1
|
Zhao X, Hu X, Wang J, Shen M, Zhou K, Han X, Thomas M, Wang K, Wang L, Wang Z. A cross-sectional study on the understanding and attitudes toward influenza and influenza vaccines among different occupational groups in China. Hum Vaccin Immunother 2024; 20:2397214. [PMID: 39286861 PMCID: PMC11409513 DOI: 10.1080/21645515.2024.2397214] [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: 04/30/2024] [Revised: 08/14/2024] [Accepted: 08/23/2024] [Indexed: 09/19/2024] Open
Abstract
This study aimed to assess the level of knowledge regarding influenza viruses and vaccines among different professional groups to investigate the reasons for vaccine hesitancy. We collected 2190 questionnaires regarding influenza vaccines in China in 2022. The respondents were categorized into the general population (GP), foreign affairs workforce population (FAWP), and veterinary workforce population (VWP) according to their job positions. Linear regression was used to assess the association between multiple factors and influenza vaccination rates. The association between work and influenza vaccination rates was also assessed by grouping different workforce populations. The vaccination rate of the GP was higher than that of the VWP (odds ratio: 1.342, 95% confidence interval: 1.025-1.853), surpassing the rates reported in previous studies. This may be attributed to heightened concerns about infectious diseases influenced by the ongoing coronavirus disease 2019 pandemic. Despite the VWP's more in-depth knowledge of the VWP on zoonotic diseases and their recognition of their importance, there was no significant difference in influenza knowledge among the three populations. This discrepancy contrasts with the observed differences in vaccination rates. Further investigation revealed that, compared with FAWP, the price of vaccines emerged as a primary influencing factor for vaccination rates (odds ratio:0.398, 95%CI; 0.280-0.564). General concerns regarding the protective effects and side effects of vaccines were also noted.
Collapse
Affiliation(s)
- Xinkun Zhao
- School of Laboratory Animal & Shandong Laboratory Animal Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Xin Hu
- School of Politicl Science and Public Administration, Shandong University, Qingdao, China
| | - Junyi Wang
- Department of Promotion, Linyi City Animal Husbandry Development and Promotion Center, Linyi, China
| | - Mingshuai Shen
- School of Laboratory Animal & Shandong Laboratory Animal Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Kaifeng Zhou
- Department of Promotion, Shandong Provincial Animal Husbandry General Station, Jinan, China
| | - Xianjie Han
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, China
| | - Milton Thomas
- Department of Microbiology and immunology, University of Louisville, Louisville, Kentucky, USA
| | - Kezhou Wang
- School of Laboratory Animal & Shandong Laboratory Animal Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Li Wang
- Physical Factors Section, Occupational Diseases Hospital of Shandong First Medical University, Jinan, China
| | - Zhao Wang
- School of Laboratory Animal & Shandong Laboratory Animal Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
2
|
Huang X, Cheng Z, Lv Y, Li W, Liu X, Huang W, Zhao C. Neutralization potency of the 2023-24 seasonal influenza vaccine against circulating influenza H3N2 strains. Hum Vaccin Immunother 2024; 20:2380111. [PMID: 39205645 PMCID: PMC11364067 DOI: 10.1080/21645515.2024.2380111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 06/27/2024] [Accepted: 07/11/2024] [Indexed: 09/04/2024] Open
Abstract
Seasonal influenza is a severe disease that significantly impacts public health, causing millions of infections and hundreds of thousands of deaths each year. Seasonal influenza viruses, particularly the H3N2 subtype, exhibit high antigenic variability, often leading to mismatch between vaccine strains and circulating strains. Therefore, rapidly assessing the alignment between existing seasonal influenza vaccine and circulating strains is crucial for enhancing vaccine efficacy. This study, based on a pseudovirus platform, evaluated the match between current influenza H3N2 vaccine strains and circulating strains through cross-neutralization assays using clinical human immune sera against globally circulating influenza virus strains. The research results show that although mutations are present in the circulating strains, the current H3N2 vaccine strain still imparting effective protection, providing a scientific basis for encouraging influenza vaccination. This research methodology can be sustainably applied for the neutralization potency assessment of subsequent circulating strains, establishing a persistent methodological framework.
Collapse
Affiliation(s)
- Xiande Huang
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing, China
| | - Ziqi Cheng
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing, China
| | - Yake Lv
- Center of Vaccine Clinical Evaluation, Institute for Immunization Program, Shaanxi Provincial Centre for Disease Control and Prevention, Xi’an, Shaanxi Province, China
| | - Weixuan Li
- Center of Vaccine Clinical Evaluation, Institute for Immunization Program, Shaanxi Provincial Centre for Disease Control and Prevention, Xi’an, Shaanxi Province, China
| | - Xiaoyu Liu
- Center of Vaccine Clinical Evaluation, Institute for Immunization Program, Shaanxi Provincial Centre for Disease Control and Prevention, Xi’an, Shaanxi Province, China
| | - Weijin Huang
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing, China
| | - Chenyan Zhao
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing, China
| |
Collapse
|
3
|
Zhang X, Skarlupka AL, Shi H, Ross TM. COBRA N2 NA vaccines induce protective immune responses against influenza viral infection. Hum Vaccin Immunother 2024; 20:2403175. [PMID: 39291424 DOI: 10.1080/21645515.2024.2403175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 08/29/2024] [Accepted: 09/08/2024] [Indexed: 09/19/2024] Open
Abstract
Influenza neuraminidase (NA) is a promising target for a broadly protective vaccine. In this study, the Computationally Optimized Broadly Reactive Antigen (COBRA) methodology was used to develop N2 NA vaccine candidates. The unique wild type (WT) N2 sequences of human and swine influenza strains isolated between 1957 and 2019 were used to design the COBRA N2-A NA vaccine, while the unique WT N2 sequences of human influenza strains isolated between 2000 and 2019 were used to design the COBRA N2-B NA vaccine. Sera collected from COBRA N2 NA vaccinated mice showed more broadly reactive antibody responses against a broad panel of H×N2 influenza virus strains than sera collected from mice vaccinated with WT N2 NA vaccines. Antibodies elicited by COBRA or WT N2 NA antigens cross react with recent human H3N2 influenza viruses from different clades, while the antibodies elicited by A/Switzerland/9715293/2013 hemagglutinin (HA) reacted with viruses from the same clade. Furthermore, mice vaccinated with COBRA N2-B NA vaccine had lower viral lung titers compared to mock vaccinated mice when challenged with human H3N2 influenza viruses. Thus, the COBRA N2 NA vaccines elicit broadly protective murine anti-NA antibodies against multiple strains across subtypes and the viral loads were significantly decreased in the lungs of the mice in the COBRA N2 NA vaccine groups, compared to the mice in the mock vaccinated group, indicating that the COBRA-based N2 subtype NA vaccines have a potential to be a component in a universal influenza vaccine.
Collapse
Affiliation(s)
- Xiaojian Zhang
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, USA
| | - Amanda L Skarlupka
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, USA
| | - Hua Shi
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, USA
| | - Ted M Ross
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, USA
- Department of Infectious Diseases, University of Georgia, Athens, GA, USA
- Florida Research and Innovation Center, Cleveland Clinic, Port Saint Lucie, FL, USA
- Department of Infection Biology, Lehner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
4
|
Riad A, Truksová V, Koščík M. Seasonal Influenza Vaccine Literacy and Hesitancy of Elderly Czechs: An Analysis Using the 5C Model of Psychological Antecedents. Int J Public Health 2024; 69:1607626. [PMID: 39469530 PMCID: PMC11513313 DOI: 10.3389/ijph.2024.1607626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Accepted: 09/30/2024] [Indexed: 10/30/2024] Open
Abstract
Objectives Seasonal influenza vaccination rates among the elderly in the Czech Republic are alarmingly low, making it one of the least vaccinated countries in Europe. This study explored the role of vaccine literacy and insurance coverage on vaccination status. Methods An analytical cross-sectional study was conducted in Summer 2023 using a self-administered questionnaire covering vaccine literacy (functional, interactive, and critical skills), negative perceptions towards influenza vaccination, and the 5C model (confidence, complacency, constraints, calculation, and collective responsibility). Individuals aged 55 and older were included in the study. Mediation analyses assessed the indirect effects of insurance coverage on vaccination status. Results Significant differences were noted in vaccination rates based on insurance coverage, chronic diseases, regular medication use, and previous COVID-19 and pneumococcal vaccinations. Vaccine literacy, especially interactive and critical skills, was higher among vaccinated individuals. Confidence and collective responsibility were significant promoters, while complacency and constraints were barriers to vaccination. Mediation analyses indicated that negative perceptions, confidence, and collective responsibility significantly mediated the relationship between insurance coverage and vaccination status. Conclusion Enhancing vaccine literacy and addressing psychological antecedents are crucial for improving influenza vaccination rates among the elderly. Policy measures should include improving vaccine literacy, building public confidence, and addressing negative perceptions.
Collapse
Affiliation(s)
- Abanoub Riad
- Department of Public Health, Faculty of Medicine, Masaryk University, Brno, Czechia
- Masaryk Centre for Global Health (MCGH), Department of Public Health, Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Veronika Truksová
- Department of Public Health, Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Michal Koščík
- Department of Public Health, Faculty of Medicine, Masaryk University, Brno, Czechia
- Masaryk Centre for Global Health (MCGH), Department of Public Health, Faculty of Medicine, Masaryk University, Brno, Czechia
| |
Collapse
|
5
|
Prada-García C, Toquero-Asensio M, Fernández-Espinilla V, Hernán-García C, Sanz-Muñoz I, Eiros JM, Castrodeza-Sanz J. Analyzing Changes in Attitudes and Behaviors towards Seasonal Influenza Vaccination in Spain's Adult Population over Three Seasons. Vaccines (Basel) 2024; 12:1162. [PMID: 39460328 PMCID: PMC11511366 DOI: 10.3390/vaccines12101162] [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: 09/12/2024] [Revised: 10/06/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024] Open
Abstract
Background/Objectives: The experience of the COVID-19 pandemic has turned the spotlight on the importance of public health measures and disease prevention. Despite this, the acceptance of influenza vaccination has remained low in most countries (and far from the 75% target set by the World Health Organization). The objective of this study has been to investigate how attitudes and behaviors regarding influenza vaccination in the Spanish adult population have changed over the last three years (from 2021 to 2024) in order to analyze trends in influenza vaccination. Methods: To this end, a cross-sectional study was conducted through 2206 telephone interviews, and the results were compared with those obtained in previous campaigns. Results: The findings indicate a significant decline in overall vaccination intent. Healthcare professionals remain the most influential factor in encouraging vaccination, yet there is a notable increase in the lack of vaccine recommendations, contributing to the decision not to vaccinate. This study also reveals low awareness of the influenza vaccine campaign, emphasizing the need for improved public health communication. Conclusions: To counteract these trends, this study recommends intensifying awareness campaigns, strengthening the role of healthcare providers in vaccine advocacy, and tailoring communication strategies. These efforts are crucial to enhancing vaccination coverage and protecting vulnerable populations against influenza.
Collapse
Affiliation(s)
- Camino Prada-García
- Department of Preventive Medicine and Public Health, University of Valladolid, 47005 Valladolid, Spain; (V.F.-E.); (C.H.-G.); (J.C.-S.)
- National Influenza Centre, Edificio Rondilla, Hospital Clínico Universitario de Valladolid, 47009 Valladolid, Spain; (M.T.-A.); (I.S.-M.); (J.M.E.)
- Dermatology Service, Complejo Asistencial Universitario de León, 24008 León, Spain
| | - Marina Toquero-Asensio
- National Influenza Centre, Edificio Rondilla, Hospital Clínico Universitario de Valladolid, 47009 Valladolid, Spain; (M.T.-A.); (I.S.-M.); (J.M.E.)
- Preventive Medicine and Public Health Service, Hospital Clínico Universitario de Valladolid, 47003 Valladolid, Spain
| | - Virginia Fernández-Espinilla
- Department of Preventive Medicine and Public Health, University of Valladolid, 47005 Valladolid, Spain; (V.F.-E.); (C.H.-G.); (J.C.-S.)
- National Influenza Centre, Edificio Rondilla, Hospital Clínico Universitario de Valladolid, 47009 Valladolid, Spain; (M.T.-A.); (I.S.-M.); (J.M.E.)
- Preventive Medicine and Public Health Service, Hospital Clínico Universitario de Valladolid, 47003 Valladolid, Spain
| | - Cristina Hernán-García
- Department of Preventive Medicine and Public Health, University of Valladolid, 47005 Valladolid, Spain; (V.F.-E.); (C.H.-G.); (J.C.-S.)
- National Influenza Centre, Edificio Rondilla, Hospital Clínico Universitario de Valladolid, 47009 Valladolid, Spain; (M.T.-A.); (I.S.-M.); (J.M.E.)
- Preventive Medicine and Public Health Service, Hospital Clínico Universitario de Valladolid, 47003 Valladolid, Spain
| | - Iván Sanz-Muñoz
- National Influenza Centre, Edificio Rondilla, Hospital Clínico Universitario de Valladolid, 47009 Valladolid, Spain; (M.T.-A.); (I.S.-M.); (J.M.E.)
- Instituto de Estudios de Ciencias de la Salud de Castilla y León (ICSCYL), 42002 Soria, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFECC), 28029 Madrid, Spain
| | - Jose M. Eiros
- National Influenza Centre, Edificio Rondilla, Hospital Clínico Universitario de Valladolid, 47009 Valladolid, Spain; (M.T.-A.); (I.S.-M.); (J.M.E.)
- Microbiology Service, Hospital Universitario Río Hortega, 47012 Valladolid, Spain
| | - Javier Castrodeza-Sanz
- Department of Preventive Medicine and Public Health, University of Valladolid, 47005 Valladolid, Spain; (V.F.-E.); (C.H.-G.); (J.C.-S.)
- National Influenza Centre, Edificio Rondilla, Hospital Clínico Universitario de Valladolid, 47009 Valladolid, Spain; (M.T.-A.); (I.S.-M.); (J.M.E.)
- Preventive Medicine and Public Health Service, Hospital Clínico Universitario de Valladolid, 47003 Valladolid, Spain
| |
Collapse
|
6
|
Mdunyelwa A, Seema C, Mabaso A, Mlambo K, Mtsweni M, Maphanga M, Rammutla E, Tempelman HA, Umunnakwe CN. Evaluation of the Seegene Allplex™ RV master assay for one-step simultaneous detection of eight respiratory viruses in nasopharyngeal specimens. J Virol Methods 2024; 331:115042. [PMID: 39384158 DOI: 10.1016/j.jviromet.2024.115042] [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: 02/22/2024] [Revised: 09/27/2024] [Accepted: 10/06/2024] [Indexed: 10/11/2024]
Abstract
BACKGROUND The Seegene Allplex™ RV Master (RVM) assay is a one-step multiplex real-time reverse transcription polymerase chain reaction (RT-PCR) system for detecting eight viral respiratory pathogens from nasopharyngeal swab, aspirate, and bronchoalveolar lavage specimens. The eight RVM targets are: severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), Influenza A (Flu A), Influenza B (Flu B), Human respiratory syncytial virus (RSV), adenovirus (AdV), rhinovirus (HRV), parainfluenza virus (PIV), and metapneumovirus (MPV). The assay is based on Seegene's multiple detection temperature (MuDT) technology and provides cycle threshold (Ct) values for each of its viral targets upon PCR completion. OBJECTIVE We aimed to evaluate the diagnostic performance of the RVM assay by calculating sensitivity, specificity, accuracy, Positive Predictive Value (PPV), Negative Predictive Value (NPV), Positive Percent Agreement (PPA), Negative Percent Agreement (NPA), and Overall Percent Agreement (OPA) compared to definite diagnosis and analogous reference assays. STUDY DESIGN Diagnostic sensitivity, specificity, accuracy, PPV, and NPV were calculated by comparing the results of the RVM assay to that of definite diagnosis assays; while PPA, NPA, and OPA were calculated by comparing results of the RVM assay to that of analogous reference products. Definite diagnosis and reference methods comprised whole genome sequencing and PCR genotyping, the Allplex™ SARS-CoV-2/FluA/FluB/RSV and Respiratory Panels 1, 2, and 3 assays (Seegene), and the Xpert® Xpress SARS-CoV-2/FluA/FluB/RSV Plus assay (Cepheid). Reproducibility of the RVM assay using fully-automated and semi-automated nucleic acid (NA) extraction workflows and as performed by independent operators was also assessed. In total, 249 positive respiratory specimens and at least 50 negative specimens for each target tested were used for this evaluation study. RESULTS Sensitivity, specificity, accuracy, PPV, NPV, PPA, NPA, and OPA ranged from 95.7 % to 100 % for detecting all eight targets tested on the RVM assay. Reproducibility PPA, NPA, and OPA between automated and semi-automated NA extraction workflows were all >97.9 %, while the reproducibility PPA, NPA and OPA between independent operators were all 100 %. CONCLUSION These results demonstrate a high level of sensitivity, specificity, accuracy and diagnostic predictive value of the RVM assay and high agreement with comparable reference assays in identifying all eight of its targets. Taken together, our study underscores the diagnostic utility of the RVM assay in detecting eight viral respiratory pathogens.
