1
|
Gaisina I, Li P, Du R, Cui Q, Dong M, Zhang C, Manicassamy B, Caffrey M, Moore T, Cooper L, Rong L. An orally active entry inhibitor of influenza A viruses protects mice and synergizes with oseltamivir and baloxavir marboxil. SCIENCE ADVANCES 2024; 10:eadk9004. [PMID: 38394202 PMCID: PMC10889430 DOI: 10.1126/sciadv.adk9004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Accepted: 01/22/2024] [Indexed: 02/25/2024]
Abstract
Seasonal or pandemic illness caused by influenza A viruses (IAVs) is a major public health concern due to the high morbidity and notable mortality. Although there are several approved drugs targeting different mechanisms, the emergence of drug resistance calls for new drug candidates that can be used alone or in combinations. Small-molecule IAV entry inhibitor, ING-1466, binds to hemagglutinin (HA) and blocks HA-mediated viral infection. Here, we show that this inhibitor demonstrates preventive and therapeutic effects in a mouse model of IAV with substantial improvement in the survival rate. When administered orally it elicits a therapeutic effect in mice, even after the well-established infection. Moreover, the combination of ING-1466 with oseltamivir phosphate or baloxavir marboxil enhances the therapeutic effect in a synergistic manner. Overall, ING-1466 has excellent oral bioavailability and in vitro absorption, distribution, metabolism, excretion, and toxicity profile, suggesting that it can be developed for monotherapy or combination therapy for the treatment of IAV infections.
Collapse
Affiliation(s)
- Irina Gaisina
- Department of Pharmaceutical Sciences, College of Pharmacy and UICentre, University of Illinois at Chicago, Chicago, IL 60612, USA
- Chicago BioSolutions Inc., Chicago, IL 60612, USA
| | - Ping Li
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, China
| | - Ruikun Du
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, China
| | - Qinghua Cui
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, China
| | - Meiyue Dong
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, China
| | - Chengcheng Zhang
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, China
| | - Balaji Manicassamy
- Department of Microbiology and Immunology, College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Michael Caffrey
- Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Terry Moore
- Department of Pharmaceutical Sciences, College of Pharmacy and UICentre, University of Illinois at Chicago, Chicago, IL 60612, USA
- University of Illinois Cancer Center, University of Illinois at Chicago, Chicago, IL 26 60612, USA
| | - Laura Cooper
- Department of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Lijun Rong
- Chicago BioSolutions Inc., Chicago, IL 60612, USA
- Department of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| |
Collapse
|
2
|
Achterbergh RCA, McGovern I, Haag M. Co-Administration of Influenza and COVID-19 Vaccines: Policy Review and Vaccination Coverage Trends in the European Union, UK, US, and Canada between 2019 and 2023. Vaccines (Basel) 2024; 12:216. [PMID: 38400199 PMCID: PMC10891656 DOI: 10.3390/vaccines12020216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 02/02/2024] [Accepted: 02/11/2024] [Indexed: 02/25/2024] Open
Abstract
Recommending co-administration of influenza and COVID-19 vaccines has emerged as a strategy to enhance vaccination coverage. This study describes the policy on co-administration and uptake of influenza and COVID-19 vaccination in Europe, the United Kingdom, the United States, and Canada between 2019 and 2023. We collected co-administration policy data from governmental websites, national health organizations, and newspapers. Influenza vaccination coverage among persons ≥65 years and COVID-19 vaccination coverage rates among persons ≥60 years or the general population were collected using national databases, the ECDC database, or ourworldindata.org between 2019 and 2023. Descriptive analyses were used. We collected data from 30/32 (94%) countries on vaccination policy in seasons 2021-2022 and 2022-2023, with most countries (25/30 to 30/30) having policies recommending co-administration. For influenza vaccination coverage, we collected data from 29/32 (91%, 2019-2020), 28/32 (88%, 2020-2021), 27/32 (84%, 2021-2022), and 6/32 (19%, 2022-2023) countries. COVID-19 vaccination was collected from 32/32 (2020-2021), 31/32 (97%, 2021-2022), and 24/32 (75%, 2022-2023) countries. Influenza vaccination coverage increased from 2019-2020 to 2021-2022. COVID-19 vaccination coverage was higher among countries with higher influenza vaccination coverage. By 2022-2023, all countries included implemented a policy supporting co-administration. A positive correlation existed between higher influenza vaccination coverage and higher COVID-19 vaccination rates.
Collapse
Affiliation(s)
| | - Ian McGovern
- Center for Outcomes Research and Epidemiology, CSL Seqirus, Waltham, MA 02451, USA;
| | - Mendel Haag
- Center for Outcomes Research and Epidemiology, CSL Seqirus, 1105 BJ Amsterdam, The Netherlands
| |
Collapse
|
3
|
Liu C, Zhang Y, Li P, Jia H, Ju H, Zhang J, Ferreira da Silva-Júnior E, Samanta S, Kar P, Huang B, Liu X, Zhan P. Development of chalcone-like derivatives and their biological and mechanistic investigations as novel influenza nuclear export inhibitors. Eur J Med Chem 2023; 261:115845. [PMID: 37804770 DOI: 10.1016/j.ejmech.2023.115845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 09/21/2023] [Accepted: 09/27/2023] [Indexed: 10/09/2023]
Abstract
Concerning the emergence of resistance to current anti-influenza drugs, our previous phenotypic-based screening study identified the compound A9 as a promising lead compound. This chalcone analog, containing a 2,6-dimethoxyphenyl moiety, exhibited significant inhibitory activity against oseltamivir-resistant strains (H1N1 pdm09), with an EC50 value of 1.34 μM. However, it also displayed notable cytotoxicity, with a CC50 value of 41.46 μM. Therefore, compound A9 was selected as a prototype structure for further structural optimization in this study. Initially, it was confirmed that the substituting the α,β-unsaturated ketone with pent-1,4-diene-3-one as a linker group significantly reduced the cytotoxicity of the final compounds. Subsequently, the penta-1,4-dien-3-one group was utilized as a privileged fragment for further structural optimization. Following two subsequent rounds of optimizations, we identified compound IIB-2, which contains a 2,6-dimethoxyphenyl- and 1,4-pentadiene-3-one moieties. This compound exhibited inhibitory effects on oseltamivir-resistant strains comparable to its precursor (compound A9), while demonstrating reduced toxicity (CC50 > 100 μM). Furthermore, we investigated its mechanism of action against anti-influenza virus through immunofluorescence, Western blot, and surface plasmon resonance (SPR) experiments. The results revealed that compound IIB-2 can impede virus proliferation by blocking the export of influenza virus nucleoprotein. Thusly, our findings further emphasize influenza nuclear export as a viable target for designing novel chalcone-like derivatives with potential inhibitory properties that could be explored in future lead optimization studies.
Collapse
Affiliation(s)
- Chuanfeng Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, 250012, Jinan, Shandong, PR China; Suzhou Research Institute of Shandong University, Room607, Building B of NUSP, NO.388 Ruoshui Road, SIP, Suzhou, Jiangsu, 215123, PR China
| | - Ying Zhang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, 250012, Jinan, Shandong, PR China
| | - Ping Li
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, 250012, Jinan, Shandong, PR China; Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Huinan Jia
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, 250012, Jinan, Shandong, PR China
| | - Han Ju
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, 250012, Jinan, Shandong, PR China
| | - Jiwei Zhang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, 250012, Jinan, Shandong, PR China
| | - Edeildo Ferreira da Silva-Júnior
- Research Group of Biological and Molecular Chemistry, Institute of Chemistry and Biotechnology, Federal University of Alagoas, Lourival Melo Mota Avenue, AC. Simões Campus, 57072-970, Alagoas, Maceió, Brazil
| | - Sunanda Samanta
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Khandwa Road, Indore, 453552, Madhya Pradesh, India
| | - Parimal Kar
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Khandwa Road, Indore, 453552, Madhya Pradesh, India.
| | - Bing Huang
- China-Belgium Collaborative Research Center for Innovative Antiviral Drugs of Shandong Province, 44 West Culture Road, 250012, Jinan, Shandong, PR China.
| | - Xinyong Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, 250012, Jinan, Shandong, PR China.
| | - Peng Zhan
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, 250012, Jinan, Shandong, PR China.
| |
Collapse
|
4
|
Gupta D, Mohan S. Influenza vaccine: a review on current scenario and future prospects. J Genet Eng Biotechnol 2023; 21:154. [PMID: 38030859 PMCID: PMC10686931 DOI: 10.1186/s43141-023-00581-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 10/28/2023] [Indexed: 12/01/2023]
Abstract
Vaccination is a crucial tool in preventing influenza, but it requires annual updates in vaccine composition due to the ever-changing nature of the flu virus. While healthcare and economic burdens have reduced, the virus remains a challenge. Research conducted over the past decade has revealed pathways for improvement through both basic and clinical studies. Viral surveillance plays a vital role in the better selection of candidate viruses for vaccines and the early detection of drug-resistant strains.This page offers a description of future vaccine developments and an overview of current vaccine options. In the coming years, we anticipate significant changes in vaccine production, moving away from traditional egg-based methods towards innovative technologies such as DNA and RNA vaccines. These newer approaches offer significant advantages over traditional egg-based and cell culture-based influenza vaccine manufacturing.
Collapse
Affiliation(s)
- Dipanshi Gupta
- Amity Institute of Biotechnology, Amity University Uttar Pradesh (AUUP), Sector-125, Noida, Uttar Pradesh, 201303, India
| | - Sumedha Mohan
- Amity Institute of Biotechnology, Amity University Uttar Pradesh (AUUP), Sector-125, Noida, Uttar Pradesh, 201303, India.
| |
Collapse
|
5
|
Molecular characterization of haemagglutinin genes of influenza B viruses circulating in Ghana during 2016 and 2017. PLoS One 2022; 17:e0271321. [PMID: 36149889 PMCID: PMC9506629 DOI: 10.1371/journal.pone.0271321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Accepted: 06/28/2022] [Indexed: 11/29/2022] Open
Abstract
Recent reports of haemagglutinin antigen (HA) mismatch between vaccine composition strains and circulating strains, have led to renewed interest in influenza B viruses. Additionally, there are concerns about resistance to neuraminidase inhibitors in new influenza B isolates. To assess the potential impact in Ghana, we characterized the lineages of influenza B viruses that circulated in Ghana between 2016 and 2017 from different regions of the country: Southern, Northern and Central Ghana. Eight representative specimens from the three regions that were positive for influenza B virus by real-time RT-PCR were sequenced and compared to reference genomes from each lineage. A total of eleven amino acids substitutions were detected in the B/Victoria lineage and six in the B/Yamagata lineage. The strains of influenza B viruses were closely related to influenza B/Brisbane/60/2008 and influenza B/Phuket/3073/2013 for the Victoria and Yamagata lineages, respectively. Three main amino acid substitutions (P31S, I117V and R151K) were found in B/Victoria lineages circulating between 2016 and 2017, while one strain of B/Victoria possessed a unique glycosylation site at amino acid position 51 in the HA2 subunit. Two main substitutions (L172Q and M251V) were detected in the HA gene of the B/Yamagata lineage. The U.S. CDC recently reported a deletion sub-group in influenza B virus, but this was not identified among the Ghanaian specimens. Close monitoring of the patterns of influenza B evolution is necessary for the efficient selection of representative viruses for the design and formulation of effective influenza vaccines.
Collapse
|
6
|
Williams BJM, Ogbunugafor CB, Althouse BM, Hébert-Dufresne L. Immunity-induced criticality of the genotype network of influenza A (H3N2) hemagglutinin. PNAS NEXUS 2022; 1:pgac143. [PMID: 36060623 PMCID: PMC9434636 DOI: 10.1093/pnasnexus/pgac143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 07/22/2022] [Indexed: 11/17/2022]
Abstract
Seasonal influenza kills hundreds of thousands every year, with multiple constantly changing strains in circulation at any given time. A high mutation rate enables the influenza virus to evade recognition by the human immune system, including immunity acquired through past infection and vaccination. Here, we capture the genetic similarity of influenza strains and their evolutionary dynamics with genotype networks. We show that the genotype networks of influenza A (H3N2) hemagglutinin are characterized by heavy-tailed distributions of module sizes and connectivity indicative of critical behavior. We argue that (i) genotype networks are driven by mutation and host immunity to explore a subspace of networks predictable in structure and (ii) genotype networks provide an underlying structure necessary to capture the rich dynamics of multistrain epidemic models. In particular, inclusion of strain-transcending immunity in epidemic models is dependent upon the structure of an underlying genotype network. This interplay is consistent with self-organized criticality where the epidemic dynamics of influenza locates critical regions of its genotype network. We conclude that this interplay between disease dynamics and network structure might be key for future network analysis of pathogen evolution and realistic multistrain epidemic models.
Collapse
Affiliation(s)
- Blake J M Williams
- Vermont Complex Systems Center, University of Vermont , Burlington, VT 05405, USA
| | - C Brandon Ogbunugafor
- Vermont Complex Systems Center, University of Vermont , Burlington, VT 05405, USA
- Department of Ecology and Evolutionary Biology, Yale University , New Haven, CT 06511, USA
- Santa Fe Institute , Santa Fe, NM 87501, USA
- Public Health Modeling Unit, Yale School of Public Health , New Haven, CT 06510, USA
| | - Benjamin M Althouse
- Institute for Disease Modeling, Global Health, Bill & Melinda Gates Foundation , Seattle, WA 98109, USA
- Information School, University of Washington , Seattle, WA 98195, USA
- Department of Biology, New Mexico State University , Las Cruces, NM 88003, USA
| | - Laurent Hébert-Dufresne
- Vermont Complex Systems Center, University of Vermont , Burlington, VT 05405, USA
- Department of Computer Science, University of Vermont , Burlington VT 05405, USA
| |
Collapse
|
7
|
Varghese PM, Kishore U, Rajkumari R. Innate and adaptive immune responses against Influenza A Virus: Immune evasion and vaccination strategies. Immunobiology 2022; 227:152279. [DOI: 10.1016/j.imbio.2022.152279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 08/31/2022] [Accepted: 09/07/2022] [Indexed: 11/25/2022]
|
8
|
Nabakooza G, Galiwango R, Frost SDW, Kateete DP, Kitayimbwa JM. Molecular Epidemiology and Evolutionary Dynamics of Human Influenza Type-A Viruses in Africa: A Systematic Review. Microorganisms 2022; 10:900. [PMID: 35630344 PMCID: PMC9145646 DOI: 10.3390/microorganisms10050900] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/21/2022] [Accepted: 03/22/2022] [Indexed: 02/01/2023] Open
Abstract
Genomic characterization of circulating influenza type-A viruses (IAVs) directs the selection of appropriate vaccine formulations and early detection of potentially pandemic virus strains. However, longitudinal data on the genomic evolution and transmission of IAVs in Africa are scarce, limiting Africa's benefits from potential influenza control strategies. We searched seven databases: African Journals Online, Embase, Global Health, Google Scholar, PubMed, Scopus, and Web of Science according to the PRISMA guidelines for studies that sequenced and/or genomically characterized Africa IAVs. Our review highlights the emergence and diversification of IAVs in Africa since 1993. Circulating strains continuously acquired new amino acid substitutions at the major antigenic and potential N-linked glycosylation sites in their hemagglutinin proteins, which dramatically affected vaccine protectiveness. Africa IAVs phylogenetically mixed with global strains forming strong temporal and geographical evolution structures. Phylogeographic analyses confirmed that viral migration into Africa from abroad, especially South Asia, Europe, and North America, and extensive local viral mixing sustained the genomic diversity, antigenic drift, and persistence of IAVs in Africa. However, the role of reassortment and zoonosis remains unknown. Interestingly, we observed substitutions and clades and persistent viral lineages unique to Africa. Therefore, Africa's contribution to the global influenza ecology may be understated. Our results were geographically biased, with data from 63% (34/54) of African countries. Thus, there is a need to expand influenza surveillance across Africa and prioritize routine whole-genome sequencing and genomic analysis to detect new strains early for effective viral control.
