1
|
Wen F, Liu S, Zhou L, Zhu Y, Wang W, Wei M, Xu X, Liu Y, Shuai Q, Yu J, Jing P, Li J, Zhu F. Immunogenicity and safety of 1 versus 2 doses of quadrivalent-inactivated influenza vaccine in children aged 3-8 years with or without previous influenza vaccination histories. Hum Vaccin Immunother 2025; 21:2468074. [PMID: 39993940 PMCID: PMC11853548 DOI: 10.1080/21645515.2025.2468074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 01/24/2025] [Accepted: 02/13/2025] [Indexed: 02/26/2025] Open
Abstract
This study assessed the immunogenicity and safety of a quadrivalent influenza split-virion vaccine (IIV4) in children with and without prior influenza vaccination. An open-label, phase IV clinical trial was conducted in healthy children aged 3-8 years in Pizhou, Jiangsu Province, China. Children with ≥2 doses of prior influenza vaccination (Influenza Vaccination Group) and those with no prior vaccination (Influenza Vaccine-naïve Group) were given two doses of IIV4, 4 weeks apart. The primary immunogenicity endpoints were seroprotection rates (SPRs) of hemagglutination inhibition antibody 28 days after each dose against influenza A/H1N1, A/H3N2, B/Yamagata (BY), and B/Victoria (BV). Safety endpoints included adverse events (AEs) within 28 days and serious adverse events (SAEs) within 6 months. Between September 19-25, 2021, 278 participants were screened, and 240 were enrolled (120 per group). In the Influenza Vaccination Group, SPRs after the 2nd dose were 98.28%, 92.24%, 99.14%, and 87.93%, similar to those after the 1st dose (all p > .05). In the Influenza Vaccine-naïve Group, SPRs after the 2nd dose were 99.12%, 96.49%, 99.12%, and 92.11%, significantly higher than after the 1st dose (p < .001 for all strains except BY, p = .070). Most AEs were mild or moderate, with no serious AEs related to the vaccine. Children aged 3-8 years with prior influenza vaccination need only one dose of IIV4, while those without prior vaccination require two doses. ClinicalTrials.gov, the identifier is NCT05144464.
Collapse
MESH Headings
- Humans
- Influenza Vaccines/administration & dosage
- Influenza Vaccines/immunology
- Influenza Vaccines/adverse effects
- Child, Preschool
- Male
- Female
- Child
- Antibodies, Viral/blood
- Influenza, Human/prevention & control
- Influenza, Human/immunology
- Vaccines, Inactivated/immunology
- Vaccines, Inactivated/administration & dosage
- Vaccines, Inactivated/adverse effects
- Hemagglutination Inhibition Tests
- Influenza A Virus, H3N2 Subtype/immunology
- Influenza A Virus, H1N1 Subtype/immunology
- China
- Influenza B virus/immunology
- Immunogenicity, Vaccine
- Vaccination/methods
- Immunization Schedule
Collapse
Affiliation(s)
- Feng Wen
- School of Public Health, Southeast University, Nanjing, China
| | - Sheng Liu
- Department of Disease Control and Prevention, Pizhou City Center for Disease Control and Prevention, Xuzhou, China
| | - Li Zhou
- Department of Vaccine Clinical Evaluation, Jiangsu Provincial Center for Disease Control and Prevention (Jiangsu Provincial Academy of Preventive Medicine), Nanjing, China
- School of Public Health, National Vaccine Innovation Platform, Nanjing Medical University, Nanjing, China
| | - Yinbiao Zhu
- Research and Development Department, Jiangsu GDK Biological Technology Co. Ltd, Taizhou, China
| | - Wenjuan Wang
- Department of Vaccine Clinical Evaluation, Jiangsu Provincial Center for Disease Control and Prevention (Jiangsu Provincial Academy of Preventive Medicine), Nanjing, China
- Jiangsu Provincial Medical Innovation Center (Jiangsu Provincial Academy of Preventive Medicine), National Health Commission Key Laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China
| | - Mingwei Wei
- Department of Vaccine Clinical Evaluation, Jiangsu Provincial Center for Disease Control and Prevention (Jiangsu Provincial Academy of Preventive Medicine), Nanjing, China
- Jiangsu Provincial Medical Innovation Center (Jiangsu Provincial Academy of Preventive Medicine), National Health Commission Key Laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China
| | - Xinglong Xu
- Research and Development Department, Jiangsu GDK Biological Technology Co. Ltd, Taizhou, China
| | - Yan Liu
- Research and Development Department, Jiangsu GDK Biological Technology Co. Ltd, Taizhou, China
| | - Qi Shuai
- Research and Development Department, Jiangsu GDK Biological Technology Co. Ltd, Taizhou, China
| | - Jun Yu
- Research and Development Department, Jiangsu GDK Biological Technology Co. Ltd, Taizhou, China
| | - Pengfei Jing
- Department of Vaccine Clinical Evaluation, Jiangsu Provincial Center for Disease Control and Prevention (Jiangsu Provincial Academy of Preventive Medicine), Nanjing, China
- Jiangsu Provincial Medical Innovation Center (Jiangsu Provincial Academy of Preventive Medicine), National Health Commission Key Laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China
| | - Jingxin Li
- School of Public Health, Southeast University, Nanjing, China
- Department of Vaccine Clinical Evaluation, Jiangsu Provincial Center for Disease Control and Prevention (Jiangsu Provincial Academy of Preventive Medicine), Nanjing, China
- Jiangsu Provincial Medical Innovation Center (Jiangsu Provincial Academy of Preventive Medicine), National Health Commission Key Laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China
| | - Fengcai Zhu
- School of Public Health, Southeast University, Nanjing, China
- Department of Vaccine Clinical Evaluation, Jiangsu Provincial Center for Disease Control and Prevention (Jiangsu Provincial Academy of Preventive Medicine), Nanjing, China
- Jiangsu Provincial Medical Innovation Center (Jiangsu Provincial Academy of Preventive Medicine), National Health Commission Key Laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China
| |
Collapse
|
2
|
Zhao T, Liu X, Huang X, Wang L, Lei Y, Luo C, Liu J, Fang S, Zou X, Yan H, Sun C, Shu Y. Development and evaluation of mosaic VLPs vaccine for enhanced broad-Spectrum immunity against influenza B virus lineages in mice. Vaccine 2025; 51:126882. [PMID: 39970593 DOI: 10.1016/j.vaccine.2025.126882] [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: 08/16/2024] [Revised: 01/03/2025] [Accepted: 02/10/2025] [Indexed: 02/21/2025]
Abstract
Influenza B virus (IBV) causes annual respiratory outbreaks, posing significant public health challenges. Traditional vaccines are limited in their effectiveness by antigenic drift and strain mismatches. This study introduces innovative mosaic virus-like particle (VLP) vaccines designed to present a broader range of epitopes, aiming for broad-spectrum immunity against both B/Victoria (BV) and B/Yamagata (BY) IBV lineages in mice. Two mosaic hemagglutinin (HAM) proteins were incorporated into VLPs, mimicking the morphology and size of the virus. Notably, the BV component of our mosaic VLPs demonstrated significant cross-reactivity against the BY strain, surpassing a commercial quadrivalent inactivated influenza vaccine (QIV) in generating broad immune responses. Immunogenicity and efficacy assessments in mice revealed that the mosaic VLPs induced Th1/Th2 cytokine responses and provided effective protection against homologous IBV challenge, notably reducing lung damage. This study highlights the potential of mosaic VLPs in universal influenza B vaccine development and emphasizes the importance of T cell immunity in influenza vaccine design.
Collapse
Affiliation(s)
- Tianyi Zhao
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen 518107, PR China
| | - Xuejie Liu
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen 518107, PR China
| | - Xiaoping Huang
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen 518107, PR China
| | - Liangliang Wang
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen 518107, PR China
| | - Yuxuan Lei
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen 518107, PR China
| | - Chuming Luo
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen 518107, PR China
| | - Jing Liu
- Center for Disease Control and Prevention of Southern Military Theatre, 510610 Guangzhou, PR China
| | - Shisong Fang
- Microorganism Testing Institute, Shenzhen Center for Disease Control and Prevention, Shenzhen 518107, PR China
| | - Xuan Zou
- Microorganism Testing Institute, Shenzhen Center for Disease Control and Prevention, Shenzhen 518107, PR China
| | - Huacheng Yan
- Center for Disease Control and Prevention of Southern Military Theatre, 510610 Guangzhou, PR China.
| | - Caijun Sun
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen 518107, PR China.
| | - Yuelong Shu
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen 518107, PR China; Key Laboratory of Pathogen Infection Prevention and Control (MOE), State Key Laboratory of Respiratory Health and Multimorbidity, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 102629, PR China.
| |
Collapse
|
3
|
Lu X, Li W, Li P, Li Y, Gou Y, Wang T, Liu Z, Wu Y. Selection and identification of an ssDNA aptamer against influenza B virus hemagglutinin protein. Virol J 2025; 22:64. [PMID: 40050943 PMCID: PMC11887071 DOI: 10.1186/s12985-025-02657-2] [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: 06/03/2024] [Accepted: 02/10/2025] [Indexed: 03/09/2025] Open
Abstract
BACKGROUND The influenza virus causes infectious respiratory disease with high morbidity and mortality worldwide. Influenza B typically goes unnoticed owing to its mild clinical symptoms and limitations. However, its increasing prevalence in recent years poses a significant health burden. Consequently, current diagnostic methods for the detection of influenza B virus are inadequate, highlighting the urgent need to develop accurate and sensitive techniques for early disease diagnosis. Aptamers, single-stranded deoxyribonucleic acid (ssDNA), or ribonucleic acid molecules primarily rely on their secondary structures, such as stem-loops and hairpins, to bind efficiently and specifically to the target through base complementary pairing, electrostatic interaction, hydrogen bonding, and van der Waals forces. Aptamers are superior to antibodies in their ability to bind targets. The objective of this study was to identify and develop aptamers against the hemagglutinin (HA) protein of influenza B virus. METHODS An enriched DNA library with strong binding to the influenza B virus HA protein was obtained using magnetic bead systematic evolution of ligands by exponential enrichment technology after nine rounds of selection. Five candidate aptamers were identified by high-throughput sequencing. The aptamers were characterized using surface plasmon resonance and enzyme-linked immunosorbent assay techniques, and the aptamer exhibiting the highest affinity and specificity for the target protein was selected. RESULTS We screened and characterized five ssDNA aptamer sequences that bind to influenza B virus HA. Among these, aptamer sequence A573 exhibited the highest sensitivity and binding affinity for the target protein. CONCLUSIONS The novel aptamer sequences selected in this study have the potential to be used as biorecognition molecules for the development of aptamer sensors to detect influenza B virus.
Collapse
Affiliation(s)
- Xing Lu
- Department of Respiratory and Critical Care Medicine, General Hospital of Southern Theater Command of PLA, No.111, Liuhua Road, Yuexiu District, Guangzhou, 510010, Guangdong, China
- Graduate School, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Weifeng Li
- Department of Respiratory and Critical Care Medicine, General Hospital of Southern Theater Command of PLA, No.111, Liuhua Road, Yuexiu District, Guangzhou, 510010, Guangdong, China
| | - Ping Li
- Department of Respiratory and Critical Care Medicine, General Hospital of Southern Theater Command of PLA, No.111, Liuhua Road, Yuexiu District, Guangzhou, 510010, Guangdong, China
| | - Yongqiang Li
- Department of Respiratory and Critical Care Medicine, General Hospital of Southern Theater Command of PLA, No.111, Liuhua Road, Yuexiu District, Guangzhou, 510010, Guangdong, China
| | - Yanni Gou
- Graduate School, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Tao Wang
- Graduate School, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhifeng Liu
- Department of Critical Care Medicine, General Hospital of Southern Theater Command of PLA, Yuexiu District, No.111, Liuhua Road, Guangzhou, 510010, Guangdong, China.
| | - Yuting Wu
- Department of Respiratory and Critical Care Medicine, General Hospital of Southern Theater Command of PLA, No.111, Liuhua Road, Yuexiu District, Guangzhou, 510010, Guangdong, China.
- Graduate School, Guangzhou University of Chinese Medicine, Guangzhou, China.
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China.
| |
Collapse
|
4
|
Alizadeh S, Edalat F, Letafati A, Pirbonyeh N, Tabibzadeh A, Mousavizadeh L, Moattari A, Karbalaie Niya MH. Genetic characterization of influenza B virus and oseltamivir resistance in pediatric patients with acute respiratory infections: a cross-sectional study. Virus Genes 2025; 61:54-63. [PMID: 39499431 DOI: 10.1007/s11262-024-02119-x] [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: 04/29/2024] [Accepted: 10/10/2024] [Indexed: 11/07/2024]
Abstract
Influenza virus neuraminidase inhibitors (NAIs) drug usage can result in NAI resistance, especially in children and individuals with weakened immune systems. The aim of the present study was to identify NAI-resistant variants of IBV and to introduce probable novel mutations, phylogenetic study, and its epitope mapping based on NA gene in patients from Shiraz, Iran. A cross-sectional study was conducted between 2017 and 2018 on symptomatic children. A real-time PCR was run for IBV screening. Then, making use of direct sequencing, amplified 1401 bases of NA gene and phylogenetic tree reconstructed. Epitopes were predicted using ABCpred server. From among a total of 235 specimens, 9.7% were identified with IBV infection. Of them, sequence of NA gene for 17 isolates were analyzed. Phylogenetic analysis showed that 15 isolates belonged to Yamagata clade 3 Wisconsin/01-like subclade and 2 were related to Victoria clade 1 Brisbane/60-like subclade (Vic-1A-2). NA gene sequence analysis showed a total of 52 substitutions in which 27 were for BVic and 37 were for BYam isolates and 19 were novel substitutions. Only one substitution (S198N) was found in NA active site and T49M, I120V, N198S, N219K, S295R, D320K N340D, E358K, D384G, and D463N were found as probable resistance variants to NAIs. Epitope mapping showed some major differences in our isolates NA gene. Present study was one of the rare comprehensive studies conducted in Shiraz/Iran on IBV resistant associated variants to NAIs. We reported 11.7% mutation in NA active site and some probable NAIs resistant mutations. Epitope mapping confirmed major changes in NA gene which needs broader studies to confirm.
