good personal health and hygiene habits, together with vaccination, is the most effective tools to reduce influenza burden both in community and in healthcare setting. However, achieving adequate vaccination rates is challenging, and vaccination does not always guarantee complete protection. Neuraminidase inhibitors represent an important measure to reduce the risk of influenza-related complications among high-risk patients developing influenza infection. Areas covered: Neuraminidase inhibitors have been proven to be safe and effective in reducing influenza severity, duration of symptoms, hospitalizations, and influenza-related-mortality. Here the authors review the available data on neuraminidase inhibitors, including the mechanism of action, pharmacokinetics, efficacy, safety and current indications for their use in clinical practice. Expert opinion: Although vaccination is the most effective tool to reduce influenza-associated morbidity and mortality, neuraminidase inhibitors represent an important option for the treatment of patients with influenza infection, particularly in high-risk categories. Moreover, antivirals play an important role in influenza prevention and prophylaxis in selected settings.
Collapse
Affiliation(s)
- Matteo Bassetti
- Infectious Diseases Clinic, Department of Medicine University of Udine and Azienda Sanitaria Universitaria Integrata di Udine , Udine , Italy
| | - Nadia Castaldo
- Infectious Diseases Clinic, Department of Medicine University of Udine and Azienda Sanitaria Universitaria Integrata di Udine , Udine , Italy
| | - Alessia Carnelutti
- Infectious Diseases Clinic, Department of Medicine University of Udine and Azienda Sanitaria Universitaria Integrata di Udine , Udine , Italy
| |
Collapse
|
10
|
Huang SY, Huang CH, Chen CJ, Chen TW, Lin CY, Lin YT, Kuo SM, Huang CG, Lee LA, Chen YH, Chen MF, Kuo RL, Shih SR. Novel Role for miR-1290 in Host Species Specificity of Influenza A Virus. MOLECULAR THERAPY-NUCLEIC ACIDS 2019; 17:10-23. [PMID: 31173947 PMCID: PMC6554369 DOI: 10.1016/j.omtn.2019.04.028] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 04/29/2019] [Accepted: 04/29/2019] [Indexed: 11/30/2022]
Abstract
The role of microRNA (miRNA) in influenza A virus (IAV) host species specificity is not well understood as yet. Here, we show that a host miRNA, miR-1290, is induced through the extracellular signal-regulated kinase (ERK) pathway upon IAV infection and is associated with increased viral titers in human cells and ferret animal models. miR-1290 was observed to target and reduce expression of the host vimentin gene. Vimentin binds with the PB2 subunit of influenza A virus ribonucleoprotein (vRNP), and knockdown of vimentin expression significantly increased vRNP nuclear retention and viral polymerase activity. Interestingly, miR-1290 was not detected in either chicken cells or mouse animal models, and the 3′ UTR of the chicken vimentin gene contains no binding site for miR-1290. These findings point to a host species-specific mechanism by which IAV upregulates miR-1290 to disrupt vimentin expression and retain vRNP in the nucleus, thereby enhancing viral polymerase activity and viral replication.
Collapse
Affiliation(s)
- Sheng-Yu Huang
- Graduate Institute of Biomedical Science, Division of Biotechnology, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Chih-Heng Huang
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; The Institute of Microbiology and Immunology, National Defense Medical Center, Taipei 11490, Taiwan; The Institute of Preventive Medicine, National Defense Medical Center, Taipei 11490, Taiwan
| | - Chi-Jene Chen
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Ting-Wen Chen
- Institute of Bioinformatics and Systems Biology, National Chiao Tung University, Hsinchu 30068, Taiwan; Department of Biological Science and Technology, National Chiao Tung University, Hsinchu, 30068, Taiwan
| | - Chun-Yuan Lin
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; Department of Computer Science and Information Engineering, College of Engineering, Chang Gung University, Taoyuan 33302, Taiwan
| | - Yueh-Te Lin
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; Department of Computer Science and Information Engineering, College of Engineering, Chang Gung University, Taoyuan 33302, Taiwan; Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Shu-Ming Kuo
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Chung-Guei Huang
- Graduate Institute of Biomedical Science, Division of Biotechnology, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; Department of Laboratory Medicine, Linkou Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan
| | - Li-Ang Lee
- Department of Otorhinolaryngology-Head and Neck Surgery, Linkou Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan; Faculty of Medicine, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Yi-Hsiang Chen
- Graduate Institute of Biomedical Science, Division of Biotechnology, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Mei-Feng Chen
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Rei-Lin Kuo
- Graduate Institute of Biomedical Science, Division of Biotechnology, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; Department of Pediatrics, Linkou Chang Gung Memorial Hospital, Taoyuan 33302, Taiwan
| | - Shin-Ru Shih
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; Department of Laboratory Medicine, Linkou Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan; Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 33303, Taiwan; Research Center for Food and Cosmetic Safety, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 33303, Taiwan; Graduate Institute of Health Industry Technology, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 33303, Taiwan.