Collapse
Affiliation(s)
- Anele Mdunyelwa
- Ndlovu Research Centre, Ndlovu Laboratories, Elandsdoorn, Dennilton, Limpopo, South Africa
| | - Colette Seema
- Ndlovu Research Centre, Ndlovu Laboratories, Elandsdoorn, Dennilton, Limpopo, South Africa
| | - Anna Mabaso
- Ndlovu Research Centre, Ndlovu Laboratories, Elandsdoorn, Dennilton, Limpopo, South Africa
| | - Khamusi Mlambo
- Ndlovu Research Centre, Ndlovu Laboratories, Elandsdoorn, Dennilton, Limpopo, South Africa
| | - Mandisa Mtsweni
- Ndlovu Research Centre, Ndlovu Laboratories, Elandsdoorn, Dennilton, Limpopo, South Africa
| | - Mathapelo Maphanga
- Ndlovu Research Centre, Ndlovu Laboratories, Elandsdoorn, Dennilton, Limpopo, South Africa
| | - Elizabeth Rammutla
- Ndlovu Research Centre, Ndlovu Laboratories, Elandsdoorn, Dennilton, Limpopo, South Africa
| | - Hugo A Tempelman
- Ndlovu Research Centre, Ndlovu Laboratories, Elandsdoorn, Dennilton, Limpopo, South Africa; Wits Reproductive Health and HIV Institute, University of the Witwatersrand, Johannesburg, South Africa; Utrecht University, Netherlands
| | - Chijioke N Umunnakwe
- Ndlovu Research Centre, Ndlovu Laboratories, Elandsdoorn, Dennilton, Limpopo, South Africa.
| |
Collapse
|
7
|
Chakraborty S, Chauhan A. Fighting the flu: a brief review on anti-influenza agents. Biotechnol Genet Eng Rev 2024; 40:858-909. [PMID: 36946567 DOI: 10.1080/02648725.2023.2191081] [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: 10/26/2022] [Accepted: 03/06/2023] [Indexed: 03/23/2023]
Abstract
The influenza virus causes one of the most prevalent and lethal infectious viral diseases of the respiratory system; the disease progression varies from acute self-limiting mild fever to disease chronicity and death. Although both the preventive and treatment measures have been vital in protecting humans against seasonal epidemics or sporadic pandemics, there are several challenges to curb the influenza virus such as limited or poor cross-protection against circulating virus strains, moderate protection in immune-compromised patients, and rapid emergence of resistance. Currently, there are four US-FDA-approved anti-influenza drugs to treat flu infection, viz. Rapivab, Relenza, Tamiflu, and Xofluza. These drugs are classified based on their mode of action against the viral replication cycle with the first three being Neuraminidase inhibitors, and the fourth one targeting the viral polymerase. The emergence of the drug-resistant strains of influenza, however, underscores the need for continuous innovation towards development and discovery of new anti-influenza agents with enhanced antiviral effects, greater safety, and improved tolerability. Here in this review, we highlighted commercially available antiviral agents besides those that are at different stages of development including under clinical trials, with a brief account of their antiviral mechanisms.
Collapse
Affiliation(s)
| | - Ashwini Chauhan
- Department of Microbiology, Tripura University, Agartala, India
| |
Collapse
|
8
|
Li Z, Zhou L, Zhang Q, Fan Z, Xiao C. Different effects of air pollutant concentrations on influenza A and B in Sichuan, China. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 284:116923. [PMID: 39213756 DOI: 10.1016/j.ecoenv.2024.116923] [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: 04/06/2024] [Revised: 07/15/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND The detrimental effects of air pollution on the respiratory system are well documented. Previous research has established a correlation between air pollutant concentration and the frequency of outpatient visits for influenza-like illness. However, studies investigating the variations in infection among different influenza subtypes remain sparse. We aimed to determine the correlation between air pollutant levels and different influenza subtypes in Sichuan Province, China. METHODS A generalized additive model and distributed lag nonlinear model were employed to assess the association between air pollutants and influenza subtypes, utilizing daily influenza data obtained from 30 hospitals across 21 cities in Sichuan Province. The analysis considered the temporal effects and meteorological factors. The study spanned from January 1, 2017, to December 31, 2019. To provide a more precise evaluation of the actual impact of air pollution on different subtypes of influenza, we also performed subgroup analyses based on factors such as gender, age, and geography within the population. RESULTS During the investigation, 17,462 specimens from Sichuan Province tested positive for influenza. Among these, 12,607 and 4855 were diagnosed with Flu A and B, respectively. The related risk of influenza A infection significantly increased following exposure to PM2.5 on Lag2 days (RR=1.008, 95 % confidence interval [CI]: 1.000-1.016), SO2 and CO on Lag1 days (RR=1.121, 95 % CI: 1.032-1.219; RR=1.151, 95 % CI: 1.030-1.289), and NO2 on Lag0 day (RR=1.089, 95 % CI: 1.035-1.145). PM10 and SO2 levels on Lag0 day, PM2.5 levels on Lag1 day, and CO levels on Lag6 day, with a reduced risk of influenza B (RR=0.987, 95 % CI: 0.976-0.997; RR=0.817, 95 % CI: 0.676-0.987; RR=0.979, 95 % CI: 0.970-0.989; RR=0.814, 95 % CI: 0.561-0.921). CONCLUSION The findings from the overall population and subgroup analyses indicated that the impact of air pollutant concentrations on influenza A and B is inconsistent, with influenza A demonstrating greater susceptibility to these pollutants. Minimizing the levels of SO2, CO, NO2, and PM2.5 can significantly decrease the likelihood of contracting influenza A. Analyzing the influence of environmental contaminants on different influenza subtypes can provide insights into seasonal influenza trends and guide the development of preventive and control strategies.
Collapse
Affiliation(s)
- Zhirui Li
- Department of Disease Control and Prevention, Sichuan provincial Center for Disease Control and Prevention, Chengdu, Sichuan 610000, PR China
| | - Lijun Zhou
- Department of Disease Control and Prevention, Sichuan provincial Center for Disease Control and Prevention, Chengdu, Sichuan 610000, PR China
| | - Qian Zhang
- Department of Oncology, Xiamen Fifth Hospital, Min'an Road, Maxiang Street, Xiang 'an District, Xiamen, Fujian 361000, PR China
| | - Zixuan Fan
- School of Health Policy and Management, Peking Union Medical College, Beijing 100730, PR China
| | - Chongkun Xiao
- Department of Disease Control and Prevention, Sichuan provincial Center for Disease Control and Prevention, Chengdu, Sichuan 610000, PR China.
| |
Collapse
|
9
|
Shen AK, Gutu V, Druc A, Ebama M, Belayneh A, Adams B, Valleau M, Paraschiv A. An evaluation of the National Influenza Vaccination Program in the Republic of Moldova, 2023-2024. Vaccine 2024; 42:126322. [PMID: 39293299 DOI: 10.1016/j.vaccine.2024.126322] [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: 05/07/2024] [Revised: 08/30/2024] [Accepted: 09/04/2024] [Indexed: 09/20/2024]
Abstract
During the 2023-2024 influenza season, the Republic of Moldova, a lower-middle income country seeking accession into the European Union, independently financed their influenza vaccine supply transitioning from external support from the Partnership for International Vaccine Initiatives, a collaboration conceived in 2015. As part of this transition, a mixed-methods evaluation was conducted from May 2023 - January 2024 to identify current strengths and weaknesses of the influenza vaccination program. A total of 157 interviews were conducted: one with the National Immunization Program (NIP), six with district health officers, 18 at health facilities, 18 with caregivers/parents, 34 with healthcare workers, 43 with adults with chronic diseases, 19 with pregnant women, and 13 vaccine observation sessions; further five expert interviews with an international organization, the insurance company, senior government officials in public health and within the ministry of health, and those involved with COVID-19 were conducted. The Republic of Moldova's NIP has benefited from decades of experience, internal commitments to progress, and contributions from external partners. Despite this progress, the evaluation recognized four areas for improvement. Recommendations from the evaluation assessment included: 1) develop a national strategy for immunization, including the establishment of national goals in consultation with the national immunization technical advisory group (NITAG); 2) expand immunization communications and advocacy initiatives, particularly to adults and pregnant individuals; 3) leverage trusted patient-doctor relationships and encourage vaccination as a healthcare norm with physician specialists; and 4) conduct operations research to better understand vaccine hesitancy in populations such as pregnant individuals. Additional thematic findings emphasized the importance of ensuring timely receipt of vaccine doses into the country no later than September, as medical providers reported difficulty administering doses when vaccines were delivered after September. Our findings outline ways to further strengthen the Republic of Moldova's self-sustained annual influenza vaccination program.
Collapse
Affiliation(s)
- Angela K Shen
- Task Force for Global Health, United States; Perelman School of Medicine, University of Pennsylvania, United States.
| | | | - Alina Druc
- National Agency for Public Health, Republic of Moldova
| | | | | | - Brittany Adams
- US Centers for Disease Control and Prevention, United States
| | - Molly Valleau
- US Centers for Disease Control and Prevention, United States
| | - Angela Paraschiv
- Nicolae Testemițanu State University of Medicine and Pharmacy, Chișinău, Republic of Moldova
| |
Collapse
|
10
|
Wang M, Chen J, Zhang ZL. Highly-Efficient Selection of Aptamers for Quantitative Fluorescence Detecting Multiple IAV Subtypes. Anal Chem 2024. [PMID: 39259665 DOI: 10.1021/acs.analchem.4c03052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/13/2024]
Abstract
Influenza A virus (IAV) can cause infectious respiratory diseases in humans and animals. IAVs mutate rapidly through antigenic drift and shift, resulting in the emergence of numerous IAV subtypes and significant challenges for IAV detection. Therefore, achieving the simultaneous detection of multiple IAVs is crucial. In this work, three specific aptamers targeting the hemagglutination (HA) protein of the influenza A H5N1, H7N9, and H9N2 viruses were screened using a multichannel magnetic microfluidic chip. The aptamers exhibit nanomolar affinity and excellent specificity for the HA protein of H5N1, H7N9, and H9N2 viruses. Furthermore, three specific aptamers were truncated and labeled with different fluorescence markers to realize fluorescence quantitative detection of influenza A H5N1, H7N9, and H9N2 viruses through an aptamer sandwich assay in 1 h. The limit of detection (LOD) of the developed method is 0.38 TCID50/mL for the H5N1 virus, 0.75 TCID50/mL for the H7N9 virus, and 1.14 TCID50/mL for the H9N2 virus. The detection method has excellent specificity, strong anti-interference ability, and good reproducibility. This work provides a sensitive quantitative detection method for the H5N1, H7N9, and H9N2 viruses, enabling quantitative fluorescence detection for multiple IAV subtypes.
Collapse
Affiliation(s)
- Meng Wang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, Hubei 430072, China
| | - Jianjun Chen
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei 430071, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhi-Ling Zhang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, Hubei 430072, China
| |
Collapse
|
11
|
Hsu D, Jayaraman A, Pucci A, Joshi R, Mancini K, Chen HL, Koslovsky K, Mao X, Choi A, Henry C, Vakil J, Stadlbauer D, Jorquera P, Arunkumar GA, Sanchez-Crespo NE, Wadsworth LT, Bhupathy V, Du E, Avanesov A, Ananworanich J, Nachbagauer R. Safety and immunogenicity of mRNA-based seasonal influenza vaccines formulated to include multiple A/H3N2 strains with or without the B/Yamagata strain in US adults aged 50-75 years: a phase 1/2, open-label, randomised trial. THE LANCET. INFECTIOUS DISEASES 2024:S1473-3099(24)00493-6. [PMID: 39245055 DOI: 10.1016/s1473-3099(24)00493-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 07/18/2024] [Accepted: 07/23/2024] [Indexed: 09/10/2024]
Abstract
BACKGROUND Inclusion of additional influenza A/H3N2 strains in seasonal influenza vaccines could expand coverage against multiple, antigenically distinct, cocirculating A/H3N2 clades and potentially replace the no longer circulating B/Yamagata strain. We aimed to evaluate the safety and immunogenicity of three next-generation seasonal influenza mRNA vaccines with different compositions that encode for haemagglutinins of multiple A/H3N2 strains, with or without the B/Yamagata strain, in adults. METHODS This randomised, open-label, phase 1/2 trial enrolled healthy adults aged 50-75 years across 22 sites in the USA. Participants were randomly assigned (1:1:1:1:1:1:1) via interactive response technology to receive a single dose of mRNA-1011.1 (pentavalent; containing one additional A/H3N2 strain [Newcastle]), mRNA-1011.2 (quadrivalent; B/Yamagata replaced with one additional A/H3N2 strain [Newcastle]), mRNA-1012 at one of two dose levels (pentavalent; B/Yamagata replaced with two additional A/H3N2 strains [Newcastle and Hong Kong]), or one of three quadrivalent mRNA-1010 controls each encoding one of the A/H3N2 study strains. The primary outcomes were safety, evaluated in all randomly assigned participants who received a study vaccination (safety population), and reactogenicity, evaluated in all participants from the safety population who contributed any solicited adverse reaction data (solicited safety population). The secondary outcome was humoral immunogenicity of investigational mRNA vaccines at day 29 versus mRNA-1010 control vaccines based on haemagglutination inhibition antibody (HAI) assay in the per-protocol population. Here, we summarise findings from the planned interim analysis after participants had completed day 29. The study is registered with ClinicalTrials.gov, NCT05827068, and is ongoing. FINDINGS Between March 27 and May 9, 2023, 1183 participants were screened for eligibility, 699 (59·1%) were randomly assigned, and 696 (58·8%) received vaccination (safety population, n=696; solicited safety population, n=694; per-protocol population, n=646). 382 (55%) of the 696 participants in the safety population self-reported as female and 314 (45%) as male. Frequencies of solicited adverse reactions were similar across vaccine groups; 551 (79%) of 694 participants reported at least one solicited adverse reaction within 7 days after vaccination and 83 (12%) of 696 participants reported at least one unsolicited adverse event within 28 days after vaccination. No vaccine-related serious adverse events or deaths were reported. All three next-generation influenza vaccines elicited robust antibody responses against vaccine-matched influenza A and B strains at day 29 that were generally similar to mRNA-1010 controls, and higher responses against additional A/H3N2 strains that were not included within respective mRNA-1010 controls. Day 29 geometric mean fold rises in HAI titres from day 1 against vaccine-matched A/H3N2 strains were 3·0 (95% CI 2·6-3·6; Darwin) and 3·1 (2·6-3·8; Newcastle) for mRNA-1011.1; 3·3 (2·7-4·1; Darwin) and 4·2 (3·4-5·2; Newcastle) for mRNA-1011.2; 3·4 (2·9-4·0; Darwin), 4·5 (3·6-5·5; Newcastle), and 5·1 (4·2-6·2; Hong Kong) for mRNA-1012 50·0 μg; and 2·6 (2·2-3·1; Darwin), 3·7 (3·0-4·6; Newcastle), and 4·1 (3·3-5·1; Hong Kong) for mRNA-1012 62·5 μg. Inclusion of additional A/H3N2 strains did not reduce responses against influenza A/H1N1 or influenza B strains, and removal of B/Yamagata did not affect responses to B/Victoria. INTERPRETATION These data support the continued clinical development of mRNA-based next-generation seasonal influenza vaccines with broadened influenza A/H3N2 strain coverage. FUNDING Moderna.
Collapse
|
12
|
Zhang M, Yang C, Wu X, Wang Y, Wang L, Cui Q, Tong J, An Y, Cai M, Cheng S, Jiang Q, Wang Y, Zhao C, Wang Y, Huang W. Antigenic analysis of the influenza B virus hemagglutinin protein. Virol Sin 2024:S1995-820X(24)00139-1. [PMID: 39233140 DOI: 10.1016/j.virs.2024.08.012] [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: 02/22/2024] [Accepted: 08/30/2024] [Indexed: 09/06/2024] Open
Abstract
Influenza B viruses (IBVs) primarily infect humans and are a common cause of respiratory infections in humans. Here, to systematically analyze the antigenicity of the IBVs Hemagglutinin (HA) protein, 31 B/Victoria and 19 B/Yamagata representative circulating strains were selected from Global Initiative of Sharing All Influenza Data (GISAID), and pseudotyped viruses were constructed with the vesicular stomatitis virus system. Guinea pigs were immunized with three doses of vaccines (one dose of DNA vaccines following two doses of pseudotyped virus vaccines) of the seven IBV vaccine strains, and neutralizing antibodies against the pseudotyped viruses were tested. By comparing differences between various vaccine strains, we constructed several pseudotyped viruses that contained various mutations based on vaccine strain BV-21. The vaccine strains showed good neutralization levels against the epidemic virus strains of the same year, with neutralization titers ranging from 370 to 840, while the level of neutralization against viruses prevalent in previous years decreased 1-10-fold. Each of the high-frequency epidemic strains of B/Victoria and B/Yamagata not only induced high neutralizing titers, but also had broadly neutralizing effects against virus strains of different years, with neutralizing titers ranging from 1000 to 7200. R141G, D197 N, and R203K were identified as affecting the antigenicity of IBV. These mutation sites provide valuable references for the selection and design of a universal IBV vaccine strain in the future.
Collapse
Affiliation(s)
- Mengyi Zhang
- Division of HIV/AIDS and Sex-Transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing, 102629, China; National Institutes for Food and Drug Control, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, 100730, China
| | - Chaoying Yang
- Division of HIV/AIDS and Sex-Transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing, 102629, China; National Vaccine & Serum Institute, Beijing, 101111, China
| | - Xi Wu
- Division of HIV/AIDS and Sex-Transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing, 102629, China
| | - Yifei Wang
- Division of HIV/AIDS and Sex-Transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing, 102629, China
| | - Lijie Wang
- Division of HIV/AIDS and Sex-Transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing, 102629, China
| | - Qianqian Cui
- Division of HIV/AIDS and Sex-Transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing, 102629, China
| | - Jincheng Tong
- Division of HIV/AIDS and Sex-Transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing, 102629, China
| | - Yimeng An
- Division of HIV/AIDS and Sex-Transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing, 102629, China
| | - Meina Cai
- Division of HIV/AIDS and Sex-Transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing, 102629, China
| | - Shishi Cheng
- Division of HIV/AIDS and Sex-Transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing, 102629, China
| | - Qi Jiang
- Division of HIV/AIDS and Sex-Transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing, 102629, China
| | - Yulin Wang
- National Vaccine & Serum Institute, Beijing, 101111, China.
| | - Chenyan Zhao
- Division of HIV/AIDS and Sex-Transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing, 102629, China.
| | - Youchun Wang
- Institute of Medical Biology, Chinese Academy of Medical Science & Peking Union Medical College, Kunming, 650031, China.
| | - Weijin Huang
- Division of HIV/AIDS and Sex-Transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing, 102629, China; National Institutes for Food and Drug Control, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|
13
|
Feys S, Carvalho A, Clancy CJ, Gangneux JP, Hoenigl M, Lagrou K, Rijnders BJA, Seldeslachts L, Vanderbeke L, van de Veerdonk FL, Verweij PE, Wauters J. Influenza-associated and COVID-19-associated pulmonary aspergillosis in critically ill patients. THE LANCET. RESPIRATORY MEDICINE 2024; 12:728-742. [PMID: 39025089 DOI: 10.1016/s2213-2600(24)00151-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 05/06/2024] [Accepted: 05/07/2024] [Indexed: 07/20/2024]
Abstract
Influenza-associated pulmonary aspergillosis (IAPA) and COVID-19-associated pulmonary aspergillosis (CAPA) are increasingly recognised as important complications in patients requiring intensive care for severe viral pneumonia. The diagnosis can typically be made in 10-20% of patients with severe influenza or COVID-19, but only when appropriate diagnostic tools are used. Bronchoalveolar lavage sampling for culture, galactomannan testing, and PCR forms the cornerstone of diagnosis, whereas visual examination of the tracheobronchial tract during bronchoscopy is required to detect invasive Aspergillus tracheobronchitis. Azoles are the first-choice antifungal drugs, with liposomal amphotericin B as an alternative in settings where azole resistance is prevalent. Despite antifungal therapy, IAPA and CAPA are associated with poor outcomes, with fatality rates often exceeding 50%. In this Review, we discuss the mechanistic and clinical aspects of IAPA and CAPA. Moreover, we identify crucial knowledge gaps and formulate directions for future research.