Collapse
Affiliation(s)
- Grace Nabakooza
- Department of Immunology and Molecular Biology, Makerere University, Old Mulago Hill Road, P.O. Box 7072, Kampala 256, Uganda
- UVRI Centre of Excellence in Infection and Immunity Research and Training (MUII-Plus), Makerere University, Plot No: 51-59 Nakiwogo Road, P.O. Box 49, Entebbe 256, Uganda
| | - Ronald Galiwango
- UVRI Centre of Excellence in Infection and Immunity Research and Training (MUII-Plus), Makerere University, Plot No: 51-59 Nakiwogo Road, P.O. Box 49, Entebbe 256, Uganda
- Centre for Computational Biology, Uganda Christian University, Plot 67-173, Bishop Tucker Road, P.O. Box 4, Mukono 256, Uganda
- African Center of Excellence in Bioinformatics and Data Intensive Sciences, Infectious Diseases Institute, Makerere University, Kampala 256, Uganda
| | - Simon D W Frost
- Microsoft Research, Redmond, 14820 NE 36th Street, Washington, DC 98052, USA
- London School of Hygiene & Tropical Medicine (LSHTM), University of London, Keppel Street, Bloomsbury, London WC1E7HT, UK
| | - David P Kateete
- Department of Immunology and Molecular Biology, Makerere University, Old Mulago Hill Road, P.O. Box 7072, Kampala 256, Uganda
- UVRI Centre of Excellence in Infection and Immunity Research and Training (MUII-Plus), Makerere University, Plot No: 51-59 Nakiwogo Road, P.O. Box 49, Entebbe 256, Uganda
| | - John M Kitayimbwa
- UVRI Centre of Excellence in Infection and Immunity Research and Training (MUII-Plus), Makerere University, Plot No: 51-59 Nakiwogo Road, P.O. Box 49, Entebbe 256, Uganda
- Centre for Computational Biology, Uganda Christian University, Plot 67-173, Bishop Tucker Road, P.O. Box 4, Mukono 256, Uganda
| |
Collapse
|
9
|
Phillips O, Tubre T, Lorenco H, Batish SD, Brown W. Limbic encephalitis in a child with ovarian teratoma and influenza B. Case report and critical review of the history of autoimmune anti-N-methyl-d-aspartate receptor encephalitis. J Neuroimmunol 2021; 360:577716. [PMID: 34517152 DOI: 10.1016/j.jneuroim.2021.577716] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 08/29/2021] [Accepted: 09/05/2021] [Indexed: 11/17/2022]
Abstract
We report the appearance of clinical symptoms and signs of N-methyl-d-Aspartate (NMDA) receptor encephalitis in a patient presenting just days after contraction of influenza B. The offending mature ovarian teratoma was identified and removed on the 10th day after the appearance of symptoms, with subsequent nearly complete resolution of symptoms over the subsequent 6 months. We provide a focused literature review of the clinical and pathophysiologic literature of anti-NMDA receptor encephalitis pertaining to influenza B virus and the pediatric population. Taken together, this study contributes to the pathophysiological understanding of anti-NMDA receptor encephalitis and aids clinicians in its early recognition and management.
Collapse
Affiliation(s)
- Oliver Phillips
- Cleveland Clinic Center for Neurological Restoration, Cleveland, OH, USA.
| | - Teddi Tubre
- Rhode Island Hospital, Department of Pathology, Providence, RI, USA
| | - Hillary Lorenco
- Rhode Island Hospital, Department of Pediatric Surgery, Providence, RI, USA
| | | | - William Brown
- Rhode Island Hospital, Department of Pediatrics, Providence, RI, USA
| |
Collapse
|
10
|
Nuwarda RF, Alharbi AA, Kayser V. An Overview of Influenza Viruses and Vaccines. Vaccines (Basel) 2021; 9:1032. [PMID: 34579269 PMCID: PMC8473132 DOI: 10.3390/vaccines9091032] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 09/12/2021] [Accepted: 09/13/2021] [Indexed: 01/12/2023] Open
Abstract
Influenza remains one of the major public health concerns because it causes annual epidemics and can potentially instigate a global pandemic. Numerous countermeasures, including vaccines and antiviral treatments, are in use against seasonal influenza infection; however, their effectiveness has always been discussed due to the ongoing resistance to antivirals and relatively low and unpredictable efficiency of influenza vaccines compared to other vaccines. The growing interest in vaccines as a promising approach to prevent and control influenza may provide alternative vaccine development options with potentially increased efficiency. In addition to currently available inactivated, live-attenuated, and recombinant influenza vaccines on the market, novel platforms such as virus-like particles (VLPs) and nanoparticles, and new vaccine formulations are presently being explored. These platforms provide the opportunity to design influenza vaccines with improved properties to maximize quality, efficacy, and safety. The influenza vaccine manufacturing process is also moving forward with advancements relating to egg- and cell-based production, purification processes, and studies into the physicochemical attributes and vaccine degradation pathways. These will contribute to the design of more stable, optimized vaccine formulations guided by contemporary analytical testing methods and via the implementation of the latest advances in the field.
Collapse
Affiliation(s)
| | | | - Veysel Kayser
- Faculty of Medicine and Health, Sydney Pharmacy School, The University of Sydney, Sydney, NSW 2006, Australia; (R.F.N.); (A.A.A.)
| |
Collapse
|
11
|
Naeem A, Elbakkouri K, Alfaiz A, Hamed ME, Alsaran H, AlOtaiby S, Enani M, Alosaimi B. Antigenic drift of hemagglutinin and neuraminidase in seasonal H1N1 influenza viruses from Saudi Arabia in 2014 to 2015. J Med Virol 2020; 92:3016-3027. [PMID: 32159230 PMCID: PMC7228267 DOI: 10.1002/jmv.25759] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Accepted: 03/04/2020] [Indexed: 01/19/2023]
Abstract
Antigenic drift of the hemagglutinin (HA) and neuraminidase (NA) proteins of the influenza virus cause a decrease in vaccine efficacy. Since the information about the evolution of these viruses in Saudi is deficient so we investigated the genetic diversity of circulating H1N1 viruses. Nasopharyngeal aspirates/swabs collected from 149 patients hospitalized with flu-like symptoms during 2014 and 2015 were analyzed. Viral RNA extraction was followed by a reverse transcription-polymerase chain reaction and genetic sequencing. We analyzed complete gene sequences of HA and NA from 80 positive isolates. Phylogenetic analysis of HA and NA genes of 80 isolates showed similar topologies and co-circulation of clades 6b. Genetic diversity was observed among circulating viruses belonging to clade 6B.1A. The amino acid residues in the HA epitope domain were under purifying selection. Amino acid changes at key antigenic sites, such as position S101N, S179N (antigenic site-Sa), I233T (antigenic site-Sb) in the head domain might have resulted in antigenic drift and emergence of variant viruses. For NA protein, 36% isolates showed the presence of amino acid changes such as V13I (n = 29), I314M (n = 29) and 12% had I34V (n = 10). However, H257Y mutation responsible for resistance to neuraminidase inhibitors was missing. The presence of amino acid changes at key antigenic sites and their topologies with structural mapping of residues under purifying selection highlights the importance of antigenic drift and warrants further characterization of recently circulating viruses in view of vaccine effectiveness. The co-circulation of several clades and the predominance of clade 6B.1 suggest multiple introductions in Saudi.
Collapse
MESH Headings
- Humans
- Neuraminidase/genetics
- Saudi Arabia/epidemiology
- Influenza, Human/virology
- Influenza, Human/epidemiology
- Phylogeny
- Influenza A Virus, H1N1 Subtype/genetics
- Influenza A Virus, H1N1 Subtype/immunology
- Influenza A Virus, H1N1 Subtype/enzymology
- Influenza A Virus, H1N1 Subtype/isolation & purification
- Hemagglutinin Glycoproteins, Influenza Virus/genetics
- Adult
- Male
- Female
- Young Adult
- Genetic Variation
- Middle Aged
- Adolescent
- Genetic Drift
- Child
- Child, Preschool
- Amino Acid Substitution
- Viral Proteins/genetics
- Nasopharynx/virology
- Antigens, Viral/genetics
- Antigens, Viral/immunology
- RNA, Viral/genetics
- Antigenic Variation
- Aged
- Sequence Analysis, DNA
Collapse
Affiliation(s)
- Asif Naeem
- Research CenterKing Fahad Medical CityRiyadhSaudi Arabia
| | | | - Ali Alfaiz
- Research CenterKing Fahad Medical CityRiyadhSaudi Arabia
| | | | - Hadel Alsaran
- Research CenterKing Fahad Medical CityRiyadhSaudi Arabia
| | | | - Mushira Enani
- Medical Specialties Department, Section of Infectious DiseasesKing Fahad Medical CityRiyadhSaudi Arabia
| | - Bandar Alosaimi
- Research CenterKing Fahad Medical CityRiyadhSaudi Arabia
- College of MedicineKing Fahad Medical CityRiyadhSaudi Arabia
| |
Collapse
|
12
|
Walter EB, Klein NP, Wodi AP, Rountree W, Todd CA, Wiesner A, Duffy J, Marquez PL, Broder KR. Fever After Influenza, Diphtheria-Tetanus-Acellular Pertussis, and Pneumococcal Vaccinations. Pediatrics 2020; 145:peds.2019-1909. [PMID: 32029684 PMCID: PMC7055925 DOI: 10.1542/peds.2019-1909] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/27/2019] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Administering inactivated influenza vaccine (IIV), 13-valent pneumococcal conjugate vaccine (PCV13), and diphtheria-tetanus-acellular pertussis (DTaP) vaccine together has been associated with increased risk for febrile seizure after vaccination. We assessed the effect of administering IIV at a separate visit from PCV13 and DTaP on postvaccination fever. METHODS In 2017-2018, children aged 12 to 16 months were randomly assigned to receive study vaccines simultaneously or sequentially. They had 2 study visits 2 weeks apart; nonstudy vaccines were permitted at visit 1. The simultaneous group received PCV13, DTaP, and quadrivalent IIV (IIV4) at visit 1 and no vaccines at visit 2. The sequential group received PCV13 and DTaP at visit 1 and IIV4 at visit 2. Participants were monitored for fever (≥38°C) and antipyretic use during the 8 days after visits. RESULTS There were 110 children randomly assigned to the simultaneous group and 111 children to the sequential group; 90% received ≥1 nonstudy vaccine at visit 1. Similar proportions of children experienced fever on days 1 to 2 after visits 1 and 2 combined (simultaneous [8.1%] versus sequential [9.3%]; adjusted relative risk = 0.87 [95% confidence interval 0.36-2.10]). During days 1 to 2 after visit 1, more children in the simultaneous group received antipyretics (37.4% vs 22.4%; P = .020). CONCLUSIONS In our study, delaying IIV4 administration by 2 weeks in children receiving DTaP and PCV13 did not reduce fever occurrence after vaccination. Reevaluating this strategy to prevent fever using an IIV4 with a different composition in a future influenza season may be considered.
Collapse
Affiliation(s)
- Emmanuel B. Walter
- Department of Pediatrics and,Duke Human Vaccine Institute, School of Medicine,
Duke University, Durham, North Carolina
| | - Nicola P. Klein
- Kaiser Permanente Vaccine Study Center, Kaiser
Permanente Northern California, Oakland, California; and
| | - A. Patricia Wodi
- Immunization Safety Office, Centers for Disease
Control and Prevention, Atlanta, Georgia
| | - Wes Rountree
- Duke Human Vaccine Institute, School of Medicine,
Duke University, Durham, North Carolina
| | - Christopher A. Todd
- Duke Human Vaccine Institute, School of Medicine,
Duke University, Durham, North Carolina
| | - Amy Wiesner
- Kaiser Permanente Vaccine Study Center, Kaiser
Permanente Northern California, Oakland, California; and
| | - Jonathan Duffy
- Immunization Safety Office, Centers for Disease
Control and Prevention, Atlanta, Georgia
| | - Paige L. Marquez
- Immunization Safety Office, Centers for Disease
Control and Prevention, Atlanta, Georgia
| | - Karen R. Broder
- Immunization Safety Office, Centers for Disease
Control and Prevention, Atlanta, Georgia
| |
Collapse
|
13
|
Yoshihara K, Minh LN, Okada T, Toizumi M, Nguyen HA, Vo HM, Hashizume M, Dang DA, Kimura H, Yoshida LM. Evolutionary dynamics of influenza B strains detected from paediatric acute respiratory infections in central Vietnam. INFECTION GENETICS AND EVOLUTION 2020; 81:104264. [PMID: 32105864 DOI: 10.1016/j.meegid.2020.104264] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 02/07/2020] [Accepted: 02/22/2020] [Indexed: 11/28/2022]
Abstract
Influenza virus B belongs to the family Orthomyxoviridae with segmented negative-sense RNA genomes. Since 1970s, influenza B has diverged intoVictoria and Yamagata, which differs in antigenic and evolutionary characteristics. Yet, molecular-epidemiological information of influenza B from developing nations is limited. In central Vietnam, influenza A subtype-specific circulation pattern and clinical characteristics were previously described. However, molecular evolutionary characteristics of influenza B has not been discussed to date. We utilized the influenza B positives obtained from paediatric ARI surveillance during 2007-2013. Influenza B HA and NA genes were amplified, sequenced, and phylogenetic/molecular evolutionary analysis was performed using Maximum Likelihood and Bayesian MCMC. Phylodynamics analysis was performed with Bayesian Skyline Plot (BSP). Furthermore, we performed selection pressure analysis and estimated N-glycosylation sites. In the current study, overall positive rate for influenza B was 3.0%, and Victoria lineage immediately became predominant in post-A/H1N1pdm09 period. The noticeable shift in Victoria lineage WHO Group occurred. With respect to the evolutionary rate (substitutions/site/year), Victoria lineage HA gene was evolving faster than Yamagata lineage (2.43 × 10-3 vs 2.00 × 10-3). Furthermore, the evolutionary rate of Victoria Group 5 was greater than Group 1. BSP presented the rapid growth in Effective Population Size (EPS) of Victoria lineage occurred soon after the 1st A/H1N1pdm09 case was detected whereas the EPS of Yamagata lineage was stable for both genes. N-glycosylation pattern between lineages and among WHO Groups were slightly different, and HA gene had a total of 6 amino acid substitutions under positive section pressure (4 for Victoria and 2 for Yamagata). The current results highlight the importance of Victoria lineage in post-A/H1N1pdm09 period. Difference in evolutionary characteristics and phylodynamics may indicate lineage and WHO Group-specific evolutionary dynamics. It is necessary to further continue the molecular-epidemiological surveillance in local setting to gain a better understanding of local evolutionary characteristics of influenza B strains.
Collapse
Affiliation(s)
- Keisuke Yoshihara
- Department of Paediatric Infectious Diseases, Institute of Tropical Medicine, Nagasaki University, Nagasaki 852-8523, Japan
| | - Le Nhat Minh
- National Institute of Hygiene and Epidemiology, Hanoi 100000, Viet Nam
| | - Takashi Okada
- Department of Paediatric Infectious Diseases, Institute of Tropical Medicine, Nagasaki University, Nagasaki 852-8523, Japan
| | - Michiko Toizumi
- Department of Paediatric Infectious Diseases, Institute of Tropical Medicine, Nagasaki University, Nagasaki 852-8523, Japan
| | - Hien Anh Nguyen
- National Institute of Hygiene and Epidemiology, Hanoi 100000, Viet Nam
| | - Hien Minh Vo
- Department of Paediatrics, Khanh Hoa General Hospital, Nha Trang 650000, Viet Nam
| | - Masahiro Hashizume
- Department of Paediatric Infectious Diseases, Institute of Tropical Medicine, Nagasaki University, Nagasaki 852-8523, Japan
| | - Duc Anh Dang
- National Institute of Hygiene and Epidemiology, Hanoi 100000, Viet Nam
| | - Hirokazu Kimura
- School of Medical Technology, Gunma Paz University, Takasaki-shi, Gunma, 370-0006, Japan
| | - Lay-Myint Yoshida
- Department of Paediatric Infectious Diseases, Institute of Tropical Medicine, Nagasaki University, Nagasaki 852-8523, Japan.
| |
Collapse
|
14
|
Quan L, Ji C, Ding X, Peng Y, Liu M, Sun J, Jiang T, Wu A. Cluster-Transition Determining Sites Underlying the Antigenic Evolution of Seasonal Influenza Viruses. Mol Biol Evol 2019; 36:1172-1186. [DOI: 10.1093/molbev/msz050] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Affiliation(s)
- Lijun Quan
- Center for Systems Medicine, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou Institute of Systems Medicine, Suzhou, China
- School of Computer Science and Technology, Soochow University, Suzhou, China
| | - Chengyang Ji
- Center for Systems Medicine, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou Institute of Systems Medicine, Suzhou, China
| | - Xiao Ding
- Center for Systems Medicine, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou Institute of Systems Medicine, Suzhou, China
| | - Yousong Peng
- College of Biology, Human University, Changsha, China
| | - Mi Liu
- Jiangsu Institute of Clinical Immunology & Jiangsu Key Laboratory of Clinical Immunology, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jiya Sun
- Center for Systems Medicine, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou Institute of Systems Medicine, Suzhou, China
| | - Taijiao Jiang
- Center for Systems Medicine, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou Institute of Systems Medicine, Suzhou, China
| | - Aiping Wu
- Center for Systems Medicine, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou Institute of Systems Medicine, Suzhou, China
| |
Collapse
|
15
|
Jorquera PA, Mishin VP, Chesnokov A, Nguyen HT, Mann B, Garten R, Barnes J, Hodges E, De La Cruz J, Xu X, Katz J, Wentworth DE, Gubareva LV. Insights into the antigenic advancement of influenza A(H3N2) viruses, 2011-2018. Sci Rep 2019; 9:2676. [PMID: 30804469 PMCID: PMC6389938 DOI: 10.1038/s41598-019-39276-1] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 01/11/2019] [Indexed: 11/12/2022] Open
Abstract
Influenza A(H3N2) viruses evade human immunity primarily by acquiring antigenic changes in the haemagglutinin (HA). HA receptor-binding features of contemporary A(H3N2) viruses hinder traditional antigenic characterization using haemagglutination inhibition and promote selection of HA mutants. Thus, alternative approaches are needed to reliably assess antigenic relatedness between circulating viruses and vaccines. We developed a high content imaging-based neutralization test (HINT) to reduce antigenic mischaracterization resulting from virus adaptation to cell culture. Ferret reference antisera were raised using clinical specimens containing viruses representing recent vaccine strains. Analysis of viruses circulating during 2011-2018 showed that gain of an N158-linked glycosylation in HA was a molecular determinant of antigenic distancing between A/Hong Kong/4801/2014-like (clade 3C.2a) and A/Texas/50/2012-like viruses (clade 3C.1), while multiple evolutionary HA F193S substitution were linked to antigenic distancing from A/Switzerland/97152963/2013-like (clade 3C.3a) and further antigenic distancing from A/Texas/50/2012-like viruses. Additionally, a few viruses carrying HA T135K and/or I192T showed reduced neutralization by A/Hong Kong/4801/2014-like antiserum. Notably, this technique elucidated the antigenic characteristics of clinical specimens, enabling direct characterization of viruses produced in vivo, and eliminating in vitro culture, which rapidly alters the genotype/phenotype. HINT is a valuable new antigenic analysis tool for vaccine strain selection.