Collapse
Affiliation(s)
- Sheida Alizadeh
- Department of Virology and Bacteriology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fahime Edalat
- Autophagy Research Center, Shiraz University of Medical Science, Shiraz, Iran
| | - Arash Letafati
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
- Research Center for Clinical Virology, Tehran University of Medical Sciences, Tehran, Iran
| | - Neda Pirbonyeh
- Department of Virology and Bacteriology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Microbiology Department, Burn and Wound Healing Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Alireza Tabibzadeh
- Department of Medical Laboratory Sciences, Faculty of Medical Sciences, Islamic Azad University, Arak Branch, Arak, Iran
| | - Leila Mousavizadeh
- Department of Virology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Afagh Moattari
- Department of Virology and Bacteriology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
| | | |
Collapse
|
5
|
Adeleke RA, Sahler J, Choi A, Roth K, Upadhye V, Ezzatpour S, Imbiakha B, Khomandiak S, Diaz A, Whittaker GR, Jager MC, August A, Buchholz DW, Aguilar HC. Replication-incompetent VSV-based vaccine elicits protective responses against SARS-CoV-2 and influenza virus. SCIENCE ADVANCES 2025; 11:eadq4545. [PMID: 39879304 PMCID: PMC11777205 DOI: 10.1126/sciadv.adq4545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 12/30/2024] [Indexed: 01/31/2025]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and influenza viruses lead to severe respiratory illnesses and death in humans, exacerbated in individuals with underlying health conditions, remaining substantial global public health concerns. Here, we developed a bivalent replication-incompetent single-cycle pseudotyped vesicular stomatitis virus vaccine that incorporates both a prefusion-stabilized SARS-CoV-2 spike protein lacking a furin cleavage site and a full-length influenza A virus neuraminidase protein. Vaccination of K18-hACE2 or C57BL/6J mouse models generated durable levels of neutralizing antibodies, T cell responses, and protection from morbidity and mortality upon challenge with either virus. Furthermore, the vaccine provided heterologous protection upon challenge with a different influenza virus strain, supporting the advantage of using NA to increase the breadth of vaccine protection. Now, no bivalent vaccine is approved for use against both SARS-CoV-2 and influenza virus. Our study supports using this platform to develop safe and efficient vaccines against multiple viruses.
Collapse
Affiliation(s)
- Richard A. Adeleke
- Department of Microbiology and Immunology, Cornell University College of Veterinary Medicine, Ithaca, NY 14853, USA
| | - Julie Sahler
- Department of Microbiology and Immunology, Cornell University College of Veterinary Medicine, Ithaca, NY 14853, USA
| | - Annette Choi
- Department of Microbiology and Immunology, Cornell University College of Veterinary Medicine, Ithaca, NY 14853, USA
| | - Kyle Roth
- Department of Microbiology and Immunology, Cornell University College of Veterinary Medicine, Ithaca, NY 14853, USA
| | - Viraj Upadhye
- Department of Microbiology and Immunology, Cornell University College of Veterinary Medicine, Ithaca, NY 14853, USA
| | - Shahrzad Ezzatpour
- Department of Microbiology and Immunology, Cornell University College of Veterinary Medicine, Ithaca, NY 14853, USA
| | - Brian Imbiakha
- Department of Microbiology and Immunology, Cornell University College of Veterinary Medicine, Ithaca, NY 14853, USA
| | - Solomiia Khomandiak
- Department of Microbiology and Immunology, Cornell University College of Veterinary Medicine, Ithaca, NY 14853, USA
| | - Annika Diaz
- Department of Population Medicine and Diagnostic Sciences, Cornell University College of Veterinary Medicine, Ithaca, NY 14853, USA
| | - Gary R. Whittaker
- Department of Microbiology and Immunology, Cornell University College of Veterinary Medicine, Ithaca, NY 14853, USA
| | - Mason C. Jager
- Department of Population Medicine and Diagnostic Sciences, Cornell University College of Veterinary Medicine, Ithaca, NY 14853, USA
| | - Avery August
- Department of Microbiology and Immunology, Cornell University College of Veterinary Medicine, Ithaca, NY 14853, USA
| | - David W. Buchholz
- Department of Microbiology and Immunology, Cornell University College of Veterinary Medicine, Ithaca, NY 14853, USA
| | - Hector C. Aguilar
- Department of Microbiology and Immunology, Cornell University College of Veterinary Medicine, Ithaca, NY 14853, USA
| |
Collapse
|
6
|
Cao Y, Han J, Xiao Y, Wang Z, Zhang H, Fang R, Li J, Dong M, Chen R, Zhu G, Han J, Sun L. Xiao-Er-Kang-Du capsules regulate autophagy against the influenza B virus (Victoria strain) through the mTOR/ULK1/Beclin1/VPS34 pathway. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118872. [PMID: 39366496 DOI: 10.1016/j.jep.2024.118872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 09/06/2024] [Accepted: 09/30/2024] [Indexed: 10/06/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Xiao-er-kang-du (XEKD) capsule is a Chinese herbal formula used for treatment of upper respiratory tract infection caused by various viruses in pediatric patients in China. XEKD is used clinically for the treatment of influenza-like symptoms, including fever, chills, cough, stuffy and runny nose, headache, and sore throat, with remarkable efficacy. However, the pharmacologic mechanism of XEKD against influenza B virus (IBV) infection is unclear. AIM OF THE STUDY The main purpose of the present work is to explore the curative effect as well as possible mechanisms of XEKD against influenza B virus (IBV) (Victoria strain). MATERIALS AND METHODS Both in vivo and in vitro experiments were performed to confirm the antiviral properties of XEKD. High-performance liquid chromatography was used to analyze the active components and assess the stability of XEKD. In addition, the mechanism of action of XEKD against IBV (Victoria) was investigated by western blot, immunofluorescence, and immunohistochemical analyses, in addition to confocal fluorescence microscopy. RESULTS The findings revealed that XEKD demonstrated antiviral effects against IBV infection in both in vivo and in vitro via the mTOR/ULK1/Beclin1/VPS34 pathway and promote cellular autophagy to mitigate IBV-induced lung tissue damage. The results of this work are expected to lead to a deeper understanding of the mechanism underlying the effect of the XEKD capsule against IBV infections. CONCLUSIONS IBV infection was found to inhibit autophagy, which exacerbated inflammatory damage. XEKD regulates autophagy through the mTOR/ULK1/Beclin1/VPS34 pathway and exerts antiviral effects, thereby laying a foundation for further development of XEKD as a potential therapeutic against IBV infection.
Collapse
Affiliation(s)
- Yan Cao
- College of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130117, PR China; College of Basic Medical Sciences, Changchun University of Chinese Medicine, Changchun, 130117, PR China
| | - Jing Han
- College of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130117, PR China; Center of Children's Clinic, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, 130022, PR China
| | - Yan Xiao
- Key Laboratory of Jilin Province for Traditional Chinese Medicine Prevention and Treatment of Infectious Diseases, College of Integrative Medicine, Changchun University of Chinese Medicine, Changchun, 130117, PR China
| | - Zhongtian Wang
- College of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130117, PR China; Center of Children's Clinic, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, 130022, PR China
| | - Haiyang Zhang
- College of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130117, PR China; Center of Children's Clinic, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, 130022, PR China
| | - Ruikang Fang
- Key Laboratory of Jilin Province for Traditional Chinese Medicine Prevention and Treatment of Infectious Diseases, College of Integrative Medicine, Changchun University of Chinese Medicine, Changchun, 130117, PR China
| | - Jingjing Li
- Key Laboratory of Jilin Province for Traditional Chinese Medicine Prevention and Treatment of Infectious Diseases, College of Integrative Medicine, Changchun University of Chinese Medicine, Changchun, 130117, PR China
| | - Meiwen Dong
- Key Laboratory of Jilin Province for Traditional Chinese Medicine Prevention and Treatment of Infectious Diseases, College of Integrative Medicine, Changchun University of Chinese Medicine, Changchun, 130117, PR China
| | - Rui Chen
- College of Basic Medical Sciences, Changchun University of Chinese Medicine, Changchun, 130117, PR China.
| | - Guangze Zhu
- Key Laboratory of Jilin Province for Traditional Chinese Medicine Prevention and Treatment of Infectious Diseases, College of Integrative Medicine, Changchun University of Chinese Medicine, Changchun, 130117, PR China.
| | - Jicheng Han
- Key Laboratory of Jilin Province for Traditional Chinese Medicine Prevention and Treatment of Infectious Diseases, College of Integrative Medicine, Changchun University of Chinese Medicine, Changchun, 130117, PR China.
| | - Liping Sun
- College of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130117, PR China; Center of Children's Clinic, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, 130022, PR China.
| |
Collapse
|
7
|
Spinardi JR, Thakkar KB, Welch VL, Jagun O, Kyaw MH. The need for novel influenza vaccines in low- and middle-income countries: A narrative review. Braz J Infect Dis 2025; 29:104465. [PMID: 39642677 PMCID: PMC11664161 DOI: 10.1016/j.bjid.2024.104465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 09/27/2024] [Accepted: 10/30/2024] [Indexed: 12/09/2024] Open
Abstract
Influenza viruses cause 3-5 million severe cases and 300,000-600,000 deaths worldwide. Most of the disease burden is in Low- and Middle-Income Countries (LMICs) owing to factors such as high population density, infrastructure challenges, poor quality healthcare, lack of consistent recommendations, less prioritization of all high-risk groups, and prevalent use of trivalent influenza vaccines. Although influenza vaccines are effective in reducing the annual influenza disease burden, existing vaccines have several limitations. In this narrative review, we address the unmet needs of existing influenza vaccines in LMICs in Africa, Asia Pacific, Latin America and the Middle East and discuss the characteristics of novel vaccines in clinical development. We also describe features of a successful vaccination program that LMICs could emulate to improve their current vaccination coverage and reduce the public health burden of influenza.
Collapse
Affiliation(s)
- Julia R Spinardi
- Vaccine Medical and Scientific Affairs, Emerging Markets, Pfizer Inc., São Paulo, SP, Brazil.
| | - Karan B Thakkar
- Vaccine Medical and Scientific Affairs, Emerging Markets, Pfizer Inc., Singapore, Singapore
| | - Verna L Welch
- Vaccine Medical and Scientific Affairs, Pfizer Inc., Pennsylvania, USA
| | - Oladayo Jagun
- Real World Strategy and Innovation, IQVIA Inc., New Jersey, USA
| | - Moe H Kyaw
- Vaccine Medical and Scientific Affairs, Emerging Markets, Pfizer Inc., Virginia, USA
| |
Collapse
|
8
|
Abu-Helalah M, Al-Shatnawi SF, Abu Lubad M, Al-Zayadneh E, Al-Hanaktah M, Harahsheh M, AL-Iede M, Nafi O, Yousef R, Almaaitah I, Ababneh M, AlZubi T, Abu Mahfouz R, Adaylah H, AlHajaj H, Al Tamimi M, Drysdale SB. The Epidemiology and Health Burdens of Influenza Infections Amongst Hospitalized Children Under 5 Years of Age in Jordan: A National Multi-Center Cross-Sectional Study. Vaccines (Basel) 2024; 13:12. [PMID: 39852790 PMCID: PMC11769506 DOI: 10.3390/vaccines13010012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 12/19/2024] [Accepted: 12/24/2024] [Indexed: 01/26/2025] Open
Abstract
BACKGROUND/OBJECTIVES Seasonal influenza is a significant global health concern, causing substantial morbidity and mortality, particularly among high-risk groups such as children under five years old. There is scarce local evidence from developing countries such as Jordan on the burden of influenza, which has limited preventive measures. This multi-center national cross-sectional study aimed to assess the epidemiological and clinical burden of influenza among hospitalized children under five years old in Jordan. METHODS Data were collected from 1000 participants across four hospitals between November 2022 and April 2023. Nasopharyngeal specimens were analyzed using multiplex RT-PCR to determine positivity for influenza A and B. RESULTS We found a 9.9% positivity rate, predominantly influenza A (8.4%), while influenza B was positive among 1.5% of the participants. Positivity rates were higher in older age groups, particularly children older than 2 years. Influenza-positive cases exhibited longer fever durations and higher rates of sore throat. There were no positive influenza cases among participants if they or any of their family members received the influenza vaccine, highlighting the vaccine's protective role. Logistic regression analysis identified maternal smoking during pregnancy as a significant predictor of influenza positivity. CONCLUSIONS The findings of this study underscore the need for enhanced vaccination efforts and public health policies targeting young children and pregnant women in Jordan. Expanding vaccination uptake could significantly mitigate the burden of influenza and its complications in this vulnerable population.