Collapse
Affiliation(s)
- Simon Feys
- Medical Intensive Care Unit, University Hospitals Leuven, Leuven, Belgium; Department of Internal Medicine, University Hospitals Leuven, Leuven, Belgium; Department of Microbiology, Immunology, and Transplantation, KU Leuven, Leuven, Belgium.
| | - Agostinho Carvalho
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal; ICVS/3B's Associate Laboratory, Braga/ Guimarães, Portugal
| | - Cornelius J Clancy
- Division of Infectious Diseases, University of Pittsburgh, PA, USA; VA Pittsburgh Healthcare System, Pittsburgh, PA, USA
| | - Jean-Pierre Gangneux
- Université de Rennes, CHU Rennes, INSERM, EHESP, IRSET, UMR_S 1085, Rennes, France; Centre Hospitalier Universitaire de Rennes, Laboratoire de Parasitologie-Mycologie, ECMM Excellence Center in Medical Mycology, French National Reference Center on Mycoses and Antifungals (CNRMA-LA AspC), Rennes, France
| | - Martin Hoenigl
- Division of Infectious Diseases, ECMM Excellence Center in Medical Mycology, Department of Internal Medicine, Medical University of Graz, Graz, Austria; Translational Medical Mycology Research Group, Medical University of Graz, Graz, Austria; Bio TechMed-Graz, Graz, Austria
| | - Katrien Lagrou
- Department of Microbiology, Immunology, and Transplantation, KU Leuven, Leuven, Belgium; Department of Laboratory Medicine and National Reference Center for Mycosis, University Hospitals Leuven, Leuven, Belgium
| | - Bart J A Rijnders
- Department of Internal Medicine, Section of Infectious Diseases and Department of Medical Microbiology and Infectious Diseases, Erasmus MC, University Medical Center, Rotterdam, Netherlands
| | | | - Lore Vanderbeke
- Department of Internal Medicine, University Hospitals Leuven, Leuven, Belgium
| | | | - Paul E Verweij
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, Netherlands; Center of Expertise for Mycology, Radboud University Medical Center/Canisius Wilhelmina Hospital, Nijmegen, Netherlands
| | - Joost Wauters
- Medical Intensive Care Unit, University Hospitals Leuven, Leuven, Belgium; Department of Internal Medicine, University Hospitals Leuven, Leuven, Belgium; Department of Microbiology, Immunology, and Transplantation, KU Leuven, Leuven, Belgium
| |
Collapse
|
14
|
Sun R, Zhang X, Hou J, Jia W, Li P, Song C. Development and validation of nomogram for predicting the risk of transferring to the ICU for children with influenza. Eur J Clin Microbiol Infect Dis 2024; 43:1795-1805. [PMID: 39002105 DOI: 10.1007/s10096-024-04898-5] [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: 05/16/2024] [Accepted: 07/08/2024] [Indexed: 07/15/2024]
Abstract
OBJECTIVE Development of a nomogram model for predicting the magnitude of risk of transferring hospitalized children with influenza to the ICU. METHODS In a single-center retrospective study, 318 children with influenza who were hospitalized in our hospital from January 2018 to August 2023 were collected as study subjects. Children with influenza were randomly assigned to the training set and validation set in a ratio of 4:1. In the training set, risk factors were identified using univariate and multivariate logistic regression analyses, and a nomogram model was created on this basis. The validation set was used to evaluate the predictive power of the model. RESULTS Multifactorial logistic regression analysis revealed six independent risk factors for transfer to the ICU in hospitalized children with influenza, including elevated peripheral white blood cell counts, elevated large platelet ratios, reduced mean platelet width, reduced complement C3, elevated serum globulin levels, and reduced total immunoglobulin M levels. Using these six metrics as predictors to construct a nomogram graphical model, the C-index was 0.970 (95% Cl: 0.953-0.988). The areas under the curve for the training and validation sets were 0.966 (95%Cl 0.947-0.985) and 0.919 (95%Cl 0.851-0.986), respectively. CONCLUSION A nomogram for predicting the risk of transferring to the ICU for children with influenza was developed and validated, which demonstrates good calibration and clinical benefits.
Collapse
Affiliation(s)
- Ruiyang Sun
- Henan Province Engineering Research Center of Diagnosis and Treatment of Pediatric Infection and Critical Care, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, 450052, China
| | - Xue Zhang
- Henan Province Engineering Research Center of Diagnosis and Treatment of Pediatric Infection and Critical Care, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, 450052, China
| | - Jiapu Hou
- Henan Province Engineering Research Center of Diagnosis and Treatment of Pediatric Infection and Critical Care, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, 450052, China
| | - Wanyu Jia
- Henan Province Engineering Research Center of Diagnosis and Treatment of Pediatric Infection and Critical Care, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, 450052, China
| | - Peng Li
- Henan Province Engineering Research Center of Diagnosis and Treatment of Pediatric Infection and Critical Care, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, 450052, China
| | - Chunlan Song
- Henan Province Engineering Research Center of Diagnosis and Treatment of Pediatric Infection and Critical Care, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, 450052, China.
| |
Collapse
|
15
|
Zhao M, Zhang B, Yan M, Zhao Z. Development and validation of a nomogram to predict severe influenza. Immun Inflamm Dis 2024; 12:e70026. [PMID: 39340342 PMCID: PMC11437489 DOI: 10.1002/iid3.70026] [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: 02/21/2024] [Revised: 09/09/2024] [Accepted: 09/11/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Influenza is an acute respiratory disease posing significant harm to human health. Early prediction and intervention in patients at risk of developing severe influenza can significantly decrease mortality. METHOD A comprehensive analysis of 146 patients with influenza was conducted using the Gene Expression Omnibus (GEO) database. We assessed the relationship between severe influenza and patients' clinical information and molecular characteristics. First, the variables of differentially expressed genes were selected using R software. Least absolute shrinkage and selection operator (LASSO) and multivariate logistic regression analysis were performed to investigate the association between clinical information and molecular characteristics and severe influenza. A nomogram was developed to predict the presence of severe influenza. At the same time, the concordance index (C-index) is adopted area under the receiver operating characteristic (ROC), area under the curve (AUC), decision curve analysis (DCA), and calibration curve to evaluate the predictive ability of the model and its clinical application. RESULTS Severe influenza was identified in 47 of 146 patients (32.20%) and was significantly related to age and duration of illness. Multivariate logistic regression demonstrated significant correlations between severe influenza and myloperoxidase (MPO) level, haptoglobin (HP) level, and duration of illness. A nomogram was formulated based on MPO level, HP level, and duration of illness. This model produced a C-index of 0.904 and AUC of 0.904. CONCLUSIONS A nomogram based on the expression levels of MPO, HP, and duration of illness is an efficient model for the early identification of patients with severe influenza. These results will be useful in guiding prevention and treatment for severe influenza disease.
Collapse
Affiliation(s)
- Mingzhen Zhao
- Pulmonary and Critical Care MedicineAffiliated Hospital of Chengde Medical UniversityChengdeHebeiChina
| | - Bo Zhang
- Pulmonary and Critical Care MedicineAffiliated Hospital of Chengde Medical UniversityChengdeHebeiChina
| | - Mingjun Yan
- Pulmonary and Critical Care MedicineAffiliated Hospital of Chengde Medical UniversityChengdeHebeiChina
| | - Zhiwei Zhao
- Pulmonary and Critical Care MedicineAffiliated Hospital of Chengde Medical UniversityChengdeHebeiChina
| |
Collapse
|
16
|
Zhu H, Li X, Li X, Chen H, Qian P. Protection against the H1N1 influenza virus using self-assembled nanoparticles formed by lumazine synthase and bearing the M2e peptide. Virology 2024; 597:110162. [PMID: 38955082 DOI: 10.1016/j.virol.2024.110162] [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: 03/28/2024] [Revised: 06/21/2024] [Accepted: 06/25/2024] [Indexed: 07/04/2024]
Abstract
There is an urgent need for influenza vaccines that offer broad cross-protection. The highly conserved ectodomain of the influenza matrix protein 2 (M2e) is a promising candidate; however, its low immunogenicity can be addressed. In this study, we developed influenza vaccines using the Lumazine synthase (LS) platform. The primary objective of this study was to determine the protective potential of M2e proteins expressed on Lumazine synthase (LS) nanoparticles. M2e-LS proteins, produced through the E. coli system, spontaneously assemble into nanoparticles. The study investigated the efficacy of the M2e-LS nanoparticle vaccine in mice. Mice immunized with M2e-LS nanoparticles exhibited significantly higher levels of intracellular cytokines than those receiving soluble M2e proteins. The M2e-LS protein exhibited robust immunogenicity and provided 100% protection against cross-clade influenza.
Collapse
Affiliation(s)
- Hechao Zhu
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei, China; Guangxi Yangxiang Co., LTD, Guigang, 537100, China
| | - Xinxin Li
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei, China
| | - Xiangmin Li
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei, China
| | - Huanchun Chen
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei, China
| | - Ping Qian
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei, China.
| |
Collapse
|
17
|
Dias RA. Towards a Comprehensive Definition of Pandemics and Strategies for Prevention: A Historical Review and Future Perspectives. Microorganisms 2024; 12:1802. [PMID: 39338476 PMCID: PMC11433773 DOI: 10.3390/microorganisms12091802] [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: 08/12/2024] [Revised: 08/22/2024] [Accepted: 08/28/2024] [Indexed: 09/30/2024] Open
Abstract
The lack of a universally accepted definition of a pandemic hinders a comprehensive understanding of and effective response to these global health crises. Current definitions often lack quantitative criteria, rendering them vague and limiting their utility. Here, we propose a refined definition that considers the likelihood of susceptible individuals contracting an infectious disease that culminates in widespread global transmission, increased morbidity and mortality, and profound societal, economic, and political consequences. Applying this definition retrospectively, we identify 22 pandemics that occurred between 165 and 2024 AD and were caused by a variety of diseases, including smallpox (Antonine and American), plague (Justinian, Black Death, and Third Plague), cholera (seven pandemics), influenza (two Russian, Spanish, Asian, Hong Kong, and swine), AIDS, and coronaviruses (SARS, MERS, and COVID-19). This work presents a comprehensive analysis of past pandemics caused by both emerging and re-emerging pathogens, along with their epidemiological characteristics, societal impact, and evolution of public health responses. We also highlight the need for proactive measures to reduce the risk of future pandemics. These strategies include prioritizing surveillance of emerging zoonotic pathogens, conserving biodiversity to counter wildlife trafficking, and minimizing the potential for zoonotic spillover events. In addition, interventions such as promoting alternative protein sources, enforcing the closure of live animal markets in biodiversity-rich regions, and fostering global collaboration among diverse stakeholders are critical to preventing future pandemics. Crucially, improving wildlife surveillance systems will require the concerted efforts of local, national and international entities, including laboratories, field researchers, wildlife conservationists, government agencies and other stakeholders. By fostering collaborative networks and establishing robust biorepositories, we can strengthen our collective capacity to detect, monitor, and mitigate the emergence and transmission of zoonotic pathogens.
Collapse
Affiliation(s)
- Ricardo Augusto Dias
- School of Veterinary Medicine, University of Sao Paulo, Av. Prof. Dr. Orlando Marques de Paiva, 87, São Paulo 05508-270, Brazil
| |
Collapse
|
18
|
Weber DJ, Zimmerman KO, Tartof SY, McLaughlin JM, Pather S. Risk of COVID-19 in Children throughout the Pandemic and the Role of Vaccination: A Narrative Review. Vaccines (Basel) 2024; 12:989. [PMID: 39340021 PMCID: PMC11435672 DOI: 10.3390/vaccines12090989] [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: 07/09/2024] [Revised: 08/20/2024] [Accepted: 08/22/2024] [Indexed: 09/30/2024] Open
Abstract
At the beginning of the coronavirus disease 2019 (COVID-19) pandemic, persons ≥65 years of age and healthcare personnel represented the most vulnerable groups with respect to risk of infection, severe illness, and death. However, as the pandemic progressed, there was an increasingly detrimental effect on young children and adolescents. Severe disease and hospitalization increased over time in pediatric populations, and containment measures created substantial psychosocial, educational, and economic challenges for young people. Vaccination of children against COVID-19 has been shown to reduce severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections and severe outcomes in pediatric populations and may also help to prevent the spread of variants of concern and improve community immunity. This review discusses the burden of COVID-19 on children throughout the pandemic, the role of children in disease transmission, and the impact of COVID-19 vaccination.
Collapse
Affiliation(s)
- David J Weber
- Division of Infectious Diseases, UNC School of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Kanecia O Zimmerman
- Duke Department of Pediatrics, Duke University School of Medicine, Durham, NC 27710, USA
| | - Sara Y Tartof
- Department of Research & Evaluation, Kaiser Permanente Southern California, Pasadena, CA 91107, USA
| | | | - Shanti Pather
- BioNTech SE, An der Goldgrube 12, 55131 Mainz, Germany
| |
Collapse
|
19
|
Zhu H, Chen S, Qin W, Aynur J, Chen Y, Wang X, Chen K, Xie Z, Li L, Liu Y, Chen G, Ou J, Zheng K. Study on the impact of meteorological factors on influenza in different periods and prediction based on artificial intelligence RF-Bi-LSTM algorithm: to compare the COVID-19 period with the non-COVID-19 period. BMC Infect Dis 2024; 24:878. [PMID: 39198754 PMCID: PMC11360838 DOI: 10.1186/s12879-024-09750-x] [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: 03/22/2024] [Accepted: 08/12/2024] [Indexed: 09/01/2024] Open
Abstract
OBJECTIVE At different times, public health faces various challenges and the degree of intervention measures varies. The research on the impact and prediction of meteorology factors on influenza is increasing gradually, however, there is currently no evidence on whether its research results are affected by different periods. This study aims to provide limited evidence to reveal this issue. METHODS Daily data on influencing factors and influenza in Xiamen were divided into three parts: overall period (phase AB), non-COVID-19 epidemic period (phase A), and COVID-19 epidemic period (phase B). The association between influencing factors and influenza was analysed using generalized additive models (GAMs). The excess risk (ER) was used to represent the percentage change in influenza as the interquartile interval (IQR) of meteorology factors increases. The 7-day average daily influenza cases were predicted using the combination of bi-directional long short memory (Bi-LSTM) and random forest (RF) through multi-step rolling input of the daily multifactor values of the previous 7-day. RESULTS In periods A and AB, air temperature below 22 °C was a risk factor for influenza. However, in phase B, temperature showed a U-shaped effect on it. Relative humidity had a more significant cumulative effect on influenza in phase AB than in phase A (peak: accumulate 14d, AB: ER = 281.54, 95% CI = 245.47 ~ 321.37; A: ER = 120.48, 95% CI = 100.37 ~ 142.60). Compared to other age groups, children aged 4-12 were more affected by pressure, precipitation, sunshine, and day light, while those aged ≥ 13 were more affected by the accumulation of humidity over multiple days. The accuracy of predicting influenza was highest in phase A and lowest in phase B. CONCLUSIONS The varying degrees of intervention measures adopted during different phases led to significant differences in the impact of meteorology factors on influenza and in the influenza prediction. In association studies of respiratory infectious diseases, especially influenza, and environmental factors, it is advisable to exclude periods with more external interventions to reduce interference with environmental factors and influenza related research, or to refine the model to accommodate the alterations brought about by intervention measures. In addition, the RF-Bi-LSTM model has good predictive performance for influenza.
Collapse
Affiliation(s)
- Hansong Zhu
- Fujian Provincial Center for Disease Control and Prevention, Fuzhou, Fujian, 350012, China.
- School of Public Health, Fujian Medical University, Fuzhou, Fujian, 350011, China.
| | - Si Chen
- Fujian Institute of Meteorological Sciences, Fuzhou, Fujian, 350007, China
- Fujian Key Laboratory of Severe Weather, Fuzhou, Fujian, 350007, China
- Key Laboratory of Straits Severe Weather, China Meteorological Administration, Fuzhou, Fujian, 350007, China
| | - Weixia Qin
- The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, 361003, China
| | - Joldosh Aynur
- School of Public Health, Xiamen University, Xiamen, Fujian, 361100, China
| | - Yuyan Chen
- Fujian Provincial Judicial Drug Rehabilitation Hospital, Fuzhou, Fujian, 350007, China
| | - Xiaoying Wang
- School of Public Health, Xiamen University, Xiamen, Fujian, 361100, China
| | - Kaizhi Chen
- Fuzhou University, Fuzhou, Fujian, 350108, China
| | - Zhonghang Xie
- Fujian Provincial Center for Disease Control and Prevention, Fuzhou, Fujian, 350012, China
- School of Public Health, Fujian Medical University, Fuzhou, Fujian, 350011, China
| | - Lingfang Li
- Fujian Provincial Center for Disease Control and Prevention, Fuzhou, Fujian, 350012, China
| | - Yu Liu
- Xiangnan University, Chenzhou, Hunan, 423001, China.
| | - Guangmin Chen
- Fujian Provincial Center for Disease Control and Prevention, Fuzhou, Fujian, 350012, China.
- School of Public Health, Fujian Medical University, Fuzhou, Fujian, 350011, China.
| | - Jianming Ou
- Fujian Provincial Center for Disease Control and Prevention, Fuzhou, Fujian, 350012, China.
- School of Public Health, Fujian Medical University, Fuzhou, Fujian, 350011, China.
| | - Kuicheng Zheng
- Fujian Provincial Center for Disease Control and Prevention, Fuzhou, Fujian, 350012, China.