| |
Collapse
|
11
|
Gubareva LV, Besselaar TG, Daniels RS, Fry A, Gregory V, Huang W, Hurt AC, Jorquera PA, Lackenby A, Leang SK, Lo J, Pereyaslov D, Rebelo-de-Andrade H, Siqueira MM, Takashita E, Odagiri T, Wang D, Zhang W, Meijer A. Global update on the susceptibility of human influenza viruses to neuraminidase inhibitors, 2015-2016. Antiviral Res 2017; 146:12-20. [PMID: 28802866 PMCID: PMC5667636 DOI: 10.1016/j.antiviral.2017.08.004] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 07/25/2017] [Accepted: 08/08/2017] [Indexed: 01/26/2023]
Abstract
Four World Health Organization (WHO) Collaborating Centres for Reference and Research on Influenza and one WHO Collaborating Centre for the Surveillance, Epidemiology and Control of Influenza (WHO CCs) assessed antiviral susceptibility of 14,330 influenza A and B viruses collected by WHO-recognized National Influenza Centres (NICs) between May 2015 and May 2016. Neuraminidase (NA) inhibition assay was used to determine 50% inhibitory concentration (IC50) data for NA inhibitors (NAIs) oseltamivir, zanamivir, peramivir and laninamivir. Furthermore, NA sequences from 13,484 influenza viruses were retrieved from public sequence databases and screened for amino acid substitutions (AAS) associated with reduced inhibition (RI) or highly reduced inhibition (HRI) by NAIs. Of the viruses tested by WHO CCs 93% were from three WHO regions: Western Pacific, the Americas and Europe. Approximately 0.8% (n = 113) exhibited either RI or HRI by at least one of four NAIs. As in previous seasons, the most common NA AAS was H275Y in A(H1N1)pdm09 viruses, which confers HRI by oseltamivir and peramivir. Two A(H1N1)pdm09 viruses carried a rare NA AAS, S247R, shown in this study to confer RI/HRI by the four NAIs. The overall frequency of A(H1N1)pdm09 viruses containing NA AAS associated with RI/HRI was approximately 1.8% (125/6915), which is slightly higher than in the previous 2014-15 season (0.5%). Three B/Victoria-lineage viruses contained a new AAS, NA H134N, which conferred HRI by zanamivir and laninamivir, and borderline HRI by peramivir. A single B/Victoria-lineage virus harboured NA G104E, which was associated with HRI by all four NAIs. The overall frequency of RI/HRI phenotype among type B viruses was approximately 0.6% (43/7677), which is lower than that in the previous season. Overall, the vast majority (>99%) of the viruses tested by WHO CCs were susceptible to all four NAIs, showing normal inhibition (NI). Hence, NAIs remain the recommended antivirals for treatment of influenza virus infections. Nevertheless, our data indicate that it is prudent to continue drug susceptibility monitoring using both NAI assay and sequence analysis. A total of 14,330 influenza viruses were collected worldwide, May 2015–May 2016. Approximately 0.8% showed reduced inhibition by at least one NA inhibitor. The frequency of viruses with reduced inhibition was slightly higher than in 2014–15 (0.5%). NA inhibitors remain an appropriate choice for influenza treatment. Global surveillance of influenza antiviral susceptibility should be continued.
Collapse
Affiliation(s)
- Larisa V Gubareva
- WHO Collaborating Center for Surveillance, Epidemiology and Control of Influenza, Centers for Disease Control and Prevention (CDC), 1600 Clifton RD NE, MS-G16, Atlanta, GA, 30329, United States.
| | - Terry G Besselaar
- Global Influenza Programme, World Health Organization, Avenue Appia 20, 1211 Geneva 27, Switzerland
| | - Rod S Daniels
- The Francis Crick Institute, Worldwide Influenza Centre (WIC), WHO Collaborating Centre for Reference and Research on Influenza, 1 Midland Road, London, NW1 1AT, United Kingdom
| | - Alicia Fry
- WHO Collaborating Center for Surveillance, Epidemiology and Control of Influenza, Centers for Disease Control and Prevention (CDC), 1600 Clifton RD NE, MS-G16, Atlanta, GA, 30329, United States
| | - Vicki Gregory
- The Francis Crick Institute, Worldwide Influenza Centre (WIC), WHO Collaborating Centre for Reference and Research on Influenza, 1 Midland Road, London, NW1 1AT, United Kingdom
| | - Weijuan Huang
- WHO Collaborating Centre for Reference and Research on Influenza, National Institute for Viral Disease Control and Prevention, Collaboration Innovation Centre for Diagnosis and Treatment of Infectious Diseases, China CDC, Beijing, China
| | - Aeron C Hurt
- WHO Collaborating Centre for Reference and Research on Influenza, At the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, 3000, Australia; Department of Microbiology and Immunology, University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Patricia A Jorquera
- WHO Collaborating Center for Surveillance, Epidemiology and Control of Influenza, Centers for Disease Control and Prevention (CDC), 1600 Clifton RD NE, MS-G16, Atlanta, GA, 30329, United States
| | - Angie Lackenby
- National Infection Service, Public Health England, London, NW9 5HT, United Kingdom
| | - Sook-Kwan Leang
- WHO Collaborating Centre for Reference and Research on Influenza, At the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, 3000, Australia
| | - Janice Lo
- Public Health Laboratory Centre, 382 Nam Cheong Street, Hong Kong, China
| | - Dmitriy Pereyaslov
- Division of Health Emergencies and Communicable Diseases, World Health Organization Regional Office for Europe, UN City, Marmorvej 51, DK-2100, Copenhagen, Denmark
| | - Helena Rebelo-de-Andrade
- Influenza Pathogenesis and Antiviral Resistance Laboratory, National Institute of Health, Av. Padre Cruz, 1649-016, Lisboa, Portugal; Faculdade de Farmácia, Universidade de Lisboa, Av. Prof Gama Pinto, 1649-016, Lisboa, Portugal
| | - Marilda M Siqueira
- National Influenza Center, Laboratorio de Virus Respiratorios, Oswaldo Cruz Institute/FIOCRUZ, Rio de Janeiro, Brazil
| | - Emi Takashita
- WHO Collaborating Centre for Reference and Research on Influenza, National Institute of Infectious Diseases, Gakuen 4-7-1, Musashimurayama, Tokyo, 208-0011, Japan
| | - Takato Odagiri
- WHO Collaborating Centre for Reference and Research on Influenza, National Institute of Infectious Diseases, Gakuen 4-7-1, Musashimurayama, Tokyo, 208-0011, Japan
| | - Dayan Wang
- WHO Collaborating Centre for Reference and Research on Influenza, National Institute for Viral Disease Control and Prevention, Collaboration Innovation Centre for Diagnosis and Treatment of Infectious Diseases, China CDC, Beijing, China
| | - Wenqing Zhang
- Global Influenza Programme, World Health Organization, Avenue Appia 20, 1211 Geneva 27, Switzerland
| | - Adam Meijer
- National Institute for Public Health and the Environment, PO Box 1, 3720 BA, Bilthoven, The Netherlands
| |
Collapse
|
12
|
Mc Mahon A, Martin-Loeches I. The pharmacological management of severe influenza infection - 'existing and emerging therapies'. Expert Rev Clin Pharmacol 2016; 10:81-95. [PMID: 27797595 DOI: 10.1080/17512433.2017.1255550] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION Over the last century several influenza outbreaks have traversed the globe, most recently the influenza A(H1N1) 2009 pandemic. On each occasion, a highly contagious, virulent pathogen has emerged, leading to significant morbidity and mortality amongst those affected. Areas covered: Early antiviral therapy and supportive care is the mainstay of treatment. Treatment should be started as soon as possible and not delayed for the results of diagnostic testing. Whilst oseltamivir is still the first choice, in case of treatment failure, oseltamivir resistance should be considered, particularly in immunosuppressed patients. Here we review the antivirals currently used for management of influenza and explore a number of investigational agents that may emerge as effective antivirals including parenteral agents, combination antiviral therapy and novel agents in order to adequately target influenza virulence. Expert Commentary: New tools for rapid diagnosis and susceptible strains will help if a patient is not improving because of a resistant strain or an inadequate immune response. Further randomized control trials will be conducted to investigate the use of new antivirals and co-adjuvant therapies that will help to elucidate the process of immune modulation, particularly in immunocompetent patients.