Collapse
Affiliation(s)
- Munir Abu-Helalah
- Department of Family and Community Medicine, Faculty of Medicine, University of Jordan, Amman 11942, Jordan
- Public Health Institute, The University of Jordan, Amman 11942, Jordan
| | - Samah F. Al-Shatnawi
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, P.O. Box 3030, Irbid 22110, Jordan;
| | - Mohammad Abu Lubad
- Department of Microbiology and Pathology, Faculty of Medicine, Mutah University, Karak 61710, Jordan;
| | - Enas Al-Zayadneh
- Department of Pediatrics, Faculty of Medicine, University of Jordan, Amman 11942, Jordan; (E.A.-Z.); (M.A.-I.)
| | | | | | - Montaha AL-Iede
- Department of Pediatrics, Faculty of Medicine, University of Jordan, Amman 11942, Jordan; (E.A.-Z.); (M.A.-I.)
| | - Omar Nafi
- Department of Pediatrics, Faculty of Medicine, Mutah University, Karak 61710, Jordan;
| | - Ruba Yousef
- Medical Department, MENA Center for Research and Development, Amman 11941, Jordan;
| | - Ihsan Almaaitah
- Pediatrics Department, Zarqa Governmental Hospital, Zarqa 13112, Jordan; (I.A.); (H.A.)
| | - Mai Ababneh
- MENA Center for Research and Development, Amman 11942, Jordan; (M.A.); (T.A.); (R.A.M.); (H.A.)
| | - Toqa AlZubi
- MENA Center for Research and Development, Amman 11942, Jordan; (M.A.); (T.A.); (R.A.M.); (H.A.)
| | - Rand Abu Mahfouz
- MENA Center for Research and Development, Amman 11942, Jordan; (M.A.); (T.A.); (R.A.M.); (H.A.)
| | - Heba Adaylah
- MENA Center for Research and Development, Amman 11942, Jordan; (M.A.); (T.A.); (R.A.M.); (H.A.)
| | - Hamzeh AlHajaj
- Pediatrics Department, Zarqa Governmental Hospital, Zarqa 13112, Jordan; (I.A.); (H.A.)
| | | | - Simon B. Drysdale
- Oxford Vaccine Group, Department of Pediatrics, University of Oxford, Oxford OX1 2JD, UK;
- The NIHR Oxford Biomedical Research Centre, Oxford OX3 7JX, UK
| |
Collapse
|
9
|
Han J, Yang C, Xiao Y, Li J, Jin N, Li Y. Influenza B virus: Target and acting mechanism of antiviral drugs. Microb Pathog 2024; 197:107051. [PMID: 39442816 DOI: 10.1016/j.micpath.2024.107051] [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: 04/07/2024] [Revised: 08/30/2024] [Accepted: 10/20/2024] [Indexed: 10/25/2024]
Abstract
The influenza B virus is one of the causes of seasonal influenza, which has a long history of existence in various populations. Adolescents, children, pregnant women, the elderly, as well as patients with major diseases such as high blood pressure, diabetes, and cancer, and those with low immunity are more susceptible to infection by the influenza virus. During the influenza seasons, the influenza B virus can cause significant harm and economic burden. At present, neuraminidase inhibitors, hemagglutinin inhibitors and RNA polymerase inhibitors are the main antiviral drugs that are used in the clinical treatment of influenza B. Due to the repeated use of antiviral drugs in recent years, the emergence of resistant strains of the influenza virus exacerbated. By combining anti-viral drugs with different mechanisms of action or using a combination of traditional Chinese medicine and chemical drugs, the problem of reduced drug sensitivity can be improved. This article introduces the drug targets of the influenza B virus and the mechanism of virus resistance. It also emphasizes the clinically used antiviral drugs and their mechanisms of action, thereby providing a reference basis for the development of new anti-influenza drugs.
Collapse
Affiliation(s)
- Jicheng Han
- Key Laboratory of Jilin Province for Traditional Chinese Medicine Prevention and Treatment of Infectious Diseases, College of Integrative Medicine, Changchun University of Chinese Medicine, Changchun, PR China
| | - Chunhui Yang
- Key Laboratory of Jilin Province for Traditional Chinese Medicine Prevention and Treatment of Infectious Diseases, College of Integrative Medicine, Changchun University of Chinese Medicine, Changchun, PR China
| | - Yan Xiao
- Key Laboratory of Jilin Province for Traditional Chinese Medicine Prevention and Treatment of Infectious Diseases, College of Integrative Medicine, Changchun University of Chinese Medicine, Changchun, PR China.
| | - Jingjing Li
- Key Laboratory of Jilin Province for Traditional Chinese Medicine Prevention and Treatment of Infectious Diseases, College of Integrative Medicine, Changchun University of Chinese Medicine, Changchun, PR China
| | - Ningyi Jin
- Key Laboratory of Jilin Province for Traditional Chinese Medicine Prevention and Treatment of Infectious Diseases, College of Integrative Medicine, Changchun University of Chinese Medicine, Changchun, PR China
| | - Yiquan Li
- Key Laboratory of Jilin Province for Traditional Chinese Medicine Prevention and Treatment of Infectious Diseases, College of Integrative Medicine, Changchun University of Chinese Medicine, Changchun, PR China.
| |
Collapse
|
10
|
Zhu A, Zhao B, Li J, Li X, Shi Q, Zhang X, Lu D, Yan D. Establishment of a Raman microsphere-based immunochromatographic method for the combined detection of influenza A and B viruses and SARS-CoV-2 antigen on a single T-line. RSC Adv 2024; 14:37498-37511. [PMID: 39582936 PMCID: PMC11582967 DOI: 10.1039/d4ra05483k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 10/15/2024] [Indexed: 11/26/2024] Open
Abstract
A simple and rapid method based on Raman microsphere immunochromatography was developed in this study for the simultaneous detection of influenza A and B viruses and SARS-CoV-2 on a single test T-line. Three types of Raman microspheres with different Raman characteristics were used as the signal sources and were labelled with monoclonal antibodies against FluA, FluB and SARS-CoV-2, respectively. A mixture of antibodies containing anti-FluA monoclonal antibody, anti-FluB monoclonal antibody and anti-SARS-CoV-2 was sprayed on the detection line (T), and goat polyclonal antibody to chicken (IgY) encapsulated on the quality control line (C), for qualitative detection of these three viruses by the double antibody sandwich method. The results demonstrated that the LOD values were 0.5 ng mL-1 for FluA, 0.25 ng mL-1 for FluB, and 0.5 ng mL-1 for SARS-CoV-2. The method showed good repeatability for the respiratory viral antigens, with CV values below 15%. Oxymetazoline and commonly used oral medications did not interfere with the test results; the strips did not cross-react with common respiratory virus antigens, demonstrating good specificity. This method does not require any complicated pre-treatment, and all three viruses can be detected simultaneously by titrating one sample, which improves the detection efficiency. The Respiratory Pathogen Multiplex provides a scientific basis for preventing and controlling the spread of respiratory diseases by analyzing data to understand epidemiological trends and the spread of pathogens.
Collapse
Affiliation(s)
- Aolin Zhu
- Pharmacy Academy of Xinjiang Medical University Urumqi 830054 People's Republic of China
| | - Binbin Zhao
- Pharmacy Academy of Xinjiang Medical University Urumqi 830054 People's Republic of China
| | - Jiutong Li
- Pharmacy Academy of Xinjiang Medical University Urumqi 830054 People's Republic of China
- Xinjiang Xingyi Bio-Science Co., Ltd Urumqi 830011 China
| | - Xinxia Li
- Pharmacy Academy of Xinjiang Medical University Urumqi 830054 People's Republic of China
- Key Laboratory of High Incidence Disease Research in Xinjiang (Xinjiang Medical University), Ministry of Education Urumqi 830054 China
| | - Qian Shi
- Department of Clinical Laboratory, Hospital of Xinjiang Production and Construction Corps No. 232, Qingnian Road, Tianshan District Urumqi Xinjiang China
| | - Xin Zhang
- Department of Clinical Laboratory, Hospital of Xinjiang Production and Construction Corps No. 232, Qingnian Road, Tianshan District Urumqi Xinjiang China
| | - Dongmei Lu
- Respiratory and Critical Care Medicine, People's Hospital of Xinjiang Uygur Autonomous Region Urumqi Xinjiang 830000 China
| | - Dong Yan
- Pharmacy Academy of Xinjiang Medical University Urumqi 830054 People's Republic of China
- Xinjiang Key Laboratory of Biopharmaceuticals and Medical Devices Urumqi Xinjiang China
| |
Collapse
|
11
|
Hayashi K, Maeda H, Hayashi H, Suzuki A, Nakaya Y, Sato M, Hayashi K, Kobayashi Y. Polymyalgia Rheumatica Following Influenza B Infection: A Case Report. Cureus 2024; 16:e70671. [PMID: 39493100 PMCID: PMC11528242 DOI: 10.7759/cureus.70671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/01/2024] [Indexed: 11/05/2024] Open
Abstract
We describe a case of polymyalgia rheumatica (PMR) following an influenza B infection. The patient was a 71-year-old woman who developed a fever above 38.0°C and was diagnosed with influenza B, confirmed by a rapid antigen test. Although the fever resolved after three days, she continued to experience neck pain, back pain, and joint pain, particularly in both shoulders and hips. She also reported morning joint stiffness lasting for more than an hour and occasional low-grade fever. She presented to our hospital 25 days after the onset of symptoms. Blood tests revealed elevated C-reactive protein and erythrocyte sedimentation rate, but levels of creatine phosphokinase, rheumatoid factor, and anti-cyclic citrullinated peptide were not elevated. She was diagnosed with PMR and treated with prednisolone (PSL) 15 mg/day. The response to steroids was remarkably good, and PSL was tapered over six months. PMR is believed to result from an immune-mediated process and may be associated with certain human leukocyte antigen haplotypes. Additionally, PMR is sometimes preceded by an infection. To date, there have been very few reports suggesting a connection between influenza B and PMR, underscoring the need for further case accumulation.
Collapse
Affiliation(s)
- Koji Hayashi
- Department of Rehabilitation Medicine, Fukui General Hospital, Fukui, JPN
| | - Hiroaki Maeda
- Department of Rehabilitation Medicine, Fukui General Hospital, Fukui, JPN
| | - Hiromi Hayashi
- Department of Rehabilitation Medicine, Fukui General Hospital, Fukui, JPN
| | - Asuka Suzuki
- Department of Rehabilitation Medicine, Fukui General Hospital, Fukui, JPN
| | - Yuka Nakaya
- Department of Rehabilitation Medicine, Fukui General Hospital, Fukui, JPN
| | - Mamiko Sato
- Department of Rehabilitation Medicine, Fukui General Hospital, Fukui, JPN
- Graduate School of Health Science, Fukui Health Science University, Fukui, JPN
| | - Kouji Hayashi
- Graduate School of Health Science, Fukui Health Science University, Fukui, JPN
| | - Yasutaka Kobayashi
- Graduate School of Health Science, Fukui Health Science University, Fukui, JPN
| |
Collapse
|
12
|
Acocal-Juárez E, Márquez-Domínguez L, Vallejo-Ruíz V, Cedillo L, Santos-López G. Baloxavir Resistance Markers in Influenza A and B Viruses in the Americas. Drug Healthc Patient Saf 2024; 16:105-113. [PMID: 39296541 PMCID: PMC11410037 DOI: 10.2147/dhps.s470868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 07/20/2024] [Indexed: 09/21/2024] Open
Abstract
Aim Influenza control demands multifaceted strategies, including antiviral drugs. Baloxavir, a recent addition to influenza treatment, acts as an inhibitor of the Polymerase acid (PA) component of the viral polymerase. However, mutations associated with resistance have been identified. Purpose This study analyzed PA gene sequences of influenza A and B viruses (IAV and IBV, respectively) reported in the Americas, retrieved from databases published until May 2023, to identify primary markers of resistance to baloxavir. Patients and Methods PA gene sequences were obtained from the GISAID and NCBI databases, focusing on countries in the Americas with 500 or more sequences for IAV, and 50 or more sequences for IBV. Results Of the 58,816 PA sequences analyzed for IAV, only 55 (0.1%) harbored resistance markers, representing approximately 1 in 1000 occurrence. The most frequent markers were I38V (21 cases) and I38M (7 cases) at position 38 of PA, followed by E199G (9 cases) at position 199. For IBV, 14,684 sequences were analyzed, of which only eight presented a resistance marker (0.05%). Five sequences had the M34I marker, while the remaining three had the I38V marker. While frequency of resistance markers in PA is comparable to other regions, these results highlight the need for enhanced sequencing efforts, particularly in Latin America. Such efforts would serve to intensify influenza surveillance and inform public health interventions. Conclusion While baloxavir demonstrates efficacy against influenza, resistance markers have been identified, including pre-existing ones. Our study adds eight (IAV: six and IBV: two) new spontaneously occurring substitutions to the existing literature, highlighting the need for continued surveillance. Among these, I38M stands out due to its significant tenfold reduction in drug susceptibility. Therefore, vigilant monitoring of these resistance markers in IAV and IBV remains crucial for maintaining baloxavir's effectiveness and informing future public health interventions.