- School of Public Health, Fujian Medical University, Fuzhou, Fujian, 350011, China.
| |
Collapse
|
20
|
Gao Y, Guyatt G, Uyeki TM, Liu M, Chen Y, Zhao Y, Shen Y, Xu J, Zheng Q, Li Z, Zhao W, Luo S, Chen X, Tian J, Hao Q. Antivirals for treatment of severe influenza: a systematic review and network meta-analysis of randomised controlled trials. Lancet 2024; 404:753-763. [PMID: 39181595 PMCID: PMC11369965 DOI: 10.1016/s0140-6736(24)01307-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 06/12/2024] [Accepted: 06/18/2024] [Indexed: 08/27/2024]
Abstract
BACKGROUND The optimal antiviral drug for treatment of severe influenza remains unclear. To support updated WHO influenza clinical guidelines, this systematic review and network meta-analysis evaluated antivirals for treatment of patients with severe influenza. METHODS We systematically searched MEDLINE, Embase, Cochrane Central Register of Controlled Trials, Cumulative Index to Nursing and Allied Health Literature, Global Health, Epistemonikos, and ClinicalTrials.gov for randomised controlled trials published up to Sept 20, 2023, that enrolled hospitalised patients with suspected or laboratory-confirmed influenza and compared direct-acting influenza antivirals against placebo, standard care, or another antiviral. Pairs of coauthors independently extracted data on study characteristics, patient characteristics, antiviral characteristics, and outcomes, with discrepancies resolved by discussion or by a third coauthor. Key outcomes of interest were time to alleviation of symptoms, duration of hospitalisation, admission to intensive care unit, progression to invasive mechanical ventilation, duration of mechanical ventilation, mortality, hospital discharge destination, emergence of antiviral resistance, adverse events, adverse events related to treatments, and serious adverse events. We conducted frequentist network meta-analyses to summarise the evidence and evaluated the certainty of evidence using the GRADE (Grading of Recommendations Assessment, Development and Evaluation) approach. This study is registered with PROSPERO, CRD42023456650. FINDINGS Of 11 878 records identified by our search, eight trials with 1424 participants (mean age 36-60 years for trials that reported mean or median age; 43-78% male patients) were included in this systematic review, of which six were included in the network meta-analysis. The effects of oseltamivir, peramivir, or zanamivir on mortality compared with placebo or standard care without placebo for seasonal and zoonotic influenza were of very low certainty. Compared with placebo or standard care, we found low certainty evidence that duration of hospitalisation for seasonal influenza was reduced with oseltamivir (mean difference -1·63 days, 95% CI -2·81 to -0·45) and peramivir (-1·73 days, -3·33 to -0·13). Compared with standard care, there was little or no difference in time to alleviation of symptoms with oseltamivir (0·34 days, -0·86 to 1·54; low certainty evidence) or peramivir (-0·05 days, -0·69 to 0·59; low certainty evidence). There were no differences in adverse events or serious adverse events with oseltamivir, peramivir, and zanamivir (very low certainty evidence). Uncertainty remains about the effects of antivirals on other outcomes for patients with severe influenza. Due to the small number of eligible trials, we could not test for publication bias. INTERPRETATION In hospitalised patients with severe influenza, oseltamivir and peramivir might reduce duration of hospitalisation compared with standard care or placebo, although the certainty of evidence is low. The effects of all antivirals on mortality and other important patient outcomes are very uncertain due to scarce data from randomised controlled trials. FUNDING World Health Organization.
Collapse
Affiliation(s)
- Ya Gao
- Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China; Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON, Canada
| | - Gordon Guyatt
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON, Canada; Department of Medicine, McMaster University, Hamilton, ON, Canada; MAGIC Evidence Ecosystem Foundation, Oslo, Norway
| | - Timothy M Uyeki
- Influenza Division, US Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Ming Liu
- Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China; Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON, Canada
| | - Yamin Chen
- Clinical Nursing Teaching and Research Section, The Second Xiangya Hospital, Central South University, Changsha, China; Xiangya School of Nursing, Central South University, Changsha, China
| | - Yunli Zhao
- Department of Geriatric Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China; Chongqing Municipality Clinical Research Center for Geriatrics, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yanjiao Shen
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON, Canada; Chinese Evidence-Based Medicine Center, West China Hospital, Sichuan University, Chengdu, China
| | - Jianguo Xu
- Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Qingyong Zheng
- Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Zhifan Li
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Wanyu Zhao
- National Clinical Research Centre for Geriatrics, West China Hospital, Sichuan University, Chengdu, China; Center of Gerontology and Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Shuyue Luo
- National Clinical Research Centre for Geriatrics, West China Hospital, Sichuan University, Chengdu, China; Center of Gerontology and Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaoyan Chen
- Department of Geriatric, Zigong Affiliated Hospital of Southwest Medical University, Zigong, China
| | - Jinhui Tian
- Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Qiukui Hao
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON, Canada; School of Rehabilitation Science, McMaster University, Hamilton, ON, Canada.
| |
Collapse
|
21
|
Qiu H, Yuan XY, Holloway K, Wood H, Cabral T, Grant C, McQueen P, Westmacott G, Beniac DR, Lin L, Carpenter M, Kobasa D, Gräfenhan T, Cheney IW. Development and characterization of monoclonal antibodies recognizing nucleocapsid protein of multiple SARS-CoV-2 variants. Heliyon 2024; 10:e35325. [PMID: 39170261 PMCID: PMC11336563 DOI: 10.1016/j.heliyon.2024.e35325] [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: 12/19/2023] [Revised: 07/25/2024] [Accepted: 07/26/2024] [Indexed: 08/23/2024] Open
Abstract
Rapid antigen test (RAT) is widely used for SARS-CoV-2 infection diagnostics. However, test sensitivity has decreased recently due to the emergence of the Omicron variant and its sublineages. Here we developed a panel of SARS-CoV-2 nucleocapsid protein (NP) specific mouse monoclonal antibodies (mAbs) and assessed their sensitivity and specificity to important SARS-CoV-2 variants. We identified seven mAbs that exhibited strong reactivity to SARS-CoV-2 variants and recombinant NP (rNP) by Western immunoblot or ELISA. Their specificity to SARS-CoV-2 was confirmed by negative or low reactivity to rNPs from SARS-CoV-1, MERS, and common human coronaviruses (HCoV-HKU1, HCoV-CO43, HCoV-NL63, and HCoV-229E). These seven mAbs were further tested by immunoplaque assay against selected variants of concern (VOCs), including two Omicron sublineages, and five mAbs (F461G13, F461G7, F459G7, F457G3, and F461G6), showed strong reactions, warranting further suitability testing for the development of diagnostic assay.
Collapse
Affiliation(s)
- Hongyu Qiu
- National Microbiology Laboratory, Public Health Agency of Canada, 1015 Arlington Street, Winnipeg, MB, R3E 3R2, Canada
| | - Xin-Yong Yuan
- National Microbiology Laboratory, Public Health Agency of Canada, 1015 Arlington Street, Winnipeg, MB, R3E 3R2, Canada
| | - Kimberly Holloway
- National Microbiology Laboratory, Public Health Agency of Canada, 1015 Arlington Street, Winnipeg, MB, R3E 3R2, Canada
| | - Heidi Wood
- National Microbiology Laboratory, Public Health Agency of Canada, 1015 Arlington Street, Winnipeg, MB, R3E 3R2, Canada
| | - Teresa Cabral
- National Microbiology Laboratory, Public Health Agency of Canada, 1015 Arlington Street, Winnipeg, MB, R3E 3R2, Canada
| | - Chris Grant
- National Microbiology Laboratory, Public Health Agency of Canada, 1015 Arlington Street, Winnipeg, MB, R3E 3R2, Canada
| | - Peter McQueen
- National Microbiology Laboratory, Public Health Agency of Canada, 1015 Arlington Street, Winnipeg, MB, R3E 3R2, Canada
| | - Garrett Westmacott
- National Microbiology Laboratory, Public Health Agency of Canada, 1015 Arlington Street, Winnipeg, MB, R3E 3R2, Canada
| | - Daniel R. Beniac
- National Microbiology Laboratory, Public Health Agency of Canada, 1015 Arlington Street, Winnipeg, MB, R3E 3R2, Canada
| | - Lisa Lin
- National Microbiology Laboratory, Public Health Agency of Canada, 1015 Arlington Street, Winnipeg, MB, R3E 3R2, Canada
| | - Michael Carpenter
- National Microbiology Laboratory, Public Health Agency of Canada, 1015 Arlington Street, Winnipeg, MB, R3E 3R2, Canada
| | - Darwyn Kobasa
- National Microbiology Laboratory, Public Health Agency of Canada, 1015 Arlington Street, Winnipeg, MB, R3E 3R2, Canada
| | | | - Ian Wayne Cheney
- National Microbiology Laboratory, Public Health Agency of Canada, 1015 Arlington Street, Winnipeg, MB, R3E 3R2, Canada
| |
Collapse
|
22
|
Häring C, Schroeder J, Jungwirth J, Löffler B, Henke A, Engert B, Ehrhardt C. ProcCluster ® and procaine hydrochloride inhibit the replication of influenza A virus in vitro. Front Microbiol 2024; 15:1422651. [PMID: 39206370 PMCID: PMC11350405 DOI: 10.3389/fmicb.2024.1422651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 07/24/2024] [Indexed: 09/04/2024] Open
Abstract
Introduction Treatment of influenza A virus infections is currently limited to few direct acting antiviral substances. Repurposing other established pharmaceuticals as antivirals could aid in improving treatment options. Methods This study investigates the antiviral properties of ProcCluster® and procaine hydrochloride, two derivatives of the local anesthetic procaine, in influenza A virus infection of A549, Calu-3 and MDCK cells. Results Both substances inhibit replication in all three of these cell lines in multi-cycle experiments. However, cell line-dependent differences in the effects of the substances on viral RNA replication and subsequent protein synthesis, as well as release of progeny viruses in single-cycle experiments can be observed. Both ProcCluster® and procaine hydrochloride delay endosome fusion of the virus early in the replication cycle, possibly due to the alkaline nature of the active component procaine. In A549 and Calu-3 cells an additional effect of the substances can be observed at late stages in the first replication cycle. Interestingly, this effect is absent in MDCK cells. We demonstrate that ProcCluster® and procaine hydrochloride inhibit phospholipase A2 (PLA2) enzymes from A549 but not MDCK cells and confirm that specific inhibition of calcium independent PLA2 but not cytosolic PLA2 has antiviral effects. Discussion We show that ProcCluster® and procaine hydrochloride inhibit influenza A virus infection at several stages of the replication cycle and have potential as antiviral substances.
Collapse
Affiliation(s)
- Clio Häring
- Section of Experimental Virology, Institute of Medical Microbiology, Center for Molecular Biomedicine (CMB), Jena University Hospital, Jena, Germany
| | - Josefine Schroeder
- Section of Experimental Virology, Institute of Medical Microbiology, Center for Molecular Biomedicine (CMB), Jena University Hospital, Jena, Germany
| | - Johannes Jungwirth
- Section of Experimental Virology, Institute of Medical Microbiology, Center for Molecular Biomedicine (CMB), Jena University Hospital, Jena, Germany
| | - Bettina Löffler
- Institute of Medical Microbiology, Jena University Hospital, Jena, Germany
| | - Andreas Henke
- Section of Experimental Virology, Institute of Medical Microbiology, Center for Molecular Biomedicine (CMB), Jena University Hospital, Jena, Germany
| | | | - Christina Ehrhardt
- Section of Experimental Virology, Institute of Medical Microbiology, Center for Molecular Biomedicine (CMB), Jena University Hospital, Jena, Germany
| |
Collapse
|
23
|
Kim J, Yuan Y, Agaronyan K, Zhao A, D Wang V, Gupta G, Essayas H, Kaminski A, McGovern J, Yu S, Woo S, Lee CJ, Gandhi S, Saber T, Saleh T, Hu B, Sun Y, Ishikawa G, Bain W, Evankovich J, Chen L, Yun H, Herzog EL, Dela Cruz CS, Ryu C, Sharma L. Damage sensing through TLR9 Regulates Inflammatory and Antiviral Responses During Influenza Infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.04.583378. [PMID: 38496452 PMCID: PMC10942338 DOI: 10.1101/2024.03.04.583378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Host response aimed at eliminating the infecting pathogen, as well as the pathogen itself, can cause tissue injury. Tissue injury leads to the release of a myriad of cellular components including mitochondrial DNA, which the host senses through pattern recognition receptors. How the sensing of tissue injury by the host shapes the anti-pathogen response remains poorly understood. In this study, we utilized mice that are deficient in toll-like receptor-9 (TLR9), which binds to unmethylated CpG DNA sequences such as those present in bacterial and mitochondrial DNA. To avoid direct pathogen sensing by TLR9, we utilized the influenza virus, which lacks ligands for TLR9, to determine how damage sensing by TLR9 contributes to anti-influenza immunity. Our data show that TLR9-mediated sensing of tissue damage promotes an inflammatory response during early infection, driven by the epithelial and myeloid cells. Along with the diminished inflammatory response, the absence of TLR9 led to impaired viral clearance manifested as a higher and prolonged influenza components in myeloid cells including monocytes and macrophages rendering them highly inflammatory. The persistent inflammation driven by infected myeloid cells led to persistent lung injury and impaired recovery in influenza-infected TLR9-/- mice. Further, we show elevated TLR9 activation in the plasma samples of patients with influenza and its association with the disease severity in hospitalized patients, demonstrating its clinical relevance. Overall, we demonstrate an essential role of damage sensing through TLR9 in promoting anti-influenza immunity and inflammatory response.
Collapse
Affiliation(s)
- Jooyoung Kim
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT
| | - Yifan Yuan
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT
- University of Maryland, MD
| | - Karen Agaronyan
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT
- Howard Hughes Medical Institute
| | - Amy Zhao
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT
| | - Victoria D Wang
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Gayatri Gupta
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT
| | - Heran Essayas
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT
| | - Ayelet Kaminski
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT
| | - John McGovern
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT
| | - Sheeline Yu
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT
| | - Samuel Woo
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT
| | - Chris J. Lee
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT
| | - Shifa Gandhi
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT
| | - Tina Saber
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT
| | - Tayebeh Saleh
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Buqu Hu
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT
| | - Ying Sun
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT
| | - Genta Ishikawa
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT
| | - William Bain
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA
- VA Medical Center, Pittsburgh, PA
| | - John Evankovich
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Lujia Chen
- Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, PA 15206, USA
| | - HongDuck Yun
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Erica L. Herzog
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT
| | - Charles S. Dela Cruz
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT
- VA Medical Center, Pittsburgh, PA
| | - Changwan Ryu
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT
| | - Lokesh Sharma
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT
| |
Collapse
|
24
|
Wang L, Guo M, Wang Y, Chen R, Wei X. The relationship between influenza vaccine hesitancy and vaccine literacy among youth and adults in China. Front Immunol 2024; 15:1444393. [PMID: 39161763 PMCID: PMC11330759 DOI: 10.3389/fimmu.2024.1444393] [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: 06/05/2024] [Accepted: 07/17/2024] [Indexed: 08/21/2024] Open
Abstract
Objectives The present study aimed to assess influenza vaccine hesitancy and vaccine literacy levels among youth and adults in China, as well as the association between them. Methods An online cross-sectional survey was conducted in Mainland China. Participants' total vaccine literacy and three sub-dimension vaccine literacy (knowledge literacy, competence literacy, and decision-making literacy) were assessed by a validated vaccine literacy scale. Having received influenza vaccination in the past three years or intending to accept it in next influenza season indicates less influenza vaccine hesitancy. Results Among 997 participants, a sub-optimal vaccine literacy was observed, with a mean score of 66.83 ± 10.27. Regression models 1-4 revealed that participants with middle (aOR: 1.431, P=0.039, 95% CI: 1.018~2.010) or high (aOR: 1.651, P=0.006, 95% CI: 1.157~2.354) total vaccine literacy, as well as those with high competence literacy (aOR: 1.533, P=0.017, 95% CI: 1.079~2.180), or high decision-making literacy (aOR: 1.822, P=0.001, 95% CI: 1.261~2.632) were more likely to have been vaccinated against influenza at least once in past three years. However, those with a high knowledge literacy were associated with a lower influenza vaccine rate (aOR: 0.676, P=0.046, 95% CI: 0.460~0.994). Regression models 5-8 revealed that participants with middle (aOR: 1.661, P=0.008, 95% CI: 1.142~2.414) or high total vaccine literacy (aOR: 2.645, P=0.000, 95% CI: 1.774~3.942), as well as those with middle (aOR: 1.703, P=0.005, 95% CI: 1.177~2.464) or high competence literacy (aOR: 2.346, P=0.000, 95% CI: 1.159~3.461), or high decision-making literacy (aOR: 2.294, P=0.000, 95% CI: 1.531~3.436) were more likely to express the willingness to receive the influenza vaccine in the next influenza season. Conclusion The participants' influenza vaccine hesitancy was negatively associated with their total vaccine literacy levels and two of the three sub-dimensions: competence literacy and decision-making literacy. Knowledge literacy suggested a positive or no relationship with influenza vaccine hesitancy.
Collapse
Affiliation(s)
- Li Wang
- School of Health Service Management, Anhui Medical University, Hefei, Anhui, China
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| | - Mengjie Guo
- School of Health Service Management, Anhui Medical University, Hefei, Anhui, China
| | - Yan Wang
- School of Health Service Management, Anhui Medical University, Hefei, Anhui, China
| | - Ren Chen
- School of Health Service Management, Anhui Medical University, Hefei, Anhui, China
| | - Xiaolin Wei
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
- Institute of Health Policy, Management, and Evaluation, Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
25
|
Zhou Y, Lin Z, Fang J, Wang Z, Guo J, Li G, Xu Q, Jin M, Chen H, Zou J, Zhou H. The recombinant vaccine of Lactobacillus plantarum elicits immune protection against H1N1 and H9N2 influenza virus infection. Int J Biol Macromol 2024; 275:133453. [PMID: 38942402 DOI: 10.1016/j.ijbiomac.2024.133453] [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: 05/06/2024] [Revised: 06/24/2024] [Accepted: 06/24/2024] [Indexed: 06/30/2024]
Abstract
Influenza A virus (IAV) causes annual epidemics and occasional pandemics, resulting in significant economic losses and numerous fatalities. Current vaccines, typically administered through injection, provide limited protection due to the frequent antigenic shift and drift of IAV strains. Therefore, the development of alternative broad-spectrum vaccine strategies is imperative. Lactic acid bacteria (LAB) represent promising candidates for vaccine engineering due to their low cost, high safety profile, and suitability for oral administration. In this study, we identified a strain of Lactobacillus plantarum (Lp) that is resistant to acid and bile salts and capable of colonizing the intestines of mice. Subsequently, we employed the RecE/T gene editing system to integrate headless hemagglutinins (mini-HA) into the genome of Lp, generating Lp-mini-HA-SP. Remarkably, immunization with Lp-mini-HA-SP elicited serum IgG antibody responses and conferred immune protection against H9N2 and H1N1 influenza virus challenges. Collectively, our findings offer a novel approach for the development of orally administered IAV vaccines and hold significant potential for future drug development endeavors.
