1
|
Xu Y, Sun F, Chuai Z, Wang J, Bai Z, Bian C, Wang X, Zhao Z, Liu Y, Yang P. Cold-adapted influenza vaccine carrying three repeats of a respiratory syncytial virus (RSV) fusion glycoprotein epitope site protects BALB/c mice and cotton rats against RSV infection. Antiviral Res 2024; 229:105960. [PMID: 38986872 DOI: 10.1016/j.antiviral.2024.105960] [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: 01/18/2024] [Revised: 07/05/2024] [Accepted: 07/07/2024] [Indexed: 07/12/2024]
Abstract
Respiratory syncytial virus is the major cause of respiratory viral infections, particularly in infants, immunocompromised populations, and the elderly (over 65 years old), the prevention of RSV infection has become a priority. In this study, we generated a chimeric influenza virus, termed LAIV/RSV/HA-3F, using reverse genetics technology which contained three repeats of the RSV fusion protein neutralizing epitope site II to the N terminal in the background of the hemagglutinin (HA) gene of cold adapted influenza vaccine A/California/7/2009 ca. LAIV/RSV/HA-3F exhibited cold-adapted (ca) and attenuated (att) phenotype. BALB/c mice immunized intranasally with LAIV/RSV/HA-3F showed robust immunogenicity, inducing viral-specific antibody responses against both influenza and RSV, eliciting RSV-specific humoral, cellular and mucosal immune responses. LAIV/RSV/HA-3F also conferred protection as indicated by reduced viral titers and improved lung histopathological alterations against live RSV virus challenge. Mechanismly, single-cell RNA sequencing (scRNA-seq) and single-cell T cell antigen receptor (TCR) sequencing were employed to characterize the immune responses triggered by chimeric RSV vaccine, displaying that LAIV/RSV/HA-3F provided protection mainly via interferon-γ (IFN-γ). Moreover, we found that LAIV/RSV/HA-3F significantly inhibited viral replication in the challenged lung and protected against subsequent RSV challenge in cotton rats without causing lung disease. Taken together, our findings demonstrated that LAIV/RSV/HA-3F has potential as a promising bivalent vaccine with dual purpose candidate for the prevention of influenza and RSV, and preclinical and clinical studies warrant further investigations.
Collapse
Affiliation(s)
- Yongru Xu
- The First Medical Center of Chinese PLA General Hospital, Faculty of Hepato-Pancreato-Biliary Surgery, Institute of Hepatobiliary Surgery of Chinese PLA, Key Laboratory of Digital Hepatobiliary Surgery, PLA, Beijing, 100853, China
| | - Fang Sun
- The First Medical Center of Chinese PLA General Hospital, Faculty of Hepato-Pancreato-Biliary Surgery, Institute of Hepatobiliary Surgery of Chinese PLA, Key Laboratory of Digital Hepatobiliary Surgery, PLA, Beijing, 100853, China; Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, 100039, China
| | - Zhengran Chuai
- Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, 100039, China
| | - Junyun Wang
- Harbin GenVista Medical Laboratory Co., Ltd, Harbin, 150001, China; Heilongjiang Hulu Institute of Precision Medicine Co., Ltd, Harbin 150001, China
| | - Zhifang Bai
- Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, 100039, China
| | - Chengrong Bian
- Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, 100039, China
| | - Xiliang Wang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, 100071, China
| | - Zhongpeng Zhao
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, 100071, China
| | - Yongzhuang Liu
- School of Computer Science and Technology, Harbin Institute of Technology, Harbin 150001, China
| | - Penghui Yang
- The First Medical Center of Chinese PLA General Hospital, Faculty of Hepato-Pancreato-Biliary Surgery, Institute of Hepatobiliary Surgery of Chinese PLA, Key Laboratory of Digital Hepatobiliary Surgery, PLA, Beijing, 100853, China.