Collapse
Affiliation(s)
- Erick Acocal-Juárez
- Centro de Investigaciones en Ciencias Microbiológicas, Benemérita Universidad Autónoma de Puebla, Puebla Pue, Mexico
- Laboratorio de Biología Molecular y Virología, Centro de Investigación Biomédica de Oriente, Instituto Mexicano del Seguro Social, Puebla, Mexico
- Centro de Detección Biomolecular, Benemérita Universidad Autónoma de Puebla México, Puebla Pue, Mexico
| | - Luis Márquez-Domínguez
- Laboratorio de Biología Molecular y Virología, Centro de Investigación Biomédica de Oriente, Instituto Mexicano del Seguro Social, Puebla, Mexico
| | - Verónica Vallejo-Ruíz
- Laboratorio de Biología Molecular y Virología, Centro de Investigación Biomédica de Oriente, Instituto Mexicano del Seguro Social, Puebla, Mexico
| | - Lilia Cedillo
- Centro de Detección Biomolecular, Benemérita Universidad Autónoma de Puebla México, Puebla Pue, Mexico
| | - Gerardo Santos-López
- Laboratorio de Biología Molecular y Virología, Centro de Investigación Biomédica de Oriente, Instituto Mexicano del Seguro Social, Puebla, Mexico
| |
Collapse
|
13
|
Pekarek MJ, Weaver EA. Influenza B Virus Vaccine Innovation through Computational Design. Pathogens 2024; 13:755. [PMID: 39338946 PMCID: PMC11434669 DOI: 10.3390/pathogens13090755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 08/26/2024] [Accepted: 08/31/2024] [Indexed: 09/30/2024] Open
Abstract
As respiratory pathogens, influenza B viruses (IBVs) cause a significant socioeconomic burden each year. Vaccine and antiviral development for influenza viruses has historically viewed IBVs as a secondary concern to influenza A viruses (IAVs) due to their lack of animal reservoirs compared to IAVs. However, prior to the global spread of SARS-CoV-2, the seasonal epidemics caused by IBVs were becoming less predictable and inducing more severe disease, especially in high-risk populations. Globally, researchers have begun to recognize the need for improved prevention strategies for IBVs as a primary concern. This review discusses what is known about IBV evolutionary patterns and the effect of the spread of SARS-CoV-2 on these patterns. We also analyze recent advancements in the development of novel vaccines tested against IBVs, highlighting the promise of computational vaccine design strategies when used to target both IBVs and IAVs and explain why these novel strategies can be employed to improve the effectiveness of IBV vaccines.
Collapse
Affiliation(s)
| | - Eric A. Weaver
- Nebraska Center for Virology, School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA;
| |
Collapse
|
14
|
Takazono T, Ito G, Hosogaya N, Iwanaga N, Komeda T, Kobayashi M, Kitanishi Y, Ogura E, Mukae H. Comparison of the Effectiveness of Baloxavir and Oseltamivir in Outpatients With Influenza B. Influenza Other Respir Viruses 2024; 18:e70002. [PMID: 39189087 PMCID: PMC11347862 DOI: 10.1111/irv.70002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 08/08/2024] [Accepted: 08/10/2024] [Indexed: 08/28/2024] Open
Abstract
This retrospective cohort study analyzed data from a Japanese health insurance database to assess the effectiveness of baloxavir (n = 4822) for preventing severe events compared with oseltamivir (n = 10,523) in patients with influenza B. The primary endpoint was hospitalization incidence (Days 2-14). The secondary endpoints included intravenous antibacterial drug use, pneumonia hospitalization, heart failure hospitalization, inhalational oxygen requirement, and use of other anti-influenza drugs. The hospitalization incidence was significantly lower with baloxavir (0.15% vs. 0.37%; risk ratio: 2.48, 95% confidence interval: 1.13-5.43). Pneumonia and additional anti-influenza therapy were also less frequent with baloxavir, thus supporting its use. Trial Registration: UMIN Clinical Trials Registry Study ID: UMIN000051382.
Collapse
Affiliation(s)
- Takahiro Takazono
- Department of Respiratory MedicineNagasaki University HospitalNagasakiJapan
- Department of Infectious DiseasesNagasaki University Graduate School of Biomedical SciencesNagasakiJapan
| | - Genta Ito
- Data Science Department, Shionogi & Co., Ltd.OsakaJapan
| | - Naoki Hosogaya
- Clinical Research CenterNagasaki University HospitalNagasakiJapan
| | - Naoki Iwanaga
- Department of Respiratory MedicineNagasaki University HospitalNagasakiJapan
| | - Takuji Komeda
- Data Science Department, Shionogi & Co., Ltd.OsakaJapan
| | | | | | - Eriko Ogura
- Global Development Division, Shionogi & Co., Ltd.TokyoJapan
| | - Hiroshi Mukae
- Department of Respiratory MedicineNagasaki University HospitalNagasakiJapan
- Department of Respiratory MedicineNagasaki University Graduate School of Biomedical SciencesNagasakiJapan
| |
Collapse
|
15
|
Xing G, Zheng Q. Insights into the specific feature of the electrostatic recognition binding mechanism between BM2 and BM1: a molecular dynamics simulation study. Phys Chem Chem Phys 2024; 26:22726-22738. [PMID: 39161312 DOI: 10.1039/d4cp01936a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/21/2024]
Abstract
Matrix protein 2 (M2) and matrix protein 1 (M1) of the influenza B virus are two important proteins, and the interactions between BM2 and BM1 play an important role in the process of virus assembly and replication. However, the interaction details between BM2 and BM1 are still unclear at the atomic level. Here, we constructed the BM2-BM1 complex system using homology modelling and molecular docking methods. Molecular dynamics (MD) simulations were used to illustrate the binding mechanism between BM2 and BM1. The results identify that the eight polar residues (E88B, E89B, H119BM1, E94B, R101BM1, K102BM1, R105BM1, and E104B) play an important role in stabilizing the binding through the formation of hydrogen bond networks and salt-bridge interactions at the binding interface. Furthermore, based on the simulation results and the experimental facts, the mutation experiments were designed to verify the influence of the mutation of residues both within and outside the effector domain. The mutations directly or indirectly disrupt interactions between polar residues, thus affecting viral assembly and replication. The results could help us understand the details of the interactions between BM2 and BM1 and provide useful information for the anti-influenza drug design.
Collapse
Affiliation(s)
- Guixuan Xing
- Institute of Theoretical Chemistry, College of Chemistry, Jilin University, Changchun 130023, China
| | - Qingchuan Zheng
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China.
- Institute of Theoretical Chemistry, College of Chemistry, Jilin University, Changchun 130023, China
| |
Collapse
|
16
|
Zhang YN, Gomes KB, Lee YZ, Ward G, Xie B, Auclair S, He L, Zhu J. A Single-Component Multilayered Self-Assembling Protein Nanoparticle Vaccine Based on Extracellular Domains of Matrix Protein 2 against Both Influenza A and B. Vaccines (Basel) 2024; 12:975. [PMID: 39340007 PMCID: PMC11435909 DOI: 10.3390/vaccines12090975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 08/14/2024] [Accepted: 08/24/2024] [Indexed: 09/30/2024] Open
Abstract
The development of an effective and broadly protective influenza vaccine against circulating and emerging strains remains elusive. In this study, we evaluated a potentially universal influenza vaccine based on single-component self-assembling protein nanoparticles (1c-SApNPs) presenting the conserved matrix protein 2 ectodomain (M2e) from influenza A and B viruses (IAV and IBV, respectively). We previously designed a tandem antigen comprising three IAV M2e domains of human, avian/swine, and human/swine origins (termed M2ex3). The M2ex3-presenting 1c-SApNPs conferred complete protection in mice against sequential lethal challenges with H1N1 and H3N2. To broaden this protection to cover IBVs, we designed a series of antigens incorporating different arrangements of three IAV M2e domains and three copies of IBV M2e. Tandem repeats of IAV and IBV (termed influenza A-B) M2e arrayed on the I3-01v9a 60-mer 1c-SApNP, when formulated with an oil-in-water emulsion adjuvant, generated greater M2e-specific immunogenicity and protective efficacy than the soluble influenza A-B M2e trimer, indicated by higher survival rates and reduced weight loss post-challenge. Importantly, one of the influenza A-B M2e SApNP constructs elicited 100% protection against a lethal influenza A/Puerto Rico/8/1934 (H1N1) challenge in mice and 70% protection against a lethal influenza B/Florida/4/2006 (Yamagata lineage) challenge, the latter of which has not been reported in the literature to date. Our study thus provides a promising M2e-based single-component universal vaccine candidate against the two major types of influenza virus circulating in humans.
Collapse
Affiliation(s)
- Yi-Nan Zhang
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA; (Y.-N.Z.); (Y.-Z.L.); (G.W.); (B.X.); (S.A.); (L.H.)
| | | | - Yi-Zong Lee
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA; (Y.-N.Z.); (Y.-Z.L.); (G.W.); (B.X.); (S.A.); (L.H.)
| | - Garrett Ward
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA; (Y.-N.Z.); (Y.-Z.L.); (G.W.); (B.X.); (S.A.); (L.H.)
| | - Bomin Xie
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA; (Y.-N.Z.); (Y.-Z.L.); (G.W.); (B.X.); (S.A.); (L.H.)
| | - Sarah Auclair
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA; (Y.-N.Z.); (Y.-Z.L.); (G.W.); (B.X.); (S.A.); (L.H.)
| | - Linling He
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA; (Y.-N.Z.); (Y.-Z.L.); (G.W.); (B.X.); (S.A.); (L.H.)
| | - Jiang Zhu
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA; (Y.-N.Z.); (Y.-Z.L.); (G.W.); (B.X.); (S.A.); (L.H.)
- Uvax Bio, LLC, Newark, DE 19702, USA;
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| |
Collapse
|
17
|
Schwab LSU, Do THT, Pilapitiya D, Koutsakos M. Dissemination of influenza B virus to the lower respiratory tract of mice is restricted by the interferon response. J Virol 2024; 98:e0160423. [PMID: 38780249 PMCID: PMC11237704 DOI: 10.1128/jvi.01604-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 04/26/2024] [Indexed: 05/25/2024] Open
Abstract
The global burden of disease caused by influenza B virus (IBV) is substantial; however, IBVs remain overlooked. Understanding host-pathogen interactions and establishing physiologically relevant models of infection are important for the development and assessment of therapeutics and vaccines against IBV. In this study, we assessed an upper respiratory tract (URT)-restricted model of mouse IBV infection, comparing it to the conventional administration of the virus to the total respiratory tract (TRT). We found that URT infections caused by different strains of IBV disseminate to the trachea but resulted in limited dissemination of IBV to the lungs. Infection of the URT did not result in weight loss or systemic inflammation even at high inoculum doses and despite robust viral replication in the nose. Dissemination of IBV to the lungs was enhanced in mice lacking functional type I IFN receptor (IFNAR2), but not IFNγ. Conversely, in mice expressing the IFN-inducible gene Mx1, we found reduced IBV replication in the lungs and reduced dissemination of IBV from the URT to the lungs. Inoculation of IBV in both the URT and TRT resulted in seroconversion against IBV. However, priming at the TRT conferred superior protection from a heterologous lethal IBV challenge compared to URT priming, as determined by improved survival rates and reduced viral replication throughout the respiratory tract. Overall, our study establishes a URT-restricted IBV infection model, highlights the critical role of IFNs in limiting dissemination of IBV to the lungs, and also demonstrates that the lack of viral replication in the lungs may impact protection from subsequent infections. IMPORTANCE Our study investigated how influenza B virus (IBV) spreads from the nose to the lungs of mice and the impact this has on disease and protection from re-infection. We found that when applied to the nose only, IBV does not spread very efficiently to the lungs in a process controlled by the interferon response. Priming immunity at the nose only resulted in less protection from re-infection than priming immunity at both the nose and lungs. These insights can guide the development of potential therapies targeting the interferon response as well as of intranasal vaccines against IBV.
Collapse
Affiliation(s)
- Lara S. U. Schwab
- Department of Microbiology and Immunology, University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Parkville, Victoria, Australia
| | - Thi H. T. Do
- Department of Microbiology and Immunology, University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Parkville, Victoria, Australia
| | - Devaki Pilapitiya
- Department of Microbiology and Immunology, University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Parkville, Victoria, Australia
| | - Marios Koutsakos
- Department of Microbiology and Immunology, University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Parkville, Victoria, Australia
| |
Collapse
|
18
|
Wolters RM, Ferguson JA, Nuñez IA, Chen EE, Sornberger T, Myers L, Oeverdieck S, Raghavan SSR, Kona C, Handal LS, Esilu TE, Davidson E, Doranz BJ, Engdahl TB, Kose N, Williamson LE, Creech CB, Gibson-Corley KN, Ward AB, Crowe JE. Isolation of human antibodies against influenza B neuraminidase and mechanisms of protection at the airway interface. Immunity 2024; 57:1413-1427.e9. [PMID: 38823390 PMCID: PMC11440431 DOI: 10.1016/j.immuni.2024.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 01/16/2024] [Accepted: 05/06/2024] [Indexed: 06/03/2024]
Abstract
Influenza B viruses (IBVs) comprise a substantial portion of the circulating seasonal human influenza viruses. Here, we describe the isolation of human monoclonal antibodies (mAbs) that recognized the IBV neuraminidase (NA) glycoprotein from an individual following seasonal vaccination. Competition-binding experiments suggested the antibodies recognized two major antigenic sites. One group, which included mAb FluB-393, broadly inhibited IBV NA sialidase activity, protected prophylactically in vivo, and bound to the lateral corner of NA. The second group contained an active site mAb, FluB-400, that broadly inhibited IBV NA sialidase activity and virus replication in vitro in primary human respiratory epithelial cell cultures and protected against IBV in vivo when administered systemically or intranasally. Overall, the findings described here shape our mechanistic understanding of the human immune response to the IBV NA glycoprotein through the demonstration of two mAb delivery routes for protection against IBV and the identification of potential IBV therapeutic candidates.