Collapse
Affiliation(s)
- Yuanbao Zhou
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, PR China
| | - Zhipeng Lin
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, PR China
| | - Jiaqing Fang
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, PR China
| | - Zhihao Wang
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, PR China
| | - Jinli Guo
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, PR China
| | - Guohong Li
- Wuhan Keqian Biology Co., Ltd, Wuhan, Hubei, PR China
| | - Qiaoxia Xu
- Wuhan Keqian Biology Co., Ltd, Wuhan, Hubei, PR China
| | - Meilin Jin
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, PR China; Hubei Hongshan Laboratory, Wuhan, Hubei, PR China
| | - Huanchun Chen
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, PR China; Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan, Hubei, PR China; Hubei Hongshan Laboratory, Wuhan, Hubei, PR China
| | - Jiahui Zou
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, PR China.
| | - Hongbo Zhou
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, PR China; Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan, Hubei, PR China; Hubei Hongshan Laboratory, Wuhan, Hubei, PR China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, PR China.
| |
Collapse
|
26
|
Zhang X, Shi H, Hendy DA, Bachelder EM, Ainslie KM, Ross TM. Multi-COBRA hemagglutinin formulated with cGAMP microparticles elicits protective immune responses against influenza viruses. mSphere 2024; 9:e0016024. [PMID: 38920382 PMCID: PMC11288037 DOI: 10.1128/msphere.00160-24] [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: 02/26/2024] [Accepted: 05/21/2024] [Indexed: 06/27/2024] Open
Abstract
In humans, seasonal influenza viruses cause epidemics. Avian influenza viruses are of particular concern because they can infect multiple species and lead to unpredictable and severe disease. Therefore, there is an urgent need for a universal influenza vaccine that provides protection against all influenza strains. The cyclic GMP-AMP (cGAMP) is a promising adjuvant for subunit vaccines, which promotes type I interferons' production through the stimulator of interferon genes (STING) pathway. The encapsulation of cGAMP in acetalated dextran (Ace-DEX) microparticles (MPs) enhances its intracellular delivery. In this study, the Computationally Optimized Broadly Reactive Antigen (COBRA) methodology was used to generate H1, H3, and H5 vaccine candidates. Monovalent and multivalent COBRA HA vaccines formulated with cGAMP Ace-DEX MPs were evaluated in mice for protective antibody responses. cGAMP MPs adjuvanted COBRA HA vaccines elicited robust antigen-specific antibodies following vaccination. Compared with COBRA HA vaccine groups with no adjuvant or blank MPs, the cGAMP MPs enhanced HAI activity elicited by COBRA HA vaccines. The HAI activity was not significantly different between cGAMP MPs adjuvanted monovalent or multivalent COBRA HA vaccines. The cGAMP MPs adjuvanted COBRA vaccine groups had higher antigen-specific IgG2a-binding titers than the COBRA vaccine groups with no adjuvant or blank MPs. The COBRA vaccines formulated with cGAMP MPs mitigated diseases caused by influenza viral challenge and decreased pulmonary viral titers in mice. Therefore, the formulation of COBRA vaccines plus cGAMP MPs is a promising universal influenza vaccine that elicits protective immune responses against human seasonal and pre-pandemic strains. IMPORTANCE Influenza viruses cause severe respiratory disease, particularly in the very young and the elderly. Next-generation influenza vaccines are needed to protect against new influenza variants. This report used a promising adjuvant, cyclic GMP-AMP (cGAMP), to enhance the elicited antibodies by an improved influenza hemagglutinin candidate and protect against influenza virus infection. Overall, adding adjuvants to influenza vaccines is an effective method to improve vaccines.
Collapse
Affiliation(s)
- Xiaojian Zhang
- Center for Vaccines and Immunology, University of Georgia, Athens, Georgia, USA
| | - Hua Shi
- Center for Vaccines and Immunology, University of Georgia, Athens, Georgia, USA
| | - Dylan A. Hendy
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Eric M. Bachelder
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Kristy M. Ainslie
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, North Carolina, USA
- Department of Microbiology and Immunology, UNC School of Medicine, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Ted M. Ross
- Center for Vaccines and Immunology, University of Georgia, Athens, Georgia, USA
- Department of Infectious Diseases, University of Georgia, Athens, Georgia, USA
- Florida Research and Innovation Center, Cleveland Clinic, Port Saint Lucie, Florida, USA
- Department of Infection Biology, Lehner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
27
|
Sey EA, Warris A. The gut-lung axis: the impact of the gut mycobiome on pulmonary diseases and infections. OXFORD OPEN IMMUNOLOGY 2024; 5:iqae008. [PMID: 39193472 PMCID: PMC11316619 DOI: 10.1093/oxfimm/iqae008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 06/12/2024] [Accepted: 06/13/2024] [Indexed: 08/29/2024] Open
Abstract
The gastrointestinal tract contains a diverse microbiome consisting of bacteria, fungi, viruses and archaea. Although these microbes usually reside as commensal organisms, it is now well established that higher abundance of specific bacterial or fungal species, or loss of diversity in the microbiome can significantly affect development, progression and outcomes in disease. Studies have mainly focused on the effects of bacteria, however, the impact of other microbes, such as fungi, has received increased attention in the last few years. Fungi only represent around 0.1% of the total gut microbial population. However, key fungal taxa such as Candida, Aspergillus and Wallemia have been shown to significantly impact health and disease. The composition of the gut mycobiome has been shown to affect immunity at distal sites, such as the heart, lung, brain, pancreas, and liver. In the case of the lung this phenomenon is referred to as the 'gut-lung axis'. Recent studies have begun to explore and unveil the relationship between gut fungi and lung immunity in diseases such as asthma and lung cancer, and lung infections caused by viruses, bacteria and fungi. In this review we will summarize the current, rapidly growing, literature describing the impact of the gut mycobiome on respiratory disease and infection.
Collapse
Affiliation(s)
- Emily A Sey
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, EX4 4QD, UK
| | - Adilia Warris
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, EX4 4QD, UK
| |
Collapse
|
28
|
Kannan N, Choi A, Rivera De Jesus MA, Wei PM, Sahler JM, Curley SM, August A, DeLisa MP, Whittaker GR, Putnam D. Intranasal Vaccination with Recombinant TLR2-Active Outer Membrane Vesicles Containing Sequential M2e Epitopes Protects against Lethal Influenza a Challenge. Vaccines (Basel) 2024; 12:724. [PMID: 39066362 PMCID: PMC11281606 DOI: 10.3390/vaccines12070724] [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: 05/23/2024] [Revised: 06/14/2024] [Accepted: 06/20/2024] [Indexed: 07/28/2024] Open
Abstract
Influenza is a highly contagious respiratory disease, resulting in an estimated 3 to 5 million cases of severe illness annually. While most influenza vaccines are administered parenterally via injection, one shortcoming is that they do not generate a strong immune response at the site of infection, which can become important in a pandemic. Intranasal vaccines can generate both local and systemic protective immune responses, can reduce costs, and enhance ease of administration. Previous studies showed that parenterally administered outer membrane vesicles (OMVs) that carry sequences of the M2e protein (OMV-M2e) protect against influenza A/PR8 challenge in mice and ferrets. In the current study, we measured the effectiveness of the intranasal route of the OMV-M2e vaccine against the influenza A/PR8 strain in mice. We observed high anti-M2e IgG and IgA titers post-challenge in mice vaccinated intranasally with OMV-M2e. In addition, we observed a Th1/Tc1 bias in the vaccinated mice, and an increased Th17/Tc17 response, both of which correlated with survival to A/PR8 challenge and significantly lower lung viral titers. We conclude that the intranasal-route administration of the OMV-M2e vaccine is a promising approach toward generating protection against influenza A as it leads to an increased proinflammatory immune response correlating with survival to viral challenge.
Collapse
Affiliation(s)
- Nisha Kannan
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA; (N.K.); (M.A.R.D.J.); (P.M.W.); (S.M.C.)
| | - Annette Choi
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853, USA; (A.C.); (J.M.S.); (A.A.); (G.R.W.)
| | - Mariela A. Rivera De Jesus
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA; (N.K.); (M.A.R.D.J.); (P.M.W.); (S.M.C.)
| | - Peter Male Wei
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA; (N.K.); (M.A.R.D.J.); (P.M.W.); (S.M.C.)
| | - Julie Marie Sahler
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853, USA; (A.C.); (J.M.S.); (A.A.); (G.R.W.)
| | - Stephanie Marie Curley
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA; (N.K.); (M.A.R.D.J.); (P.M.W.); (S.M.C.)
| | - Avery August
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853, USA; (A.C.); (J.M.S.); (A.A.); (G.R.W.)
| | - Matthew P. DeLisa
- Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY 14853, USA;
| | - Gary R. Whittaker
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853, USA; (A.C.); (J.M.S.); (A.A.); (G.R.W.)
| | - David Putnam
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA; (N.K.); (M.A.R.D.J.); (P.M.W.); (S.M.C.)
- Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY 14853, USA;
| |
Collapse
|
29
|
Sanchez PL, Staats HF, Abraham SN, Ross TM. Mastoparan-7 adjuvanted COBRA H1 and H3 hemagglutinin influenza vaccines. Sci Rep 2024; 14:13800. [PMID: 38877101 PMCID: PMC11178843 DOI: 10.1038/s41598-024-64351-7] [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: 01/18/2024] [Accepted: 06/07/2024] [Indexed: 06/16/2024] Open
Abstract
Adjuvants enhance, prolong, and modulate immune responses by vaccine antigens to maximize protective immunity and enable more effective immunization in the young and elderly. Most adjuvants are formulated with injectable vaccines. However, an intranasal route of vaccination may induce mucosal and systemic immune responses for enhancing protective immunity in individuals and be easier to administer compared to injectable vaccines. In this study, a next generation of broadly-reactive influenza hemagglutinin (HA) vaccines were developed using the Computationally Optimized Broadly Reactive Antigen (COBRA) methodology. These HA vaccines were formulated with Mastoparan 7 (M7-NH2) mast cell degranulating peptide adjuvant and administered intranasally to determine vaccine-induced seroconversion of antibodies against a panel of influenza viruses and protection following infection with H1N1 and H3N2 viruses in mice. Mice vaccinated intranasally with M7-NH2-adjuvanted COBRA HA vaccines had high HAIs against a panel of H1N1 and H3N2 influenza viruses and were protected against both morbidity and mortality, with reduced viral lung titers, following challenge with an H1N1 influenza virus. Additionally, M7-NH2 adjuvanted COBRA HA vaccines induced Th2 skewed immune responses with robust IgG and isotype antibodies in the serum and mucosal lung lavages. Overall, this intranasally delivered M7-NH2 -adjuvanted COBRA HA vaccine provides effective protection against drifted H1N1 and H3N2 viruses.
Collapse
Affiliation(s)
- Pedro L Sanchez
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, USA
- Department of Infectious Diseases, University of Georgia, Athens, GA, USA
- Florida Research and Innovation Center, Cleveland Clinic, Port Saint Lucie, FL, USA
| | - Herman F Staats
- Pathology Department, School of Medicine, Duke University Medical Center, Durham, NC, USA
- Department of Immunology, School of Medicine, Duke University Medical Center, Durham, NC, USA
- Duke Human Vaccine Institute, Duke University, Duke University Medical Center, Durham, NC, USA
| | - Soman N Abraham
- Pathology Department, School of Medicine, Duke University Medical Center, Durham, NC, USA
- Department of Immunology, School of Medicine, Duke University Medical Center, Durham, NC, USA
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC, USA
- Program in Emerging Infectious Diseases, Duke-National University of Singapore, Singapore, Singapore
| | - Ted M Ross
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, USA.
- Department of Infectious Diseases, University of Georgia, Athens, GA, USA.
- Florida Research and Innovation Center, Cleveland Clinic, Port Saint Lucie, FL, USA.
- Department of Infection Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.
| |
Collapse
|
30
|
Cipelli R, Falato S, Lusito E, Maifredi G, Montedoro M, Valpondi P, Zucchi A, Azzi MV, Zanetta L, Gualano MR, Xoxi E, Marchisio PG, Castaldi S. The Hospital Burden of Flu in Italy: a retrospective study on administrative data from season 2014-2015 to 2018-2019. BMC Infect Dis 2024; 24:572. [PMID: 38851739 PMCID: PMC11162570 DOI: 10.1186/s12879-024-09446-2] [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: 02/22/2024] [Accepted: 05/29/2024] [Indexed: 06/10/2024] Open
Abstract
BACKGROUND Every year in Italy, influenza affects about 4 million people. Almost 5% of them are hospitalised. During peak illness, enormous pressure is placed on healthcare and economic systems. This study aims to quantify the clinical and economic burden of severe influenza during 5 epidemic seasons (2014-2019) from administrative claims data. METHODS Patients hospitalized with a diagnosis of influenza between October 2014, and April 2019, were analyzed. Clinical characteristics and administrative information were retrieved from health-related Administrative Databases (ADs) of 4 Italian Local Health Units (LHUs). The date of first admission was set as the Index Date (ID). A follow-up period of six months after ID was considered to account for complications and re-hospitalizations, while a lookback period (2 years before ID) was set to assess the prevalence of underlying comorbidities. RESULTS Out of 2,333 patients with severe influenza, 44.1% were adults ≥ 65, and 25.6% young individuals aged 0-17. 46.8% had comorbidities (i.e., were at risk), mainly cardiovascular and metabolic diseases (45.3%), and chronic conditions (24.7%). The highest hospitalization rates were among the elderly (≥ 75) and the young individuals (0-17), and were 37.6 and 19.5/100,000 inhabitants/year, respectively. The average hospital stay was 8 days (IQR: 14 - 4). It was higher for older individuals (≥ 65 years, 11 days, [17 - 6]) and for those with comorbidities (9 days, [16 - 6]), p-value < 0.001. Similarly, mortality was higher in elderly and those at risk (p-value < 0.001). Respiratory complications occurred in 12.7% of patients, and cardiovascular disorders in 5.9%. Total influenza-related costs were €9.7 million with hospitalization accounting for 95% of them. 47.3% of hospitalization costs were associated with individuals ≥ 65 and 52.9% with patients at risk. The average hospitalisation cost per patient was € 4,007. CONCLUSIONS This retrospective study showed that during the 2014-2019 influenza seasons in Italy, individuals of extreme ages and those with pre-existing medical conditions, were more likely to be hospitalized with severe influenza. Together with complications and ageing, they worsen patient's outcome and may lead to a prolonged hospitalization, thus increasing healthcare utilization and costs. Our data generate real-world evidence on the burden of influenza, useful to inform public health decision-making.
Collapse
Affiliation(s)
- Riccardo Cipelli
- IQVIA Solutions Italy Srl, Via Fabio Filzi 29, Milan, 20124, Italy.
| | - Serena Falato
- IQVIA Solutions Italy Srl, Via Fabio Filzi 29, Milan, 20124, Italy
| | - Eleonora Lusito
- IQVIA Solutions Italy Srl, Via Fabio Filzi 29, Milan, 20124, Italy
| | - Giovanni Maifredi
- SS Epidemiologia, Agenzia di Tutela della Salute di Brescia, Brescia, Italy
| | | | | | - Alberto Zucchi
- UOC Servizio Epidemiologia presso ATS di Bergamo, Bergamo, Italy
| | | | | | | | - Entela Xoxi
- Università Cattolica del Sacro Cuore, Alta Scuola di Economia e Management dei Sistemi Sanitari (ALTEMS), Rome, Italy
| | - Paola Giovanna Marchisio
- Dipartimento di Fisiopatologia Medico-Chirurgica e dei Trapianti, University of Milan, Milan, Italy
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Silvana Castaldi
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
31
|
Jayaraman AS, Darekar I, Dadhich NV, Tadepalli LSM, Gongwang Y, Singh S, Gavor E. Effect of the COVID-19 Pandemic on Respiratory Diseases and Their Economic Impacts. Pathogens 2024; 13:491. [PMID: 38921789 PMCID: PMC11206581 DOI: 10.3390/pathogens13060491] [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: 04/24/2024] [Revised: 05/23/2024] [Accepted: 05/29/2024] [Indexed: 06/27/2024] Open
Abstract
COVID-19 is an airborne respiratory disease that mainly affects the lungs. To date, COVID-19 has infected 580 million people with a mortality of approximately 7 million people worldwide. The emergence of COVID-19 has also affected the infectivity, diagnosis, and disease outcomes of existing diseases such as influenza, TB, and asthma in human populations. These are airborne respiratory diseases with symptoms and mode of transmission similar to those of COVID-19. It was speculated that the protracted nature of the COVID-19 pandemic coupled with vaccination could impact other respiratory diseases and mortality. In this study, we analyzed the impact of COVID-19 on flu, tuberculosis (TB), and asthma. Our analyses suggest that COVID-19 has a potential impact on the mortality of flu, TB, and asthma. These impacts vary across before the COVID-19 era, during the peak period of the pandemic, and after vaccinations/preventive measures were implemented, as well as across different regions of the world. Overall, the spread of flu generally reduced during the pandemic, resulting in a reduced expenditure on flu-related hospitalizations, although there were sporadic spikes at setting times. In contrast, TB deaths generally increased perhaps due to the disruption in access to TB services and reduction in resources. Asthma deaths, on the other hand, only marginally varied. Collectively, the emergence of COVID-19 added extra cost to the overall expenditure on some respiratory infectious diseases, while the cost for other infectious diseases was either reduced or somewhat unaffected.