| |
Collapse
|
2
|
Xu Y, Sun F, Bai Z, Bian C, Wang X, Zhao Z, Yang P. Cold-adapted influenza-vectored RSV vaccine protects BALB/c mice and cotton rats from RSV challenge. J Med Virol 2024; 96:e29308. [PMID: 39007405 DOI: 10.1002/jmv.29308] [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: 03/23/2023] [Revised: 11/03/2023] [Accepted: 11/27/2023] [Indexed: 07/16/2024]
Abstract
Respiratory syncytial virus (RSV) remains the primary cause of lower respiratory tract infections, particularly in infants and the elderly. In this study, we employed reverse genetics to generate a chimeric influenza virus expressing neuraminidase-3F protein conjugate with three repeats of the RSV F protein protective epitope inserted into the NA gene of A/California/7/2009 ca (CA/AA ca), resulting in rFlu/RSV/NA-3F (hereafter, rFRN3). The expression of NA-3F protein was confirmed by Western blotting. The morphology and temperature-sensitive phenotype of rFRN3 were similar to CA/AA ca. Its immunogenicity and protective efficiency were evaluated in BALB/c mice and cotton rats. Intranasal administration of rFRN3 elicited robust humoral, cellular, and to some extent, mucosal immune responses. Compared to controls, rFRN3 protected animals from RSV infection, attenuated lung injury, and reduced viral titers in the nose and lungs post-RSV challenge. These results demonstrate that rFRN3 can trigger RSV-specific immune responses and thus exhibits potent protective efficacy. The "dual vaccine" approach of a cold-adapted influenza vector RSV vaccine will improve the prophylaxis of influenza and RSV infection. rFRN3 thus warrants further clinical investigations as a candidate RSV vaccine.
Collapse
Affiliation(s)
- Yongru Xu
- The First Medical Center of Chinese PLA General Hospital, Beijing, China
- Faculty of Hepato-Pancreato-Biliary Surgery, Chinese PLA General Hospital, Beijing, China
- Key Laboratory of Digital Hepatobiliary Surgery, PLA, Institute of Hepatobiliary Surgery of Chinese PLA, Beijing, China
| | - Fang Sun
- Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Zhifang Bai
- Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Chengrong Bian
- Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Xiliang Wang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
| | - Zhongpeng Zhao
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
| | - Penghui Yang
- The First Medical Center of Chinese PLA General Hospital, Beijing, China
- Faculty of Hepato-Pancreato-Biliary Surgery, Chinese PLA General Hospital, Beijing, China
- Key Laboratory of Digital Hepatobiliary Surgery, PLA, Institute of Hepatobiliary Surgery of Chinese PLA, Beijing, China
| |
Collapse
|
3
|
Vieira Antão A, Oltmanns F, Schmidt A, Viherlehto V, Irrgang P, Rameix-Welti MA, Bayer W, Lapuente D, Tenbusch M. Filling two needs with one deed: a combinatory mucosal vaccine against influenza A virus and respiratory syncytial virus. Front Immunol 2024; 15:1376395. [PMID: 38975350 PMCID: PMC11224462 DOI: 10.3389/fimmu.2024.1376395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 06/10/2024] [Indexed: 07/09/2024] Open
Abstract
Influenza A Virus (IAV) and Respiratory Syncytial Virus (RSV) are both responsible for millions of severe respiratory tract infections every year worldwide. Effective vaccines able to prevent transmission and severe disease, are important measures to reduce the burden for the global health system. Despite the strong systemic immune responses induced upon current parental immunizations, this vaccination strategy fails to promote a robust mucosal immune response. Here, we investigated the immunogenicity and efficacy of a mucosal adenoviral vector vaccine to tackle both pathogens simultaneously at their entry site. For this purpose, BALB/c mice were immunized intranasally with adenoviral vectors (Ad) encoding the influenza-derived proteins, hemagglutinin (HA) and nucleoprotein (NP), in combination with an Ad encoding for the RSV fusion (F) protein. The mucosal combinatory vaccine induced neutralizing antibodies as well as local IgA responses against both viruses. Moreover, the vaccine elicited pulmonary CD8+ and CD4+ tissue resident memory T cells (TRM) against the immunodominant epitopes of RSV-F and IAV-NP. Furthermore, the addition of Ad-TGFβ or Ad-CCL17 as mucosal adjuvant enhanced the formation of functional CD8+ TRM responses against the conserved IAV-NP. Consequently, the combinatory vaccine not only provided protection against subsequent infections with RSV, but also against heterosubtypic challenges with pH1N1 or H3N2 strains. In conclusion, we present here a potent combinatory vaccine for mucosal applications, which provides protection against two of the most relevant respiratory viruses.