Collapse
Affiliation(s)
- Rachael M Wolters
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - James A Ferguson
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Ivette A Nuñez
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Elaine E Chen
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Ty Sornberger
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Luke Myers
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Svearike Oeverdieck
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Sai Sundar Rajan Raghavan
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Chandrahaas Kona
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Laura S Handal
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | | | | | | | - Taylor B Engdahl
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Nurgun Kose
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Lauren E Williamson
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - C Buddy Creech
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Katherine N Gibson-Corley
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| | - Andrew B Ward
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA.
| | - James E Crowe
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| |
Collapse
|
19
|
Yahyavi I, Edalat F, Pirbonyeh N, Letafati A, Sattarahmady N, Heli H, Moattari A. Nucleic acid-based electrochemical biosensor for detection of influenza B by gold nanoparticles. J Mol Recognit 2024; 37:e3073. [PMID: 38126612 DOI: 10.1002/jmr.3073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 12/03/2023] [Accepted: 12/05/2023] [Indexed: 12/23/2023]
Abstract
The influenza virus is a pervasive pathogen that exhibits increased prevalence during colder seasons, resulting in a significant annual occurrence of infections. Notably, pharmaceutical interventions effective against influenza A strains often exhibit limited efficacy against influenza B variants. Against this backdrop, the need for innovative approaches to accurately and swiftly differentiate and detect influenza B becomes evident. Biosensors play a pivotal role in this detection process, offering rapid, specific, and sensitive identification of the virus, facilitating timely intervention and containment efforts. Oligonucleotide sequences targeting the conserved B/Victoria/2/87 influenza virus NP region were designed. Nasopharyngeal swabs were collected from patients suspected of influenza virus infection, and viral RNA was extracted. RNA quality was assessed through one-step PCR. cDNA synthesis was performed using random hexamers, and real-time PCR quantified the influenza genome. Gold nanoparticles were immobilized on a surface to immobilize the specific DNA probe, and electrochemical hybridization was electrochemically followed. The biosensor exhibited high selectivity and effective distinction of complementary sequences from mismatches and influenza virus cDNA genome. The biosensor successfully detected the influenza B virus genome in real samples. Non-influenza samples yielded no significant hybridization signals. The comparison between the results obtained from the biosensor and real-time PCR revealed full agreement of these methods. The biosensor utilized electrochemical detection of hybridization and proved effective in detecting the influenza B virus genome with high specificity, sensitivity, and selectivity. Comparative analysis with real-time PCR underscored the accuracy and potential applicability of the biosensor in rapid and specific virus detection. This innovative approach holds promise for future diagnostic and epidemiological applications in detecting influenza B virus and other pathogens.
Collapse
Affiliation(s)
- Isar Yahyavi
- Department of Bacteriology and Virology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Ceinge Biotechnologie Avanzate, Naples, Italy
| | - Fahime Edalat
- Department of Bacteriology and Virology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Neda Pirbonyeh
- Department of Bacteriology and Virology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Microbiology, Burn and Wound Healing Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Arash Letafati
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Naghmeh Sattarahmady
- Department of Medical Physics, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Nanomedicine and Nanobiology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hossein Heli
- Nanomedicine and Nanobiology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Afagh Moattari
- Department of Bacteriology and Virology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
20
|
Liu A, Zhang H, Zheng Q, Wang S. The Potential of Cyclodextrins as Inhibitors for the BM2 Protein: An In Silico Investigation. Molecules 2024; 29:620. [PMID: 38338365 PMCID: PMC10856705 DOI: 10.3390/molecules29030620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 01/23/2024] [Accepted: 01/26/2024] [Indexed: 02/12/2024] Open
Abstract
The influenza BM2 transmembrane domain (BM2TM), an acid-activated proton channel, is an attractive antiviral target due to its essential roles during influenza virus replication, whereas no effective inhibitors have been reported for BM2. In this study, we draw inspiration from the properties of cyclodextrins (CDs) and hypothesize that CDs of appropriate sizes may possess the potential to act as inhibitors of the BM2TM proton channel. To explore this possibility, molecular dynamics simulations were employed to assess their inhibitory capabilities. Our findings reveal that CD4, CD5, and CD6 are capable of binding to the BM2TM proton channel, resulting in disrupted water networks and reduced hydrogen bond occupancy between H19 and the solvent within the BM2TM channel necessary for proton conduction. Notably, CD4 completely obstructs the BM2TM water channel. Based on these observations, we propose that CD4, CD5, and CD6 individually contribute to diminishing the proton transfer efficiency of the BM2 protein, and CD4 demonstrates promising potential as an inhibitor for the BM2 proton channel.
Collapse
Affiliation(s)
- Aijun Liu
- Institute of Theoretical Chemistry, College of Chemistry, Jilin University, Changchun 130023, China; (A.L.); (H.Z.)
| | - Hao Zhang
- Institute of Theoretical Chemistry, College of Chemistry, Jilin University, Changchun 130023, China; (A.L.); (H.Z.)
| | - Qingchuan Zheng
- School of Pharmaceutical Sciences, Jilin University, Changchun, 130021, China
| | - Song Wang
- Institute of Theoretical Chemistry, College of Chemistry, Jilin University, Changchun 130023, China; (A.L.); (H.Z.)
| |
Collapse
|
21
|
Luo R, Lv C, Wang T, Deng X, Sima M, Guo J, Qi J, Sun W, Shen B, Li Y, Yue D, Gao Y. A potential Chinese medicine monomer against influenza A virus and influenza B virus: isoquercitrin. Chin Med 2023; 18:144. [PMID: 37919750 PMCID: PMC10621105 DOI: 10.1186/s13020-023-00843-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 09/27/2023] [Indexed: 11/04/2023] Open
Abstract
BACKGROUND Influenza viruses, especially Influenza A virus and Influenza B virus, are respiratory pathogens and can cause seasonal epidemics and pandemics. Severe influenza viruses infection induces strong host-defense response and excessive inflammatory response, resulting in acute lung damage, multiple organ failure and high mortality. Isoquercitrin is a Chinese medicine monomer, which was reported to have multiple biological activities, including antiviral activity against HSV, IAV, SARS-CoV-2 and so on. Aims of this study were to assess the in vitro anti-IAV and anti-IBV activity, evaluate the in vivo protective efficacy against lethal infection of the influenza virus and searched for the more optimal method of drug administration of isoquercitrin. METHODS In vitro infection model (MDCK and A549 cells) and mouse lethal infection model of Influenza A virus and Influenza B virus were used to evaluate the antiviral activity of isoquercitrin. RESULTS Isoquercitrin could significantly suppress the replication in vitro and in vivo and reduced the mortality of mouse lethal infection models. Compared with virus infection group, isoquercitrin mitigated lung and multiple organ damage. Moreover, isoquercitrin blocked hyperproduction of cytokines induced by virus infection via inactivating NF-κB signaling. Among these routes of isoquercitrin administration, intramuscular injection is a better drug delivery method. CONCLUSION Isoquercitrin is a potential Chinese medicine monomer Against Influenza A Virus and Influenza B Virus infection.
Collapse
Affiliation(s)
- Rongbo Luo
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130122, China
| | - Chaoxiang Lv
- The Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Tiecheng Wang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130122, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, 225009, People's Republic of China
| | - Xiuwen Deng
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130122, China
- College of Integrated Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, 130117, China
| | - Mingwei Sima
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130122, China
- College of Integrated Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, 130117, China
| | - Jin Guo
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130122, China
- College of Life Sciences, Shandong Normal University, Jinan, 250014, China
| | - Jing Qi
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130122, China
- College of Life Sciences, Northeast Normal University, Changchun, 130021, China
| | - Weiyang Sun
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130122, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, 225009, People's Republic of China
| | - Beilei Shen
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130122, China
| | - Yuanguo Li
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130122, China
| | - Donghui Yue
- School of Medical Sciences, Changchun University of Chinese Medicine, Changchun, Jilin, 130117, China.
| | - Yuwei Gao
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130122, China.
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, 225009, People's Republic of China.
- College of Integrated Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, 130117, China.
- College of Life Sciences, Shandong Normal University, Jinan, 250014, China.
| |
Collapse
|
22
|
Pekarek MJ, Weaver EA. Existing Evidence for Influenza B Virus Adaptations to Drive Replication in Humans as the Primary Host. Viruses 2023; 15:2032. [PMID: 37896807 PMCID: PMC10612074 DOI: 10.3390/v15102032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 09/25/2023] [Accepted: 09/28/2023] [Indexed: 10/29/2023] Open
Abstract
Influenza B virus (IBV) is one of the two major types of influenza viruses that circulate each year. Unlike influenza A viruses, IBV does not harbor pandemic potential due to its lack of historical circulation in non-human hosts. Many studies and reviews have highlighted important factors for host determination of influenza A viruses. However, much less is known about the factors driving IBV replication in humans. We hypothesize that similar factors influence the host restriction of IBV. Here, we compile and review the current understanding of host factors crucial for the various stages of the IBV viral replication cycle. While we discovered the research in this area of IBV is limited, we review known host factors that may indicate possible host restriction of IBV to humans. These factors include the IBV hemagglutinin (HA) protein, host nuclear factors, and viral immune evasion proteins. Our review frames the current understanding of IBV adaptations to replication in humans. However, this review is limited by the amount of research previously completed on IBV host determinants and would benefit from additional future research in this area.
Collapse
Affiliation(s)
| | - Eric A. Weaver
- Nebraska Center for Virology, School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA;
| |
Collapse
|
23
|
Momont C, Dang HV, Zatta F, Hauser K, Wang C, di Iulio J, Minola A, Czudnochowski N, De Marco A, Branch K, Donermeyer D, Vyas S, Chen A, Ferri E, Guarino B, Powell AE, Spreafico R, Yim SS, Balce DR, Bartha I, Meury M, Croll TI, Belnap DM, Schmid MA, Schaiff WT, Miller JL, Cameroni E, Telenti A, Virgin HW, Rosen LE, Purcell LA, Lanzavecchia A, Snell G, Corti D, Pizzuto MS. A pan-influenza antibody inhibiting neuraminidase via receptor mimicry. Nature 2023:10.1038/s41586-023-06136-y. [PMID: 37258672 DOI: 10.1038/s41586-023-06136-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 04/26/2023] [Indexed: 06/02/2023]
Abstract
Rapidly evolving influenza A viruses (IAVs) and influenza B viruses (IBVs) are major causes of recurrent lower respiratory tract infections. Current influenza vaccines elicit antibodies predominantly to the highly variable head region of haemagglutinin and their effectiveness is limited by viral drift1 and suboptimal immune responses2. Here we describe a neuraminidase-targeting monoclonal antibody, FNI9, that potently inhibits the enzymatic activity of all group 1 and group 2 IAVs, as well as Victoria/2/87-like, Yamagata/16/88-like and ancestral IBVs. FNI9 broadly neutralizes seasonal IAVs and IBVs, including the immune-evading H3N2 strains bearing an N-glycan at position 245, and shows synergistic activity when combined with anti-haemagglutinin stem-directed antibodies. Structural analysis reveals that D107 in the FNI9 heavy chain complementarity-determinant region 3 mimics the interaction of the sialic acid carboxyl group with the three highly conserved arginine residues (R118, R292 and R371) of the neuraminidase catalytic site. FNI9 demonstrates potent prophylactic activity against lethal IAV and IBV infections in mice. The unprecedented breadth and potency of the FNI9 monoclonal antibody supports its development for the prevention of influenza illness by seasonal and pandemic viruses.