Collapse
MESH Headings
- Animals
- Antibodies, Viral/immunology
- Antigens, Viral/immunology
- Ferrets/immunology
- Ferrets/virology
- Glycosylation
- Hemagglutination Inhibition Tests/methods
- Hemagglutinin Glycoproteins, Influenza Virus/genetics
- Hemagglutinin Glycoproteins, Influenza Virus/immunology
- Hemagglutinin Glycoproteins, Influenza Virus/metabolism
- Humans
- Immune Sera/immunology
- Influenza A Virus, H3N2 Subtype/classification
- Influenza A Virus, H3N2 Subtype/genetics
- Influenza A Virus, H3N2 Subtype/physiology
- Influenza Vaccines/immunology
- Influenza, Human/diagnosis
- Influenza, Human/immunology
- Influenza, Human/virology
- Neutralization Tests/methods
- Phylogeny
Collapse
Affiliation(s)
- Patricia A Jorquera
- Influenza Division, Centers for Disease Control and Prevention (CDC), 1600 Clifton Road, Atlanta, GA, 30329, USA
- CNI Advantage, LLC. 17 Executive Park Dr NE, Atlanta, GA, 30329, USA
| | - Vasiliy P Mishin
- Influenza Division, Centers for Disease Control and Prevention (CDC), 1600 Clifton Road, Atlanta, GA, 30329, USA
| | - Anton Chesnokov
- Influenza Division, Centers for Disease Control and Prevention (CDC), 1600 Clifton Road, Atlanta, GA, 30329, USA
| | - Ha T Nguyen
- Influenza Division, Centers for Disease Control and Prevention (CDC), 1600 Clifton Road, Atlanta, GA, 30329, USA
- Battelle Memorial Institute, 2987 Clairmont Rd, Suite 450, Atlanta, GA, 30329, USA
| | - Brian Mann
- Influenza Division, Centers for Disease Control and Prevention (CDC), 1600 Clifton Road, Atlanta, GA, 30329, USA
- Battelle Memorial Institute, 2987 Clairmont Rd, Suite 450, Atlanta, GA, 30329, USA
| | - Rebecca Garten
- Influenza Division, Centers for Disease Control and Prevention (CDC), 1600 Clifton Road, Atlanta, GA, 30329, USA
| | - John Barnes
- Influenza Division, Centers for Disease Control and Prevention (CDC), 1600 Clifton Road, Atlanta, GA, 30329, USA
| | - Erin Hodges
- Influenza Division, Centers for Disease Control and Prevention (CDC), 1600 Clifton Road, Atlanta, GA, 30329, USA
- CNI Advantage, LLC. 17 Executive Park Dr NE, Atlanta, GA, 30329, USA
| | - Juan De La Cruz
- Influenza Division, Centers for Disease Control and Prevention (CDC), 1600 Clifton Road, Atlanta, GA, 30329, USA
- Battelle Memorial Institute, 2987 Clairmont Rd, Suite 450, Atlanta, GA, 30329, USA
| | - Xiyan Xu
- Influenza Division, Centers for Disease Control and Prevention (CDC), 1600 Clifton Road, Atlanta, GA, 30329, USA
| | - Jackie Katz
- Influenza Division, Centers for Disease Control and Prevention (CDC), 1600 Clifton Road, Atlanta, GA, 30329, USA
| | - David E Wentworth
- Influenza Division, Centers for Disease Control and Prevention (CDC), 1600 Clifton Road, Atlanta, GA, 30329, USA
| | - Larisa V Gubareva
- Influenza Division, Centers for Disease Control and Prevention (CDC), 1600 Clifton Road, Atlanta, GA, 30329, USA.
| |
Collapse
|
16
|
Khan A, AlBalwi MA, AlAbdulkareem I, AlMasoud A, AlAsiri A, AlHarbi W, AlSehile F, El-Saed A, Balkhy HH. Atypical influenza A(H1N1)pdm09 strains caused an influenza virus outbreak in Saudi Arabia during the 2009-2011 pandemic season. J Infect Public Health 2019; 12:557-567. [PMID: 30799182 DOI: 10.1016/j.jiph.2019.01.067] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 01/22/2019] [Accepted: 01/30/2019] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND The triple assortment influenza A(H1N1) virus emerged in spring 2009 and disseminated worldwide, including Saudi Arabia. This study was carried out to characterize Saudi influenza isolates in relation to the global strains and to evaluate the potential role of mutated residues in transmission, adaptation, and the pathogenicity of the virus. METHODS Nasopharyngeal samples (n = 6492) collected between September 2009 to March 2011 from patients with influenza-like illness were screened by PCR for influenza A(H1N1). Phylogenetic and Molecular evolutionary analysis were carried out to place the Saudi strains in relation to the global strains followed by Mutation analysis of surface and internal proteins. RESULTS Concatenated whole-genome phylogenetic analysis along with hemagglutinin (HA) signature changes, that is, Aspartic Acid (D) at position 187, P83S, S203T, and R223Q confirmed that the Saudi strains belong to the antigenic category of A/California/07/2009. However, phylogenetic analysis revealed unusual strains of A(H1N1) circulating in Saudi Arabia, not belonging to any of known clades, appearing in five distinct groups well supported by group-specific mutations and novel mutation complexes. These cases had characteristic inter- and intragroup substitution patterns while few of their closest matches showed up as sporadic cases the world over. Specific mutation patterns were detected within the functional domains of internal proteins PB2, PB1, PA, NP, NS1, and M2 having a putative role in viral fitness and virulence. Bayesian coalescent MCMC analysis revealed that Saudi strains belonged to cluster 2 of A(H1N1)pdm09 and spread a month later as compared to other strains of this cluster. CONCLUSION Influenza outbreak in Saudi Arabia during 2009-2011 was caused by atypical strains of influenza A(H1N1)pdm09, probably introduced in this community on multiple occasions. To understand the antigenic significance of these novel point mutations and mutation complexes require functional studies, which will be crucial for risk assessment of emergent strains and defining infection control measures.
Collapse
Affiliation(s)
- Anis Khan
- Department of Medical Genomics Research, King Abdullah International Medical Research Center, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Mohammed A AlBalwi
- Department of Medical Genomics Research, King Abdullah International Medical Research Center, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia; Department of Pathology & Laboratory Medicine, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia; King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia.
| | - Ibraheem AlAbdulkareem
- Intramural health sciences research, Princess Nourah Bint Abdulrahman university, Riyadh, Saudi Arabia
| | - Abdulrahman AlMasoud
- Department of Medical Genomics Research, King Abdullah International Medical Research Center, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Abdulrahman AlAsiri
- Department of Medical Genomics Research, King Abdullah International Medical Research Center, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Wardah AlHarbi
- Department of Medical Genomics Research, King Abdullah International Medical Research Center, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Faisal AlSehile
- King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Aiman El-Saed
- Department of Infection Prevention & Control Department, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Hanan H Balkhy
- King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia; Department of Infection Prevention & Control Department, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| |
Collapse
|
17
|
Santos JJS, Abente EJ, Obadan AO, Thompson AJ, Ferreri L, Geiger G, Gonzalez-Reiche AS, Lewis NS, Burke DF, Rajão DS, Paulson JC, Vincent AL, Perez DR. Plasticity of Amino Acid Residue 145 Near the Receptor Binding Site of H3 Swine Influenza A Viruses and Its Impact on Receptor Binding and Antibody Recognition. J Virol 2019; 93:e01413-18. [PMID: 30355680 PMCID: PMC6321904 DOI: 10.1128/jvi.01413-18] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 10/13/2018] [Indexed: 11/20/2022] Open
Abstract
The hemagglutinin (HA), a glycoprotein on the surface of influenza A virus (IAV), initiates the virus life cycle by binding to terminal sialic acid (SA) residues on host cells. The HA gradually accumulates amino acid substitutions that allow IAV to escape immunity through a mechanism known as antigenic drift. We recently confirmed that a small set of amino acid residues are largely responsible for driving antigenic drift in swine-origin H3 IAV. All identified residues are located adjacent to the HA receptor binding site (RBS), suggesting that substitutions associated with antigenic drift may also influence receptor binding. Among those substitutions, residue 145 was shown to be a major determinant of antigenic evolution. To determine whether there are functional constraints to substitutions near the RBS and their impact on receptor binding and antigenic properties, we carried out site-directed mutagenesis experiments at the single-amino-acid level. We generated a panel of viruses carrying substitutions at residue 145 representing all 20 amino acids. Despite limited amino acid usage in nature, most substitutions at residue 145 were well tolerated without having a major impact on virus replication in vitro All substitution mutants retained receptor binding specificity, but the substitutions frequently led to decreased receptor binding. Glycan microarray analysis showed that substitutions at residue 145 modulate binding to a broad range of glycans. Furthermore, antigenic characterization identified specific substitutions at residue 145 that altered antibody recognition. This work provides a better understanding of the functional effects of amino acid substitutions near the RBS and the interplay between receptor binding and antigenic drift.IMPORTANCE The complex and continuous antigenic evolution of IAVs remains a major hurdle for vaccine selection and effective vaccination. On the hemagglutinin (HA) of the H3N2 IAVs, the amino acid substitution N 145 K causes significant antigenic changes. We show that amino acid 145 displays remarkable amino acid plasticity in vitro, tolerating multiple amino acid substitutions, many of which have not yet been observed in nature. Mutant viruses carrying substitutions at residue 145 showed no major impairment in virus replication in the presence of lower receptor binding avidity. However, their antigenic characterization confirmed the impact of the 145 K substitution in antibody immunodominance. We provide a better understanding of the functional effects of amino acid substitutions implicated in antigenic drift and its consequences for receptor binding and antigenicity. The mutation analyses presented in this report represent a significant data set to aid and test the ability of computational approaches to predict binding of glycans and in antigenic cartography analyses.
Collapse
Affiliation(s)
- Jefferson J S Santos
- Department of Population Health, Poultry Diagnostic and Research Center, University of Georgia, Athens, Georgia, USA
| | - Eugenio J Abente
- Virus and Prion Research Unit, National Animal Disease Center, Agricultural Research Service, U.S. Department of Agriculture, Ames, Iowa, USA
| | - Adebimpe O Obadan
- Department of Population Health, Poultry Diagnostic and Research Center, University of Georgia, Athens, Georgia, USA
| | - Andrew J Thompson
- Department of Molecular Medicine and Immunology & Microbiology, The Scripps Research Institute, La Jolla, California, USA
| | - Lucas Ferreri
- Department of Population Health, Poultry Diagnostic and Research Center, University of Georgia, Athens, Georgia, USA
| | - Ginger Geiger
- Department of Population Health, Poultry Diagnostic and Research Center, University of Georgia, Athens, Georgia, USA
| | - Ana S Gonzalez-Reiche
- Department of Population Health, Poultry Diagnostic and Research Center, University of Georgia, Athens, Georgia, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Nicola S Lewis
- Department of Pathobiology and Population Sciences, Royal Veterinary College, Hatfield, Hertfordshire, United Kingdom
| | - David F Burke
- Department of Zoology, University of Cambridge, Cambridge, United Kingdom
| | - Daniela S Rajão
- Department of Population Health, Poultry Diagnostic and Research Center, University of Georgia, Athens, Georgia, USA
| | - James C Paulson
- Department of Molecular Medicine and Immunology & Microbiology, The Scripps Research Institute, La Jolla, California, USA
| | - Amy L Vincent
- Virus and Prion Research Unit, National Animal Disease Center, Agricultural Research Service, U.S. Department of Agriculture, Ames, Iowa, USA
| | - Daniel R Perez
- Department of Population Health, Poultry Diagnostic and Research Center, University of Georgia, Athens, Georgia, USA
| |
Collapse
|
18
|
Kinetic, Thermodynamic, and Structural Analysis of Drug Resistance Mutations in Neuraminidase from the 2009 Pandemic Influenza Virus. Viruses 2018; 10:v10070339. [PMID: 29933553 PMCID: PMC6071225 DOI: 10.3390/v10070339] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2018] [Revised: 06/14/2018] [Accepted: 06/19/2018] [Indexed: 12/25/2022] Open
Abstract
Neuraminidase is the main target for current influenza drugs. Reduced susceptibility to oseltamivir, the most widely prescribed neuraminidase inhibitor, has been repeatedly reported. The resistance substitutions I223V and S247N, alone or in combination with the major oseltamivir-resistance mutation H275Y, have been observed in 2009 pandemic H1N1 viruses. We overexpressed and purified the ectodomain of wild-type neuraminidase from the A/California/07/2009 (H1N1) influenza virus, as well as variants containing H275Y, I223V, and S247N single mutations and H275Y/I223V and H275Y/S247N double mutations. We performed enzymological and thermodynamic analyses and structurally examined the resistance mechanism. Our results reveal that the I223V or S247N substitution alone confers only a moderate reduction in oseltamivir affinity. In contrast, the major oseltamivir resistance mutation H275Y causes a significant decrease in the enzyme’s ability to bind this drug. Combination of H275Y with an I223V or S247N mutation results in extreme impairment of oseltamivir’s inhibition potency. Our structural analyses revealed that the H275Y substitution has a major effect on the oseltamivir binding pose within the active site while the influence of other studied mutations is much less prominent. Our crystal structures also helped explain the augmenting effect on resistance of combining H275Y with both substitutions.
Collapse
|
19
|
Antigenically Diverse Swine Origin H1N1 Variant Influenza Viruses Exhibit Differential Ferret Pathogenesis and Transmission Phenotypes. J Virol 2018. [PMID: 29540597 DOI: 10.1128/jvi.00095-18] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Influenza A(H1) viruses circulating in swine represent an emerging virus threat, as zoonotic infections occur sporadically following exposure to swine. A fatal infection caused by an H1N1 variant (H1N1v) virus was detected in a patient with reported exposure to swine and who presented with pneumonia, respiratory failure, and cardiac arrest. To understand the genetic and phenotypic characteristics of the virus, genome sequence analysis, antigenic characterization, and ferret pathogenesis and transmissibility experiments were performed. Antigenic analysis of the virus isolated from the fatal case, A/Ohio/09/2015, demonstrated significant antigenic drift away from the classical swine H1N1 variant viruses and H1N1 pandemic 2009 viruses. A substitution in the H1 hemagglutinin (G155E) was identified that likely impacted antigenicity, and reverse genetics was employed to understand the molecular mechanism of antibody escape. Reversion of the substitution to 155G, in a reverse genetics A/Ohio/09/2015 virus, showed that this residue was central to the loss of hemagglutination inhibition by ferret antisera raised against a prototypical H1N1 pandemic 2009 virus (A/California/07/2009), as well as gamma lineage classical swine H1N1 viruses, demonstrating the importance of this residue for antibody recognition of this H1 lineage. When analyzed in the ferret model, A/Ohio/09/2015 and another H1N1v virus, A/Iowa/39/2015, as well as A/California/07/2009, replicated efficiently in the respiratory tract of ferrets. The two H1N1v viruses transmitted efficiently among cohoused ferrets, but respiratory droplet transmission studies showed that A/California/07/2009 transmitted through the air more efficiently. Preexisting immunity to A/California/07/2009 did not fully protect ferrets from challenge with A/Ohio/09/2015.IMPORTANCE Human infections with classical swine influenza A(H1N1) viruses that circulate in pigs continue to occur in the United States following exposure to swine. To understand the genetic and virologic characteristics of a virus (A/Ohio/09/2015) associated with a fatal infection and a virus associated with a nonfatal infection (A/Iowa/39/2015), we performed genome sequence analysis, antigenic testing, and pathogenicity and transmission studies in a ferret model. Reverse genetics was employed to identify a single antigenic site substitution (HA G155E) responsible for antigenic variation of A/Ohio/09/2015 compared to related classical swine influenza A(H1N1) viruses. Ferrets with preexisting immunity to the pandemic A(H1N1) virus were challenged with A/Ohio/09/2015, demonstrating decreased protection. These data illustrate the potential for currently circulating swine influenza viruses to infect and cause illness in humans with preexisting immunity to H1N1 pandemic 2009 viruses and a need for ongoing risk assessment and development of candidate vaccine viruses for improved pandemic preparedness.