Collapse
Affiliation(s)
- Ana Vieira Antão
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Friederike Oltmanns
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Anna Schmidt
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Vera Viherlehto
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Pascal Irrgang
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Marie-Anne Rameix-Welti
- Université Paris-Saclay – Université de Versailles St. Quentin, UMR 1173 (2I), Institut national de la santé et de la recherche médicale (INSERM), Montigny-le-Bretonneux, France
| | - Wibke Bayer
- Institute for Virology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Dennis Lapuente
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Matthias Tenbusch
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
- FAU Profile Center Immunomedicine (FAU I-MED), Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
4
|
Shi P, Wang R, Liu E, Deng Y. Timing patterns of initial respiratory syncytial virus infection and factors influencing disease severity in hospitalized infants with different health status. J Med Virol 2024; 96:e29719. [PMID: 38873911 DOI: 10.1002/jmv.29719] [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: 03/26/2024] [Revised: 05/14/2024] [Accepted: 05/23/2024] [Indexed: 06/15/2024]
Abstract
This study aimed to determine the timing patterns of the initial respiratory syncytial virus (RSV) infection and to identify the factors influencing disease severity in infants of varying health status. A retrospective study was conducted at the Affiliated Children's Hospital of Chongqing Medical University from 2012 to 2022. The timing of the first RSV infection was estimated in infants with differing health status using correlation analysis, considering their birth time. Logistic regression was utilized to identify factors influencing severe RSV infection in these infants. RSV detection primarily occurred in the winter and spring. Epidemic season and peak timing of RSV were not significantly affected by health status or the COVID-19 pandemic. A strong positive correlation was observed between the age at RSV infection and the interval from birth to the RSV peak season. Infants born during the RSV epidemic season exhibited a higher likelihood of infection within the first 2 months postbirth. In contrast, those born outside the RSV epidemic season were more susceptible to infection during the subsequent peak. Notably, infants with pre-existing health conditions contracted RSV at an earlier age compared to their healthy counterparts. Among healthy infants, severe RSV infection was associated with sex, age, and timing of infection. For infants with underlying conditions, severe RSV infection was primarily related to age and timing of infection. The initial timing of RSV infection in infants varied depending on their health status. Young age and infection timing during the RSV epidemic season were significant risk factors for severe RSV infection. These findings provide a theoretical basis for optimizing immunization strategies for infants with diverse health conditions.