Collapse
Affiliation(s)
- Ananya Sivaraman Jayaraman
- Global Indian International School, 27 Punggol Field Walk, Singapore 828649, Singapore; (A.S.J.); (I.D.); (N.V.D.); (L.S.M.T.)
| | - Ishita Darekar
- Global Indian International School, 27 Punggol Field Walk, Singapore 828649, Singapore; (A.S.J.); (I.D.); (N.V.D.); (L.S.M.T.)
| | - Nidhi Vijayprakash Dadhich
- Global Indian International School, 27 Punggol Field Walk, Singapore 828649, Singapore; (A.S.J.); (I.D.); (N.V.D.); (L.S.M.T.)
| | - Lakshmi Sai Manasvi Tadepalli
- Global Indian International School, 27 Punggol Field Walk, Singapore 828649, Singapore; (A.S.J.); (I.D.); (N.V.D.); (L.S.M.T.)
| | - Yao Gongwang
- Department of Biological Sciences, National University of Singapore, Singapore 117543, Singapore; (Y.G.); (S.S.)
| | - Sunil Singh
- Department of Biological Sciences, National University of Singapore, Singapore 117543, Singapore; (Y.G.); (S.S.)
| | - Edem Gavor
- Department of Biological Sciences, National University of Singapore, Singapore 117543, Singapore; (Y.G.); (S.S.)
| |
Collapse
|
32
|
Gambadauro A, Galletta F, Li Pomi A, Manti S, Piedimonte G. Immune Response to Respiratory Viral Infections. Int J Mol Sci 2024; 25:6178. [PMID: 38892370 PMCID: PMC11172738 DOI: 10.3390/ijms25116178] [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: 05/07/2024] [Revised: 05/29/2024] [Accepted: 05/31/2024] [Indexed: 06/21/2024] Open
Abstract
The respiratory system is constantly exposed to viral infections that are responsible for mild to severe diseases. In this narrative review, we focalized the attention on respiratory syncytial virus (RSV), influenza virus, and severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2) infections, responsible for high morbidity and mortality in the last decades. We reviewed the human innate and adaptive immune responses in the airways following infection, focusing on a particular population: newborns and pregnant women. The recent Coronavirus disease-2019 (COVID-19) pandemic has highlighted how our interest in viral pathologies must not decrease. Furthermore, we must increase our knowledge of infection mechanisms to improve our future defense strategies.
Collapse
Affiliation(s)
- Antonella Gambadauro
- Pediatric Unit, Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, University of Messina, Via Consolare Valeria 1, 98124 Messina, Italy; (A.G.); (F.G.); (A.L.P.)
| | - Francesca Galletta
- Pediatric Unit, Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, University of Messina, Via Consolare Valeria 1, 98124 Messina, Italy; (A.G.); (F.G.); (A.L.P.)
| | - Alessandra Li Pomi
- Pediatric Unit, Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, University of Messina, Via Consolare Valeria 1, 98124 Messina, Italy; (A.G.); (F.G.); (A.L.P.)
| | - Sara Manti
- Pediatric Unit, Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, University of Messina, Via Consolare Valeria 1, 98124 Messina, Italy; (A.G.); (F.G.); (A.L.P.)
| | - Giovanni Piedimonte
- Office for Research and Departments of Pediatrics, Biochemistry, and Molecular Biology, Tulane University, New Orleans, LA 70112, USA;
| |
Collapse
|
33
|
Martin MA, Berg N, Koelle K. Influenza A genomic diversity during human infections underscores the strength of genetic drift and the existence of tight transmission bottlenecks. Virus Evol 2024; 10:veae042. [PMID: 38883977 PMCID: PMC11179161 DOI: 10.1093/ve/veae042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 05/06/2024] [Accepted: 05/21/2024] [Indexed: 06/18/2024] Open
Abstract
Influenza infections result in considerable public health and economic impacts each year. One of the contributing factors to the high annual incidence of human influenza is the virus's ability to evade acquired immunity through continual antigenic evolution. Understanding the evolutionary forces that act within and between hosts is therefore critical to interpreting past trends in influenza virus evolution and in predicting future ones. Several studies have analyzed longitudinal patterns of influenza A virus genetic diversity in natural human infections to assess the relative contributions of selection and genetic drift on within-host evolution. However, in these natural infections, within-host viral populations harbor very few single-nucleotide variants, limiting our resolution in understanding the forces acting on these populations in vivo. Furthermore, low levels of within-host viral genetic diversity limit the ability to infer the extent of drift across transmission events. Here, we propose to use influenza virus genomic diversity as an alternative signal to better understand within- and between-host patterns of viral evolution. Specifically, we focus on the dynamics of defective viral genomes (DVGs), which harbor large internal deletions in one or more of influenza virus's eight gene segments. Our longitudinal analyses of DVGs show that influenza A virus populations are highly dynamic within hosts, corroborating previous findings based on viral genetic diversity that point toward the importance of genetic drift in driving within-host viral evolution. Furthermore, our analysis of DVG populations across transmission pairs indicates that DVGs rarely appeared to be shared, indicating the presence of tight transmission bottlenecks. Our analyses demonstrate that viral genomic diversity can be used to complement analyses based on viral genetic diversity to reveal processes that drive viral evolution within and between hosts.
Collapse
Affiliation(s)
- Michael A Martin
- Department of Pathology, Johns Hopkins School of Medicine, 600 N. Wolfe Street, Baltimore, MD 21287, USA
- Graduate Program in Population Biology, Ecology, and Evolution, Emory University, 1462 Clifton Road NE, Atlanta, GA 30322, USA
- Department of Biology, Emory University, 1510 Clifton Road NE, Atlanta, GA 30322, USA
| | - Nick Berg
- Department of Biology, Emory University, 1510 Clifton Road NE, Atlanta, GA 30322, USA
- Department of Biochemistry, Brandeis University, 415 South Street, Waltham, MA 02453, USA
- National Institute of Allergy and Infectious Diseases Laboratory of Viral Disease, National Institutes of Health, 33 North Drive, Bethesda, MD 20814, USA
| | - Katia Koelle
- Department of Biology, Emory University, 1510 Clifton Road NE, Atlanta, GA 30322, USA
- Emory Center of Excellence for Influenza Research and Response (Emory-CEIRR), 1510 Clifton Road NE, Atlanta, GA 30322, USA
| |
Collapse
|
34
|
Liu Z, Kabir MT, Chen S, Zhang H, Wakim LM, Rehm BHA. Intranasal Epitope-Polymer Vaccine Lodges Resident Memory T Cells Protecting Against Influenza Virus. Adv Healthc Mater 2024; 13:e2304188. [PMID: 38411375 PMCID: PMC11469178 DOI: 10.1002/adhm.202304188] [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: 11/27/2023] [Revised: 02/22/2024] [Indexed: 02/28/2024]
Abstract
Intranasal vaccines, unlike injectable vaccines, boost immunity along the respiratory tract; this can significantly limit respiratory virus replication and shedding. There remains a need to develop mucosal adjuvants and vaccine delivery systems that are both safe and effective following intranasal administration. Here, biopolymer particles (BP) densely coated with repeats of MHC class I restricted immunodominant epitopes derived from influenza A virus namely NP366, a nucleoprotein-derived epitope and PA224, a polymerase acidic subunit derived epitope, are bioengineered. These BP-NP366/PA224 can be manufactured at a high yield and are obtained at ≈93% purity, exhibiting ambient-temperature stability. Immunological characterization includes comparing systemic and mucosal immune responses mounted following intramuscular or intranasal immunization. Immunization with BP-NP366/PA224 without adjuvant triggers influenza-specific CD8+ T cell priming and memory CD8+ T cell development. Co-delivery with the adjuvant poly(I:C) significantly boosts the size and functionality of the influenza-specific pulmonary resident memory CD8+ T cell pool. Intranasal, but not intramuscular delivery of BP-NP366/PA224 with poly(I:C), provides protection against influenza virus challenge. Overall, the BP approach demonstrates as a suitable antigen formulation for intranasal delivery toward induction of systemic protective T cell responses against influenza virus.
Collapse
Affiliation(s)
- Ziyang Liu
- Department of Microbiology and ImmunologyThe University of MelbourneThe Peter Doherty Institute for Infection and ImmunityMelbourneVictoria3000Australia
| | - Md. Tanvir Kabir
- Centre for Cell Factories and BiopolymersGriffith Institute for Drug DiscoveryGriffith UniversityDon Young RoadNathanQueensland4111Australia
| | - Shuxiong Chen
- Centre for Cell Factories and BiopolymersGriffith Institute for Drug DiscoveryGriffith UniversityDon Young RoadNathanQueensland4111Australia
| | - Heran Zhang
- Department of Microbiology and ImmunologyThe University of MelbourneThe Peter Doherty Institute for Infection and ImmunityMelbourneVictoria3000Australia
| | - Linda M. Wakim
- Department of Microbiology and ImmunologyThe University of MelbourneThe Peter Doherty Institute for Infection and ImmunityMelbourneVictoria3000Australia
| | - Bernd H. A. Rehm
- Centre for Cell Factories and BiopolymersGriffith Institute for Drug DiscoveryGriffith UniversityDon Young RoadNathanQueensland4111Australia
| |
Collapse
|
35
|
Ma Y, Li W, Li J, Qian J, Jiang M, Sun Y, Ma Y, Yang W, Feng L. Association between influenza vaccination and one-year all-cause and cardiovascular mortality risk: A self-controlled case series and matched case-control study. J Med Virol 2024; 96:e29722. [PMID: 38837255 DOI: 10.1002/jmv.29722] [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: 02/16/2024] [Revised: 04/29/2024] [Accepted: 05/23/2024] [Indexed: 06/07/2024]
Abstract
Debates surrounding the efficacy of influenza vaccination for survival benefits persist, and there is a lack of data regarding its duration of protection. A self-controlled case series (SCCS) and a 1:4 matched case-control study were conducted using the National Health Interview Survey (NHIS) and public-use mortality data from 2005 to 2018 in the United States. The SCCS study identified participants who received influenza vaccination within 12 months before the survey and subsequently died within 1 year of postvaccination. The matched case-control study paired participants who died during the influenza season at the time of survey with four survivors. Among 1167 participants in the SCCS study, there was a 46% reduction in all-cause mortality and a 43% reduction in cardiovascular mortality within 29-196 days of postvaccination. The greatest protection was observed during days 29-56 (all-cause mortality: RI: 0.19; 95% CI: 0.12-0.29; cardiovascular mortality: RI: 0.28; 95% CI: 0.14-0.56). Among 626 cases and 2504 controls included in the matched case-control study, influenza vaccination was associated with a reduction in all-cause mortality (OR: 0.74, 95% CI: 0.60-0.92) and cardiovascular mortality (OR: 0.64, 95% CI: 0.44-0.93) during the influenza season. This study highlights the importance of influenza vaccination in reducing the risks of all-cause and cardiovascular mortality, with effects lasting for approximately 6 months.
Collapse
Affiliation(s)
- Yuan Ma
- School of Population Medicine and Public Health, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Beijing Center for Disease Prevention and Control, Beijing Research Center for Preventive Medicine, Beijing, China
| | - Wei Li
- Center for Applied Statistics and School of Statistics, Renmin University of China, Beijing, China
| | - Juan Li
- Beijing Center for Disease Prevention and Control, Beijing Research Center for Preventive Medicine, Beijing, China
| | - Jie Qian
- School of Population Medicine and Public Health, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Beijing Center for Disease Prevention and Control, Beijing Research Center for Preventive Medicine, Beijing, China
| | - Mingyue Jiang
- School of Population Medicine and Public Health, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Beijing Center for Disease Prevention and Control, Beijing Research Center for Preventive Medicine, Beijing, China
| | - Yanxia Sun
- School of Population Medicine and Public Health, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Beijing Center for Disease Prevention and Control, Beijing Research Center for Preventive Medicine, Beijing, China
| | - Yue Ma
- School of Public Health, Southeast University, Nanjing, China
| | - Weizhong Yang
- School of Population Medicine and Public Health, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Beijing Center for Disease Prevention and Control, Beijing Research Center for Preventive Medicine, Beijing, China
| | - Luzhao Feng
- School of Population Medicine and Public Health, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- State Key Laboratory of Respiratory Health and Multimorbidity, Beijing, China
- Key Laboratory of Pathogen Infection Prevention and Control (Peking Union Medical College), Ministry of Education, Beijing, China
| |
Collapse
|
36
|
Martínez JL, Lemus N, Lai TY, Mishra M, González-Domínguez I, Puente-Massaguer E, Loganathan M, Francis B, Samanovic MI, Krammer F, Mulligan MJ, Simon V, Palese P, Sun W. The immunodominance of antigenic site Sb on the H1 influenza virus hemagglutinin increases with high immunoglobulin titers of the cohorts and with young age, but not sex. Vaccine 2024; 42:3365-3373. [PMID: 38627145 PMCID: PMC11145762 DOI: 10.1016/j.vaccine.2024.04.037] [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: 11/28/2023] [Revised: 02/20/2024] [Accepted: 04/11/2024] [Indexed: 05/12/2024]
Abstract
The head domain of the hemagglutinin of influenza viruses plays a dominant role in the antibody response due to the presence of immunodominant antigenic sites that are the main targets of host neutralizing antibodies. For the H1 hemagglutinin, five major antigenic sites defined as Sa, Sb, Ca1, Ca2, and Cb have been described. Although previous studies have focused on defining the hierarchy of the antigenic sites of the hemagglutinin in different human cohorts, it is still unclear if the immunodominance profile of the antigenic sites might change with the antibody levels of individuals or if other demographic factors (such as exposure history, sex, or age) could also influence the importance of the antigenic sites. The major antigenic sites of influenza viruses hemagglutinins are responsible for eliciting most of the hemagglutination inhibition antibodies in the host. To determine the antibody prevalence towards each major antigenic site, we evaluated the hemagglutination inhibition against a panel of mutant H1 viruses, each one lacking one of the "classic" antigenic sites. Our results showed that the individuals from the Stop Flu NYU cohort had an immunodominant response towards the sites Sb and Ca2 of H1 hemagglutinin. A simple logistic regression analysis of the immunodominance profiles and the hemagglutination inhibition titers displayed by each donor revealed that individuals with high hemagglutination inhibition titers against the wild-type influenza virus exhibited higher probabilities of displaying an immunodominance profile dominated by Sb, followed by Ca2 (Sb > Ca2 profile), while individuals with low hemagglutination inhibition titers presented a higher chance of displaying an immunodominance profile in which Sb and Ca2 presented the same level of immunodominance (Sb = Ca2 profile). Finally, while age exhibited an influence on the immunodominance of the antigenic sites, biological sex was not related to displaying a specific immunodominance profile.
Collapse
Affiliation(s)
- Jose L Martínez
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States.
| | - Nicholas Lemus
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States
| | - Tsoi Ying Lai
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States
| | - Mitali Mishra
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States
| | - Irene González-Domínguez
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States
| | - Eduard Puente-Massaguer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States; Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, New York, United States
| | - Madhumathi Loganathan
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States; Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, New York, United States
| | - Benjamin Francis
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States; Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, New York, United States
| | - Marie I Samanovic
- Department of Medicine, NYU Grossman School of Medicine, New York, New York, United States; NYU Langone Vaccine Center, NYU Grossman School of Medicine, New York, New York, United States
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States; Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, New York, United States; Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, United States
| | - Mark J Mulligan
- Department of Medicine, NYU Grossman School of Medicine, New York, New York, United States; NYU Langone Vaccine Center, NYU Grossman School of Medicine, New York, New York, United States
| | - Viviana Simon
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States; Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, New York, United States; Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, United States
| | - Peter Palese
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States; Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, United States
| | - Weina Sun
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States.
| |
Collapse
|
37
|
Yang T, Tang L, Li P, Li B, Ye L, Zhou J. Effectiveness of inactivated influenza vaccine against laboratory-confirmed influenza among Chinese elderly: a test-negative design. BMC Geriatr 2024; 24:404. [PMID: 38714944 PMCID: PMC11077745 DOI: 10.1186/s12877-024-05003-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 04/22/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND Evidence on the effectiveness of influenza vaccination in the elderly is limited, and results are controversial. There are also few reports from China. METHODS We conducted a test-negative case-control study design to estimate influenza vaccine effectiveness (VE) against laboratory-confirmed influenza-associated visits among elderly (aged ≥ 60 years) across four influenza seasons in Ningbo, China, from 2018 to 19 to 2021-22. Influenza-positive cases and negative controls were randomly matched in a 1:1 ratio according to age, sex, hospital, and date of influenza testing. We used logistic regression models to compare vaccination odds ratios (ORs) in cases to controls. We calculated the VE as [100% × (1-adjusted OR)] and calculated the 95% confidence interval (CI) around the estimate. RESULTS A total of 30,630 elderly patients tested for influenza with virus nucleic acid or antigen during the study period. After exclusions, we included 1 825 influenza-positive cases and 1 825 influenza-negative controls. Overall, the adjusted VE for influenza-related visits was 63.5% (95% CI, 56.3-69.5%), but varied by season. Influenza VE was 59.8% (95% CI, 51.5-66.7%) for influenza A and 89.6% (95% CI, 77.1-95.3%) for influenza B. The VE for ages 60-69 and 70-79 was 65.2% (95% CI, 55.4-72.9%) and 69.8% (95% CI, 58.7-77.9%), respectively, but only 45.4% (95% CI, 6.2-68.2%) for ages 80 and over. CONCLUSIONS Standard-dose inactivated influenza vaccine has shown good protection in the elderly in China. However, protection may not be satisfactory in people aged 80 years and older.