Collapse
|
20
|
Montomoli E, Torelli A, Manini I, Gianchecchi E. Immunogenicity and Safety of the New Inactivated Quadrivalent Influenza Vaccine Vaxigrip Tetra: Preliminary Results in Children ≥6 Months and Older Adults. Vaccines (Basel) 2018; 6:E14. [PMID: 29518013 PMCID: PMC5874655 DOI: 10.3390/vaccines6010014] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 02/26/2018] [Accepted: 03/05/2018] [Indexed: 12/31/2022] Open
Abstract
Since the mid-1980s, two lineages of influenza B viruses have been distinguished. These can co-circulate, limiting the protection provided by inactivated trivalent influenza vaccines (TIVs). This has prompted efforts to formulate quadrivalent influenza vaccines (QIVs), to enhance protection against circulating influenza B viruses. This review describes the results obtained from seven phase III clinical trials evaluating the immunogenicity, safety, and lot-to-lot consistency of a new quadrivalent split-virion influenza vaccine (Vaxigrip Tetra®) formulated by adding a second B strain to the already licensed TIV. Since Vaxigrip Tetra was developed by means of a manufacturing process strictly related to that used for TIV, the data on the safety profile of TIV are considered supportive of that of Vaxigrip Tetra. The safety and immunogenicity of Vaxigrip Tetra were similar to those of the corresponding licensed TIV. Moreover, the new vaccine elicits a superior immune response towards the additional strain, without affecting immunogenicity towards the other three strains. Vaxigrip Tetra is well tolerated, has aroused no safety concerns, and is recommended for the active immunization of individuals aged ≥6 months. In addition, preliminary data confirm its immunogenicity and safety even in children aged 6-35 months and its immunogenicity in older subjects (aged 66-80 years).
Collapse
Affiliation(s)
- Emanuele Montomoli
- VisMederi srl, Strada del Petriccio e Belriguardo, 35, 53100 Siena, Italy.
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy.
| | - Alessandro Torelli
- VisMederi srl, Strada del Petriccio e Belriguardo, 35, 53100 Siena, Italy.
- Department of Life Sciences, University of Siena, 53100 Siena, Italy.
| | - Ilaria Manini
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy.
| | - Elena Gianchecchi
- VisMederi srl, Strada del Petriccio e Belriguardo, 35, 53100 Siena, Italy.
| |
Collapse
|
21
|
Morris DH, Gostic KM, Pompei S, Bedford T, Łuksza M, Neher RA, Grenfell BT, Lässig M, McCauley JW. Predictive Modeling of Influenza Shows the Promise of Applied Evolutionary Biology. Trends Microbiol 2018; 26:102-118. [PMID: 29097090 PMCID: PMC5830126 DOI: 10.1016/j.tim.2017.09.004] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 09/06/2017] [Accepted: 09/19/2017] [Indexed: 01/16/2023]
Abstract
Seasonal influenza is controlled through vaccination campaigns. Evolution of influenza virus antigens means that vaccines must be updated to match novel strains, and vaccine effectiveness depends on the ability of scientists to predict nearly a year in advance which influenza variants will dominate in upcoming seasons. In this review, we highlight a promising new surveillance tool: predictive models. Based on data-sharing and close collaboration between the World Health Organization and academic scientists, these models use surveillance data to make quantitative predictions regarding influenza evolution. Predictive models demonstrate the potential of applied evolutionary biology to improve public health and disease control. We review the state of influenza predictive modeling and discuss next steps and recommendations to ensure that these models deliver upon their considerable biomedical promise.
Collapse
Affiliation(s)
- Dylan H Morris
- Department of Ecology and Evolutionary Biology, Princeton University, Princeton, NJ, USA.
| | - Katelyn M Gostic
- Department of Ecology and Evolutionary Biology, University of California, Los Angeles, CA, USA
| | - Simone Pompei
- Institute for Theoretical Physics, University of Cologne, Cologne, Germany
| | - Trevor Bedford
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Marta Łuksza
- Institute for Advanced Study, Princeton, NJ, USA
| | - Richard A Neher
- Biozentrum, University of Basel and Swiss Institute of Bioinformatics, Basel, Switzerland
| | - Bryan T Grenfell
- Department of Ecology and Evolutionary Biology, Princeton University, Princeton, NJ, USA; Fogarty International Center, National Institutes of Health, Bethesda, MD, USA
| | - Michael Lässig
- Institute for Theoretical Physics, University of Cologne, Cologne, Germany
| | - John W McCauley
- Worldwide Influenza Centre, Francis Crick Institute, London, UK
| |
Collapse
|
22
|
Diversity of antigenic mutants of influenza A(H1N1)pdm09 virus escaped from human monoclonal antibodies. Sci Rep 2017; 7:17735. [PMID: 29255273 PMCID: PMC5735164 DOI: 10.1038/s41598-017-17986-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 12/04/2017] [Indexed: 12/21/2022] Open
Abstract
Since the 2017 Southern Hemisphere influenza season, the A(H1N1)pdm09-like virus recommended for use in the vaccine was changed because human, but not ferret, sera distinguish A(H1N1)pdm09 viruses isolated after 2013 from the previously circulating strains. An amino acid substitution, lysine to glutamine, at position 166 (H3 numbering) in the major antigenic site of HA was reported to be responsible for the antigenic drift. Here, we obtained two anti-A(H1N1)pdm09 HA monoclonal antibodies that failed to neutralize viruses isolated after 2013 from a vaccinated volunteer. Escape mutations were identified at position 129, 165, or 166 in the major antigenic site of HA. Competitive growth of the escape mutant viruses with the wild-type virus revealed that some escape mutants possessing an amino acid substitution other than K166Q showed superior growth to that of the wild-type virus. These results suggest that in addition to the K166Q mutation that occurred in epidemic strains, other HA mutations can confer resistance to antibodies that recognize the K166 area, leading to emergence of epidemic strains with such mutations.
Collapse
|
23
|
Canche-Pech JR, Conde-Ferraez L, Puerto-Solis M, Gonzalez-Losa R, Granja-Pérez P, Villanueva-Jorge S, Chan-Gasca M, Gómez-Carballo J, López-Ochoa L, Jiménez-Delgadillo B, Rodríguez-Sánchez I, Ramírez-Prado J, Ayora-Talavera G. Temporal distribution and genetic variants in influenza A(H1N1)pdm09 virus circulating in Mexico, seasons 2012 and 2013. PLoS One 2017; 12:e0189363. [PMID: 29220381 PMCID: PMC5722308 DOI: 10.1371/journal.pone.0189363] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 11/26/2017] [Indexed: 12/15/2022] Open
Abstract
The 2012 and 2013 annual influenza epidemics in Mexico were characterized by presenting different seasonal patterns. In 2012 the A(H1N1)pdm09 virus caused a high incidence of influenza infections after a two-year period of low circulation; whereas the 2013 epidemic presented circulation of the A(H1N1)pdm09 virus throughout the year. We have characterized the molecular composition of the Hemagglutinin (HA) and Neuraminidase (NA) genes of the A(H1N1)pdm09 virus from both epidemic seasons, emphasizing the genetic characteristics of viruses isolated from Yucatan in Southern Mexico. The molecular analysis of viruses from the 2012 revealed that all viruses from Mexico were predominantly grouped in clade 7. Strikingly, the molecular characterization of viruses from 2013 revealed that viruses circulating in Yucatan were genetically different to viruses from other regions of Mexico. In fact, we identified the occurrence of two genetic variants containing relevant mutations at both the HA and NA surface antigens. There was a difference on the temporal circulation of each genetic variant, viruses containing the mutations HA-A141T / NA-N341S were detected in May, June and July; whereas viruses containing the mutations HA-S162I / NA-L206S circulated in August and September. We discuss the significance of these novel genetic changes.
Collapse
Affiliation(s)
- Jose Reyes Canche-Pech
- Universidad Autonoma de Yucatan. Centro de Investigaciones Regionales Dr.Hideyo Noguchi. Av. Centro. C.P. Merida, Yucatan, Mexico
| | - Laura Conde-Ferraez
- Universidad Autonoma de Yucatan. Centro de Investigaciones Regionales Dr.Hideyo Noguchi. Av. Centro. C.P. Merida, Yucatan, Mexico
| | - Marylin Puerto-Solis
- Universidad Autonoma de Yucatan. Centro de Investigaciones Regionales Dr.Hideyo Noguchi. Av. Centro. C.P. Merida, Yucatan, Mexico
| | - Refugio Gonzalez-Losa
- Universidad Autonoma de Yucatan. Centro de Investigaciones Regionales Dr.Hideyo Noguchi. Av. Centro. C.P. Merida, Yucatan, Mexico
| | - Pilar Granja-Pérez
- Laboratorio Estatal de Salud Publica. Servicios de Salud de Yucatan, Yucatan, México
| | | | - Maria Chan-Gasca
- Laboratorio Estatal de Salud Publica. Servicios de Salud de Yucatan, Yucatan, México
| | - Jesus Gómez-Carballo
- Universidad Autonoma de Yucatan. Centro de Investigaciones Regionales Dr.Hideyo Noguchi. Av. Centro. C.P. Merida, Yucatan, Mexico
| | - Luisa López-Ochoa
- Unidad de Bioquimica y Biologia Molecular de Plantas, Centro de Investigacion Cientifica de Yucatan, A.C., Calle, Col. Chuburna de Hidalgo, C.P. Merida, Yucatan, Mexico
| | | | - Iram Rodríguez-Sánchez
- Departamento de Genética, Facultad de Medicina, Universidad Autonoma de Nuevo Leon. Av. Gonzalitos s/n cruce con Av. Madero. Col. Mitras Centro. C.P. Monterrey, Nuevo Leon, Mexico
| | | | - Guadalupe Ayora-Talavera
- Universidad Autonoma de Yucatan. Centro de Investigaciones Regionales Dr.Hideyo Noguchi. Av. Centro. C.P. Merida, Yucatan, Mexico
| |
Collapse
|
24
|
|
25
|
Seleka M, Treurnicht FK, Tempia S, Hellferscee O, Mtshali S, Cohen AL, Buys A, McAnerney JM, Besselaar TG, Pretorius M, von Gottberg A, Walaza S, Cohen C, Madhi SA, Venter M. Epidemiology of influenza B/Yamagata and B/Victoria lineages in South Africa, 2005-2014. PLoS One 2017; 12:e0177655. [PMID: 28542324 PMCID: PMC5444647 DOI: 10.1371/journal.pone.0177655] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 05/01/2017] [Indexed: 12/29/2022] Open
Abstract
Background Studies describing the epidemiology of influenza B lineages in South Africa are lacking. Methods We conducted a prospective study to describe the circulation of influenza B/Victoria and B/Yamagata lineages among patients of all ages enrolled in South Africa through three respiratory illness surveillance systems between 2005 and 2014: (i) the Viral Watch (VW) program enrolled outpatients with influenza-like illness (ILI) from private healthcare facilities during 2005–2014; (ii) the influenza-like illnesses program enrolled outpatients in public healthcare clinics (ILI/PHC) during 2012–2014; and (iii) the severe acute respiratory illnesses (SARI) program enrolled inpatients from public hospitals during 2009–2014. Influenza B viruses were detected by virus isolation during 2005 to 2009 and by real-time reverse transcription polymerase chain reaction from 2009–2014. Clinical and epidemiological characteristics of patients hospitalized with SARI and infected with different influenza B lineages were also compared using unconditional logistic regression. Results Influenza viruses were detected in 22% (8,706/39,804) of specimens from patients with ILI or SARI during 2005–2014, of which 24% (2,087) were positive for influenza B. Influenza B viruses predominated in all three surveillance systems in 2010. B/Victoria predominated prior to 2011 (except 2008) whereas B/Yamagata predominated thereafter (except 2012). B lineages co-circulated in all seasons, except in 2013 and 2014 for SARI and ILI/PHC surveillance. Among influenza B-positive SARI cases, the detection of influenza B/Yamagata compared to influenza B/Victoria was significantly higher in individuals aged 45–64 years (adjusted odds ratio [aOR]: 4.2; 95% confidence interval [CI]: 1.1–16.5) and ≥65 years (aOR: 12.2; 95% CI: 2.3–64.4) compared to children aged 0–4 years, but was significantly lower in HIV-infected patients (aOR: 0.4; 95% CI: 0.2–0.9). Conclusion B lineages co-circulated in most seasons except in 2013 and 2014. Hospitalized SARI cases display differential susceptibility for the two influenza B lineages, with B/Victoria being more prevalent among children and HIV-infected persons.
Collapse
Affiliation(s)
- Mpho Seleka
- Centre for Respiratory Diseases and Meningitis, National Institute for Communicable Diseases (NICD) of the National Health Laboratory Services (NHLS), Johannesburg, South Africa
| | - Florette K. Treurnicht
- Centre for Respiratory Diseases and Meningitis, National Institute for Communicable Diseases (NICD) of the National Health Laboratory Services (NHLS), Johannesburg, South Africa
- * E-mail:
| | - Stefano Tempia
- Centre for Respiratory Diseases and Meningitis, National Institute for Communicable Diseases (NICD) of the National Health Laboratory Services (NHLS), Johannesburg, South Africa
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
- Influenza Program, Centers for Disease Control and Prevention, Pretoria, South Africa
| | - Orienka Hellferscee
- Centre for Respiratory Diseases and Meningitis, National Institute for Communicable Diseases (NICD) of the National Health Laboratory Services (NHLS), Johannesburg, South Africa
- School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johanneburg, South Africa
| | - Senzo Mtshali
- Sequencing Core Facility, National Institute for Communicable Diseases (NICD) of the National Health Laboratory Services (NHLS), Johannesburg, South Africa
| | - Adam L. Cohen
- Global Influenza Program, World Health Organization (WHO), Geneva, Switzerland
| | - Amelia Buys
- Centre for Respiratory Diseases and Meningitis, National Institute for Communicable Diseases (NICD) of the National Health Laboratory Services (NHLS), Johannesburg, South Africa
| | - Johanna M. McAnerney
- Centre for Respiratory Diseases and Meningitis, National Institute for Communicable Diseases (NICD) of the National Health Laboratory Services (NHLS), Johannesburg, South Africa
| | - Terry G. Besselaar
- Global Influenza Program, World Health Organization (WHO), Geneva, Switzerland
| | - Marthi Pretorius
- Centre for Respiratory Diseases and Meningitis, National Institute for Communicable Diseases (NICD) of the National Health Laboratory Services (NHLS), Johannesburg, South Africa
| | - Anne von Gottberg
- Centre for Respiratory Diseases and Meningitis, National Institute for Communicable Diseases (NICD) of the National Health Laboratory Services (NHLS), Johannesburg, South Africa
- School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johanneburg, South Africa
| | - Sibongile Walaza
- Centre for Respiratory Diseases and Meningitis, National Institute for Communicable Diseases (NICD) of the National Health Laboratory Services (NHLS), Johannesburg, South Africa
- Medical Research Council, Respiratory and Meningeal Pathogens Research Unit, University of the Witwatersrand, Johannesburg, South Africa
| | - Cheryl Cohen
- Centre for Respiratory Diseases and Meningitis, National Institute for Communicable Diseases (NICD) of the National Health Laboratory Services (NHLS), Johannesburg, South Africa
- School of Public Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Shabir A. Madhi
- Centre for Respiratory Diseases and Meningitis, National Institute for Communicable Diseases (NICD) of the National Health Laboratory Services (NHLS), Johannesburg, South Africa
- Medical Research Council, Respiratory and Meningeal Pathogens Research Unit, University of the Witwatersrand, Johannesburg, South Africa
- Department of Science and Technology/National Research Foundation: Vaccine Preventable Diseases, University of the Witwatersrand, Johannesburg, South Africa
| | - Marietjie Venter
- Zoonoses Research Unit, Department of Medical Virology, University of Pretoria, Pretoria, South Africa
| |
Collapse
|
26
|
Influenza A(H1N1)pdm 2009 and influenza B virus co-infection in hospitalized and non-hospitalized patients during the 2015–2016 epidemic season in Israel. J Clin Virol 2017; 88:12-16. [DOI: 10.1016/j.jcv.2017.01.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2016] [Revised: 11/24/2016] [Accepted: 01/03/2017] [Indexed: 01/23/2023]
|
27
|
Gonzalez-Reiche AS, Ramírez AL, Müller ML, Orellana D, Sosa SM, Ola P, Paniagua J, Ortíz L, Hernandez J, Cordón-Rosales C, Perez DR. Origin, distribution, and potential risk factors associated with influenza A virus in swine in two production systems in Guatemala. Influenza Other Respir Viruses 2017; 11:182-192. [PMID: 27860313 PMCID: PMC5304577 DOI: 10.1111/irv.12437] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/08/2016] [Indexed: 01/02/2023] Open
Abstract
Background Guatemala is the country with the largest swine production in Central America; however, evidence of influenza A virus (IAV) in pigs has not been clearly delineated. Objectives In this study, we analyzed the presence and spatial distribution of IAV in commercial and backyard swine populations. Methods Samples from two nationwide surveys conducted in 2010 and 2011 were tested using virological (rRT‐PCR and virus isolation) and serological (ELISA and hemagglutination inhibition) assays to detect IAV. Results Influenza A virus was detected in 15.7% of the sampled pigs (30.6% of herds) in 2010 and in 11.7% (24.2% of herds) in 2011. The percentage of seropositive pigs was 10.6% (16.1% of herds) and 1.4% (3.1% of herds) for each year, respectively. Three pandemic H1N1 and one seasonal human‐like H3N2 viruses were isolated. Antibodies against viruses from different genetic clusters were detected. No reassortant strains with swine viruses were detected. The H3N2 virus was closely related to human viruses that circulated in Central America in 2010, distinct to the most recent human seasonal vaccine lineages. Spatial clusters of rRT‐PCR positive herds were detected each year by scan statistics. Conclusions Our results demonstrate circulation of IAV throughout Guatemala and identify commercial farms, animal health status, and age as potential risk factors associated with IAV infection and exposure. Detection of human‐origin viruses in pigs suggests a role for humans in the molecular epidemiology of IAV in swine in Guatemala and evidences gaps in local animal and human surveillance.