Collapse
Affiliation(s)
- Peiru Shi
- Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Chongqing, China
- Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Rong Wang
- Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Chongqing, China
- Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Enmei Liu
- Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Chongqing, China
| | - Yu Deng
- Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Chongqing, China
| |
Collapse
|
5
|
Losa-Martin O, Frisuelos-Garcia A, Delgado-Iribarren A, Martin-deCabo MR, Martin-Segarra O, Vegas-Serrano A, Hervas-Gomez R, Moreno-Nuñez L, Velasco-Arribas M, Losa-Garcia JE. Respiratory syncytial virus infection in adults: Differences with influenza. ENFERMEDADES INFECCIOSAS Y MICROBIOLOGIA CLINICA (ENGLISH ED.) 2024; 42:62-68. [PMID: 36624032 DOI: 10.1016/j.eimce.2022.12.013] [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: 04/19/2022] [Revised: 07/19/2022] [Accepted: 07/26/2022] [Indexed: 06/17/2023]
Abstract
INTRODUCTION Respiratory syncytial virus (RSV) causes an acute respiratory illness similar to influenza, although there are few data comparing both of them in adults. The existence of clinical differences between these two infections could have implications for their management. MATERIALS AND METHODS Retrospective observational cohort study including 63 adults with positive PCR for RSV and 221 for influenza during winter 2018-2019. Epidemiological, clinical characteristics and outcomes were contrasted between both groups. RESULTS Compared to influenza, RSV-positive patients presented a higher association with active neoplasia (OR=2.9; 95% CI: 1.2-6.9), dependence for basic activities of daily living (OR=3.4; 95% CI: 1.4-8.2) and immunosuppression due to chronic glucocorticoid administration (OR=7.6; 95% CI: 1.6-36.1). At diagnosis, fever was less common (OR=0.3; 95% CI: 0.2-0.7), and C-reactive protein level ≥100mg/l was more frequent (OR=2.1; 95% CI: 1.0-4.5). They developed bacterial co-infection by Staphylococcus aureus in a higher proportion (OR=8.3; 95% CI: 1.5-46.9) and presented a greater need for admission to the intensive care unit (OR=5.4; 95% CI: 1.4-19.2). CONCLUSION RSV is an important cause of respiratory illness in adults during the influenza season. It especially affects vulnerable patients with chronic underlying diseases, and has a higher morbidity than influenza. For all these reasons, specific detection, prevention and treatment of RSV is necessary in order to reduce the consumption of health care resources due to RSV disease in adults.
Collapse
Affiliation(s)
- Oscar Losa-Martin
- Facultad de Medicina, Universidad Rey Juan Carlos, Madrid, Spain; Cirugía Plástica Estética y Reparadora, Hospital Universitario La Paz, Madrid, Spain.
| | | | - Alberto Delgado-Iribarren
- Unidad de Enfermedades Infecciosas, Hospital Universitario Fundación Alcorcón, Alcorcón, Madrid, Spain
| | | | - Oriol Martin-Segarra
- Unidad de Enfermedades Infecciosas, Hospital Universitario Fundación Alcorcón, Alcorcón, Madrid, Spain
| | - Ana Vegas-Serrano
- Unidad de Enfermedades Infecciosas, Hospital Universitario Fundación Alcorcón, Alcorcón, Madrid, Spain
| | - Rafael Hervas-Gomez
- Unidad de Enfermedades Infecciosas, Hospital Universitario Fundación Alcorcón, Alcorcón, Madrid, Spain
| | - Leonor Moreno-Nuñez
- Unidad de Enfermedades Infecciosas, Hospital Universitario Fundación Alcorcón, Alcorcón, Madrid, Spain
| | - Maria Velasco-Arribas
- Facultad de Medicina, Universidad Rey Juan Carlos, Madrid, Spain; Unidad de Enfermedades Infecciosas, Hospital Universitario Fundación Alcorcón, Alcorcón, Madrid, Spain
| | - Juan E Losa-Garcia
- Facultad de Medicina, Universidad Rey Juan Carlos, Madrid, Spain; Unidad de Enfermedades Infecciosas, Hospital Universitario Fundación Alcorcón, Alcorcón, Madrid, Spain
| |
Collapse
|
6
|