Collapse
Affiliation(s)
- Tianchi Yang
- Institute of Immunization and Prevention, Ningbo Municipal Center for Disease Control and Prevention, Ningbo, Zhejiang, China
| | - Ling Tang
- Ningbo Health Information Center, Ningbo, Zhejiang, China
| | - Pingping Li
- Jiangbei District Center for Disease Control and Prevention, Ningbo, Zhejiang, China
| | - Baojun Li
- Haishu District Center for Disease Control and Prevention, Ningbo, Zhejiang, China
| | - Lixia Ye
- Institute of Immunization and Prevention, Ningbo Municipal Center for Disease Control and Prevention, Ningbo, Zhejiang, China.
| | - Jifang Zhou
- School of International Pharmaceutical Business, China Pharmaceutical University, Jiangsu, China.
| |
Collapse
|
38
|
Lopuhaä BV, Guzel C, van der Lee A, van den Bosch TPP, van Kemenade FJ, Huisman MV, Kruip MJHA, Luider TM, von der Thüsen JH. Increase in venous thromboembolism in SARS-CoV-2 infected lung tissue: proteome analysis of lung parenchyma, isolated endothelium, and thrombi. Histopathology 2024; 84:967-982. [PMID: 38253958 DOI: 10.1111/his.15143] [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: 07/13/2023] [Revised: 12/22/2023] [Accepted: 01/04/2024] [Indexed: 01/24/2024]
Abstract
AIMS COVID-19 pneumonia is characterized by an increased rate of deep venous thrombosis and pulmonary embolism. To better understand the pathophysiology behind thrombosis in COVID-19, we performed proteomics analysis on SARS-CoV-2 infected lung tissue. METHODS Liquid chromatography mass spectrometry was performed on SARS-CoV-2 infected postmortem lung tissue samples. Five protein profiling analyses were performed: whole slide lung parenchyma analysis, followed by analysis of isolated thrombi and endothelium, both stratified by disease (COVID-19 versus influenza) and thrombus morphology (embolism versus in situ). Influenza autopsy cases with pulmonary thrombi were used as controls. RESULTS Compared to influenza controls, both analyses of COVID-19 whole-tissue and isolated endothelium showed upregulation of proteins and pathways related to liver metabolism including urea cycle activation, with arginase being among the top upregulated proteins in COVID-19 lung tissue. Analysis of isolated COVID-19 thrombi showed significant downregulation of pathways related to platelet activation compared to influenza thrombi. Analysis of isolated thrombi based on histomorphology shows that in situ thrombi have significant upregulation of coronavirus pathogenesis proteins. CONCLUSIONS The decrease in platelet activation pathways in severe COVID-19 thrombi suggests a relative increase in venous thromboembolism, as thrombi from venous origin tend to contain fewer platelets than arterial thrombi. Based on histomorphology, in situ thrombi show upregulation of various proteins related to SARS-CoV-2 pathogenesis compared to thromboemboli, which may indicate increased in situ pulmonary thrombosis in COVID-19. Therefore, this study supports the increase of venous thromboembolism without undercutting the involvement of in situ thrombosis in severe COVID-19.
Collapse
Affiliation(s)
- Boaz V Lopuhaä
- Department of Pathology, Erasmus University Medical Centre, Rotterdam, the Netherlands
| | - Coşkun Guzel
- Laboratory of Neuro-Oncology, Clinical and Cancer Proteomics, Department of Neurology, Erasmus University Medical Centre, Rotterdam, the Netherlands
| | | | | | | | - Menno V Huisman
- Department of Thrombosis and Hemostasis, Leiden University Medical Center, Leiden, the Netherlands
| | - Marieke J H A Kruip
- Department of Haematology, Erasmus University Medical Centre, Rotterdam, the Netherlands
| | - Theo M Luider
- Laboratory of Neuro-Oncology, Clinical and Cancer Proteomics, Department of Neurology, Erasmus University Medical Centre, Rotterdam, the Netherlands
| | - Jan H von der Thüsen
- Department of Pathology, Erasmus University Medical Centre, Rotterdam, the Netherlands
| |
Collapse
|
39
|
Guo W, Luo J, Zhao S, Li L, Xing W, Gao R. The critical role of RAGE in severe influenza infection: A target for control of inflammatory response in the disease. Clin Immunol 2024; 262:110178. [PMID: 38460892 DOI: 10.1016/j.clim.2024.110178] [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: 12/24/2023] [Revised: 01/30/2024] [Accepted: 02/21/2024] [Indexed: 03/11/2024]
Abstract
Controlling the excessive inflammatory response is one of the key ways to reduce the severity and mortality of severe influenza virus infections. RAGE is involved in inflammatory responses and acute lung injuries. Here, we investigated the role of RAGE and its potential application as a target for severe influenza treatment through serological correlation analysis for influenza patients, and treatment with the RAGE inhibitor FPS-ZM1 on A549 cells or mice with influenza A (H1N1) infection. The results showed high levels of RAGE were correlated with immunopathological injury and severity of influenza, and FPS-ZM1 treatment increased the viability of A549 cells with influenza A infection and decreased morbidity and mortality of influenza A virus infection in mice. The RAGE/NF-κb inflammatory signaling pathway is a major targeting pathway for FPS-ZM1 treatment in severe influenza. These findings provide further insights into the immune injury of severe influenza and a potential targeting candidate for the disease treatment.
Collapse
Affiliation(s)
- Wenhui Guo
- NHC Key Laboratory of Biosafety, NHC Key Laboratory of Medical Virology and Viral Diseases, Chinese National Influenza Center, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China; National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Junhao Luo
- NHC Key Laboratory of Biosafety, NHC Key Laboratory of Medical Virology and Viral Diseases, Chinese National Influenza Center, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Song Zhao
- NHC Key Laboratory of Biosafety, NHC Key Laboratory of Medical Virology and Viral Diseases, Chinese National Influenza Center, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Li Li
- NHC Key Laboratory of Biosafety, NHC Key Laboratory of Medical Virology and Viral Diseases, Chinese National Influenza Center, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Wenge Xing
- National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China.
| | - Rongbao Gao
- NHC Key Laboratory of Biosafety, NHC Key Laboratory of Medical Virology and Viral Diseases, Chinese National Influenza Center, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China.
| |
Collapse
|
40
|
Ongun M, Lokras AG, Baghel S, Shi Z, Schmidt ST, Franzyk H, Rades T, Sebastiani F, Thakur A, Foged C. Lipid nanoparticles for local delivery of mRNA to the respiratory tract: Effect of PEG-lipid content and administration route. Eur J Pharm Biopharm 2024; 198:114266. [PMID: 38499255 DOI: 10.1016/j.ejpb.2024.114266] [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: 12/18/2023] [Revised: 03/11/2024] [Accepted: 03/13/2024] [Indexed: 03/20/2024]
Abstract
Design of inhalable mRNA therapeutics is promising because local administration in the respiratory tract is minimally invasive and induces a local response. However, several challenges related to administration via inhalation and respiratory tract barriers have so far prevented the progress of inhaled mRNA therapeutics. Here, we investigated factors of importance for lipid nanoparticle (LNP)-mediated delivery of mRNA to the respiratory tract. We hypothesized that: (i) the PEG-lipid content is important for providing colloidal stability during aerosolization and for mucosal delivery, (ii) the PEG-lipid contentinfluences the expression of mRNA-encoded protein in the lungs, and (iii) the route of administration (nasal versus pulmonary) affects mRNA delivery in the lungs. In this study, we aimed to optimize the PEG-lipid content for mucosal delivery and to investigatethe effect of administration route on the kinetics of protein expression. Our results show that increasing the PEG-lipid content improves the colloidal stability during the aerosolization process, but has a negative impact on the transfection efficiencyin vitro. The kinetics of protein expressionin vivois dependent on the route of administration, and we found that pulmonaryadministration of mRNA-LNPs to mice results inmore durable protein expression than nasaladministration. These results demonstrate that the design of the delivery system and the route of administration are importantfor achieving high mRNA transfection efficiency in the respiratory tract.
Collapse
Affiliation(s)
- Melike Ongun
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen Ø, Denmark
| | - Abhijeet Girish Lokras
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen Ø, Denmark
| | - Saahil Baghel
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen Ø, Denmark
| | - Zhenning Shi
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen Ø, Denmark
| | - Signe Tandrup Schmidt
- Department of Infectious Disease Immunology, Statens Serum Institut, Artillerivej 5, 2300 Copenhagen S, Denmark
| | - Henrik Franzyk
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Jagtvej 162, DK-2100 Copenhagen Ø, Denmark
| | - Thomas Rades
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen Ø, Denmark
| | - Federica Sebastiani
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen Ø, Denmark; Division of Physical Chemistry, Department of Chemistry, Lund University, 22100 Lund, Sweden
| | - Aneesh Thakur
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen Ø, Denmark
| | - Camilla Foged
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen Ø, Denmark.
| |
Collapse
|
41
|
Sanchez PL, Andre G, Antipov A, Petrovsky N, Ross TM. Advax-SM™-Adjuvanted COBRA (H1/H3) Hemagglutinin Influenza Vaccines. Vaccines (Basel) 2024; 12:455. [PMID: 38793706 PMCID: PMC11125990 DOI: 10.3390/vaccines12050455] [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: 02/08/2024] [Revised: 03/25/2024] [Accepted: 04/22/2024] [Indexed: 05/26/2024] Open
Abstract
Adjuvants enhance immune responses stimulated by vaccines. To date, many seasonal influenza vaccines are not formulated with an adjuvant. In the present study, the adjuvant Advax-SM™ was combined with next generation, broadly reactive influenza hemagglutinin (HA) vaccines that were designed using a computationally optimized broadly reactive antigen (COBRA) methodology. Advax-SM™ is a novel adjuvant comprising inulin polysaccharide and CpG55.2, a TLR9 agonist. COBRA HA vaccines were combined with Advax-SM™ or a comparator squalene emulsion (SE) adjuvant and administered to mice intramuscularly. Mice vaccinated with Advax-SM™ adjuvanted COBRA HA vaccines had increased serum levels of anti-influenza IgG and IgA, high hemagglutination inhibition activity against a panel of H1N1 and H3N2 influenza viruses, and increased anti-influenza antibody secreting cells isolated from spleens. COBRA HA plus Advax-SM™ immunized mice were protected against both morbidity and mortality following viral challenge and, at postmortem, had no detectable lung viral titers or lung inflammation. Overall, the Advax-SM™-adjuvanted COBRA HA formulation provided effective protection against drifted H1N1 and H3N2 influenza viruses.
Collapse
Affiliation(s)
- Pedro L. Sanchez
- Center for Vaccines and Immunology, University of Georgia, Athens, GA 30602, USA;
- Department of Infectious Diseases, University of Georgia, Athens, GA 30602, USA
- Florida Research and Innovation Center, Cleveland Clinic, Port Saint Lucie, FL 34987, USA
| | - Greiciely Andre
- Vaxine Pty Ltd., Adelaide, SA 5046, Australia; (G.A.); (A.A.); (N.P.)
| | - Anna Antipov
- Vaxine Pty Ltd., Adelaide, SA 5046, Australia; (G.A.); (A.A.); (N.P.)
| | - Nikolai Petrovsky
- Vaxine Pty Ltd., Adelaide, SA 5046, Australia; (G.A.); (A.A.); (N.P.)
| | - Ted M. Ross
- Center for Vaccines and Immunology, University of Georgia, Athens, GA 30602, USA;
- Department of Infectious Diseases, University of Georgia, Athens, GA 30602, USA
- Florida Research and Innovation Center, Cleveland Clinic, Port Saint Lucie, FL 34987, USA
- Department of Infection Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| |
Collapse
|
42
|
Cohen LE, Hansen CL, Andrew MK, McNeil SA, Vanhems P, Kyncl J, Domingo JD, Zhang T, Dbaibo G, Laguna-Torres VA, Draganescu A, Baumeister E, Gomez D, Raboni SM, Giamberardino HIG, Nunes MC, Burtseva E, Sominina A, Medić S, Coulibaly D, Salah AB, Otieno NA, Koul PA, Unal S, Tanriover MD, Mazur M, Bresee J, Viboud C, Chaves SS. Predictors of Severity of Influenza-Related Hospitalizations: Results From the Global Influenza Hospital Surveillance Network (GIHSN). J Infect Dis 2024; 229:999-1009. [PMID: 37527470 PMCID: PMC11011157 DOI: 10.1093/infdis/jiad303] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 07/13/2023] [Accepted: 07/26/2023] [Indexed: 08/03/2023] Open
Abstract
BACKGROUND The Global Influenza Hospital Surveillance Network (GIHSN) has since 2012 provided patient-level data on severe influenza-like-illnesses from >100 participating clinical sites worldwide based on a core protocol and consistent case definitions. METHODS We used multivariable logistic regression to assess the risk of intensive care unit admission, mechanical ventilation, and in-hospital death among hospitalized patients with influenza and explored the role of patient-level covariates and country income level. RESULTS The data set included 73 121 patients hospitalized with respiratory illness in 22 countries, including 15 660 with laboratory-confirmed influenza. After adjusting for patient-level covariates we found a 7-fold increase in the risk of influenza-related intensive care unit admission in lower middle-income countries (LMICs), compared with high-income countries (P = .01). The risk of mechanical ventilation and in-hospital death also increased by 4-fold in LMICs, though these differences were not statistically significant. We also find that influenza mortality increased significantly with older age and number of comorbid conditions. Across all severity outcomes studied and after controlling for patient characteristics, infection with influenza A/H1N1pdm09 was more severe than with A/H3N2. CONCLUSIONS Our study provides new information on influenza severity in underresourced populations, particularly those in LMICs.
Collapse
Affiliation(s)
- Lily E Cohen
- Ready2Respond p/o The Task Force for Global Health, Decatur, Georgia, USA
- Department of Medical Education, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Chelsea L Hansen
- Division of International Epidemiology and Population Studies, Fogarty International Center, National Institutes of Health, Bethesda, Maryland, USA
- Brotman Baty Institute, University of Washington, Seattle, Washington, USA
- PandemiX Center, Department of Science & Environment, Roskilde University, Denmark
| | - Melissa K Andrew
- Canadian Center for Vaccinology, IWK Health Centre and Nova Scotia Health, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Shelly A McNeil
- Canadian Center for Vaccinology, IWK Health Centre and Nova Scotia Health, Dalhousie University, Halifax, Nova Scotia, Canada
| | | | - Jan Kyncl
- Department of Infectious Diseases Epidemiology, National Institute of Public Health, Prague, Czech Republic
- Department of Epidemiology and Biostatistics, Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Javier Díez Domingo
- Fundación Para el Fomento de la Investigación Sanitaria y Biomédica de la Comunitat Valenciana (FISABIO–Public Health), Valencia, Spain
| | - Tao Zhang
- School of Public Health, Fudan University, Shanghai, China
| | - Ghassan Dbaibo
- Center for Infectious Diseases Research, American University of Beirut, Beirut, Lebanon
| | | | - Anca Draganescu
- National Institute for Infectious Diseases “Prof Dr Matei Bals”, Bucharest, Romania
| | - Elsa Baumeister
- Respiratory Virus Laboratory, Virology Department, INEI-ANLIS, Buenos Aires, Argentina
| | - Doris Gomez
- Grupo de Investigación UNIMOL, Facultad de Medicina, Universidad de Cartagena, Cartagena de Indias, Colombia
| | - Sonia M Raboni
- Virology Laboratory, Infectious Diseases Division, Universidade Federal do Paraná, Hospital Pequeno Principe, Curitiba, Paraná, Brazil
| | - Heloisa I G Giamberardino
- Virology Laboratory, Infectious Diseases Division, Universidade Federal do Paraná, Hospital Pequeno Principe, Curitiba, Paraná, Brazil
| | - Marta C Nunes
- South African Medical Research Council, Vaccines and Infectious Diseases Analytics Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Department of Science and Technology/National Research Foundation, South African Research Chair Initiative in Vaccine Preventable Diseases, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Elena Burtseva
- Gamaleya Federal Research Center for Epidemiology and Microbiology, Ministry of Health of Russian Federation, Moscow, Russia
| | - Anna Sominina
- Smorodintsev Research Institute of Influenza, St Petersburg, Russia
| | - Snežana Medić
- Institute for Public Health of Vojvodina, Novi Sad, Serbia
- Faculty of Medicine, University of Novi Sad, Novi Sad, Serbia
| | | | - Afif Ben Salah
- Institut Pasteur de Tunis, Tunis, Tunisia
- Arabian Gulf University, Manama, Bahrain
| | - Nancy A Otieno
- Kenya Medical Research Institute (KEMRI), Nairobi, Kenya
| | - Parvaiz A Koul
- Sheri Kashmir Institute of Medical Sciences, Srinagar, India
| | - Serhat Unal
- Department of Infectious Diseases and Clinical Microbiology, Hacettepe University School of Medicine, Ankara, Turkey
- Turkish Society of Internal Medicine, Ankara, Turkey
| | - Mine Durusu Tanriover
- Turkish Society of Internal Medicine, Ankara, Turkey
- Department of Internal Medicine, Hacettepe University School of Medicine, Ankara, Turkey
| | - Marie Mazur
- Ready2Respond p/o The Task Force for Global Health, Decatur, Georgia, USA
| | - Joseph Bresee
- Ready2Respond p/o The Task Force for Global Health, Decatur, Georgia, USA
| | - Cecile Viboud
- Division of International Epidemiology and Population Studies, Fogarty International Center, National Institutes of Health, Bethesda, Maryland, USA
| | - Sandra S Chaves
- Foundation for Influenza Epidemiology, Fondation de France, Paris, France
| |
Collapse
|
43
|
Unione L, Ammerlaan ANA, Bosman GP, Uslu E, Liang R, Broszeit F, van der Woude R, Liu Y, Ma S, Liu L, Gómez-Redondo M, Bermejo IA, Valverde P, Diercks T, Ardá A, de Vries RP, Boons GJ. Probing altered receptor specificities of antigenically drifting human H3N2 viruses by chemoenzymatic synthesis, NMR, and modeling. Nat Commun 2024; 15:2979. [PMID: 38582892 PMCID: PMC10998905 DOI: 10.1038/s41467-024-47344-y] [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: 06/08/2023] [Accepted: 03/25/2024] [Indexed: 04/08/2024] Open
Abstract
Prototypic receptors for human influenza viruses are N-glycans carrying α2,6-linked sialosides. Due to immune pressure, A/H3N2 influenza viruses have emerged with altered receptor specificities that bind α2,6-linked sialosides presented on extended N-acetyl-lactosamine (LacNAc) chains. Here, binding modes of such drifted hemagglutinin's (HAs) are examined by chemoenzymatic synthesis of N-glycans having 13C-labeled monosaccharides at strategic positions. The labeled glycans are employed in 2D STD-1H by 13C-HSQC NMR experiments to pinpoint which monosaccharides of the extended LacNAc chain engage with evolutionarily distinct HAs. The NMR data in combination with computation and mutagenesis demonstrate that mutations distal to the receptor binding domain of recent HAs create an extended binding site that accommodates with the extended LacNAc chain. A fluorine containing sialoside is used as NMR probe to derive relative binding affinities and confirms the contribution of the extended LacNAc chain for binding.
Collapse
Affiliation(s)
- Luca Unione
- Department of Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG, Utrecht, The Netherlands.
- CICbioGUNE, Basque Research & Technology Alliance (BRTA), Bizkaia Technology Park, Building 800, 48160, Derio, Bizkaia, Spain.