Collapse
Affiliation(s)
- Ana S Gonzalez-Reiche
- Department of Population Health, Poultry Diagnostic and Research Center Athens, University of Georgia, Athens, GA, USA.,Universidad del Valle de Guatemala, Guatemala City, Guatemala
| | - Ana L Ramírez
- Universidad del Valle de Guatemala, Guatemala City, Guatemala.,Prince Leopold Institute of Tropical Medicine, Antwerp, Belgium
| | - María L Müller
- Department of Population Health, Poultry Diagnostic and Research Center Athens, University of Georgia, Athens, GA, USA.,Universidad del Valle de Guatemala, Guatemala City, Guatemala
| | - David Orellana
- Ministerio de Agricultura Ganadería y Alimentación, Guatemala city, Guatemala
| | - Silvia M Sosa
- Universidad del Valle de Guatemala, Guatemala City, Guatemala
| | - Pablo Ola
- Ministerio de Agricultura Ganadería y Alimentación, Guatemala city, Guatemala
| | - Jorge Paniagua
- Universidad del Valle de Guatemala, Guatemala City, Guatemala
| | - Lucía Ortíz
- Universidad del Valle de Guatemala, Guatemala City, Guatemala
| | | | | | - Daniel R Perez
- Department of Population Health, Poultry Diagnostic and Research Center Athens, University of Georgia, Athens, GA, USA
| |
Collapse
|
28
|
Cao P, Wong CM, Chan KH, Wang X, Chan KP, Peiris JSM, Poon LLM, Yang L. Age-specific genetic and antigenic variations of influenza A viruses in Hong Kong, 2013-2014. Sci Rep 2016; 6:30260. [PMID: 27453320 PMCID: PMC4958999 DOI: 10.1038/srep30260] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 07/04/2016] [Indexed: 11/18/2022] Open
Abstract
Age-specific genetic and antigenic variations of influenza viruses have not been documented in tropical and subtropical regions. We implemented a systematic surveillance program in two tertiary hospitals in Hong Kong Island, to collect 112 A(H1N1)pdm09 and 254 A(H3N2) positive specimens from 2013 to 2014. Of these, 56 and 72 were identified as genetic variants of the WHO recommended vaccine composition strains, respectively. A subset of these genetic variants was selected for hemagglutination-inhibition (HI) tests, but none appeared to be antigenic variants of the vaccine composition strains. We also found that genetic and antigenicity variations were similar across sex and age groups of ≤18 yrs, 18 to 65 yrs, and ≥65 yrs. Our findings suggest that none of the age groups led other age groups in genetic evolution of influenza virus A strains. Future studies from different regions and longer study periods are needed to further investigate the age and sex heterogeneity of influenza viruses.
Collapse
Affiliation(s)
- Peihua Cao
- Division of Epidemiology and Biostatistics, School of Public Health, The University of Hong Kong, Hong Kong SAR, China
| | - Chit-Ming Wong
- Division of Epidemiology and Biostatistics, School of Public Health, The University of Hong Kong, Hong Kong SAR, China
| | - Kwok-Hung Chan
- Department of Microbiology, The University of Hong Kong, Hong Kong SAR, China
| | - Xiling Wang
- Department of Biostatistics, School of Public Health and Key Laboratory of Public Health Safety, Fudan University, Shanghai, China
| | - King-Pan Chan
- Division of Epidemiology and Biostatistics, School of Public Health, The University of Hong Kong, Hong Kong SAR, China
| | - Joseph Sriyal Malik Peiris
- Division of Public Health Laboratory Sciences, School of Public Health, The University of Hong Kong, Hong Kong SAR, China
| | - Leo Lit-Man Poon
- Division of Public Health Laboratory Sciences, School of Public Health, The University of Hong Kong, Hong Kong SAR, China
| | - Lin Yang
- School of Nursing, The Hong Kong Polytechnic University, Hong Kong SAR, China
| |
Collapse
|
29
|
Petrie JG, Ohmit SE, Cheng CK, Martin ET, Malosh RE, Lauring AS, Lamerato LE, Reyes KC, Flannery B, Ferdinands JM, Monto AS. Influenza Vaccine Effectiveness Against Antigenically Drifted Influenza Higher Than Expected in Hospitalized Adults: 2014-2015. Clin Infect Dis 2016; 63:1017-25. [PMID: 27369320 DOI: 10.1093/cid/ciw432] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 06/21/2016] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND The 2014-2015 influenza season was severe, with circulating influenza A (H3N2) viruses that were antigenically drifted from the vaccine virus. Reported vaccine effectiveness (VE) estimates from ambulatory care settings were markedly decreased. METHODS Adults, hospitalized at 2 hospitals in southeast Michigan for acute respiratory illnesses, defined by admission diagnoses, of ≤10 days duration were prospectively enrolled. Throat and nasal swab specimens were collected, combined, and tested for influenza by real-time reverse transcription polymerase chain reaction. VE was estimated by comparing the vaccination status of those testing positive for influenza with those testing negative in logistic regression models adjusted for age, sex, hospital, calendar time, time from illness onset to specimen collection, frailty score, and Charlson comorbidity index (CCI). RESULTS Among 624 patients included in the analysis, 421 (68%) were vaccinated, 337 (54%) were female, 220 (35%) were age ≥65 years, and 92% had CCI > 0, indicating ≥1 comorbid conditions. Ninety-eight (16%) patients tested positive for influenza A (H3N2); among 60 (61%) A (H3N2) viruses tested by pyrosequencing, 53 (88%) belonged to the drifted 3C.2a genetic group. Adjusted VE was 43% (95% confidence interval [CI], 4-67) against influenza A (H3N2); 40% (95% CI, -13 to 68) for those <65 years, and 48% (95% CI, -33 to 80) for those ≥65 years. Sensitivity analyses largely supported these estimates. CONCLUSIONS VE estimates appeared higher than reports from similar studies in ambulatory care settings, suggesting that the 2014-2015 vaccine may have been more effective in preventing severe illness requiring hospitalization.
Collapse
Affiliation(s)
- Joshua G Petrie
- Department of Epidemiology, University of Michigan, School of Public Health
| | - Suzanne E Ohmit
- Department of Epidemiology, University of Michigan, School of Public Health
| | - Caroline K Cheng
- Department of Epidemiology, University of Michigan, School of Public Health
| | - Emily T Martin
- Department of Epidemiology, University of Michigan, School of Public Health
| | - Ryan E Malosh
- Department of Epidemiology, University of Michigan, School of Public Health
| | - Adam S Lauring
- Department of Microbiology and Immunology Department of Internal Medicine, Division of Infectious Diseases, University of Michigan, Ann Arbor
| | | | - Katherine C Reyes
- Department of Medicine, Division of Infectious Diseases, Henry Ford Health System, Detroit, Michigan
| | - Brendan Flannery
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Jill M Ferdinands
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Arnold S Monto
- Department of Epidemiology, University of Michigan, School of Public Health
| |
Collapse
|
30
|
Li C, Hatta M, Burke DF, Ping J, Zhang Y, Ozawa M, Taft AS, Das SC, Hanson AP, Song J, Imai M, Wilker PR, Watanabe T, Watanabe S, Ito M, Iwatsuki-Horimoto K, Russell CA, James SL, Skepner E, Maher EA, Neumann G, Klimov AI, Kelso A, McCauley J, Wang D, Shu Y, Odagiri T, Tashiro M, Xu X, Wentworth DE, Katz JM, Cox NJ, Smith DJ, Kawaoka Y. Selection of antigenically advanced variants of seasonal influenza viruses. Nat Microbiol 2016; 1:16058. [PMID: 27572841 PMCID: PMC5087998 DOI: 10.1038/nmicrobiol.2016.58] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 03/30/2016] [Indexed: 11/21/2022]
Abstract
Influenza viruses mutate frequently, necessitating constant updates of vaccine viruses. To establish experimental approaches that may complement the current vaccine strain selection process, we selected antigenic variants from human H1N1 and H3N2 influenza virus libraries possessing random mutations in the globular head of the haemagglutinin protein (which includes the antigenic sites) by incubating them with human and/or ferret convalescent sera to human H1N1 and H3N2 viruses. We also selected antigenic escape variants from human viruses treated with convalescent sera and from mice that had been previously immunized against human influenza viruses. Our pilot studies with past influenza viruses identified escape mutants that were antigenically similar to variants that emerged in nature, establishing the feasibility of our approach. Our studies with contemporary human influenza viruses identified escape mutants before they caused an epidemic in 2014-2015. This approach may aid in the prediction of potential antigenic escape variants and the selection of future vaccine candidates before they become widespread in nature.
Collapse
MESH Headings
- Amino Acid Substitution
- Animals
- Antigenic Variation
- Antigens, Viral/genetics
- Antigens, Viral/immunology
- Evolution, Molecular
- Ferrets/immunology
- Hemagglutinin Glycoproteins, Influenza Virus/chemistry
- Hemagglutinin Glycoproteins, Influenza Virus/genetics
- Hemagglutinin Glycoproteins, Influenza Virus/immunology
- Humans
- Immune Evasion
- Influenza A Virus, H1N1 Subtype/genetics
- Influenza A Virus, H1N1 Subtype/immunology
- Influenza A Virus, H3N2 Subtype/genetics
- Influenza A Virus, H3N2 Subtype/immunology
- Influenza Vaccines/genetics
- Influenza Vaccines/immunology
- Influenza, Human/epidemiology
- Influenza, Human/prevention & control
- Mice
- Orthomyxoviridae Infections/prevention & control
- Seasons
Collapse
Affiliation(s)
- Chengjun Li
- Department of Pathobiological Sciences, Influenza Research Institute, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, 53711 Wisconsin USA
| | - Masato Hatta
- Department of Pathobiological Sciences, Influenza Research Institute, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, 53711 Wisconsin USA
| | - David F. Burke
- Department of Zoology, University of Cambridge, Downing Street, Cambridge CB2 3EJ, UK
- World Health Organization Collaborating Centre for Modelling, Evolution, and Control of Emerging Infectious Diseases, Cambridge CB2 3EJ, UK
| | - Jihui Ping
- Department of Pathobiological Sciences, Influenza Research Institute, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, 53711 Wisconsin USA
| | - Ying Zhang
- Department of Pathobiological Sciences, Influenza Research Institute, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, 53711 Wisconsin USA
| | - Makoto Ozawa
- Department of Pathobiological Sciences, Influenza Research Institute, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, 53711 Wisconsin USA
- Department of Special Pathogens, International Research Center for Infectious Diseases, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
| | - Andrew S. Taft
- Department of Pathobiological Sciences, Influenza Research Institute, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, 53711 Wisconsin USA
| | - Subash C. Das
- Department of Pathobiological Sciences, Influenza Research Institute, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, 53711 Wisconsin USA
| | - Anthony P. Hanson
- Department of Pathobiological Sciences, Influenza Research Institute, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, 53711 Wisconsin USA
| | - Jiasheng Song
- Department of Pathobiological Sciences, Influenza Research Institute, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, 53711 Wisconsin USA
| | - Masaki Imai
- Department of Pathobiological Sciences, Influenza Research Institute, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, 53711 Wisconsin USA
- Department of Veterinary Medicine, Faculty of Agriculture, Iwate University, Iwate 020-8550, Japan
| | - Peter R. Wilker
- Department of Pathobiological Sciences, Influenza Research Institute, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, 53711 Wisconsin USA
| | - Tokiko Watanabe
- ERATO Infection-Induced Host Responses Project, Saitama 332-0012, Japan
| | - Shinji Watanabe
- ERATO Infection-Induced Host Responses Project, Saitama 332-0012, Japan
| | - Mutsumi Ito
- Division of Virology, Department of Microbiology and Immunology, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
| | - Kiyoko Iwatsuki-Horimoto
- Division of Virology, Department of Microbiology and Immunology, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
| | - Colin A. Russell
- World Health Organization Collaborating Centre for Modelling, Evolution, and Control of Emerging Infectious Diseases, Cambridge CB2 3EJ, UK
- Fogarty International Center, National Institutes of Health, Bethesda, 20892 Maryland USA
- Department of Veterinary Medicine, University of Cambridge, Cambridge CB3 0ES, UK
| | - Sarah L. James
- Department of Zoology, University of Cambridge, Downing Street, Cambridge CB2 3EJ, UK
- World Health Organization Collaborating Centre for Modelling, Evolution, and Control of Emerging Infectious Diseases, Cambridge CB2 3EJ, UK
| | - Eugene Skepner
- Department of Zoology, University of Cambridge, Downing Street, Cambridge CB2 3EJ, UK
- World Health Organization Collaborating Centre for Modelling, Evolution, and Control of Emerging Infectious Diseases, Cambridge CB2 3EJ, UK
| | - Eileen A. Maher
- Department of Pathobiological Sciences, Influenza Research Institute, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, 53711 Wisconsin USA
| | - Gabriele Neumann
- Department of Pathobiological Sciences, Influenza Research Institute, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, 53711 Wisconsin USA
| | - Alexander I. Klimov
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, 30033 Georgia USA
| | - Anne Kelso
- WHO Collaborating Centre for Reference and Research on Influenza (VIDRL) at the Peter Doherty Institute for Infection and Immunity, Melbourne, 3000 Victoria Australia
| | - John McCauley
- Division of Virology, MRC National Institute for Medical Research, The Ridgeway, Mill Hill, London NW7 1AA, UK
| | - Dayan Wang
- Chinese National Influenza Center, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Yuelong Shu
- Chinese National Influenza Center, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Takato Odagiri
- Influenza Virus Research Center, National Institute of Infectious Diseases, Musashi-Murayama, 208-0011 Tokyo Japan
| | - Masato Tashiro
- Influenza Virus Research Center, National Institute of Infectious Diseases, Musashi-Murayama, 208-0011 Tokyo Japan
| | - Xiyan Xu
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, 30033 Georgia USA
| | - David E. Wentworth
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, 30033 Georgia USA
| | - Jacqueline M. Katz
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, 30033 Georgia USA
| | - Nancy J. Cox
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, 30033 Georgia USA
| | - Derek J. Smith
- Department of Zoology, University of Cambridge, Downing Street, Cambridge CB2 3EJ, UK
- World Health Organization Collaborating Centre for Modelling, Evolution, and Control of Emerging Infectious Diseases, Cambridge CB2 3EJ, UK
- Department of Virology, Erasmus Medical Center, Rotterdam 3000 CA, Netherlands
| | - Yoshihiro Kawaoka
- Department of Pathobiological Sciences, Influenza Research Institute, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, 53711 Wisconsin USA
- Department of Special Pathogens, International Research Center for Infectious Diseases, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
- ERATO Infection-Induced Host Responses Project, Saitama 332-0012, Japan
- Division of Virology, Department of Microbiology and Immunology, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
| |
Collapse
|
31
|
Flannery B, Zimmerman RK, Gubareva LV, Garten RJ, Chung JR, Nowalk MP, Jackson ML, Jackson LA, Monto AS, Ohmit SE, Belongia EA, McLean HQ, Gaglani M, Piedra PA, Mishin VP, Chesnokov AP, Spencer S, Thaker SN, Barnes JR, Foust A, Sessions W, Xu X, Katz J, Fry AM. Enhanced Genetic Characterization of Influenza A(H3N2) Viruses and Vaccine Effectiveness by Genetic Group, 2014-2015. J Infect Dis 2016; 214:1010-9. [PMID: 27190176 DOI: 10.1093/infdis/jiw181] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 03/22/2016] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND During the 2014-2015 US influenza season, expanded genetic characterization of circulating influenza A(H3N2) viruses was used to assess the impact of the genetic variability of influenza A(H3N2) viruses on influenza vaccine effectiveness (VE). METHODS A novel pyrosequencing assay was used to determine genetic group, based on hemagglutinin (HA) gene sequences, of influenza A(H3N2) viruses from patients enrolled at US Influenza Vaccine Effectiveness Network sites. VE was estimated using a test-negative design comparing vaccination among patients infected with influenza A(H3N2) viruses and uninfected patients. RESULTS Among 9710 enrollees, 1868 (19%) tested positive for influenza A(H3N2) virus; genetic characterization of 1397 viruses showed that 1134 (81%) belonged to 1 HA genetic group (3C.2a) of antigenically drifted influenza A(H3N2) viruses. Effectiveness of 2014-2015 influenza vaccination varied by influenza A(H3N2) virus genetic group from 1% (95% confidence interval [CI], -14% to 14%) against illness caused by antigenically drifted influenza A(H3N2) virus group 3C.2a viruses versus 44% (95% CI, 16%-63%) against illness caused by vaccine-like influenza A(H3N2) virus group 3C.3b viruses. CONCLUSIONS Effectiveness of 2014-2015 influenza vaccination varied by genetic group of influenza A(H3N2) virus. Changes in HA genes related to antigenic drift were associated with reduced VE.