Collapse
Affiliation(s)
| | - Ha V Dang
- Vir Biotechnology, San Francisco, CA, USA
| | - Fabrizia Zatta
- Humabs Biomed SA, a subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | | | | | | | - Andrea Minola
- Humabs Biomed SA, a subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | | | - Anna De Marco
- Humabs Biomed SA, a subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | | | | | | | - Alex Chen
- Vir Biotechnology, San Francisco, CA, USA
| | | | - Barbara Guarino
- Humabs Biomed SA, a subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | | | | | | | | | | | | | - Tristan I Croll
- Cambridge Institute for Medical Research, Department of Haematology, University of Cambridge, Cambridge, UK
| | - David M Belnap
- School of Biological Sciences, Department of Biochemistry, University of Utah, Salt Lake City, UT, USA
| | - Michael A Schmid
- Humabs Biomed SA, a subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | | | | | - Elisabetta Cameroni
- Humabs Biomed SA, a subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | | | - Herbert W Virgin
- Vir Biotechnology, San Francisco, CA, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | | | | | | | | | - Davide Corti
- Humabs Biomed SA, a subsidiary of Vir Biotechnology, Bellinzona, Switzerland.
| | | |
Collapse
|
24
|
Zong H, Zhang S, Shang X, Jiang H, Zhao Z, Chen S, Wang X, Wang Y, Jiang Y, Li X, Tan L, Liu P, Lv Q, Li Y. Development of an AlphaLISA assay for sensitive and accurate detection of influenza B virus. Front Med (Lausanne) 2023; 10:1155551. [PMID: 37215702 PMCID: PMC10196263 DOI: 10.3389/fmed.2023.1155551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 04/20/2023] [Indexed: 05/24/2023] Open
Abstract
Objective Influenza B virus (IBV) is highly contagious, spreads rapidly, and causes seasonal epidemic respiratory disease in the human population, especially in immunocompromised people and young children. Clinical manifestations in this high-risk population are often more severe than in immunocompetent hosts and sometimes atypical. Therefore, rapid, and accurate detection of IBV is important. Methods An amplified luminescent proximity homogeneous assay linked immunosorbent assay (AlphaLISA) was developed for detection of IBV by optimizing the ratio of IBV antibody-labeled receptor beads, streptavidin-conjugated donor beads and biotinylated IBV antibody, as well as the optimal temperature and time conditions for incubation. Assay sensitivity, specificity and reproducibility were evaluated. A total of 228 throat swab samples and inactivated influenza B virus were tested by AlphaLISA and lateral flow colloidal gold-based immunoassay (LFIA). Results AlphaLISA produced the best results for detection of inactivated influenza B virus when IBV antibody-labeled acceptor beads were 50 μg/ mL, streptavidin-conjugated donor beads were 40 μg/mL, and biotinylated IBV antibody was 0.5 μg/mL at 37°C for 15-10 min. Under these conditions, AlphaLISA had a limit of detection of 0.24 ng/mL for the detection of influenza B nucleoprotein, did not cross react with other common respiratory viruses, and showed good reproducibility with inter-assay coefficient of variation (CV) and intra-assay CV < 5%. The results of 228 clinical throat swab samples showed good agreement between AlphaLISA and LFIA (Kappa = 0.982), and AlphaLISA showed better sensitivity than LFIA for detecting inactivated influenza B virus. Conclusion AlphaLISA showed higher sensitivity and throughput in the detection of IBV and can be used for IBV diagnosis and epidemic control.
Collapse
Affiliation(s)
- Huijun Zong
- The PLA 307 Clinical College of Anhui Medical University, The Fifth Clinical Medical College of Anhui Medical University, Hefei, China
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
- Department of Intensive Care Unit, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Shengwei Zhang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| | - Xueyi Shang
- Department of Intensive Care Unit, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Hua Jiang
- The PLA 307 Clinical College of Anhui Medical University, The Fifth Clinical Medical College of Anhui Medical University, Hefei, China
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| | - Zhongpeng Zhao
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| | - Shaolong Chen
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| | - Xin Wang
- Department of Intensive Care Unit, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Ye Wang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| | - Yongqiang Jiang
- The PLA 307 Clinical College of Anhui Medical University, The Fifth Clinical Medical College of Anhui Medical University, Hefei, China
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| | - Xinyu Li
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| | - Lingyun Tan
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| | - Peng Liu
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| | - Qingyu Lv
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| | - Yan Li
- The PLA 307 Clinical College of Anhui Medical University, The Fifth Clinical Medical College of Anhui Medical University, Hefei, China
- Department of Intensive Care Unit, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
25
|
Saim-Mamoun A, Carbonneau J, Rhéaume C, Abed Y, Boivin G. Viral Fitness of Baloxavir-Resistant Recombinant Influenza B/Victoria- and B/Yamagata-like Viruses Harboring the I38T PA Change, In Vitro, Ex Vivo and in Guinea Pigs. Microorganisms 2023; 11:1095. [PMID: 37317069 DOI: 10.3390/microorganisms11051095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 04/06/2023] [Accepted: 04/20/2023] [Indexed: 06/16/2023] Open
Abstract
Seasonal influenza A and B viruses may cause severe infections requiring therapeutic interventions. Baloxavir, the latest antiviral drug approved against those infections, targets the endonuclease activity encoded by the polymerase acidic (PA) protein. While appearing effective at cessation of viral shedding, baloxavir demonstrated a low barrier of resistance. Herein, we aimed to assess the impact of PA-I38T substitution, a major marker of baloxavir-resistance, on the fitness of contemporary influenza B viruses. Recombinant wild-type (WT) influenza B/Phuket/2073/13 (B/Yamagata/16/88-like) and B/Washington/02/19 (B/Victoria/2/87-like) viruses and their respective PA-I38T mutants were used to evaluate replication kinetics in vitro, using A549 and Calu3 cells, and ex vivo, using nasal human airway epithelium (HAE) cells. Infectivity was also assessed in guinea pigs. In the B/Washington/02/19 background, there were no major differences between the recombinant WT virus and its I38T mutant when viral replication kinetics were evaluated in human lung cell lines and HAE as well as in nasal washes of experimentally infected guinea pigs. By contrast, the I38T mutation moderately impacted the B/Phuket/2073/13 viral fitness. In conclusion, contemporary influenza B viruses that may acquire baloxavir-resistance through the PA-I38T substitution could retain a significant level of fitness, highlighting the importance of monitoring the emergence of such variant.
Collapse
Affiliation(s)
- Amel Saim-Mamoun
- Research Center, Infectious Diseases of the CHU de Québec-CHUL, Laval University, Québec City, QC G1V 4G2, Canada
| | - Julie Carbonneau
- Research Center, Infectious Diseases of the CHU de Québec-CHUL, Laval University, Québec City, QC G1V 4G2, Canada
| | - Chantal Rhéaume
- Research Center, Infectious Diseases of the CHU de Québec-CHUL, Laval University, Québec City, QC G1V 4G2, Canada
| | - Yacine Abed
- Research Center, Infectious Diseases of the CHU de Québec-CHUL, Laval University, Québec City, QC G1V 4G2, Canada
| | - Guy Boivin
- Research Center, Infectious Diseases of the CHU de Québec-CHUL, Laval University, Québec City, QC G1V 4G2, Canada
| |
Collapse
|
26
|
Li J, Wagatsuma K, Sun Y, Sato I, Kawashima T, Saito T, Shimada Y, Ono Y, Kakuya F, Nagata N, Minato M, Kodo N, Suzuki E, Kitano A, Tanaka T, Aoki S, Chon I, Phyu WW, Watanabe H, Saito R. Factors associated with viral RNA shedding and evaluation of potential viral infectivity at returning to school in influenza outpatients after treatment with baloxavir marboxil and neuraminidase inhibitors during 2013/2014-2019/2020 seasons in Japan: an observational study. BMC Infect Dis 2023; 23:188. [PMID: 36991360 PMCID: PMC10054210 DOI: 10.1186/s12879-023-08140-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 03/07/2023] [Indexed: 03/31/2023] Open
Abstract
BACKGROUND This study assessed the differences in daily virus reduction and the residual infectivity after the recommended home stay period in Japan in patients infected with influenza and treated with baloxavir (BA), laninamivir (LA), oseltamivir (OS), and zanamivir (ZA). METHODS We conducted an observational study on children and adults at 13 outpatient clinics in 11 prefectures in Japan during seven influenza seasons from 2013/2014 to 2019/2020. Virus samples were collected twice from influenza rapid test-positive patients at the first and second visit 4-5 days after the start of treatment. The viral RNA shedding was quantified using quantitative RT-PCR. Neuraminidase (NA) and polymerase acidic (PA) variant viruses that reduce susceptibility to NA inhibitors and BA, respectively, were screened using RT-PCR and genetic sequencing. Daily estimated viral reduction was evaluated using univariate and multivariate analyses for the factors such as age, treatment, vaccination status, or the emergence of PA or NA variants. The potential infectivity of the viral RNA shedding at the second visit samples was determined using the Receiver Operator Curve based on the positivity of virus isolation. RESULTS Among 518 patients, 465 (80.0%) and 116 (20.0%) were infected with influenza A (189 with BA, 58 with LA, 181 with OS, 37 with ZA) and influenza B (39 with BA, 10 with LA, 52 with OS, 15 with ZA). The emergence of 21 PA variants in influenza A was detected after BA treatment, but NA variants were not detected after NAIs treatment. Multiple linear regression analysis showed that the daily viral RNA shedding reduction in patients was slower in the two NAIs (OS and LA) than in BA, influenza B infection, aged 0-5 years, or the emergence of PA variants. The residual viral RNA shedding potentially infectious was detected in approximately 10-30% of the patients aged 6-18 years after five days of onset. CONCLUSIONS Viral clearance differed by age, type of influenza, choice of treatment, and susceptibility to BA. Additionally, the recommended homestay period in Japan seemed insufficient, but reduced viral spread to some extent since most school-age patients became non-infectious after 5 days of onset.
Collapse
Affiliation(s)
- Jiaming Li
- Division of International Health (Public Health), Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Niigata, 951-8510, Japan.
| | - Keita Wagatsuma
- Division of International Health (Public Health), Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Niigata, 951-8510, Japan
| | - Yuyang Sun
- Division of International Health (Public Health), Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Niigata, 951-8510, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Irina Chon
- Division of International Health (Public Health), Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Niigata, 951-8510, Japan
| | - Wint Wint Phyu
- Division of International Health (Public Health), Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Niigata, 951-8510, Japan
| | - Hisami Watanabe
- Division of International Health (Public Health), Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Niigata, 951-8510, Japan
| | - Reiko Saito
- Division of International Health (Public Health), Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Niigata, 951-8510, Japan
| |
Collapse
|
27
|
Kula A, Makuch E, Lisowska M, Reniewicz P, Lipiński T, Siednienko J. Pellino3 ligase negatively regulates influenza B dependent RIG-I signalling through downregulation of TRAF3-mediated induction of the transcription factor IRF3 and IFNβ production. Immunology 2023. [PMID: 36861386 DOI: 10.1111/imm.13637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 02/19/2023] [Indexed: 03/03/2023] Open
Abstract
Viral infection activates the innate immune system, which recognizes viral components by a variety of pattern recognition receptors and initiates signalling cascades leading to the production of pro-inflammatory cytokines. To date, signalling cascades triggered after virus recognition are not fully characterized and are investigated by many research groups. The critical role of the E3 ubiquitin ligase Pellino3 in antibacterial and antiviral response is now widely accepted, but the precise mechanism remains elusive. In this study, we sought to explore Pellino3 role in the retinoic acid-inducible gene I (RIG-I)-dependent signalling pathway. In this work, the molecular mechanisms of the innate immune response, regulated by Pellino3, were investigated in lung epithelial cells during influenza B virus infection. We used wild-type and Pellino3-deficient A549 cells as model cell lines to examine the role of Pellino3 ligase in the type I interferon (IFN) signalling pathway. Our results indicate that Pellino3 is involved in direct ubiquitination and degradation of the TRAF3, suppressing interferon regulatory factor 3 (IRF3) activation and interferon beta (IFNβ) production.
Collapse
Affiliation(s)
- Anna Kula
- Bioengineering Research Group, Łukasiewicz Research Network - PORT Polish Center for Technology Development, Wroclaw, Poland.,Laboratory of Medical Microbiology, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Edyta Makuch
- Bioengineering Research Group, Łukasiewicz Research Network - PORT Polish Center for Technology Development, Wroclaw, Poland
| | - Marta Lisowska
- Bioengineering Research Group, Łukasiewicz Research Network - PORT Polish Center for Technology Development, Wroclaw, Poland
| | - Patryk Reniewicz
- Bioengineering Research Group, Łukasiewicz Research Network - PORT Polish Center for Technology Development, Wroclaw, Poland
| | - Tomasz Lipiński
- Bioengineering Research Group, Łukasiewicz Research Network - PORT Polish Center for Technology Development, Wroclaw, Poland
| | - Jakub Siednienko
- Bioengineering Research Group, Łukasiewicz Research Network - PORT Polish Center for Technology Development, Wroclaw, Poland
| |
Collapse
|
28
|
da Costa VG, Gomes AJC, Bittar C, Geraldini DB, Previdelli da Conceição PJ, Cabral ÁS, Carvalho T, Biselli JM, Provazzi PJS, Campos GRF, Sanches PRDS, Costa PI, Nogueira ML, Araujo JP, Spilki FR, Calmon MF, Rahal P. Burden of Influenza and Respiratory Syncytial Viruses in Suspected COVID-19 Patients: A Cross-Sectional and Meta-Analysis Study. Viruses 2023; 15:665. [PMID: 36992374 PMCID: PMC10055802 DOI: 10.3390/v15030665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 02/11/2023] [Accepted: 02/27/2023] [Indexed: 03/06/2023] Open
Abstract
Non-SARS-CoV-2 respiratory viral infections, such as influenza virus (FluV) and human respiratory syncytial virus (RSV), have contributed considerably to the burden of infectious diseases in the non-COVID-19 era. While the rates of co-infection in SARS-CoV-2-positive group (SCPG) patients have been determined, the burden of other respiratory viruses in the SARS-CoV-2-negative group (SCNG) remains unclear. Here, we conducted a cross-sectional study (São José do Rio Preto county, Brazil), and we collected our data using a meta-analysis to evaluate the pooled prevalence of FluV and RSV among SCNG patients. Out of the 901 patients suspected of COVID-19, our molecular results showed positivity of FluV and RSV in the SCNG was 2% (15/733) and 0.27% (2/733), respectively. Co-infection with SARS-CoV-2 and FluV, or RSV, was identified in 1.7% of the patients (3/168). Following our meta-analysis, 28 studies were selected (n = 114,318 suspected COVID-19 patients), with a pooled prevalence of 4% (95% CI: 3-6) for FluV and 2% (95% CI: 1-3) for RSV among SCNG patients were observed. Interestingly, FluV positivity in the SCNG was four times higher (OR = 4, 95% CI: 3.6-5.4, p < 0.01) than in the SCPG. Similarly, RSV positivity was significantly associated with SCNG patients (OR = 2.9, 95% CI: 2-4, p < 0.01). For subgroup analysis, cold-like symptoms, including fever, cough, sore throat, headache, myalgia, diarrhea, and nausea/vomiting, were positively associated (p < 0.05) with the SCPG. In conclusion, these results show that the pooled prevalence of FluV and RSV were significantly higher in the SCNG than in the SCPG during the early phase of the COVID-19 pandemic.