Collapse
Affiliation(s)
- Aisling Mc Mahon
- a Multidisciplinary Intensive Care Research Organization (MICRO) , St James's University Hospital , Dublin , Ireland
| | - Ignacio Martin-Loeches
- a Multidisciplinary Intensive Care Research Organization (MICRO) , St James's University Hospital , Dublin , Ireland.,b Department of Clinical Medicine , Trinity College, Welcome Trust-HRB Clinical Research Facility, St Jame's Hospital , Dublin , Ireland
| |
Collapse
|
13
|
Yuan S, Chu H, Ye J, Singh K, Ye Z, Zhao H, Kao RYT, Chow BKC, Zhou J, Zheng BJ. Identification of a novel small-molecule compound targeting the influenza A virus polymerase PB1-PB2 interface. Antiviral Res 2016; 137:58-66. [PMID: 27840201 PMCID: PMC7113721 DOI: 10.1016/j.antiviral.2016.11.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Revised: 11/02/2016] [Accepted: 11/07/2016] [Indexed: 01/06/2023]
Abstract
The PB1 C-terminal domain and PB2 N-terminal domain interaction of the influenza A polymerase, which modulates the assembly of PB1 and PB2 subunits, may serve as a valuable target for the development of novel anti-influenza therapeutics. In this study, we performed a systematic screening of a chemical library, followed by the antiviral evaluation of primary hits and their analogues. Eventually, a novel small-molecule compound PP7 that abrogated the PB1-PB2 association and impaired viral polymerase activity was identified. PP7 exhibited antiviral activities against influenza virus subtypes A (H1N1)pdm09, A(H7N9) and A(H9N2) in cell cultures and partially protected mice against lethal challenge of mouse-adapted influenza A (H1N1)pdm09 virus. Surprisingly, a panel of other subtypes of influenza virus, including A(H5N1) and A(H7N7), showed various degrees of resistance to the compound. Biochemical studies revealed a similar pattern of resistance on the impairment of polymerase activity. Molecular docking analyses suggested a PP7-binding site that appeared to be completely conserved among the subtypes of the virus mentioned above. Thus, we propose that alternative/additional binding site (s) may exist for the regulation of PB1-PB2 subunits assembly of influenza A virus. A novel small-molecule compound was identified to provide anti-influenza effect in vitro and in vivo. An RT-qPCR based assay was modified to evaluate the polymerase activity of various subtypes of influenza viruses. The PB1-PB2 assembly strategies of the trimeric polymerase complex might be stain/subtype specific.
Collapse
Affiliation(s)
- Shuofeng Yuan
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Hin Chu
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Jiahui Ye
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Kailash Singh
- School of Biological Sciences, Faculty of Science, The University of Hong Kong, Hong Kong, China
| | - Ziwei Ye
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Hanjun Zhao
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Richard Y T Kao
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Billy K C Chow
- School of Biological Sciences, Faculty of Science, The University of Hong Kong, Hong Kong, China
| | - Jie Zhou
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
| | - Bo-Jian Zheng
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
14
|
Li F, Ma C, Hu Y, Wang Y, Wang J. Discovery of Potent Antivirals against Amantadine-Resistant Influenza A Viruses by Targeting the M2-S31N Proton Channel. ACS Infect Dis 2016; 2:726-733. [PMID: 27657178 DOI: 10.1021/acsinfecdis.6b00130] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Despite the existence of flu vaccines and small-molecule antiviral drugs, influenza virus infection remains a public health concern that warrants immediate attention. As resistance to the only orally bioavailable drug, oseltamivir, has been continuously reported, there is a clear need to develop the next-generation of anti-influenza drugs. We chose the influenza A virus M2-S31N mutant proton channel as the drug target to address this need as it is one of the most conserved viral proteins and persist in >95% of currently circulating influenza A viruses. In this study, we report the development of a late-stage diversification strategy for the expeditious synthesis of M2-S31N inhibitors. The channel blockage and antiviral activity of the synthesized compounds were tested in two-electrode voltage clamp assays and antiviral assays, respectively. Several M2-S31N inhibitors were identified to have potent M2-S31N channel blockage and micromolar antiviral efficacy against several M2-S31N-containing influenza A viruses.
Collapse
Affiliation(s)
- Fang Li
- Department of Pharmacology
and Toxicology, College of Pharmacy, and BIO5 Institute, The University of Arizona, Tucson, Arizona 85721, United States
| | - Chunlong Ma
- Department of Pharmacology
and Toxicology, College of Pharmacy, and BIO5 Institute, The University of Arizona, Tucson, Arizona 85721, United States
| | - Yanmei Hu
- Department of Pharmacology
and Toxicology, College of Pharmacy, and BIO5 Institute, The University of Arizona, Tucson, Arizona 85721, United States
| | - Yuanxiang Wang
- Department of Pharmacology
and Toxicology, College of Pharmacy, and BIO5 Institute, The University of Arizona, Tucson, Arizona 85721, United States
| | - Jun Wang
- Department of Pharmacology
and Toxicology, College of Pharmacy, and BIO5 Institute, The University of Arizona, Tucson, Arizona 85721, United States
| |
Collapse
|
15
|
Ma C, Zhang J, Wang J. Pharmacological Characterization of the Spectrum of Antiviral Activity and Genetic Barrier to Drug Resistance of M2-S31N Channel Blockers. Mol Pharmacol 2016; 90:188-98. [PMID: 27385729 PMCID: PMC4998667 DOI: 10.1124/mol.116.105346] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 06/30/2016] [Indexed: 12/11/2022] Open
Abstract
Adamantanes (amantadine and rimantadine) are one of the two classes of Food and Drug Administration-approved antiviral drugs used for the prevention and treatment of influenza A virus infections. They inhibit viral replication by blocking the wild-type (WT) M2 proton channel, thus preventing viral uncoating. However, their use was discontinued due to widespread drug resistance. Among a handful of drug-resistant mutants, M2-S31N is the predominant mutation and persists in more than 95% of currently circulating influenza A strains. We recently designed two classes of M2-S31N inhibitors, S31N-specific inhibitors and S31N/WT dual inhibitors, which are represented by N-[(5-cyclopropyl-1,2-oxazol-3-yl)methyl]adamantan-1-amine (WJ379) and N-[(5-bromothiophen-2-yl)methyl]adamantan-1-amine (BC035), respectively. However, their antiviral activities against currently circulating influenza A viruses and their genetic barrier to drug resistance are unknown. In this report, we evaluated the therapeutic potential of these two classes of M2-S31N inhibitors (WJ379 and BC035) by profiling their antiviral efficacy against multidrug-resistant influenza A viruses, in vitro drug resistance barrier, and synergistic effect with oseltamivir. We found that M2-S31N inhibitors were active against several influenza A viruses that are resistant to one or both classes of Food and Drug Administration-approved anti-influenza drugs. In addition, M2-S31N inhibitors display a higher in vitro genetic barrier to drug resistance than amantadine. The antiviral effect of WJ379 was also synergistic with oseltamivir carboxylate. Overall, these results reaffirm that M2-S31N inhibitors are promising antiviral drug candidates that warrant further development.