- Ikerbasque, Basque Foundation for Science, Euskadi Plaza 5, 48009, Bilbao, Bizkaia, Spain.
| | - Augustinus N A Ammerlaan
- Department of Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - Gerlof P Bosman
- Department of Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - Elif Uslu
- Department of Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - Ruonan Liang
- Department of Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - Frederik Broszeit
- Department of Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - Roosmarijn van der Woude
- Department of Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - Yanyan Liu
- Department of Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - Shengzhou Ma
- Complex Carbohydrate Research Center, University of Georgia, 315 Riverbend Rd, Athens, GA, 30602, USA
| | - Lin Liu
- Complex Carbohydrate Research Center, University of Georgia, 315 Riverbend Rd, Athens, GA, 30602, USA
| | - Marcos Gómez-Redondo
- CICbioGUNE, Basque Research & Technology Alliance (BRTA), Bizkaia Technology Park, Building 800, 48160, Derio, Bizkaia, Spain
| | - Iris A Bermejo
- CICbioGUNE, Basque Research & Technology Alliance (BRTA), Bizkaia Technology Park, Building 800, 48160, Derio, Bizkaia, Spain
| | - Pablo Valverde
- CICbioGUNE, Basque Research & Technology Alliance (BRTA), Bizkaia Technology Park, Building 800, 48160, Derio, Bizkaia, Spain
| | - Tammo Diercks
- CICbioGUNE, Basque Research & Technology Alliance (BRTA), Bizkaia Technology Park, Building 800, 48160, Derio, Bizkaia, Spain
| | - Ana Ardá
- CICbioGUNE, Basque Research & Technology Alliance (BRTA), Bizkaia Technology Park, Building 800, 48160, Derio, Bizkaia, Spain
- Ikerbasque, Basque Foundation for Science, Euskadi Plaza 5, 48009, Bilbao, Bizkaia, Spain
| | - Robert P de Vries
- Department of Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG, Utrecht, The Netherlands.
| | - Geert-Jan Boons
- Department of Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG, Utrecht, The Netherlands.
- Complex Carbohydrate Research Center, University of Georgia, 315 Riverbend Rd, Athens, GA, 30602, USA.
- Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, The Netherlands.
- Department of Chemistry, University of Georgia, Athens, GA, 30602, USA.
| |
Collapse
|
44
|
Paget J, Chaves SS, Li Y, Nair H, Spreeuwenberg P. Revisiting influenza-hospitalisation estimates from the Burden of Influenza and Respiratory Syncytial Virus Disease (BIRD) project using different extrapolation methods. J Glob Health 2024; 14:03017. [PMID: 38574354 PMCID: PMC10994668 DOI: 10.7189/jogh.14.03017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/06/2024] Open
Affiliation(s)
| | - Sandra S Chaves
- Foundation for Influenza Epidemiology, Fondation de France, Paris, France
| | - You Li
- University of Edinburgh, Edinburgh, UK
- Nanjing Medical University, Nanjing, China
| | - Harish Nair
- University of Edinburgh, Edinburgh, UK
- Nanjing Medical University, Nanjing, China
- School of Public Health, University of the Witwatersrand, Johannesburg, South Africa
| | | |
Collapse
|
45
|
Owusu D, Ndegwa LK, Ayugi J, Kinuthia P, Kalani R, Okeyo M, Otieno NA, Kikwai G, Juma B, Munyua P, Kuria F, Okunga E, Moen AC, Emukule GO. Use of Sentinel Surveillance Platforms for Monitoring SARS-CoV-2 Activity: Evidence From Analysis of Kenya Influenza Sentinel Surveillance Data. JMIR Public Health Surveill 2024; 10:e50799. [PMID: 38526537 PMCID: PMC11002741 DOI: 10.2196/50799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 12/19/2023] [Accepted: 02/02/2024] [Indexed: 03/26/2024] Open
Abstract
BACKGROUND Little is known about the cocirculation of influenza and SARS-CoV-2 viruses during the COVID-19 pandemic and the use of respiratory disease sentinel surveillance platforms for monitoring SARS-CoV-2 activity in sub-Saharan Africa. OBJECTIVE We aimed to describe influenza and SARS-CoV-2 cocirculation in Kenya and how the SARS-CoV-2 data from influenza sentinel surveillance correlated with that of universal national surveillance. METHODS From April 2020 to March 2022, we enrolled 7349 patients with severe acute respiratory illness or influenza-like illness at 8 sentinel influenza surveillance sites in Kenya and collected demographic, clinical, underlying medical condition, vaccination, and exposure information, as well as respiratory specimens, from them. Respiratory specimens were tested for influenza and SARS-CoV-2 by real-time reverse transcription polymerase chain reaction. The universal national-level SARS-CoV-2 data were also obtained from the Kenya Ministry of Health. The universal national-level SARS-CoV-2 data were collected from all health facilities nationally, border entry points, and contact tracing in Kenya. Epidemic curves and Pearson r were used to describe the correlation between SARS-CoV-2 positivity in data from the 8 influenza sentinel sites in Kenya and that of the universal national SARS-CoV-2 surveillance data. A logistic regression model was used to assess the association between influenza and SARS-CoV-2 coinfection with severe clinical illness. We defined severe clinical illness as any of oxygen saturation <90%, in-hospital death, admission to intensive care unit or high dependence unit, mechanical ventilation, or a report of any danger sign (ie, inability to drink or eat, severe vomiting, grunting, stridor, or unconsciousness in children younger than 5 years) among patients with severe acute respiratory illness. RESULTS Of the 7349 patients from the influenza sentinel surveillance sites, 76.3% (n=5606) were younger than 5 years. We detected any influenza (A or B) in 8.7% (629/7224), SARS-CoV-2 in 10.7% (768/7199), and coinfection in 0.9% (63/7165) of samples tested. Although the number of samples tested for SARS-CoV-2 from the sentinel surveillance was only 0.2% (60 per week vs 36,000 per week) of the number tested in the universal national surveillance, SARS-CoV-2 positivity in the sentinel surveillance data significantly correlated with that of the universal national surveillance (Pearson r=0.58; P<.001). The adjusted odds ratios (aOR) of clinical severe illness among participants with coinfection were similar to those of patients with influenza only (aOR 0.91, 95% CI 0.47-1.79) and SARS-CoV-2 only (aOR 0.92, 95% CI 0.47-1.82). CONCLUSIONS Influenza substantially cocirculated with SARS-CoV-2 in Kenya. We found a significant correlation of SARS-CoV-2 positivity in the data from 8 influenza sentinel surveillance sites with that of the universal national SARS-CoV-2 surveillance data. Our findings indicate that the influenza sentinel surveillance system can be used as a sustainable platform for monitoring respiratory pathogens of pandemic potential or public health importance.
Collapse
Affiliation(s)
- Daniel Owusu
- Influenza Division, US Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - Linus K Ndegwa
- Global Influenza Branch, Influenza Division, US Centers for Disease Control and Prevention, Nairobi, Kenya
| | - Jorim Ayugi
- Centre for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| | | | - Rosalia Kalani
- Disease Surveillance and Response Unit, Ministry of Health, Nairobi, Kenya
| | - Mary Okeyo
- National Influenza Centre Laboratory, National Public Health Laboratories, Ministry of Health, Nairobi, Kenya
| | - Nancy A Otieno
- Centre for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| | - Gilbert Kikwai
- Centre for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| | - Bonventure Juma
- Global Influenza Branch, Influenza Division, US Centers for Disease Control and Prevention, Nairobi, Kenya
| | - Peninah Munyua
- Global Influenza Branch, Influenza Division, US Centers for Disease Control and Prevention, Nairobi, Kenya
| | - Francis Kuria
- Directorate of Public Health, Ministry of Health, Nairobi, Kenya
| | - Emmanuel Okunga
- Disease Surveillance and Response Unit, Ministry of Health, Nairobi, Kenya
| | - Ann C Moen
- Influenza Division, US Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - Gideon O Emukule
- Global Influenza Branch, Influenza Division, US Centers for Disease Control and Prevention, Nairobi, Kenya
| |
Collapse
|
46
|
Foo IJH, Chua BY, Clemens EB, Chang SY, Jia X, McQuilten HA, Yap AHY, Cabug AF, Ashayeripanah M, McWilliam HEG, Villadangos JA, Evrard M, Mackay LK, Wakim LM, Fazakerley JK, Kedzierska K, Kedzierski L. Prior infection with unrelated neurotropic virus exacerbates influenza disease and impairs lung T cell responses. Nat Commun 2024; 15:2619. [PMID: 38521764 PMCID: PMC10960853 DOI: 10.1038/s41467-024-46822-7] [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: 07/28/2023] [Accepted: 03/07/2024] [Indexed: 03/25/2024] Open
Abstract
Immunity to infectious diseases is predominantly studied by measuring immune responses towards a single pathogen, although co-infections are common. In-depth mechanisms on how co-infections impact anti-viral immunity are lacking, but are highly relevant to treatment and prevention. We established a mouse model of co-infection with unrelated viruses, influenza A (IAV) and Semliki Forest virus (SFV), causing disease in different organ systems. SFV infection eight days before IAV infection results in prolonged IAV replication, elevated cytokine/chemokine levels and exacerbated lung pathology. This is associated with impaired lung IAV-specific CD8+ T cell responses, stemming from suboptimal CD8+ T cell activation and proliferation in draining lymph nodes, and dendritic cell paralysis. Prior SFV infection leads to increased blood brain barrier permeability and presence of IAV RNA in brain, associated with increased trafficking of IAV-specific CD8+ T cells and establishment of long-term tissue-resident memory. Relative to lung IAV-specific CD8+ T cells, brain memory IAV-specific CD8+ T cells have increased TCR repertoire diversity within immunodominant DbNP366+CD8+ and DbPA224+CD8+ responses, featuring suboptimal TCR clonotypes. Overall, our study demonstrates that infection with an unrelated neurotropic virus perturbs IAV-specific immune responses and exacerbates IAV disease. Our work provides key insights into therapy and vaccine regimens directed against unrelated pathogens.
Collapse
Affiliation(s)
- Isabelle Jia-Hui Foo
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
- Department of Veterinary Biosciences, Faculty of Science, The University of Melbourne, Melbourne, VIC 3000, Australia
| | - Brendon Y Chua
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - E Bridie Clemens
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - So Young Chang
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Xiaoxiao Jia
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Hayley A McQuilten
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Ashley Huey Yiing Yap
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Aira F Cabug
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Mitra Ashayeripanah
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Hamish E G McWilliam
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Jose A Villadangos
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
- Department of Biochemistry and Pharmacology; Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Maximilien Evrard
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Laura K Mackay
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Linda M Wakim
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - John K Fazakerley
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
- Department of Veterinary Biosciences, Faculty of Science, The University of Melbourne, Melbourne, VIC 3000, Australia
| | - Katherine Kedzierska
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia.
| | - Lukasz Kedzierski
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia.
| |
Collapse
|
47
|
Zou Y, Sun X, Wang Y, Wang Y, Ye X, Tu J, Yu R, Huang P. Integrating single-cell RNA sequencing data to genome-wide association analysis data identifies significant cell types in influenza A virus infection and COVID-19. Brief Funct Genomics 2024; 23:110-117. [PMID: 37340787 DOI: 10.1093/bfgp/elad025] [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: 07/05/2022] [Revised: 02/23/2023] [Accepted: 06/01/2023] [Indexed: 06/22/2023] Open
Abstract
With the global pandemic of COVID-19, the research on influenza virus has entered a new stage, but it is difficult to elucidate the pathogenesis of influenza disease. Genome-wide association studies (GWASs) have greatly shed light on the role of host genetic background in influenza pathogenesis and prognosis, whereas single-cell RNA sequencing (scRNA-seq) has enabled unprecedented resolution of cellular diversity and in vivo following influenza disease. Here, we performed a comprehensive analysis of influenza GWAS and scRNA-seq data to reveal cell types associated with influenza disease and provide clues to understanding pathogenesis. We downloaded two GWAS summary data, two scRNA-seq data on influenza disease. After defining cell types for each scRNA-seq data, we used RolyPoly and LDSC-cts to integrate GWAS and scRNA-seq. Furthermore, we analyzed scRNA-seq data from the peripheral blood mononuclear cells (PBMCs) of a healthy population to validate and compare our results. After processing the scRNA-seq data, we obtained approximately 70 000 cells and identified up to 13 cell types. For the European population analysis, we determined an association between neutrophils and influenza disease. For the East Asian population analysis, we identified an association between monocytes and influenza disease. In addition, we also identified monocytes as a significantly related cell type in a dataset of healthy human PBMCs. In this comprehensive analysis, we identified neutrophils and monocytes as influenza disease-associated cell types. More attention and validation should be given in future studies.
Collapse
Affiliation(s)
- Yixin Zou
- Department of Epidemiology, National Vaccine Innovation Platform, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Xifang Sun
- Department of Mathematics, School of Science, Xi'an Shiyou University, Xi'an, China
| | - Yifan Wang
- Department of Infectious Disease, Jurong Hospital Affiliated to Jiangsu University, Jurong, China
| | - Yidi Wang
- Department of Epidemiology, National Vaccine Innovation Platform, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Xiangyu Ye
- Department of Epidemiology, National Vaccine Innovation Platform, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Junlan Tu
- Department of Epidemiology, National Vaccine Innovation Platform, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Rongbin Yu
- Department of Epidemiology, National Vaccine Innovation Platform, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Peng Huang
- Department of Epidemiology, National Vaccine Innovation Platform, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| |
Collapse
|
48
|
Nesteruk I. Should we ignore SARS-CoV-2 disease? Epidemiol Infect 2024; 152:e57. [PMID: 38506229 PMCID: PMC11022254 DOI: 10.1017/s0950268824000487] [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: 12/07/2023] [Revised: 03/01/2024] [Accepted: 03/07/2024] [Indexed: 03/21/2024] Open
Abstract
Current World Health Organization (WHO) reports claim a decline in COVID-19 testing and reporting of new infections. To discuss the consequences of ignoring severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection, the endemic characteristics of the disease in 2023 with the ones estimated before using 2022 data sets are compared. The accumulated numbers of cases and deaths reported to the WHO by the 10 most infected countries and global figures were used to calculate the average daily numbers of cases DCC and deaths DDC per capita and case fatality rates (CFRs = DDC/DCC) for two periods in 2023. In some countries, the DDC values can be higher than the upper 2022 limit and exceed the seasonal influenza mortality. The increase in CFR in 2023 shows that SARS-CoV-2 infection is still dangerous. The numbers of COVID-19 cases and deaths per capita in 2022 and 2023 do not demonstrate downward trends with the increase in the percentages of fully vaccinated people and boosters. The reasons may be both rapid mutations of the coronavirus, which reduced the effectiveness of vaccines and led to a large number of re-infections, and inappropriate management.
Collapse
Affiliation(s)
- Igor Nesteruk
- Institute of Hydromechanics, National Academy of Sciences of Ukraine, Kyiv, Ukraine
| |
Collapse
|
49
|
Gomes Z, Lee GS, Mesfin S, Rocha R, Vervoort D. Viral cardiovascular surgical diseases: global burdens, challenges and opportunities. Future Cardiol 2024; 20:229-239. [PMID: 39049768 DOI: 10.1080/14796678.2024.2348382] [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: 10/20/2023] [Accepted: 04/24/2024] [Indexed: 07/27/2024] Open
Abstract
Globally, more than one billion people are vulnerable to neglected tropical diseases, many of which have viral origins and cardiovascular implications. Access to cardiovascular care is limited in countries where these conditions are endemic. Six billion people lack access to safe, timely and affordable cardiac surgical care, whereby over 100 countries and territories lack a single cardiac surgeon. Moreover, while clinically unique, the surgical consequences of neglected cardiovascular diseases with viral origins have been poorly described in the current literature. This review provides an overview of the global burden of viral cardiovascular disease, describes access to cardiac surgical care in regions where these conditions are endemic, and further highlights surgical consequences and considerations to manage patients requiring cardiac surgical care.
Collapse
Affiliation(s)
- Zoya Gomes
- Faculty of Medicine, Dalhousie University, Halifax, Nova B3H 4R2, Scotia, B3H 4R2, Canada
| | - Grace S Lee
- Temerty Faculty of Medicine, University of Toronto, Toronto, M5S 1A1, Ontario, M5S 1A1, Canada
| | - Samuel Mesfin
- College of Health Sciences, Addis Ababa University, Addis NBH1, Ababa, NBH1, Ethiopia
| | - Rodolfo Rocha
- Division of Cardiac Surgery, University of Toronto, Toronto, M5S 1A1, Ontario, M5S 1A1, Canada
| | - Dominique Vervoort
- Division of Cardiac Surgery, University of Toronto, Toronto, M5S 1A1, Ontario, M5S 1A1, Canada
- Institute of Health Policy, Management & Evaluation, University of Toronto, Toronto, M5T 3M6, M5T 3M6, Ontario,Canada
| |
Collapse
|
50
|
Kumar G, Sakharam KA. Tackling Influenza A virus by M2 ion channel blockers: Latest progress and limitations. Eur J Med Chem 2024; 267:116172. [PMID: 38330869 DOI: 10.1016/j.ejmech.2024.116172] [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: 11/09/2023] [Revised: 01/18/2024] [Accepted: 01/22/2024] [Indexed: 02/10/2024]
Abstract
Influenza outbreaks cause pandemics in millions of people. The treatment of influenza remains a challenge due to significant genetic polymorphism in the influenza virus. Also, developing vaccines to protect against seasonal and pandemic influenza infections is constantly impeded. Thus, antibiotics are the only first line of defense against antigenically distinct strains or new subtypes of influenza viruses. Among several anti-influenza targets, the M2 protein of the influenza virus performs several activities. M2 protein is an ion channel that permits proton conductance through the virion envelope and the deacidification of the Golgi apparatus. Both these functions are critical for viral replication. Thus, targeting the M2 protein of the influenza virus is an essential target. Rimantadine and amantadine are two well-known drugs that act on the M2 protein. However, these drugs acquired resistance to influenza and thus are not recommended to treat influenza infections. This review discusses an overview of anti-influenza therapy, M2 ion channel functions, and its working principle. It also discusses the M2 structure and its role, and the change in the structure leads to mutant variants of influenza A virus. We also shed light on the recently identified compounds acting against wild-type and mutated M2 proteins of influenza virus A. These scaffolds could be an alternative to M2 inhibitors and be developed as antibiotics for treating influenza infections.
Collapse
Affiliation(s)
- Gautam Kumar
- Department of Natural Products, Chemical Sciences, National Institute of Pharmaceutical Education and Research-Hyderabad, Hyderabad, Balanagar, 500037, India.
| | - Kakade Aditi Sakharam
- Department of Natural Products, Chemical Sciences, National Institute of Pharmaceutical Education and Research-Hyderabad, Hyderabad, Balanagar, 500037, India
| |
Collapse
|