Collapse
Affiliation(s)
- Vivaldo Gomes da Costa
- Laboratório de Estudos Genômicos, Departamento de Biologia, Instituto de Biociências Letras e Ciências Exatas, Universidade Estadual Paulista Júlio de Mesquita Filho (UNESP), São José do Rio Preto 15054-000, SP, Brazil
| | - Ana Júlia Chaves Gomes
- Laboratório de Estudos Genômicos, Departamento de Biologia, Instituto de Biociências Letras e Ciências Exatas, Universidade Estadual Paulista Júlio de Mesquita Filho (UNESP), São José do Rio Preto 15054-000, SP, Brazil
| | - Cíntia Bittar
- Laboratório de Estudos Genômicos, Departamento de Biologia, Instituto de Biociências Letras e Ciências Exatas, Universidade Estadual Paulista Júlio de Mesquita Filho (UNESP), São José do Rio Preto 15054-000, SP, Brazil
| | - Dayla Bott Geraldini
- Laboratório de Estudos Genômicos, Departamento de Biologia, Instituto de Biociências Letras e Ciências Exatas, Universidade Estadual Paulista Júlio de Mesquita Filho (UNESP), São José do Rio Preto 15054-000, SP, Brazil
| | - Pâmela Jóyce Previdelli da Conceição
- Laboratório de Estudos Genômicos, Departamento de Biologia, Instituto de Biociências Letras e Ciências Exatas, Universidade Estadual Paulista Júlio de Mesquita Filho (UNESP), São José do Rio Preto 15054-000, SP, Brazil
| | - Ágata Silva Cabral
- Laboratório de Estudos Genômicos, Departamento de Biologia, Instituto de Biociências Letras e Ciências Exatas, Universidade Estadual Paulista Júlio de Mesquita Filho (UNESP), São José do Rio Preto 15054-000, SP, Brazil
| | - Tamara Carvalho
- Laboratório de Estudos Genômicos, Departamento de Biologia, Instituto de Biociências Letras e Ciências Exatas, Universidade Estadual Paulista Júlio de Mesquita Filho (UNESP), São José do Rio Preto 15054-000, SP, Brazil
| | - Joice Matos Biselli
- Laboratório de Estudos Genômicos, Departamento de Biologia, Instituto de Biociências Letras e Ciências Exatas, Universidade Estadual Paulista Júlio de Mesquita Filho (UNESP), São José do Rio Preto 15054-000, SP, Brazil
| | - Paola Jocelan Scarin Provazzi
- Laboratório de Estudos Genômicos, Departamento de Biologia, Instituto de Biociências Letras e Ciências Exatas, Universidade Estadual Paulista Júlio de Mesquita Filho (UNESP), São José do Rio Preto 15054-000, SP, Brazil
| | - Guilherme Rodrigues Fernandes Campos
- Laboratório de Pesquisas em Virologia (LPV), Departamento de Doenças Dermatológicas, Infecciosas e Parasitárias, Faculdade de Medicina de São José do Rio Preto (FAMERP), São José do Rio Preto 15090-000, SP, Brazil
| | - Paulo Ricardo da Silva Sanches
- Laboratório de Virologia Molecular, Departamento de Ciências Biológicas, Faculdade de Ciências Farmacêuticas (UNESP), Araraquara 14800-903, SP, Brazil
| | - Paulo Inácio Costa
- Departamento de Análises Clínicas, Faculdade de Ciências Farmacêuticas (UNESP), Araraquara 14801-360, SP, Brazil
| | - Maurício Lacerda Nogueira
- Laboratório de Pesquisas em Virologia (LPV), Departamento de Doenças Dermatológicas, Infecciosas e Parasitárias, Faculdade de Medicina de São José do Rio Preto (FAMERP), São José do Rio Preto 15090-000, SP, Brazil
| | - João Pessoa Araujo
- Instituto de Biotecnologia, Universidade Estadual Paulista Júlio de Mesquita Filho (UNESP), Botucatu 18607-440, SP, Brazil
| | - Fernando Rosado Spilki
- Laboratório de Microbiologia Molecular, Instituto de Ciências da Saúde, Universidade Feevale, Novo Hamburgo 93525-075, RS, Brazil
| | - Marília Freitas Calmon
- Laboratório de Estudos Genômicos, Departamento de Biologia, Instituto de Biociências Letras e Ciências Exatas, Universidade Estadual Paulista Júlio de Mesquita Filho (UNESP), São José do Rio Preto 15054-000, SP, Brazil
| | - Paula Rahal
- Laboratório de Estudos Genômicos, Departamento de Biologia, Instituto de Biociências Letras e Ciências Exatas, Universidade Estadual Paulista Júlio de Mesquita Filho (UNESP), São José do Rio Preto 15054-000, SP, Brazil
| |
Collapse
|
29
|
Chan-Zapata I, Borges-Argáez R, Ayora-Talavera G. Quinones as Promising Compounds against Respiratory Viruses: A Review. Molecules 2023; 28:1981. [PMID: 36838969 PMCID: PMC9967002 DOI: 10.3390/molecules28041981] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 02/14/2023] [Accepted: 02/15/2023] [Indexed: 02/22/2023] Open
Abstract
Respiratory viruses represent a world public health problem, giving rise to annual seasonal epidemics and several pandemics caused by some of these viruses, including the COVID-19 pandemic caused by the novel SARS-CoV-2, which continues to date. Some antiviral drugs have been licensed for the treatment of influenza, but they cause side effects and lead to resistant viral strains. Likewise, aerosolized ribavirin is the only drug approved for the therapy of infections by the respiratory syncytial virus, but it possesses various limitations. On the other hand, no specific drugs are licensed to treat other viral respiratory diseases. In this sense, natural products and their derivatives have appeared as promising alternatives in searching for new compounds with antiviral activity. Besides their chemical properties, quinones have demonstrated interesting biological activities, including activity against respiratory viruses. This review summarizes the activity against respiratory viruses and their molecular targets by the different types of quinones (both natural and synthetic). Thus, the present work offers a general overview of the importance of quinones as an option for the future pharmacological treatment of viral respiratory infections, subject to additional studies that support their effectiveness and safety.
Collapse
Affiliation(s)
- Ivan Chan-Zapata
- Unidad de Biotecnología, Centro de Investigación Científica de Yucatán, Chuburná de Hidalgo, Merida 97205, Mexico
| | - Rocío Borges-Argáez
- Unidad de Biotecnología, Centro de Investigación Científica de Yucatán, Chuburná de Hidalgo, Merida 97205, Mexico
| | - Guadalupe Ayora-Talavera
- Departamento de Virología, Centro de Investigaciones Regionales “Dr. Hideyo Noguchi”, Universidad Autónoma de Yucatán, Paseo de Las Fuentes, Merida 97225, Mexico
| |
Collapse
|
30
|
Portela Catani JP, Ysenbaert T, Smet A, Vuylsteke M, Vogel TU, Saelens X. Anti-neuraminidase and anti-hemagglutinin immune serum can confer inter-lineage cross protection against recent influenza B. PLoS One 2023; 18:e0280825. [PMID: 36689429 PMCID: PMC9870131 DOI: 10.1371/journal.pone.0280825] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 01/09/2023] [Indexed: 01/24/2023] Open
Abstract
Influenza B viruses (IBV) are responsible for a considerable part of the burden caused by influenza virus infections. Since their emergence in the 1980s, the Yamagata and Victoria antigenic lineages of influenza B circulate in alternate patterns across the globe. Furthermore, their evolutionary divergence and the appearance of new IBV subclades complicates the prediction of future influenza vaccines compositions. It has been proposed that the addition of the neuraminidase (NA) antigen could potentially induce a broader protection and compensate for hemagglutinin (HA) mismatches in the current vaccines. Here we show that anti-NA and -HA sera against both Victoria and Yamagata lineages have limited inter-lineage cross-reactivity. When transferred to mice prior to infection with a panel of IBVs, anti-NA sera were as potent as anti-HA sera in conferring protection against homologous challenge and, in some cases, conferred superior protection against challenge with heterologous IBV strains.
Collapse
Affiliation(s)
- João Paulo Portela Catani
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium
- Department of Biochemistry and Microbiology, Ghent University, Ghent, Belgium
| | - Tine Ysenbaert
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium
- Department of Biochemistry and Microbiology, Ghent University, Ghent, Belgium
| | - Anouk Smet
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium
- Department of Biochemistry and Microbiology, Ghent University, Ghent, Belgium
| | | | - Thorsten U. Vogel
- Sanofi, Research North America, Cambridge, Massachusetts, United States of America
| | - Xavier Saelens
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium
- Department of Biochemistry and Microbiology, Ghent University, Ghent, Belgium
| |
Collapse
|
31
|
Kang HJ, Chu KB, Yoon KW, Eom GD, Mao J, Quan FS. Cross-Protection Induced by Virus-like Particles Derived from the Influenza B Virus. Biomedicines 2022; 10:1618. [PMID: 35884922 PMCID: PMC9313027 DOI: 10.3390/biomedicines10071618] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 06/21/2022] [Accepted: 07/05/2022] [Indexed: 12/16/2022] Open
Abstract
The mismatch between the circulating influenza B virus (IBV) and the vaccine strain contributes to the rapid emergence of IBV infection cases throughout the globe, which necessitates the development of effective vaccines conferring broad protection. Here, we generated influenza B virus-like particle (VLP) vaccines expressing hemagglutinin, neuraminidase, or both antigens derived from the influenza B virus (B/Washington/02/2019 (B/Victoria lineage)-like virus, B/Phuket/3073/2013 (B/Yamagata lineage)-like virus. We found that irrespective of the derived antigen lineage, immunizing mice with the IBV VLPs significantly reduced lung viral loads, minimized bodyweight loss, and ensured 100% survival upon Victoria lineage virus B/Colorado/06/2017 challenge infection. These results were closely correlated with the vaccine-induced antibody responses and HI titer in sera, IgG, IgA antibody responses, CD4+ and CD8+ T cell responses, germinal center B cell responses, and inflammatory cytokine responses in the lungs. We conclude that hemagglutinin, neuraminidase, or both antigen-expressing VLPs derived from these influenza B viruses that were circulating during the 2020/21 season provide cross-protections against mismatched Victoria lineage virus (B/Colorado/06/2017) challenge infections.
Collapse
Affiliation(s)
- Hae-Ji Kang
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Korea; (H.-J.K.); (K.-W.Y.); (G.-D.E.); (J.M.)
| | - Ki-Back Chu
- Department of Medical Zoology, School of Medicine, Kyung Hee University, Seoul 02447, Korea;
- Medical Research Center for Bioreaction to Reactive Oxygen Species and Biomedical Science Institute, School of Medicine, Graduate School, Kyung Hee University, Seoul 02447, Korea
| | - Keon-Woong Yoon
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Korea; (H.-J.K.); (K.-W.Y.); (G.-D.E.); (J.M.)
| | - Gi-Deok Eom
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Korea; (H.-J.K.); (K.-W.Y.); (G.-D.E.); (J.M.)
| | - Jie Mao
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Korea; (H.-J.K.); (K.-W.Y.); (G.-D.E.); (J.M.)
| | - Fu-Shi Quan
- Department of Medical Zoology, School of Medicine, Kyung Hee University, Seoul 02447, Korea;
- Medical Research Center for Bioreaction to Reactive Oxygen Species and Biomedical Science Institute, School of Medicine, Graduate School, Kyung Hee University, Seoul 02447, Korea
| |
Collapse
|
32
|
Abo Shama NM, Mahmoud SH, Bagato O, AbdElsalam ET, Alkhazindar M, Kandeil A, McKenzie PP, Webby RJ, Ali MA, Kayali G, El-Shesheny R. Incidence and neutralizing antibody seroprevalence of influenza B virus in Egypt: Results of a community-based cohort study. PLoS One 2022; 17:e0269321. [PMID: 35767564 PMCID: PMC9242516 DOI: 10.1371/journal.pone.0269321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 05/19/2022] [Indexed: 11/27/2022] Open
Abstract
Since 2000, two lineages of influenza B viruses, Victoria and Yamagata, have been circulating at similar frequencies worldwide. Little is known about the circulation of those viruses in Egypt. This study aims to describe the epidemiology of influenza B virus infections in Egypt, 2017–2019. This was performed through a household prospective cohort study on influenza infections among 2400 individuals from five villages. When a study participant had influenza like symptoms, a nasal swab and an oropharyngeal swab were obtained and tested by RT-PCR for influenza B infections. A serum sample was obtained from all participants annually to detect neutralizing antibodies using microneutralization assay. 9.1% of subjects were positive for influenza B viruses during season 2017–2018 mostly among preschoolers and 7.6% were positive during the season 2018–2019 with higher risk in females, potentially due to mothers being infected after contact with their children. The overall seroprevalence among the participants was 53.2% and 52.2% against the Victoria and Yamagata lineages respectively, the majority of seropositive participants were students. Multivariate analysis showed that age and having chronic diseases were the strongest predictors of infection. Our results show that both influenza B lineages circulated between 2017 and 2020 in Egypt almost in equal proportion. Encouraging the uptake of seasonal influenza vaccines is recommended.