Collapse
Affiliation(s)
- Chunlong Ma
- Department of Pharmacology and Toxicology, College of Pharmacy, and BIO5 Institute, University of Arizona, Tucson, Arizona
| | - Jiantao Zhang
- Department of Pharmacology and Toxicology, College of Pharmacy, and BIO5 Institute, University of Arizona, Tucson, Arizona
| | - Jun Wang
- Department of Pharmacology and Toxicology, College of Pharmacy, and BIO5 Institute, University of Arizona, Tucson, Arizona
| |
Collapse
|
16
|
Sriwilaijaroen N, Magesh S, Imamura A, Ando H, Ishida H, Sakai M, Ishitsubo E, Hori T, Moriya S, Ishikawa T, Kuwata K, Odagiri T, Tashiro M, Hiramatsu H, Tsukamoto K, Miyagi T, Tokiwa H, Kiso M, Suzuki Y. A Novel Potent and Highly Specific Inhibitor against Influenza Viral N1-N9 Neuraminidases: Insight into Neuraminidase-Inhibitor Interactions. J Med Chem 2016; 59:4563-77. [PMID: 27095056 DOI: 10.1021/acs.jmedchem.5b01863] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
People throughout the world continue to be at risk for death from influenza A virus, which is always creating a new variant. Here we present a new effective and specific anti-influenza viral neuraminidase (viNA) inhibitor, 9-cyclopropylcarbonylamino-4-guanidino-Neu5Ac2en (cPro-GUN). Like zanamivir, it is highly effective against N1-N9 avian and N1-N2 human viNAs, including H274Y oseltamivir-resistant N1 viNA, due to its C-6 portion still being anchored in the active site, different from the disruption of oseltamivir's C-6 anchoring by H274Y mutation. Unlike zanamivir, no sialidase inhibitory activity has been observed for cPro-GUN against huNeu1-huNeu4 enzymes. Broad efficacy of cPro-GUN against avian and human influenza viruses in cell cultures comparable to its sialidase inhibitory activities makes cPro-GUN ideal for further development for safe therapeutic or prophylactic use against both seasonal and pandemic influenza.
Collapse
Affiliation(s)
- Nongluk Sriwilaijaroen
- Department of Preclinical Sciences, Faculty of Medicine, Thammasat University , Pathumthani 12120, Thailand.,Health Science Hills, College of Life and Health Sciences, Chubu University , Aichi 487-8501, Japan
| | | | | | - Hiromune Ando
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University , Kyoto 606-8501, Japan
| | | | | | | | | | - Setsuko Moriya
- Division of Cancer Glycosylation Research, Institute of Molecular Biomembrane and Glycobiology, Tohoku Pharmaceutical University , Sendai 981-8558, Japan
| | | | | | - Takato Odagiri
- Influenza Virus Research Center, National Institute of Infectious Diseases , Tokyo 208-0011, Japan
| | - Masato Tashiro
- Influenza Virus Research Center, National Institute of Infectious Diseases , Tokyo 208-0011, Japan
| | - Hiroaki Hiramatsu
- Health Science Hills, College of Life and Health Sciences, Chubu University , Aichi 487-8501, Japan
| | - Kenji Tsukamoto
- Research Team for Zoonotic Diseases, National Institute of Animal Health , Ibaraki 305-0856, Japan
| | - Taeko Miyagi
- Division of Cancer Glycosylation Research, Institute of Molecular Biomembrane and Glycobiology, Tohoku Pharmaceutical University , Sendai 981-8558, Japan
| | | | - Makoto Kiso
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University , Kyoto 606-8501, Japan
| | - Yasuo Suzuki
- Health Science Hills, College of Life and Health Sciences, Chubu University , Aichi 487-8501, Japan
| |
Collapse
|
17
|
Clinical Implications of Antiviral Resistance in Influenza. Viruses 2015; 7:4929-44. [PMID: 26389935 PMCID: PMC4584294 DOI: 10.3390/v7092850] [Citation(s) in RCA: 139] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Revised: 07/28/2015] [Accepted: 07/28/2015] [Indexed: 01/30/2023] Open
Abstract
Influenza is a major cause of severe respiratory infections leading to excessive hospitalizations and deaths globally; annual epidemics, pandemics, and sporadic/endemic avian virus infections occur as a result of rapid, continuous evolution of influenza viruses. Emergence of antiviral resistance is of great clinical and public health concern. Currently available antiviral treatments include four neuraminidase inhibitors (oseltamivir, zanamivir, peramivir, laninamivir), M2-inibitors (amantadine, rimantadine), and a polymerase inhibitor (favipiravir). In this review, we focus on resistance issues related to the use of neuraminidase inhibitors (NAIs). Data on primary resistance, as well as secondary resistance related to NAI exposure will be presented. Their clinical implications, detection, and novel therapeutic options undergoing clinical trials are discussed.