Collapse
Affiliation(s)
| | | | | | | | - Jessie R Chung
- Influenza Division, Centers for Disease Control and Prevention Atlanta Research and Education Foundation, Georgia
| | | | | | | | - Arnold S Monto
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor
| | - Suzanne E Ohmit
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor
| | | | | | - Manjusha Gaglani
- Baylor Scott & White Health, Texas A&M Health Science Center College of Medicine, Temple
| | - Pedro A Piedra
- Department of Molecular Virology and Microbiology, and Pediatrics, Baylor College of Medicine, Houston, Texas
| | | | | | - Sarah Spencer
- Influenza Division, Centers for Disease Control and Prevention Atlanta Research and Education Foundation, Georgia
| | - Swathi N Thaker
- Influenza Division, Centers for Disease Control and Prevention
| | - John R Barnes
- Influenza Division, Centers for Disease Control and Prevention
| | - Angie Foust
- Influenza Division, Centers for Disease Control and Prevention
| | - Wendy Sessions
- Influenza Division, Centers for Disease Control and Prevention
| | - Xiyan Xu
- Influenza Division, Centers for Disease Control and Prevention
| | - Jacqueline Katz
- Influenza Division, Centers for Disease Control and Prevention
| | - Alicia M Fry
- Influenza Division, Centers for Disease Control and Prevention
| |
Collapse
|
32
|
Harvey WT, Benton DJ, Gregory V, Hall JPJ, Daniels RS, Bedford T, Haydon DT, Hay AJ, McCauley JW, Reeve R. Identification of Low- and High-Impact Hemagglutinin Amino Acid Substitutions That Drive Antigenic Drift of Influenza A(H1N1) Viruses. PLoS Pathog 2016; 12:e1005526. [PMID: 27057693 PMCID: PMC4825936 DOI: 10.1371/journal.ppat.1005526] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 03/04/2016] [Indexed: 12/20/2022] Open
Abstract
Determining phenotype from genetic data is a fundamental challenge. Identification of emerging antigenic variants among circulating influenza viruses is critical to the vaccine virus selection process, with vaccine effectiveness maximized when constituents are antigenically similar to circulating viruses. Hemagglutination inhibition (HI) assay data are commonly used to assess influenza antigenicity. Here, sequence and 3-D structural information of hemagglutinin (HA) glycoproteins were analyzed together with corresponding HI assay data for former seasonal influenza A(H1N1) virus isolates (1997–2009) and reference viruses. The models developed identify and quantify the impact of eighteen amino acid substitutions on the antigenicity of HA, two of which were responsible for major transitions in antigenic phenotype. We used reverse genetics to demonstrate the causal effect on antigenicity for a subset of these substitutions. Information on the impact of substitutions allowed us to predict antigenic phenotypes of emerging viruses directly from HA gene sequence data and accuracy was doubled by including all substitutions causing antigenic changes over a model incorporating only the substitutions with the largest impact. The ability to quantify the phenotypic impact of specific amino acid substitutions should help refine emerging techniques that predict the evolution of virus populations from one year to the next, leading to stronger theoretical foundations for selection of candidate vaccine viruses. These techniques have great potential to be extended to other antigenically variable pathogens. Influenza A viruses are characterized by rapid antigenic drift: structural changes in B-cell epitopes that facilitate escape from pre-existing immunity. Consequently, seasonal influenza continues to impose a major burden on human health. Accurate quantification of the antigenic impact of specific amino acid substitutions is a pre-requisite for predicting the fitness and evolutionary outcome of variant viruses. Using assays to attribute antigenic variation to amino acid sequence changes we identify substitutions that contribute to antigenic drift and quantify their impact. We show that substitutions identified as low-impact are a critical component of virus antigenic evolution and by including these, as well as the high-impact substitutions often focused on, the accuracy of predicting antigenic phenotypes of emerging viruses from genotype is doubled.
Collapse
Affiliation(s)
- William T. Harvey
- Boyd Orr Centre for Population and Ecosystem Health and Institute of Biodiversity, Animal Health and Comparative Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Donald J. Benton
- The Crick Worldwide Influenza Centre, The Francis Crick Institute, Mill Hill Laboratory, The Ridgeway, Mill Hill, London, United Kingdom (formerly WHO Collaborating Centre for Reference and Research on Influenza, Division of Virology, MRC National Institute for Medical Research, The Ridgeway, Mill Hill, London, United Kingdom)
| | - Victoria Gregory
- The Crick Worldwide Influenza Centre, The Francis Crick Institute, Mill Hill Laboratory, The Ridgeway, Mill Hill, London, United Kingdom (formerly WHO Collaborating Centre for Reference and Research on Influenza, Division of Virology, MRC National Institute for Medical Research, The Ridgeway, Mill Hill, London, United Kingdom)
| | - James P. J. Hall
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Rodney S. Daniels
- The Crick Worldwide Influenza Centre, The Francis Crick Institute, Mill Hill Laboratory, The Ridgeway, Mill Hill, London, United Kingdom (formerly WHO Collaborating Centre for Reference and Research on Influenza, Division of Virology, MRC National Institute for Medical Research, The Ridgeway, Mill Hill, London, United Kingdom)
| | - Trevor Bedford
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Daniel T. Haydon
- Boyd Orr Centre for Population and Ecosystem Health and Institute of Biodiversity, Animal Health and Comparative Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Alan J. Hay
- The Crick Worldwide Influenza Centre, The Francis Crick Institute, Mill Hill Laboratory, The Ridgeway, Mill Hill, London, United Kingdom (formerly WHO Collaborating Centre for Reference and Research on Influenza, Division of Virology, MRC National Institute for Medical Research, The Ridgeway, Mill Hill, London, United Kingdom)
| | - John W. McCauley
- The Crick Worldwide Influenza Centre, The Francis Crick Institute, Mill Hill Laboratory, The Ridgeway, Mill Hill, London, United Kingdom (formerly WHO Collaborating Centre for Reference and Research on Influenza, Division of Virology, MRC National Institute for Medical Research, The Ridgeway, Mill Hill, London, United Kingdom)
| | - Richard Reeve
- Boyd Orr Centre for Population and Ecosystem Health and Institute of Biodiversity, Animal Health and Comparative Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
- The Pirbright Institute, Pirbright, Woking, Surrey, United Kingdom
- * E-mail:
| |
Collapse
|
33
|
Mosterín Höpping A, Fonville JM, Russell CA, James S, Smith DJ. Influenza B vaccine lineage selection--an optimized trivalent vaccine. Vaccine 2016; 34:1617-1622. [PMID: 26896685 PMCID: PMC4793086 DOI: 10.1016/j.vaccine.2016.01.042] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 01/20/2016] [Indexed: 11/27/2022]
Abstract
Epidemics of seasonal influenza viruses cause considerable morbidity and mortality each year. Various types and subtypes of influenza circulate in humans and evolve continuously such that individuals at risk of serious complications need to be vaccinated annually to keep protection up to date with circulating viruses. The influenza vaccine in most parts of the world is a trivalent vaccine, including an antigenically representative virus of recently circulating influenza A/H3N2, A/H1N1, and influenza B viruses. However, since the 1970s influenza B has split into two antigenically distinct lineages, only one of which is represented in the annual trivalent vaccine at any time. We describe a lineage selection strategy that optimizes protection against influenza B using the standard trivalent vaccine as a potentially cost effective alternative to quadrivalent vaccines.
Collapse
Affiliation(s)
| | - Judith M Fonville
- Department of Zoology, University of Cambridge, Cambridge, United Kingdom; Department of Virology, Erasmus MC, Rotterdam, The Netherlands
| | - Colin A Russell
- Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Sarah James
- Department of Zoology, University of Cambridge, Cambridge, United Kingdom
| | - Derek J Smith
- Department of Zoology, University of Cambridge, Cambridge, United Kingdom; Department of Virology, Erasmus MC, Rotterdam, The Netherlands.
| |
Collapse
|
34
|
BENEDETTI E, DANIELS RS, PONTORIERO A, RUSSO M, AVARO M, CZECH A, CAMPOS A, PERIOLO N, GREGORY V, McCAULEY JW, BAUMEISTER EG. Influenza virus surveillance in Argentina during the 2012 season: antigenic characterization, genetic analysis and antiviral susceptibility. Epidemiol Infect 2016; 144:751-67. [PMID: 26345289 PMCID: PMC4762242 DOI: 10.1017/s0950268815001806] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Revised: 05/22/2015] [Accepted: 07/15/2015] [Indexed: 12/18/2022] Open
Abstract
The activity and circulation of influenza viruses in Argentina was studied during 2012 as part of the Argentinean Surveillance for Influenza and other Respiratory Viruses, in the context of Global Influenza Surveillance. The antigenicity and molecular characteristics of haemagglutinins (HA) of circulating influenza A and B viruses were analysed to assess the emergence of virus variants. Susceptibility to oseltamivir and zanamivir was evaluated by enzymatic assay and results were backed-up by sequencing of the neuraminidase (NA) genes. During the 2012 season, influenza virus circulation in Argentina was detected from weeks 24 to 51. The HA sequences of the studied A(H1N1)pdm09 subtype viruses segregated in a different genetic group compared to those identified during the 2009 pandemic, although they were still closely related antigenically to the vaccine virus A/California/07/2009. The HA sequences of the A(H3N2) viruses analysed fell into the A/Victoria/208/2009 clade, genetic group 3C. A mixed circulation of virus variants belonging to B/Victoria and B/Yamagata lineages was detected, with B/Victoria being dominant. All viruses tested were sensitive to oseltamivir and zanamivir except one. This isolate, an A(H1N1)pdm09 virus possessing the substitution NA-N295S, showed highly reduced inhibition by oseltamivir and reduced inhibition by zanamivir. Virological and epidemiological surveillance remains critical for detection of evolving influenza viruses.
Collapse
Affiliation(s)
- E. BENEDETTI
- National Influenza Centre PAHO/WHO, Servicio Virosis Respiratorias, Departamento Virología, Instituto Nacional de Enfermedades Infecciosas, ANLIS ‘Carlos G. Malbrán’, Buenos Aires, Argentina
| | - R. S. DANIELS
- The Francis Crick Institute, Mill Hill Laboratory, The Ridgeway, Mill Hill, London, UK
| | - A. PONTORIERO
- National Influenza Centre PAHO/WHO, Servicio Virosis Respiratorias, Departamento Virología, Instituto Nacional de Enfermedades Infecciosas, ANLIS ‘Carlos G. Malbrán’, Buenos Aires, Argentina
| | - M. RUSSO
- National Influenza Centre PAHO/WHO, Servicio Virosis Respiratorias, Departamento Virología, Instituto Nacional de Enfermedades Infecciosas, ANLIS ‘Carlos G. Malbrán’, Buenos Aires, Argentina
| | - M. AVARO
- National Influenza Centre PAHO/WHO, Servicio Virosis Respiratorias, Departamento Virología, Instituto Nacional de Enfermedades Infecciosas, ANLIS ‘Carlos G. Malbrán’, Buenos Aires, Argentina
| | - A. CZECH
- National Influenza Centre PAHO/WHO, Servicio Virosis Respiratorias, Departamento Virología, Instituto Nacional de Enfermedades Infecciosas, ANLIS ‘Carlos G. Malbrán’, Buenos Aires, Argentina
| | - A. CAMPOS
- National Influenza Centre PAHO/WHO, Servicio Virosis Respiratorias, Departamento Virología, Instituto Nacional de Enfermedades Infecciosas, ANLIS ‘Carlos G. Malbrán’, Buenos Aires, Argentina
| | - N. PERIOLO
- National Influenza Centre PAHO/WHO, Servicio Virosis Respiratorias, Departamento Virología, Instituto Nacional de Enfermedades Infecciosas, ANLIS ‘Carlos G. Malbrán’, Buenos Aires, Argentina
| | - V. GREGORY
- The Francis Crick Institute, Mill Hill Laboratory, The Ridgeway, Mill Hill, London, UK
| | - J. W. McCAULEY
- The Francis Crick Institute, Mill Hill Laboratory, The Ridgeway, Mill Hill, London, UK
| | - E. G. BAUMEISTER
- National Influenza Centre PAHO/WHO, Servicio Virosis Respiratorias, Departamento Virología, Instituto Nacional de Enfermedades Infecciosas, ANLIS ‘Carlos G. Malbrán’, Buenos Aires, Argentina
| |
Collapse
|
35
|
Matos-Patrón A, Byrd-Leotis L, Steinhauer DA, Barclay WS, Ayora-Talavera G. Amino acid substitution D222N from fatal influenza infection affects receptor-binding properties of the influenza A(H1N1)pdm09 virus. Virology 2015; 484:15-21. [DOI: 10.1016/j.virol.2015.05.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2015] [Revised: 03/15/2015] [Accepted: 05/12/2015] [Indexed: 12/12/2022]
|
36
|
Nguyen HKL, Nguyen PTK, Nguyen TC, Hoang PVM, Le TT, Vuong CD, Nguyen AP, Tran LTT, Nguyen BG, Lê MQ. Virological characterization of influenza H1N1pdm09 in Vietnam, 2010-2013. Influenza Other Respir Viruses 2015; 9:216-24. [PMID: 25966032 PMCID: PMC4474498 DOI: 10.1111/irv.12323] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/30/2015] [Indexed: 11/26/2022] Open
Abstract
OBJECTIVES Influenza A/H1N1pdm09 virus was first detected in Vietnam on May 31, 2009, and continues to circulate in Vietnam as a seasonal influenza virus. This study has monitored genotypic and phenotypic changes in this group of viruses during 2010-2013 period. DESIGN AND SETTING We sequenced hemagglutinin (HA) and neuraminidase (NA) genes from representative influenza A/H1N1pdm09 and compared with vaccine strain A/California/07/09 and other contemporary isolates from neighboring countries. Hemagglutination inhibition (HI) and neuraminidase inhibition (NAI) assays also were performed on these isolates. SAMPLE Representative influenza A/H1N1pdm09 isolates (n = 61) from ILI and SARI surveillances in northern Vietnam between 2010 and 2013. MAIN OUTCOME MEASURES AND RESULTS The HA and NA phylogenies revealed six and seven groups, respectively. Five isolates (8·2%) had substitutions G155E and N156K in the HA, which were associated with reduced HI titers by antiserum raised against the vaccine virus A/California/07/2009. One isolate from 2011 and one isolate from 2013 had a predicted H275Y substitution in the neuraminidase molecule, which was associated with reduced susceptibility to oseltamivir in a NAI assay. We also identified a D222N change in the HA of a virus isolated from a fatal case in 2013. CONCLUSIONS Significant genotypic and phenotypic changes in A/ H1N1pdm09 influenza viruses were detected by the National Influenza Surveillance System (NISS) in Vietnam between 2010 and 2013 highlighting the value of this system to Vietnam and to the region. Sustained NISS and continued virological monitoring of seasonal influenza viruses are required for vaccine policy development in Vietnam. 3.
Collapse
Affiliation(s)
- Hang K L Nguyen
- National Institute of Hygiene and EpidemiologyHanoi, Vietnam
| | | | - Thach C Nguyen
- National Institute of Hygiene and EpidemiologyHanoi, Vietnam
| | | | - Thanh T Le
- National Institute of Hygiene and EpidemiologyHanoi, Vietnam
| | - Cuong D Vuong
- National Institute of Hygiene and EpidemiologyHanoi, Vietnam
| | - Anh P Nguyen
- National Institute of Hygiene and EpidemiologyHanoi, Vietnam
| | - Loan T T Tran
- National Hospital of Traditional MedicineHanoi, Vietnam
| | | | - Mai Q Lê
- National Institute of Hygiene and EpidemiologyHanoi, Vietnam
| |
Collapse
|
37
|
Lee HK, Tang JWT, Loh TP, Oon LLE, Koay ESC. Predicting clinical severity based on substitutions near epitope A of influenza A/H3N2. INFECTION GENETICS AND EVOLUTION 2015; 34:292-7. [PMID: 26118307 DOI: 10.1016/j.meegid.2015.06.025] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Revised: 06/15/2015] [Accepted: 06/25/2015] [Indexed: 01/03/2023]
Abstract
Epitopes are the main targets for specific antibodies in the host defense systems. Recent studies have shown that amino acid (aa) substitutions located within the influenza A/H3N2 hemagglutinin 1 (HA1) epitopes A-E, particularly in A and B, result in antigenic drift. Viruses with such drift mutations may have resulted in more severe influenza-related illness during influenza epidemics between late 2012 and early 2015. We sought to quantify vaccine mismatches in epitopes A-E of the HA1 protein, and correlate these with the severity of the patient's illness. The influenza A/H3N2 clinical samples were collected between April 2009 and November 2013 (n=206). Patients were clinically stratified into groups with mild, moderate, and severe influenza-like illness (ILI). The impact of the number of aa mismatches in each of epitopes A-E, gender, age groups (⩽18, 19-64, ⩾65 years), and comorbidities on the likelihood that patients would suffer moderate and/or severe ILI due to influenza A/H3N2 infection were assessed. A higher number of aa mismatches in epitope A between the vaccine and locally circulating viruses correlated with more severe influenza infection, although this correlation was most significant with pre-existing comorbidities. A practical application of this finding would be to monitor patients (especially those in high-risk groups) infected with such viruses more closely, as they are at increased risk of developing more serious disease. Epidemiologically, it was of interest to note that viruses from subclade 3A of Victoria/208 strain were not detected in Singapore between 2009 and 2012. By contrast, these viruses were detected at a prevalence of up to 40% in the 2011-2012 influenza seasons in other regions of the Northern and Southern hemispheres. Such findings support the rationale for more regionally customized seasonal influenza vaccine compositions to optimize the protection of the population against locally circulating virus strains.