Collapse
Affiliation(s)
- Noura M. Abo Shama
- Center of Scientific Excellence for Influenza Virus, Environmental Research Division, National Research Centre, Giza, Egypt
| | - Sara H. Mahmoud
- Center of Scientific Excellence for Influenza Virus, Environmental Research Division, National Research Centre, Giza, Egypt
| | - Ola Bagato
- Center of Scientific Excellence for Influenza Virus, Environmental Research Division, National Research Centre, Giza, Egypt
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Riems, Germany
| | - Elsayed Tarek AbdElsalam
- Department of Botany and Microbiology, Faculty of Science, Cairo University, Gamaa Street, Giza, Egypt
| | - Maha Alkhazindar
- Department of Botany and Microbiology, Faculty of Science, Cairo University, Gamaa Street, Giza, Egypt
| | - Ahmed Kandeil
- Center of Scientific Excellence for Influenza Virus, Environmental Research Division, National Research Centre, Giza, Egypt
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, TN, United States of America
| | - Pamela P. McKenzie
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, TN, United States of America
| | - Richard J. Webby
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, TN, United States of America
| | - Mohamed A. Ali
- Center of Scientific Excellence for Influenza Virus, Environmental Research Division, National Research Centre, Giza, Egypt
| | | | - Rabeh El-Shesheny
- Center of Scientific Excellence for Influenza Virus, Environmental Research Division, National Research Centre, Giza, Egypt
- * E-mail: (GK); (RE)
| |
Collapse
|
33
|
Pekarek MJ, Petro-Turnquist EM, Rubrum A, Webby RJ, Weaver EA. Expanding Mouse-Adapted Yamagata-like Influenza B Viruses in Eggs Enhances In Vivo Lethality in BALB/c Mice. Viruses 2022; 14:v14061299. [PMID: 35746770 PMCID: PMC9229684 DOI: 10.3390/v14061299] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 06/10/2022] [Indexed: 01/23/2023] Open
Abstract
Despite the yearly global impact of influenza B viruses (IBVs), limited host range has been a hurdle to developing a readily accessible small animal disease model for vaccine studies. Mouse-adapting IBV can produce highly pathogenic viruses through serial lung passaging in mice. Previous studies have highlighted amino acid changes throughout the viral genome correlating with increased pathogenicity, but no consensus mutations have been determined. We aimed to show that growth system can play a role in mouse-adapted IBV lethality. Two Yamagata-lineage IBVs were serially passaged 10 times in mouse lungs before expansion in embryonated eggs or Madin-Darby canine kidney cells (London line) for use in challenge studies. We observed that virus grown in embryonated eggs was significantly more lethal in mice than the same virus grown in cell culture. Ten additional serial lung passages of one strain again showed virus grown in eggs was more lethal than virus grown in cells. Additionally, no mutations in the surface glycoprotein amino acid sequences correlated to differences in lethality. Our results suggest growth system can influence lethality of mouse-adapted IBVs after serial lung passaging. Further research can highlight improved mechanisms for developing animal disease models for IBV vaccine research.
Collapse
Affiliation(s)
- Matthew J. Pekarek
- Nebraska Center for Virology, School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA; (M.J.P.); (E.M.P.-T.)
| | - Erika M. Petro-Turnquist
- Nebraska Center for Virology, School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA; (M.J.P.); (E.M.P.-T.)
| | - Adam Rubrum
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (A.R.); (R.J.W.)
| | - Richard J. Webby
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (A.R.); (R.J.W.)
| | - Eric A. Weaver
- Nebraska Center for Virology, School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA; (M.J.P.); (E.M.P.-T.)
- Correspondence:
| |
Collapse
|
34
|
Wagatsuma K, Saito R, Chon I, Phyu WW, Fujio K, Kawashima T, Sato I, Saito T, Minato M, Kodo N, Suzuki E, Ono Y, Masaki H, Shirahige Y, Kitano A, Hamabata H, Yuyang S, Jiaming L, Watanabe H. Duration of fever and symptoms in influenza-infected children treated with baloxavir marboxil during the 2019–2020 season in Japan and detection of influenza virus with the PA E23K substitution. Antiviral Res 2022; 201:105310. [DOI: 10.1016/j.antiviral.2022.105310] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 03/22/2022] [Accepted: 03/25/2022] [Indexed: 12/26/2022]
|
35
|
Kumar D, Ison MG, Mira JP, Welte T, Hwan Ha J, Hui DS, Zhong N, Saito T, Katugampola L, Collinson N, Williams S, Wildum S, Ackrill A, Clinch B, Lee N. Combining baloxavir marboxil with standard-of-care neuraminidase inhibitor in patients hospitalised with severe influenza (FLAGSTONE): a randomised, parallel-group, double-blind, placebo-controlled, superiority trial. THE LANCET INFECTIOUS DISEASES 2022; 22:718-730. [DOI: 10.1016/s1473-3099(21)00469-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 05/28/2021] [Accepted: 07/29/2021] [Indexed: 10/19/2022]
|
36
|
Xu D, Zheng QC. Theoretical investigations on the effects of mutations in important residues of NS1B on its RNA-binding using molecular dynamics simulations. Comput Biol Med 2022; 145:105412. [PMID: 35344866 DOI: 10.1016/j.compbiomed.2022.105412] [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: 02/21/2022] [Revised: 03/07/2022] [Accepted: 03/14/2022] [Indexed: 11/28/2022]
Abstract
NS1B protein plays an important role in countering host antiviral defense and virulence of influenza virus B, considered as the promising target. The first experimental structure of the NS1B protein has recently been determined, was able to bind to double-stranded RNA (dsRNA). However, few studies attempt to investigate the RNA-binding mechanism of the NS1B. In this study, we provide our understanding of the structure-function relationship, dynamics and RNA-binding mechanism of the NS1B protein by performing molecular dynamics simulations combined and MM-GBSA calculations on the NS1B-dsRNA complex. 12 key residues are identified for RNA-binding by forming hydrogen bonds with the. Our results also demonstrate that mutations (R156A, K160A, R208A and K221A) can cause the local structure changes of NS1B CTD and the hydrogen bonds between NS1B CTD and RNA disappearance, which may be the main reasons for the decrease in RNA-binding affinity. These results mentioned will help us understanding the RNA-binding mechanism and could provide some medicinal chemistry insights chances for rational drug design targeting NS1B protein.
Collapse
Affiliation(s)
- Dan Xu
- Institute of Theoretical Chemistry, College of Chemistry, Jilin University, Changchun, 130023, China
| | - Qing-Chuan Zheng
- Institute of Theoretical Chemistry, College of Chemistry, Jilin University, Changchun, 130023, China; Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, Jilin University, Changchun, 130023, China.
| |
Collapse
|
37
|
Rattan A, White CL, Nelson S, Eismann M, Padilla-Quirarte H, Glover MA, Dileepan T, Marathe BM, Govorkova EA, Webby RJ, Richards KA, Sant AJ. Development of a Mouse Model to Explore CD4 T Cell Specificity, Phenotype, and Recruitment to the Lung after Influenza B Infection. Pathogens 2022; 11:251. [PMID: 35215193 PMCID: PMC8875387 DOI: 10.3390/pathogens11020251] [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: 12/27/2021] [Revised: 02/05/2022] [Accepted: 02/08/2022] [Indexed: 01/30/2023] Open
Abstract
The adaptive T cell response to influenza B virus is understudied, relative to influenza A virus, for which there has been considerable attention and progress for many decades. Here, we have developed and utilized the C57BL/6 mouse model of intranasal infection with influenza B (B/Brisbane/60/2008) virus and, using an iterative peptide discovery strategy, have identified a series of robustly elicited individual CD4 T cell peptide specificities. The CD4 T cell repertoire encompassed at least eleven major epitopes distributed across hemagglutinin, nucleoprotein, neuraminidase, and non-structural protein 1 and are readily detected in the draining lymph node, spleen, and lung. Within the lung, the CD4 T cells are localized to both lung vasculature and tissue but are highly enriched in the lung tissue after infection. When studied by flow cytometry and MHC class II: peptide tetramers, CD4 T cells express prototypical markers of tissue residency including CD69, CD103, and high surface levels of CD11a. Collectively, our studies will enable more sophisticated analyses of influenza B virus infection, where the fate and function of the influenza B-specific CD4 T cells elicited by infection and vaccination can be studied as well as the impact of anti-viral reagents and candidate vaccines on the abundance, functionality, and localization of the elicited CD4 T cells.
Collapse
Affiliation(s)
- Ajitanuj Rattan
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA; (A.R.); (C.L.W.); (S.N.); (M.E.); (M.A.G.); (K.A.R.)
| | - Chantelle L. White
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA; (A.R.); (C.L.W.); (S.N.); (M.E.); (M.A.G.); (K.A.R.)
| | - Sean Nelson
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA; (A.R.); (C.L.W.); (S.N.); (M.E.); (M.A.G.); (K.A.R.)
| | - Max Eismann
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA; (A.R.); (C.L.W.); (S.N.); (M.E.); (M.A.G.); (K.A.R.)
| | - Herbey Padilla-Quirarte
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30322, USA;
| | - Maryah A. Glover
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA; (A.R.); (C.L.W.); (S.N.); (M.E.); (M.A.G.); (K.A.R.)
| | - Thamotharampillai Dileepan
- Department of Microbiology and Immunology, University of Minnesota Medical School, Minneapolis, MN 55455, USA;
| | - Bindumadhav M. Marathe
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (B.M.M.); (E.A.G.); (R.J.W.)
| | - Elena A. Govorkova
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (B.M.M.); (E.A.G.); (R.J.W.)
| | - Richard J. Webby
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (B.M.M.); (E.A.G.); (R.J.W.)
| | - Katherine A. Richards
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA; (A.R.); (C.L.W.); (S.N.); (M.E.); (M.A.G.); (K.A.R.)
- Center for Influenza Disease and Emergence Response (CIDER), University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Andrea J. Sant
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA; (A.R.); (C.L.W.); (S.N.); (M.E.); (M.A.G.); (K.A.R.)
- Center for Influenza Disease and Emergence Response (CIDER), University of Rochester Medical Center, Rochester, NY 14642, USA
| |
Collapse
|
38
|
|
39
|
Abstract
Influenza poses a significant burden on society and health care systems. Although antivirals are an integral tool in effective influenza management, the potential for the emergence of antiviral-resistant viruses can lead to uncertainty and hesitation among front-line prescribers and policy makers. Here, we provide an overview of influenza antiviral resistance in context, exploring the key concepts underlying its development and clinical impact. Due to the acute nature of influenza in immunocompetent patients, resistant viruses that develop during antiviral treatment of a single patient ("treatment-emergent resistance") are usually cleared in a relatively short time, with no impact on future antiviral efficacy. In addition, although available data are limited by small numbers of patients, they show that antiviral treatment still provides clinical benefit to the patient within whom resistance emerges. In contrast, the sustained community transmission of resistant variants in the absence of treatment ("acquired resistance") is of greater concern and can potentially render front-line antivirals ineffective. Importantly, however, resistant viruses are usually associated with reduced fitness such that their widespread transmission is relatively rare. Influenza antivirals are an essential part of effective influenza management due to their ability to reduce the risk of complications and death in infected patients. Although antiviral resistance should be taken seriously and requires continuous careful monitoring, it is not comparable to antibiotic resistance in bacteria, which can become permanent and widespread, with far-reaching medical consequences. The benefits of antiviral treatment far outweigh concerns of potential resistance, which in the vast majority of cases does not have a significant clinical impact.
Collapse
|
40
|
Koutsakos M, Kent SJ. Influenza B viruses: underestimated and overlooked. MICROBIOLOGY AUSTRALIA 2021. [DOI: 10.1071/ma21033] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Influenza B viruses circulate globally every year causing respiratory disease with significant clinical and socio-economic impacts. IBV are considered exclusive human pathogens with no established animal reservoirs, which suggests with concerted effort it may be possible to eradicate this virus from human circulation. However, this requires a deeper understanding of IBV virology and immunology and the design of vaccines that induce universal immunity to antigenic variants of IBV.
Collapse
|