Collapse
|
18
|
Petrie SM, Butler J, Barr IG, McVernon J, Hurt AC, McCaw JM. Quantifying relative within-host replication fitness in influenza virus competition experiments. J Theor Biol 2015; 382:259-71. [PMID: 26188087 DOI: 10.1016/j.jtbi.2015.07.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Revised: 07/02/2015] [Accepted: 07/06/2015] [Indexed: 01/26/2023]
Abstract
Through accumulation of genetic mutations in the neuraminidase gene, the influenza virus can become resistant to antiviral drugs such as oseltamivir. Quantifying the fitness of emergent drug-resistant influenza viruses, relative to contemporary circulating viruses, provides valuable information to complement existing efforts in the surveillance of drug-resistance. We have previously developed a co-infection based method for the assessment of the relative in vivo fitness of two competing viruses. We have also introduced a model of within-host co-infection dynamics that enables relative within-host fitness to be quantified in these competitive-mixtures experiments. The model assumed that fitness differences between co-infecting strains were mediated by strain-dependent viral production rates from infected epithelial cells. Here we extend the model to enable a more complete exploration of biological processes that may differ between virus pairs and hence generate fitness differences. We use the extended model to re-analyse data from competitive-mixtures experiments that investigated the fitness of oseltamivir-resistant (OR) H1N1 pandemic 2009 ("H1N1pdm09") viruses that emerged during a community outbreak in Australia in 2011. Results are consistent with those of our previous analysis, suggesting that the within-host replication fitness of these OR viruses is not compromised relative to that of related oseltamivir-susceptible (OS) strains, and that potentially permissive mutations in the neuraminidase gene (V241I and N369K) significantly enhance the fitness of H1N1pdm09 OR viruses. These results are consistent regardless of the hypothesised biological cause of fitness difference.
Collapse
Affiliation(s)
- Stephen M Petrie
- Melbourne School of Population and Global Health, The University of Melbourne, Parkville, Victoria, Australia; Centre for Transformative Innovation, Swinburne University of Technology, Hawthorn, Victoria, Australia
| | - Jeff Butler
- WHO Collaborating Centre for Reference and Research on Influenza, Victorian Infectious Diseases Reference Laboratory at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Ian G Barr
- WHO Collaborating Centre for Reference and Research on Influenza, Victorian Infectious Diseases Reference Laboratory at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia; School of Applied Sciences, Monash University, Churchill, Victoria, Australia
| | - Jodie McVernon
- Melbourne School of Population and Global Health, The University of Melbourne, Parkville, Victoria, Australia; Murdoch Childrens Research Institute, The Royal Children׳s Hospital, Parkville, Victoria, Australia
| | - Aeron C Hurt
- WHO Collaborating Centre for Reference and Research on Influenza, Victorian Infectious Diseases Reference Laboratory at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia; School of Applied Sciences, Monash University, Churchill, Victoria, Australia
| | - James M McCaw
- Melbourne School of Population and Global Health, The University of Melbourne, Parkville, Victoria, Australia; Murdoch Childrens Research Institute, The Royal Children׳s Hospital, Parkville, Victoria, Australia; School of Mathematics and Statistics, The University of Melbourne, Parkville, Victoria, Australia.
| |
Collapse
|
19
|
Sugiyama MG, Armstrong SM, Wang C, Hwang D, Leong-Poi H, Advani A, Advani S, Zhang H, Szaszi K, Tabuchi A, Kuebler WM, Van Slyke P, Dumont DJ, Lee WL. The Tie2-agonist Vasculotide rescues mice from influenza virus infection. Sci Rep 2015; 5:11030. [PMID: 26046800 PMCID: PMC4457136 DOI: 10.1038/srep11030] [Citation(s) in RCA: 54] [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: 02/20/2015] [Accepted: 05/14/2015] [Indexed: 12/13/2022] Open
Abstract
Seasonal influenza virus infections cause hundreds of thousands of deaths annually while viral mutation raises the threat of a novel pandemic strain. Antiviral drugs exhibit limited efficacy unless administered early and may induce viral resistance. Thus, targeting the host response directly has been proposed as a novel therapeutic strategy with the added potential benefit of not eliciting viral resistance. Severe influenza virus infections are complicated by respiratory failure due to the development of lung microvascular leak and acute lung injury. We hypothesized that enhancing lung endothelial barrier integrity could improve the outcome. Here we demonstrate that the Tie2-agonist tetrameric peptide Vasculotide improves survival in murine models of severe influenza, even if administered as late as 72 hours after infection; the benefit was observed using three strains of the virus and two strains of mice. The effect required Tie2, was independent of viral replication and did not impair lung neutrophil recruitment. Administration of the drug decreased lung edema, arterial hypoxemia and lung endothelial apoptosis; importantly, Vasculotide is inexpensive to produce, is chemically stable and is unrelated to any Tie2 ligands. Thus, Vasculotide may represent a novel and practical therapy for severe infections with influenza.
Collapse
Affiliation(s)
- Michael G. Sugiyama
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON
- Department of Laboratory Medicine and Pathobiology, University of Toronto
| | - Susan M. Armstrong
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON
| | - Changsen Wang
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON
| | - David Hwang
- Department of Laboratory Medicine and Pathobiology, University of Toronto
- Department of Pathology, Toronto General Hospital, University Health Network, Toronto, ON
| | - Howard Leong-Poi
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON
| | - Andrew Advani
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON
| | - Suzanne Advani
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON
| | - Haibo Zhang
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON
| | - Katalin Szaszi
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON
| | - Arata Tabuchi
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON
| | - Wolfgang M. Kuebler
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON
| | - Paul Van Slyke
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON
| | - Dan J. Dumont
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON
| | - Warren L. Lee
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON
- Department of Laboratory Medicine and Pathobiology, University of Toronto
- Interdepartmental Division of Critical Care and Department of Medicine, University of Toronto.
| |
Collapse
|
20
|
Paradis EG, Pinilla LT, Holder BP, Abed Y, Boivin G, Beauchemin CA. Impact of the H275Y and I223V Mutations in the Neuraminidase of the 2009 Pandemic Influenza Virus In Vitro and Evaluating Experimental Reproducibility. PLoS One 2015; 10:e0126115. [PMID: 25992792 PMCID: PMC4439092 DOI: 10.1371/journal.pone.0126115] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 03/30/2015] [Indexed: 11/18/2022] Open
Abstract
The 2009 pandemic H1N1 (H1N1pdm09) influenza virus is naturally susceptible to neuraminidase (NA) inhibitors, but mutations in the NA protein can cause oseltamivir resistance. The H275Y and I223V amino acid substitutions in the NA of the H1N1pdm09 influenza strain have been separately observed in patients exhibiting oseltamivir-resistance. Here, we apply mathematical modelling techniques to compare the fitness of the wild-type H1N1pdm09 strain relative to each of these two mutants. We find that both the H275Y and I223V mutations in the H1N1pdm09 background significantly lengthen the duration of the eclipse phase (by 2.5 h and 3.6 h, respectively), consistent with these NA mutations delaying the release of viral progeny from newly infected cells. Cells infected by H1N1pdm09 virus carrying the I223V mutation display a disadvantageous, shorter infectious lifespan (17 h shorter) than those infected with the wild-type or MUT-H275Y strains. In terms of compensating traits, the H275Y mutation in the H1N1pdm09 background results in increased virus infectiousness, as we reported previously, whereas the I223V exhibits none, leaving it overall less fit than both its wild-type counterpart and the MUT-H275Y strain. Using computer simulated competition experiments, we determine that in the presence of oseltamivir at doses even below standard therapy, both the MUT-H275Y and MUT-I223V dominate their wild-type counterpart in all aspects, and the MUT-H275Y outcompetes the MUT-I223V. The H275Y mutation should therefore be more commonly observed than the I223V mutation in circulating H1N1pdm09 strains, assuming both mutations have a similar impact or no significant impact on between-host transmission. We also show that mathematical modelling offers a relatively inexpensive and reliable means to quantify inter-experimental variability and assess the reproducibility of results.