Collapse
Affiliation(s)
- Hong Kai Lee
- Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Department of Laboratory Medicine, National University Hospital, National University Health System, Singapore
| | - Julian Wei-Tze Tang
- Clinical Microbiology, University Hospitals of Leicester NHS Trust, Leicester, United Kingdom.
| | - Tze Ping Loh
- Department of Laboratory Medicine, National University Hospital, National University Health System, Singapore
| | | | - Evelyn Siew-Chuan Koay
- Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Department of Laboratory Medicine, National University Hospital, National University Health System, Singapore.
| |
Collapse
|
38
|
New genetic variants of influenza A(H1N1)pdm09 detected in Cuba during 2011-2013. INFECTION GENETICS AND EVOLUTION 2015; 32:322-6. [PMID: 25839842 DOI: 10.1016/j.meegid.2015.03.028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Revised: 03/23/2015] [Accepted: 03/24/2015] [Indexed: 11/20/2022]
Abstract
Influenza A(H1N1)pdm09 virus has evolved continually since its emergence in 2009. For influenza virus strains, genetic changes occurring in HA1 domain of the hemagglutinin cause the emergence of new variants. The aim of our study is to establish genetic associations between 35 A(H1N1)pdm09 viruses circulating in Cuba in 2011-2012 and 2012-2013 seasons, and A/California/07/2009 strain recommended by WHO as the H1N1 component of the influenza vaccine. The phylogenetic analysis revealed the circulation of clades 3, 6A, 6B, 6C and 7. Mutations were detected in the antigenic site or in the receptor-binding domains of HA1 segment, including S174P, S179N, K180Q, S202T, S220T and R222K. Substitutions S174P, S179N, K180Q and R222K were detected in Cuban strains for the first time.
Collapse
|
39
|
Kelly HA. Safety and effectiveness of influenza vaccines. Med J Aust 2015; 201:560-1. [PMID: 25390245 DOI: 10.5694/mja14.01281] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Accepted: 10/07/2014] [Indexed: 11/17/2022]
Affiliation(s)
- Heath A Kelly
- Victorian Infectious Diseases Reference Laboratory, Peter Doherty Institute, Melbourne, VIC, Australia.
| |
Collapse
|
40
|
Lee HK, Tang JWT, Loh TP, Hurt AC, Oon LLE, Koay ESC. Molecular surveillance of antiviral drug resistance of influenza A/H3N2 virus in Singapore, 2009-2013. PLoS One 2015; 10:e0117822. [PMID: 25635767 PMCID: PMC4311985 DOI: 10.1371/journal.pone.0117822] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2014] [Accepted: 01/02/2015] [Indexed: 01/10/2023] Open
Abstract
Adamantanes and neuraminidase inhibitors (NAIs) are two classes of antiviral drugs available for the chemoprophylaxis and treatment of influenza infections. To determine the frequency of drug resistance in influenza A/H3N2 viruses in Singapore, large-scale sequencing of neuraminidase (NA) and matrix protein (MP) genes was performed directly without initial culture amplification. 241 laboratory-confirmed influenza A/H3N2 clinical samples, collected between May 2009 and November 2013 were included. In total, 229 NA (95%) and 241 MP (100%) complete sequences were obtained. Drug resistance mutations in the NA and MP genes were interpreted according to published studies. For the NAIs, a visual inspection of the aligned NA sequences revealed no known drug resistant genotypes (DRGs). For the adamantanes, the well-recognised S31N DRG was identified in all 241 MP genes. In addition, there was an increasing number of viruses carrying the combination of D93G+Y155F+D251V (since May 2013) or D93G (since March 2011) mutations in the NA gene. However, in-vitro NAI testing indicated that neither D93G+Y155F+D251V nor D93G alone conferred any changes in NAI susceptibility. Lastly, an I222T mutation in the NA gene that has previously been reported to cause oseltamivir-resistance in influenza A/H1N1/2009, B, and A/H5N1, was detected from a treatment-naïve patient. Further in-vitro NAI testing is required to confirm the effect of this mutation in A/H3N2 virus.
Collapse
Affiliation(s)
- Hong Kai Lee
- Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Laboratory Medicine, National University Hospital, National University Health System, Singapore, Singapore
| | - Julian Wei-Tze Tang
- Clinical Microbiology, Leicester Royal Infirmary, Leicester, United Kingdom
- * E-mail: (JWT); (ESK)
| | - Tze Ping Loh
- Department of Laboratory Medicine, National University Hospital, National University Health System, Singapore, Singapore
| | - Aeron C. Hurt
- WHO Collaborating Centre for Reference and Research on Influenza, Melbourne, VIC, Australia
- Melbourne School of Population and Global Health, University of Melbourne, VIC, Australia
| | | | - Evelyn Siew-Chuan Koay
- Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Laboratory Medicine, National University Hospital, National University Health System, Singapore, Singapore
- * E-mail: (JWT); (ESK)
| |
Collapse
|
41
|
Vijaykrishna D, Holmes EC, Joseph U, Fourment M, Su YCF, Halpin R, Lee RTC, Deng YM, Gunalan V, Lin X, Stockwell TB, Fedorova NB, Zhou B, Spirason N, Kühnert D, Bošková V, Stadler T, Costa AM, Dwyer DE, Huang QS, Jennings LC, Rawlinson W, Sullivan SG, Hurt AC, Maurer-Stroh S, Wentworth DE, Smith GJD, Barr IG. The contrasting phylodynamics of human influenza B viruses. eLife 2015; 4:e05055. [PMID: 25594904 PMCID: PMC4383373 DOI: 10.7554/elife.05055] [Citation(s) in RCA: 154] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Accepted: 01/15/2015] [Indexed: 11/13/2022] Open
Abstract
A complex interplay of viral, host, and ecological factors shapes the spatio-temporal incidence and evolution of human influenza viruses. Although considerable attention has been paid to influenza A viruses, a lack of equivalent data means that an integrated evolutionary and epidemiological framework has until now not been available for influenza B viruses, despite their significant disease burden. Through the analysis of over 900 full genomes from an epidemiological collection of more than 26,000 strains from Australia and New Zealand, we reveal fundamental differences in the phylodynamics of the two co-circulating lineages of influenza B virus (Victoria and Yamagata), showing that their individual dynamics are determined by a complex relationship between virus transmission, age of infection, and receptor binding preference. In sum, this work identifies new factors that are important determinants of influenza B evolution and epidemiology.
Collapse
Affiliation(s)
| | - Edward C Holmes
- Marie Bashir Institute for Infectious Diseases and Biosecurity, University of Sydney, Sydney, Australia
| | - Udayan Joseph
- Duke-NUS Graduate Medical School, Singapore, Singapore
| | - Mathieu Fourment
- Marie Bashir Institute for Infectious Diseases and Biosecurity, University of Sydney, Sydney, Australia
| | - Yvonne C F Su
- Duke-NUS Graduate Medical School, Singapore, Singapore
| | | | - Raphael T C Lee
- Bioinformatics Institute, Agency for Science, Technology and Research, Singapore, Singapore
| | - Yi-Mo Deng
- World Health Organisation Collaborating Centre for Reference and Research on Influenza, Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Vithiagaran Gunalan
- Bioinformatics Institute, Agency for Science, Technology and Research, Singapore, Singapore
| | - Xudong Lin
- J Craig Venter Institute, Rockville, United States
| | | | | | - Bin Zhou
- J Craig Venter Institute, Rockville, United States
| | - Natalie Spirason
- World Health Organisation Collaborating Centre for Reference and Research on Influenza, Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Denise Kühnert
- Department of Environmental Systems Science, Eidgenössische Technische Hochschule Zürich, Zürich, Switzerland
| | - Veronika Bošková
- Department of Biosystems Science and Engineering, Eidgenössische Technische Hochschule Zürich, Zurich, Switzerland
| | - Tanja Stadler
- Department of Biosystems Science and Engineering, Eidgenössische Technische Hochschule Zürich, Zurich, Switzerland
| | | | - Dominic E Dwyer
- Centre for Infectious Diseases and Microbiology Laboratory Services, Westmead Hospital and University of Sydney, Westmead, Australia
| | - Q Sue Huang
- Institute of Environmental Science and Research, National Centre for Biosecurity and Infectious Disease, Upper Hutt, New Zealand
| | - Lance C Jennings
- Microbiology Department, Canterbury Health Laboratories, Christchurch, New Zealand
| | - William Rawlinson
- Virology Division, SEALS Microbiology, Prince of Wales Hospital, Sydney, Australia
| | - Sheena G Sullivan
- World Health Organisation Collaborating Centre for Reference and Research on Influenza, Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Aeron C Hurt
- World Health Organisation Collaborating Centre for Reference and Research on Influenza, Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Sebastian Maurer-Stroh
- Bioinformatics Institute, Agency for Science, Technology and Research, Singapore, Singapore
| | | | | | - Ian G Barr
- World Health Organisation Collaborating Centre for Reference and Research on Influenza, Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| |
Collapse
|
42
|
Zhang XS, Pebody R, De Angelis D, White PJ, Charlett A, McCauley JW. The Possible Impact of Vaccination for Seasonal Influenza on Emergence of Pandemic Influenza via Reassortment. PLoS One 2014; 9:e114637. [PMID: 25494180 PMCID: PMC4262424 DOI: 10.1371/journal.pone.0114637] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Accepted: 11/12/2014] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND One pathway through which pandemic influenza strains might emerge is reassortment from coinfection of different influenza A viruses. Seasonal influenza vaccines are designed to target the circulating strains, which intuitively decreases the prevalence of coinfection and the chance of pandemic emergence due to reassortment. However, individual-based analyses on 2009 pandemic influenza show that the previous seasonal vaccination may increase the risk of pandemic A(H1N1) pdm09 infection. In view of pandemic influenza preparedness, it is essential to understand the overall effect of seasonal vaccination on pandemic emergence via reassortment. METHODS AND FINDINGS In a previous study we applied a population dynamics approach to investigate the effect of infection-induced cross-immunity on reducing such a pandemic risk. Here the model was extended by incorporating vaccination for seasonal influenza to assess its potential role on the pandemic emergence via reassortment and its effect in protecting humans if a pandemic does emerge. The vaccination is assumed to protect against the target strains but only partially against other strains. We find that a universal seasonal vaccine that provides full-spectrum cross-immunity substantially reduces the opportunity of pandemic emergence. However, our results show that such effectiveness depends on the strength of infection-induced cross-immunity against any novel reassortant strain. If it is weak, the vaccine that induces cross-immunity strongly against non-target resident strains but weakly against novel reassortant strains, can further depress the pandemic emergence; if it is very strong, the same kind of vaccine increases the probability of pandemic emergence. CONCLUSIONS Two types of vaccines are available: inactivated and live attenuated, only live attenuated vaccines can induce heterosubtypic immunity. Current vaccines are effective in controlling circulating strains; they cannot always help restrain pandemic emergence because of the uncertainty of the oncoming reassortant strains, however. This urges the development of universal vaccines for prevention of pandemic influenza.
Collapse
Affiliation(s)
- Xu-Sheng Zhang
- Modelling and Economics Unit, Centre for Infectious Disease Surveillance and Control, Public Health England, London, United Kingdom
- Medical Research Council Centre for Outbreak Analysis and Modelling, Department of Infectious Disease Epidemiology, Imperial College School of Public Health, London, United Kingdom
- * E-mail:
| | - Richard Pebody
- Respiratory Diseases Department, Centre for Infectious Disease Surveillance and Control, Public Health England, London, United Kingdom
| | - Daniela De Angelis
- Statistics Unit, Centre for Infectious Disease Surveillance and Control, Public Health England, London, United Kingdom
- Medical Research Council Biostatistics Unit, University Forvie Site, Cambridge, United Kingdom
| | - Peter J. White
- Modelling and Economics Unit, Centre for Infectious Disease Surveillance and Control, Public Health England, London, United Kingdom
- Medical Research Council Centre for Outbreak Analysis and Modelling, Department of Infectious Disease Epidemiology, Imperial College School of Public Health, London, United Kingdom
- NIHR Health Protection Research Unit in Modelling Methodology, Department of Infectious Disease Epidemiology, Imperial College School of Public Health, London, United Kingdom
| | - Andre Charlett
- Statistics Unit, Centre for Infectious Disease Surveillance and Control, Public Health England, London, United Kingdom
| | - John W. McCauley
- Medical Research Council National Institute for Medical Research, Mill Hill, London, United Kingdom
| |
Collapse
|
43
|
Russo ML, Pontoriero AV, Benedetti E, Czech A, Avaro M, Periolo N, Campos AM, Savy VL, Baumeister EG. Antigenic and genomic characterization of human influenza A and B viruses circulating in Argentina after the introduction of influenza A(H1N1)pdm09. J Med Microbiol 2014; 63:1626-1637. [DOI: 10.1099/jmm.0.076208-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
This study was conducted as part of the Argentinean Influenza and other Respiratory Viruses Surveillance Network, in the context of the Global Influenza Surveillance carried out by the World Health Organization (WHO). The objective was to study the activity and the antigenic and genomic characteristics of circulating viruses for three consecutive seasons (2010, 2011 and 2012) in order to investigate the emergence of influenza viral variants. During the study period, influenza virus circulation was detected from January to December. Influenza A and B, and all current subtypes of human influenza viruses, were present each year. Throughout the 2010 post-pandemic season, influenza A(H1N1)pdm09, unexpectedly, almost disappeared. The haemagglutinin (HA) of the A(H1N1)pdm09 viruses studied were segregated in a different genetic group to those identified during the 2009 pandemic, although they were still antigenically closely related to the vaccine strain A/California/07/2009. Influenza A(H3N2) viruses were the predominant strains circulating during the 2011 season, accounting for nearly 76 % of influenza viruses identified. That year, all HA sequences of the A(H3N2) viruses tested fell into the A/Victoria/208/2009 genetic clade, but remained antigenically related to A/Perth/16/2009 (reference vaccine recommended for this three-year period). A(H3N2) viruses isolated in 2012 were antigenically closely related to A/Victoria/361/2011, recommended by the WHO as the H3 component for the 2013 Southern Hemisphere formulation. B viruses belonging to the B/Victoria lineage circulated in 2010. A mixed circulation of viral variants of both B/Victoria and B/Yamagata lineages was detected in 2012, with the former being predominant. A(H1N1)pdm09 viruses remained antigenically closely related to the vaccine virus A/California/7/2009; A(H3N2) viruses continually evolved into new antigenic clusters and both B lineages, B/Victoria/2/87-like and B/Yamagata/16/88-like viruses, were observed during the study period. The virological surveillance showed that the majority of the circulating strains during the study period were antigenically related to the corresponding Southern Hemisphere vaccine strains except for the 2012 A(H3N2) viruses.
Collapse
Affiliation(s)
- Mara L. Russo
- National Influenza Centre PAHO/WHO, Servicio Virosis Respiratorias, Departamento Virología, Instituto Nacional de Enfermedades Infecciosas, ANLIS ‘Carlos G. Malbrán’, Buenos Aires, Argentina
| | - Andrea V. Pontoriero
- National Influenza Centre PAHO/WHO, Servicio Virosis Respiratorias, Departamento Virología, Instituto Nacional de Enfermedades Infecciosas, ANLIS ‘Carlos G. Malbrán’, Buenos Aires, Argentina
| | - Estefania Benedetti
- National Influenza Centre PAHO/WHO, Servicio Virosis Respiratorias, Departamento Virología, Instituto Nacional de Enfermedades Infecciosas, ANLIS ‘Carlos G. Malbrán’, Buenos Aires, Argentina
| | - Andrea Czech
- National Influenza Centre PAHO/WHO, Servicio Virosis Respiratorias, Departamento Virología, Instituto Nacional de Enfermedades Infecciosas, ANLIS ‘Carlos G. Malbrán’, Buenos Aires, Argentina
| | - Martin Avaro
- National Influenza Centre PAHO/WHO, Servicio Virosis Respiratorias, Departamento Virología, Instituto Nacional de Enfermedades Infecciosas, ANLIS ‘Carlos G. Malbrán’, Buenos Aires, Argentina
| | - Natalia Periolo
- National Influenza Centre PAHO/WHO, Servicio Virosis Respiratorias, Departamento Virología, Instituto Nacional de Enfermedades Infecciosas, ANLIS ‘Carlos G. Malbrán’, Buenos Aires, Argentina
| | - Ana M. Campos
- National Influenza Centre PAHO/WHO, Servicio Virosis Respiratorias, Departamento Virología, Instituto Nacional de Enfermedades Infecciosas, ANLIS ‘Carlos G. Malbrán’, Buenos Aires, Argentina
| | - Vilma L. Savy
- National Influenza Centre PAHO/WHO, Servicio Virosis Respiratorias, Departamento Virología, Instituto Nacional de Enfermedades Infecciosas, ANLIS ‘Carlos G. Malbrán’, Buenos Aires, Argentina
| | - Elsa G. Baumeister
- National Influenza Centre PAHO/WHO, Servicio Virosis Respiratorias, Departamento Virología, Instituto Nacional de Enfermedades Infecciosas, ANLIS ‘Carlos G. Malbrán’, Buenos Aires, Argentina
| |
Collapse
|
44
|
Hussain S, Miller JL, Harvey DJ, Gu Y, Rosenthal PB, Zitzmann N, McCauley JW. Strain-specific antiviral activity of iminosugars against human influenza A viruses. J Antimicrob Chemother 2014; 70:136-52. [PMID: 25223974 PMCID: PMC4267503 DOI: 10.1093/jac/dku349] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Objectives Drugs that target host cell processes can be employed to complement drugs that specifically target viruses, and iminosugar compounds that inhibit host α-glucosidases have been reported to show antiviral activity against multiple viruses. Here the effect and mechanism of two iminosugar α-glucosidase inhibitors, N-butyl-deoxynojirimycin (NB-DNJ) and N-nonyl-deoxynojirimycin (NN-DNJ), on human influenza A viruses was examined. Methods The viruses examined were a recently circulating seasonal influenza A(H3N2) virus strain A/Brisbane/10/2007, an older H3N2 strain A/Udorn/307/72, and A/Lviv/N6/2009, a strain representative of the currently circulating pandemic influenza A(H1N1)pdm09 virus. Results The inhibitors had the strongest effect on Brisbane/10 and NN-DNJ was more potent than NB-DNJ. Both compounds showed antiviral activity in cell culture against three human influenza A viruses in a strain-specific manner. Consistent with its action as an α-glucosidase inhibitor, NN-DNJ treatment resulted in an altered glycan processing of influenza haemagglutinin (HA) and neuraminidase (NA), confirmed by MS. NN-DNJ treatment was found to reduce the cell surface expression of the H3 subtype HA. The level of sialidase activity of NA was reduced in infected cells, but the addition of exogenous sialidase to the cells did not complement the NN-DNJ-mediated inhibition of virus replication. Using reassortant viruses, the drug susceptibility profile was determined to correlate with the origin of the HA. Conclusions NN-DNJ inhibits influenza A virus replication in a strain-specific manner that is dependent on the HA.