Collapse
Affiliation(s)
- Eric G. Paradis
- Department of Physics, Ryerson University, Toronto, ON, Canada
| | - Lady Tatiana Pinilla
- Infectious Disease Research Centre, CHUQ-CHUL and Laval University, Québec, QC, Canada
| | | | - Yacine Abed
- Infectious Disease Research Centre, CHUQ-CHUL and Laval University, Québec, QC, Canada
| | - Guy Boivin
- Infectious Disease Research Centre, CHUQ-CHUL and Laval University, Québec, QC, Canada
| | | |
Collapse
|
21
|
Lopes e Souza TM, Fintelman-Rodrigues N, Resende PC, Mesquita M, Gregianini TS, Bozza FA, Pecego AC, Fernandes SB, Cury ALF, Riediger IN, Siqueira MM. Oseltamivir-resistant influenza A(H1N1)pdm2009 strains found in Brazil are endowed with permissive mutations, which compensate the loss of fitness imposed by antiviral resistance. Mem Inst Oswaldo Cruz 2015; 110:101-5. [PMID: 25742269 PMCID: PMC4371223 DOI: 10.1590/0074-02760140330] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Accepted: 01/12/2015] [Indexed: 11/21/2022] Open
Abstract
The 2009 pandemic influenza A virus outbreak led to the systematic use of the neuraminidase (NA) inhibitor oseltamivir (OST). Consequently, OST-resistant strains, carrying the mutation H275Y, emerged in the years after the pandemics, with a prevalence of 1-2%. Currently, OST-resistant strains have been found in community settings, in untreated individuals. To spread in community settings, H275Y mutants must contain additional mutations, collectively called permissive mutations. We display the permissive mutations in NA of OST-resistant A(H1N1)pdm09 virus found in Brazilian community settings. The NAs from 2013 are phylogenetically distinct from those of 2012, indicating a tendency of positive selection of NAs with better fitness. Some previously predicted permissive mutations, such as V241I and N369K, found in different countries, were also detected in Brazil. Importantly, the change D344N, also predicted to compensate loss of fitness imposed by H275Y mutation, was found in Brazil, but not in other countries in 2013. Our results reinforce the notion that OST-resistant A(H1N1)pdm09 strains with compensatory mutations may arise in an independent fashion, with samples being identified in different states of Brazil and in different countries. Systematic circulation of these viral strains may jeopardise the use of the first line of anti-influenza drugs in the future.
Collapse
|
22
|
Competitive fitness of influenza B viruses with neuraminidase inhibitor-resistant substitutions in a coinfection model of the human airway epithelium. J Virol 2015; 89:4575-87. [PMID: 25673705 DOI: 10.1128/jvi.02473-14] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
UNLABELLED Influenza A and B viruses are human pathogens that are regarded to cause almost equally significant disease burdens. Neuraminidase (NA) inhibitors (NAIs) are the only class of drugs available to treat influenza A and B virus infections, so the development of NAI-resistant viruses with superior fitness is a public health concern. The fitness of NAI-resistant influenza B viruses has not been widely studied. Here we examined the replicative capacity and relative fitness in normal human bronchial epithelial (NHBE) cells of recombinant influenza B/Yamanashi/166/1998 viruses containing a single amino acid substitution in NA generated by reverse genetics (rg) that is associated with NAI resistance. The replication in NHBE cells of viruses with reduced inhibition by oseltamivir (recombinant virus with the E119A mutation generated by reverse genetics [rg-E119A], rg-D198E, rg-I222T, rg-H274Y, rg-N294S, and rg-R371K, N2 numbering) or zanamivir (rg-E119A and rg-R371K) failed to be inhibited by the presence of the respective NAI. In a fluorescence-based assay, detection of rg-E119A was easily masked by the presence of NAI-susceptible virus. We coinfected NHBE cells with NAI-susceptible and -resistant viruses and used next-generation deep sequencing to reveal the order of relative fitness compared to that of recombinant wild-type (WT) virus generated by reverse genetics (rg-WT): rg-H274Y > rg-WT > rg-I222T > rg-N294S > rg-D198E > rg-E119A ≫ rg-R371K. Based on the lack of attenuated replication of rg-E119A in NHBE cells in the presence of oseltamivir or zanamivir and the fitness advantage of rg-H274Y over rg-WT, we emphasize the importance of these substitutions in the NA glycoprotein. Human infections with influenza B viruses carrying the E119A or H274Y substitution could limit the therapeutic options for those infected; the emergence of such viruses should be closely monitored. IMPORTANCE Influenza B viruses are important human respiratory pathogens contributing to a significant portion of seasonal influenza virus infections worldwide. The development of resistance to a single class of available antivirals, the neuraminidase (NA) inhibitors (NAIs), is a public health concern. Amino acid substitutions in the NA glycoprotein of influenza B virus not only can confer antiviral resistance but also can alter viral fitness. Here we used normal human bronchial epithelial (NHBE) cells, a model of the human upper respiratory tract, to examine the replicative capacities and fitness of NAI-resistant influenza B viruses. We show that virus with an E119A NA substitution can replicate efficiently in NHBE cells in the presence of oseltamivir or zanamivir and that virus with the H274Y NA substitution has a relative fitness greater than that of the wild-type NAI-susceptible virus. This study is the first to use NHBE cells to determine the fitness of NAI-resistant influenza B viruses.
Collapse
|
23
|
Pires DEV, Blundell TL, Ascher DB. Platinum: a database of experimentally measured effects of mutations on structurally defined protein-ligand complexes. Nucleic Acids Res 2014; 43:D387-91. [PMID: 25324307 PMCID: PMC4384026 DOI: 10.1093/nar/gku966] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Drug resistance is a major challenge for the treatment of many diseases and a significant concern throughout the drug development process. The ability to understand and predict the effects of mutations on protein–ligand affinities and their roles in the emergence of resistance would significantly aid treatment and drug design strategies. In order to study and understand the impacts of missense mutations on the interaction of ligands with the proteome, we have developed Platinum (http://structure.bioc.cam.ac.uk/platinum). This manually curated, literature-derived database, comprising over 1000 mutations, associates for the first time experimental information on changes in affinity with three-dimensional structures of protein–ligand complexes. To minimize differences arising from experimental techniques and to directly compare binding affinities, Platinum considers only changes measured by the same group and with the same amino-acid sequence used for structure determination, providing a direct link between protein structure, how a ligand binds and how mutations alter the affinity of the ligand of the protein. We believe Platinum will be an invaluable resource for understanding the effects of mutations that give rise to drug resistance, a major problem emerging in pandemics including those caused by the influenza virus, in infectious diseases such as tuberculosis, in cancer and in many other life-threatening illnesses.
Collapse
Affiliation(s)
- Douglas E V Pires
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1GA, UK
| | - Tom L Blundell
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1GA, UK
| | - David B Ascher
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1GA, UK
| |
Collapse
|
24
|
Butler J, Hooper KA, Petrie S, Lee R, Maurer-Stroh S, Reh L, Guarnaccia T, Baas C, Xue L, Vitesnik S, Leang SK, McVernon J, Kelso A, Barr IG, McCaw JM, Bloom JD, Hurt AC. Estimating the fitness advantage conferred by permissive neuraminidase mutations in recent oseltamivir-resistant A(H1N1)pdm09 influenza viruses. PLoS Pathog 2014; 10:e1004065. [PMID: 24699865 PMCID: PMC3974874 DOI: 10.1371/journal.ppat.1004065] [Citation(s) in RCA: 100] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Accepted: 02/27/2014] [Indexed: 01/06/2023] Open
Abstract
Oseltamivir is relied upon worldwide as the drug of choice for the treatment of human influenza infection. Surveillance for oseltamivir resistance is routinely performed to ensure the ongoing efficacy of oseltamivir against circulating viruses. Since the emergence of the pandemic 2009 A(H1N1) influenza virus (A(H1N1)pdm09), the proportion of A(H1N1)pdm09 viruses that are oseltamivir resistant (OR) has generally been low. However, a cluster of OR A(H1N1)pdm09 viruses, encoding the neuraminidase (NA) H275Y oseltamivir resistance mutation, was detected in Australia in 2011 amongst community patients that had not been treated with oseltamivir. Here we combine a competitive mixtures ferret model of influenza infection with a mathematical model to assess the fitness, both within and between hosts, of recent OR A(H1N1)pdm09 viruses. In conjunction with data from in vitro analyses of NA expression and activity we demonstrate that contemporary A(H1N1)pdm09 viruses are now more capable of acquiring H275Y without compromising their fitness, than earlier A(H1N1)pdm09 viruses circulating in 2009. Furthermore, using reverse engineered viruses we demonstrate that a pair of permissive secondary NA mutations, V241I and N369K, confers robust fitness on recent H275Y A(H1N1)pdm09 viruses, which correlated with enhanced surface expression and enzymatic activity of the A(H1N1)pdm09 NA protein. These permissive mutations first emerged in 2010 and are now present in almost all circulating A(H1N1)pdm09 viruses. Our findings suggest that recent A(H1N1)pdm09 viruses are now more permissive to the acquisition of H275Y than earlier A(H1N1)pdm09 viruses, increasing the risk that OR A(H1N1)pdm09 will emerge and spread worldwide.
Collapse
Affiliation(s)
- Jeff Butler
- World Health Organization Collaborating Centre for Reference and Research on Influenza, North Melbourne, Australia
| | - Kathryn A. Hooper
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- Molecular and Cellular Biology Program, University of Washington, Seattle, Washington, United States of America
| | - Stephen Petrie
- Melbourne School of Population and Global Health, The University of Melbourne, Parkville, Australia
| | - Raphael Lee
- Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Sebastian Maurer-Stroh
- Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), Singapore
- National Public Health Laboratory, Communicable Diseases Division Ministry of Health, Singapore
- School of Biological Sciences (SBS), Nanyang Technological University (NTU), Singapore
| | - Lucia Reh
- World Health Organization Collaborating Centre for Reference and Research on Influenza, North Melbourne, Australia
| | - Teagan Guarnaccia
- World Health Organization Collaborating Centre for Reference and Research on Influenza, North Melbourne, Australia
| | - Chantal Baas
- World Health Organization Collaborating Centre for Reference and Research on Influenza, North Melbourne, Australia
- Monash University, School of Applied Sciences, Churchill, Victoria, Australia
| | - Lumin Xue
- World Health Organization Collaborating Centre for Reference and Research on Influenza, North Melbourne, Australia
| | - Sophie Vitesnik
- World Health Organization Collaborating Centre for Reference and Research on Influenza, North Melbourne, Australia
| | - Sook-Kwan Leang
- World Health Organization Collaborating Centre for Reference and Research on Influenza, North Melbourne, Australia
| | - Jodie McVernon
- Melbourne School of Population and Global Health, The University of Melbourne, Parkville, Australia
- Murdoch Childrens Research Institute, The Royal Children's Hospital, Melbourne, Victoria, Australia
| | - Anne Kelso
- World Health Organization Collaborating Centre for Reference and Research on Influenza, North Melbourne, Australia
| | - Ian G. Barr
- World Health Organization Collaborating Centre for Reference and Research on Influenza, North Melbourne, Australia
- Monash University, School of Applied Sciences, Churchill, Victoria, Australia
| | - James M. McCaw
- Melbourne School of Population and Global Health, The University of Melbourne, Parkville, Australia
- Murdoch Childrens Research Institute, The Royal Children's Hospital, Melbourne, Victoria, Australia
| | - Jesse D. Bloom
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Aeron C. Hurt
- World Health Organization Collaborating Centre for Reference and Research on Influenza, North Melbourne, Australia
- Monash University, School of Applied Sciences, Churchill, Victoria, Australia
| |
Collapse
|
25
|
Abstract
The threat of a virulent, highly transmissible pandemic virus has motivated an escalating research effort to identify the transmissible genotypes of animal viruses that cross over into the human population (animal–human transmission) and sustain human–human transmission. In addition to the pursuit of the viral genotype, a greater understanding of the host-virus phenotype of infectiousness, transmissibility and susceptibility will be required. This review examines experimental animal transmission of influenza for insights into human influenza transmission. Transmission is viewed as sequential steps that the virus must pass critical thresholds to achieve transmission and ultimately survival in the human host. In particular, a quantitative understanding in animal models of viral replication efficiency, airway viral load, exhaled viral aerosol load, environmental virus survival and host susceptibility will likely yield important insights. Computational modeling will enhance animal model data, as well as guide the use of pandemic mitigation strategies.