Collapse
Affiliation(s)
- S Hussain
- Division of Virology, Medical Research Council National Institute for Medical Research, Mill Hill, London NW7 1AA, UK Division of Physical Biochemistry, Medical Research Council National Institute for Medical Research, Mill Hill, London NW7 1AA, UK
| | - J L Miller
- Oxford Glycobiology Institute, Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, UK
| | - D J Harvey
- Oxford Glycobiology Institute, Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, UK Department of Biological Sciences, Gibbet Hill Campus, University of Warwick, Coventry CV4 7AL, UK
| | - Y Gu
- Confocal Imaging and Analysis Laboratory, Medical Research Council National Institute for Medical Research, Mill Hill, London NW7 1AA, UK
| | - P B Rosenthal
- Division of Physical Biochemistry, Medical Research Council National Institute for Medical Research, Mill Hill, London NW7 1AA, UK
| | - N Zitzmann
- Oxford Glycobiology Institute, Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, UK
| | - J W McCauley
- Division of Virology, Medical Research Council National Institute for Medical Research, Mill Hill, London NW7 1AA, UK
| |
Collapse
|
45
|
Abstract
Planning, implementing, and evaluating interventions against infectious diseases depend on the nature of the infectious disease; the availability of intervention measures; and logistical, economic, and political constraints. Infectious diseases and vaccine- or drug-based interventions can be loosely categorized by the degree to which the infectious disease and the intervention are well established. Pertussis, polio, and measles are three examples of long-known infectious diseases for which global vaccination has dramatically reduced the public health burden. Pertussis vaccination was introduced in the 1940s, polio vaccination in the 1950s, and measles vaccination in the 1960s, nearly eliminating these diseases in many places.
Collapse
Affiliation(s)
- M Elizabeth Halloran
- Center for Statistics and Quantitative Infectious Diseases, Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue North, Seattle, WA 98109, USA. Department of Biostatistics, University of Washington, Seattle, WA 98105, USA.
| | - Ira M Longini
- Center for Statistics and Quantitative Infectious Diseases, Department of Biostatistics, College of Public Health and Health Professions, and College of Medicine, University of Florida, Gainesville, FL 32611, USA
| |
Collapse
|
46
|
Huber VC. Influenza vaccines: from whole virus preparations to recombinant protein technology. Expert Rev Vaccines 2014; 13:31-42. [PMID: 24192014 DOI: 10.1586/14760584.2014.852476] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Vaccination against influenza represents our most effective form of prevention. Historical approaches toward vaccine creation and production have yielded highly effective vaccines that are safe and immunogenic. Despite their effectiveness, these historical approaches do not allow for the incorporation of changes into the vaccine in a timely manner. In 2013, a recombinant protein-based vaccine that induces immunity toward the influenza virus hemagglutinin was approved for use in the USA. This vaccine represents the first approved vaccine formulation that does not require an influenza virus intermediate for production. This review presents a brief history of influenza vaccines, with insight into the potential future application of vaccines generated using recombinant technology.
Collapse
Affiliation(s)
- Victor C Huber
- Division of Basic Biomedical Sciences, University of South Dakota, 414 E Clark Street, Vermillion, SD 57069, USA
| |
Collapse
|
47
|
Lee BJ, Kwon HI, Kim EH, Park SJ, Lee SH, Choi YK, Kim SH. Assessment of mOMV adjuvant efficacy in the pathogenic H1N1 influenza virus vaccine. Clin Exp Vaccine Res 2014; 3:194-201. [PMID: 25003093 PMCID: PMC4083072 DOI: 10.7774/cevr.2014.3.2.194] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2014] [Revised: 04/02/2014] [Accepted: 04/10/2014] [Indexed: 01/15/2023] Open
Abstract
Purpose Since the pandemic (H1N1) 2009 virus has been a seasonal flu which still poses great human health concerns worldwide, vaccination would be considered as the most effective strategy to control the influenza virus spreading. Here, we assessed adjuvant efficacy of modified outer membrane vesicle (mOMV) towards the pandemic H1N1 split antigen. Materials and Methods For this study, mice were vaccinated twice with various amount of antigen (0.05, 0.1, and 0.5 µg/dose hemagglutinin [HA]) that were mixed with mOMV, aluminum hydroxide (alum), and MF59, as well as the combined adjuvant comprising the mOMV plus alum. Results We found that all the adjuvanted vaccines of A/California/04/09 (CA04, H1N1) containing HA antigen more than 0.1 µg/dose protected effectively from lethal challenge (maCA04, H1N1) virus, compared to the antigen only group. Furthermore, vaccinated mice received as low as 0.05 µg/dose of the split vaccine containing the combined adjuvant (10 µg of mOMV plus alum) showed a full protection against lethal challenge with H1N1 virus. Taken together, these results suggest that mOMV can exert not only the self-adjuvanticity but also a synergy effect for the vaccine efficacy when combined with alum. Conclusion Our results indicate that mOMV could be a promising vaccine adjuvant by itself and it could be used as a vaccine platform for development of various vaccine formulations to prepare future influenza pandemic.
Collapse
Affiliation(s)
- Byeong-Jae Lee
- College of Medicine and Medical Research Institute, Chungbuk National University, Cheongju, Korea
| | - Hyeok-Il Kwon
- College of Medicine and Medical Research Institute, Chungbuk National University, Cheongju, Korea
| | - Eun-Ha Kim
- College of Medicine and Medical Research Institute, Chungbuk National University, Cheongju, Korea
| | - Su-Jin Park
- College of Medicine and Medical Research Institute, Chungbuk National University, Cheongju, Korea
| | - Sang-Ho Lee
- Viral Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Korea
| | - Young Ki Choi
- College of Medicine and Medical Research Institute, Chungbuk National University, Cheongju, Korea
| | - Sang-Hyun Kim
- Viral Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Korea
| |
Collapse
|
48
|
Escuret V, Collins PJ, Casalegno JS, Vachieri SG, Cattle N, Ferraris O, Sabatier M, Frobert E, Caro V, Skehel JJ, Gamblin S, Valla F, Valette M, Ottmann M, McCauley JW, Daniels RS, Lina B. A novel I221L substitution in neuraminidase confers high-level resistance to oseltamivir in influenza B viruses. J Infect Dis 2014; 210:1260-9. [PMID: 24795482 PMCID: PMC4176448 DOI: 10.1093/infdis/jiu244] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
UNLABELLED Influenza B viruses with a novel I221L substitution in neuraminidase (NA) conferring high-level resistance to oseltamivir were isolated from an immunocompromised patient after prolonged oseltamivir treatment. METHODS Enzymatic characterization of the NAs (Km, Ki) and the in vitro fitness of viruses carrying wild-type or mutated (I221L) NA genes were evaluated. Proportions of wild-type and mutated NA genes were directly quantified in the patient samples. Structural characterizations by X-ray crystallography of a wild-type and I221L variant NA were performed. RESULTS The Km and Ki revealed that the I221L variant NA had approximately 84 and 51 times lower affinity for oseltamivir carboxylate and zanamivir, respectively, compared with wild-type NA. Viruses with a wild-type or I221L variant NA had similar growth kinetics in Madin-Darby canine kidney (MDCK) cells, and 5 passages in MDCK cells revealed no reversion of the I221L substitution. The crystal structure of the I221L NA and oseltamivir complex showed that the leucine side chain protrudes into the hydrophobic pocket of the active site that accommodates the pentyloxy substituent of oseltamivir. CONCLUSIONS Enzyme kinetic and NA structural analyses provide an explanation for the high level of resistance to oseltamivir while retaining good fitness of viruses carrying I221L variant NA.
Collapse
Affiliation(s)
- Vanessa Escuret
- Laboratoire de Virologie et Centre National de Référence virus influenzae Laboratoire Virpath EA4610, Faculté de Médecine Lyon Est, Université Claude Bernard Lyon 1, Université de Lyon, and
| | | | - Jean-Sébastien Casalegno
- Laboratoire de Virologie et Centre National de Référence virus influenzae Laboratoire Virpath EA4610, Faculté de Médecine Lyon Est, Université Claude Bernard Lyon 1, Université de Lyon, and
| | | | - Nicholas Cattle
- WHO Collaborating Centre for Reference and Research on Influenza, Division of Virology, Medical Research Council National Institute for Medical Research, London, United Kingdom
| | - Olivier Ferraris
- Laboratoire Virpath EA4610, Faculté de Médecine Lyon Est, Université Claude Bernard Lyon 1, Université de Lyon, and
| | - Murielle Sabatier
- Laboratoire Virpath EA4610, Faculté de Médecine Lyon Est, Université Claude Bernard Lyon 1, Université de Lyon, and
| | - Emilie Frobert
- Laboratoire de Virologie et Centre National de Référence virus influenzae Laboratoire Virpath EA4610, Faculté de Médecine Lyon Est, Université Claude Bernard Lyon 1, Université de Lyon, and
| | - Valérie Caro
- Genotyping of Pathogens and Public Health Platform, Institut Pasteur, Paris, France
| | | | | | - Frédéric Valla
- Service de Réanimation Pédiatrique, Hôpital Femme Mère Enfant, Groupement Hospitalier Est, Hospices Civils de Lyon, Bron
| | - Martine Valette
- Laboratoire de Virologie et Centre National de Référence virus influenzae
| | - Michèle Ottmann
- Laboratoire Virpath EA4610, Faculté de Médecine Lyon Est, Université Claude Bernard Lyon 1, Université de Lyon, and
| | - John W McCauley
- WHO Collaborating Centre for Reference and Research on Influenza, Division of Virology, Medical Research Council National Institute for Medical Research, London, United Kingdom
| | - Rodney S Daniels
- WHO Collaborating Centre for Reference and Research on Influenza, Division of Virology, Medical Research Council National Institute for Medical Research, London, United Kingdom
| | - Bruno Lina
- Laboratoire de Virologie et Centre National de Référence virus influenzae Laboratoire Virpath EA4610, Faculté de Médecine Lyon Est, Université Claude Bernard Lyon 1, Université de Lyon, and
| |
Collapse
|
49
|
Delangue J, Roca Sanchez Y, Piorkowski G, Bessaud M, Baronti C, Thirion-Perrier L, Mafayle RL, Ardaya CA, Aguilera GA, Guzman JR, Riera JL, de Lamballerie X. Viral aetiology influenza like illnesses in Santa Cruz, Bolivia (2010-2012). Virol J 2014; 11:35. [PMID: 24564892 PMCID: PMC4015617 DOI: 10.1186/1743-422x-11-35] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Accepted: 02/10/2014] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Acute respiratory infections represent a serious public health issue worldwide but virological aetiologies of Influenza Like Illnesses (ILIs) remain largely unknown in developing countries. This study represents the first attempt to characterise viral aetiologies of ILIs in Bolivia. METHODS It was performed in Santa Cruz city from January 2010 to September 2012, based on 564 naso-pharyngeal swabs collected in a National Reference Laboratory and real-time PCR techniques, viral cultures and phylogenetic analyses. RESULTS 50.2% of samples were positive for at least one virus with influenza viruses (Flu A: ~15%; Flu B: ~9%), rhinoviruses (~8%), coronaviruses (~5%) and hRSV (~4%) being the most frequently identified. The pattern of viral infections varied according to age groups. The elucidation rate was the highest (>60%) amongst patients under 10 yo and the lowest (<40%) amongst patients ≥60 yo. Nearly 3% of samples showed dual viral infections. Epidemiological peaks were associated with a predominant virus but generally included 30-50% of infections by different viruses. Unexpectedly, the frequency of influenza in the 0-4 yo population was very low and a complete hRSV eclipse occurred in 2011. Genetic analyses indicated that distinct evolutionary lineages of Flu A(H1N1)pdm2009, Flu A/H3N2 and Flu B have co-circulated in Bolivia in the study period, originating from Central and North America, Europe, Asia and Australia. CONCLUSION Our results emphasise the requirement for a reinforced epidemiological and genetic follow-up of influenza and other ILIs in Bolivia to further inform the preparation of vaccines used in the region, guide vaccination campaigns and improve the medical management of patients.
Collapse
Affiliation(s)
- Julie Delangue
- IRD French Institute of Research for Development, EHESP French School of Public Health, UMR_D 190 "Emergence des Pathologies Virales", Aix Marseille Univ, 13005 Marseille, France.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Petridis D, Zarogoulidis P, Kallianos A, Kioumis I, Trakada G, Spyratos D, Papaiwannou A, Porpodis K, Huang H, Rapti A, Hohenforst-Schmidt W, Zarogoulidis K. Clinical differences between H3N2 and H1N1 influenza 2012 and lower respiratory tract infection found using a statistical classification approach. Ther Clin Risk Manag 2014; 10:77-86. [PMID: 24532970 PMCID: PMC3923611 DOI: 10.2147/tcrm.s57429] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Background Influenza A H1N1 and H3N2 are two influenza waves that have been identified in past years. Methods Data from 77 inpatients from three tertiary hospitals were included and statistical analysis was performed in three different clusters. Results Thirty-four patients (44.2%) had respiratory distress upon admission, 31.2% had a smoking history or were active smokers, 37.7% manifested disease symptoms, and 7.8% were obese (body mass index >41). The mean age of patients was 51.1 years. Cough was the most common symptom observed in 77.9% of the patients, accompanied by sputum production (51.9%) and fatigue (42.9%). Hemoptysis and vomiting were rarely recorded in the patients (9.1% and 16.9%, respectively). Oseltamivir administration varied between 0 and 10 days, giving a mean value of 2.2 days. In particular, 19 patients received no drug, 31 patients received drug for only for 1 day, 19 patients for 5 days, and 8 patients from 2 to 10 days. Conclusion Clusters of symptoms can be used to identify different types of influenza and disease severity. Patients with vaccination had pneumonia, whereas patients without vaccination had influenza A. Patients more than 54.5 years old had H3N2 and patients less than 54.5 years had H1N1. White blood cell count values increased from normal to elevated in H3N2 patients but still remained abnormal in lower tract infection and H1N1 patients.
Collapse
Affiliation(s)
- Dimitris Petridis
- Department of Food Technology, Alexander Technological Educational Institute of Thessaloniki, Thessaloniki, Greece
| | - Paul Zarogoulidis
- Pulmonary Department, "G Papanikolaou" General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece ; Department of Interventional Pneumology, Ruhrlandklinik, West German Lung Center, University Hospital, University Duisburg-Essen, Essen, Germany
| | | | - Ioannis Kioumis
- Pulmonary Department, "G Papanikolaou" General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Georgia Trakada
- Department of Clinical Therapeutics, Division of Pneumonology, Medical School, National University of Athens, Athens, Greece
| | - Dionysios Spyratos
- Pulmonary Department, "G Papanikolaou" General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Antonis Papaiwannou
- Pulmonary Department, "G Papanikolaou" General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Konstantinos Porpodis
- Pulmonary Department, "G Papanikolaou" General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Haidong Huang
- Department of Respiratory Diseases, Changhai Hospital/First Affiliated Hospital of the Second Military Medical University, Shanghai, People's Republic of China
| | - Aggeliki Rapti
- 2nd Pulmonary Department, "Sotiria" Hospital for Chest Diseases, Athens, Greece
| | | | - Konstantinos Zarogoulidis
- Pulmonary Department, "G Papanikolaou" General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|