Collapse
Affiliation(s)
- Frederick Koster
- Department of Computer Science, University of New Mexico, Albuquerque, NM, USA and The Lovelace Respiratory Research Institute, Albuquerque, NM, USA
| |
Collapse
|
26
|
van der Vries E, Stittelaar KJ, van Amerongen G, Veldhuis Kroeze EJB, de Waal L, Fraaij PLA, Meesters RJ, Luider TM, van der Nagel B, Koch B, Vulto AG, Schutten M, Osterhaus ADME. Prolonged influenza virus shedding and emergence of antiviral resistance in immunocompromised patients and ferrets. PLoS Pathog 2013; 9:e1003343. [PMID: 23717200 PMCID: PMC3662664 DOI: 10.1371/journal.ppat.1003343] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Accepted: 03/21/2013] [Indexed: 01/17/2023] Open
Abstract
Immunocompromised individuals tend to suffer from influenza longer with more serious complications than otherwise healthy patients. Little is known about the impact of prolonged infection and the efficacy of antiviral therapy in these patients. Among all 189 influenza A virus infected immunocompromised patients admitted to ErasmusMC, 71 were hospitalized, since the start of the 2009 H1N1 pandemic. We identified 11 (15%) cases with prolonged 2009 pandemic virus replication (longer than 14 days), despite antiviral therapy. In 5 out of these 11 (45%) cases oseltamivir resistant H275Y viruses emerged. Given the inherent difficulties in studying antiviral efficacy in immunocompromised patients, we have infected immunocompromised ferrets with either wild-type, or oseltamivir-resistant (H275Y) 2009 pandemic virus. All ferrets showed prolonged virus shedding. In wild-type virus infected animals treated with oseltamivir, H275Y resistant variants emerged within a week after infection. Unexpectedly, oseltamivir therapy still proved to be partially protective in animals infected with resistant virus. Immunocompromised ferrets offer an attractive alternative to study efficacy of novel antiviral therapies. Immunocompromised patients, such as transplant recipients on immune suppressive therapy, are a substantial and gradually expanding patient group. Upon influenza virus infection, these patients clear the virus less efficiently and are more likely to develop severe pneumonia than immunocompetent individuals. Existing antiviral strategies are far from satisfactory for this patient group, as they show limited effectiveness with frequent emergence of antiviral resistance. For ethical and practical reasons antiviral efficacy studies are hard to conduct in these patients. Therefore, we developed an immunocompromised ferret, mimicking an immune suppressive regimen used for solid organ transplant recipients. Upon infection with 2009 pandemic influenza A/H1N1 virus these animals, like immunocompromised patients, develop severe respiratory disease with prolonged virus excretion. Interestingly, all immunocompromised ferrets on oseltamivir therapy excreted oseltamivir resistant viruses (H275Y) within one week after start of treatment. Furthermore, high dose oseltamivir therapy still proved to be partially effective against these oseltamivir resistant viruses. These immunocompromised ferrets provide a useful tool in the development of novel antiviral approaches for immunocompromised patients suffering from influenza.
Collapse
Affiliation(s)
| | | | | | | | - Leon de Waal
- Viroclinics Biosciences B.V., Rotterdam, The Netherlands
| | - Pieter L. A. Fraaij
- Department of Virology, ErasmusMC, Rotterdam, The Netherlands
- Department of Paediatrics, ErasmusMC-Sophia, Rotterdam, The Netherlands
| | | | - Theo M. Luider
- Department of Neurology, ErasmusMC, Rotterdam, The Netherlands
| | | | - Birgit Koch
- Department of Hospital Pharmacy, ErasmusMC, Rotterdam, The Netherlands
| | - Arnold G. Vulto
- Department of Hospital Pharmacy, ErasmusMC, Rotterdam, The Netherlands
| | - Martin Schutten
- Department of Virology, ErasmusMC, Rotterdam, The Netherlands
| | - Albert D. M. E. Osterhaus
- Department of Virology, ErasmusMC, Rotterdam, The Netherlands
- Viroclinics Biosciences B.V., Rotterdam, The Netherlands
- * E-mail:
| |
Collapse
|
27
|
Affiliation(s)
- Christopher B Brooke
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland, USA
| | | |
Collapse
|
28
|
Drivers and consequences of influenza antiviral resistant-strain emergence in a capacity-constrained pandemic response. Epidemics 2012; 4:219-26. [PMID: 23351374 DOI: 10.1016/j.epidem.2012.12.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2012] [Revised: 11/06/2012] [Accepted: 12/17/2012] [Indexed: 01/19/2023] Open
Abstract
Antiviral agents remain a key component of most pandemic influenza preparedness plans, but there is considerable uncertainty regarding their optimal use. In particular, concerns exist regarding the likelihood of wide-scale distribution to select for drug-resistant variants. We used a model that considers the influence of logistical constraints on diagnosis and drug delivery to consider achievable 'reach' of alternative antiviral intervention strategies targeted at cases of varying severity, with or without pre-exposure prophylaxis of contacts. To identify key drivers of epidemic mitigation and resistance emergence, we used Latin hypercube sampling to explore plausible ranges of parameters describing characteristics of wild type and resistant viruses, along with intervention efficacy, target coverage and distribution capacity. Within our model framework, 'real world' constraints substantially reduced achievable drug coverage below stated targets as the epidemic progressed. In consequence, predictions of both intervention impact and selection for resistance were more modest than earlier work that did not consider such limitations. Definitive containment of transmission was unlikely but, where observed, achieved through early liberal post-exposure prophylaxis of known contacts of treated cases. Predictors of resistant strain dominance were high intrinsic fitness relative to the wild type virus, and early emergence in the course of the epidemic into a largely susceptible population, even when drug use was restricted to severe case treatment. Our work demonstrates the importance of consideration of 'real world' constraints in scenario analysis modeling, and highlights the utility of models to guide surveillance activities in preparedness and response.
Collapse
|