Wang D, Deng Y, Zhou J, Wang W, Huang B, Wang W, Wei L, Ren J, Han R, Bing J, Zhai C, Guo X, Tan W. Single-Dose Intranasal Immunisation with Novel Chimeric H1N1 Expressing the Receptor-Binding Domain of SARS-CoV-2 Induces Robust Mucosal Immunity, Tissue-Resident Memory T Cells, and Heterologous Protection in Mice. Vaccines (Basel) 2023; 11:1453. [PMID: 37766130 PMCID: PMC10537001 DOI: 10.3390/vaccines11091453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 09/01/2023] [Accepted: 09/02/2023] [Indexed: 09/29/2023] Open
Abstract
Current COVID-19 vaccines can effectively reduce disease severity and hospitalisation; however, they are not considerably effective in preventing infection and transmission. In this context, mucosal vaccines are pertinent to prevent SARS-CoV-2 infection and spread. In this study, we generated a replication-competent recombinant chimeric influenza A virus (IAV) expressing the receptor-binding domain (RBD) of a SARS-CoV-2 prototype in the C-terminus of the neuraminidase (NA) of A/Puerto Rico/08/1934 H1N1 (PR8). The remaining seven segments from A/WSN/1933 H1N1 (WSN) were named PR8NARBD/WSN. We observed that the recombinant virus with the WSN backbone demonstrated improved expression of NA and RBD. A single intranasal dose of PR8NARBD/WSN(103PFU) in mice generated robust mucosal immunity, neutralising antibodies, cellular immunity, and tissue-resident memory T cells specific to SARS-CoV-2 and IAV. Importantly, immunisation with PR8NARBD/WSN viruses effectively protected mice against lethal challenges with H1N1, H3N2 IAV, and SARS-CoV-2 Beta variant and significantly reduced lung viral loads. Overall, our research demonstrates the promising potential of PR8NARBD/WSN as an attractive vaccine against emerging SARS-CoV-2 variants and influenza A virus infections.
Collapse
Affiliation(s)
- Donghong Wang
- Key Laboratory of Biosafety, National Health Commissions, National Institute for Viral Disease Control and Prevention, China CDC, 155 Changbai Road, Beijing 102206, China; (D.W.)
| | - Yao Deng
- Key Laboratory of Biosafety, National Health Commissions, National Institute for Viral Disease Control and Prevention, China CDC, 155 Changbai Road, Beijing 102206, China; (D.W.)
| | - Jianfang Zhou
- State Key Laboratory for Molecular Virology and Genetic Engineering, Chinese National Influenza Center, National Institute for Viral Disease Control and Prevention, China CDC, 155 Changbai Road, Beijing 102206, China
| | - Wen Wang
- Key Laboratory of Biosafety, National Health Commissions, National Institute for Viral Disease Control and Prevention, China CDC, 155 Changbai Road, Beijing 102206, China; (D.W.)
| | - Baoying Huang
- Key Laboratory of Biosafety, National Health Commissions, National Institute for Viral Disease Control and Prevention, China CDC, 155 Changbai Road, Beijing 102206, China; (D.W.)
| | - Wenling Wang
- Key Laboratory of Biosafety, National Health Commissions, National Institute for Viral Disease Control and Prevention, China CDC, 155 Changbai Road, Beijing 102206, China; (D.W.)
| | - Lan Wei
- Key Laboratory of Biosafety, National Health Commissions, National Institute for Viral Disease Control and Prevention, China CDC, 155 Changbai Road, Beijing 102206, China; (D.W.)
| | - Jiao Ren
- Key Laboratory of Biosafety, National Health Commissions, National Institute for Viral Disease Control and Prevention, China CDC, 155 Changbai Road, Beijing 102206, China; (D.W.)
| | - Ruiwen Han
- Key Laboratory of Biosafety, National Health Commissions, National Institute for Viral Disease Control and Prevention, China CDC, 155 Changbai Road, Beijing 102206, China; (D.W.)
- Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Jialuo Bing
- Key Laboratory of Biosafety, National Health Commissions, National Institute for Viral Disease Control and Prevention, China CDC, 155 Changbai Road, Beijing 102206, China; (D.W.)
- School of Public Health, Xinxiang Medical University, Xinxiang 453003, China
| | - Chengcheng Zhai
- Key Laboratory of Biosafety, National Health Commissions, National Institute for Viral Disease Control and Prevention, China CDC, 155 Changbai Road, Beijing 102206, China; (D.W.)
| | - Xiaoyan Guo
- Key Laboratory of Biosafety, National Health Commissions, National Institute for Viral Disease Control and Prevention, China CDC, 155 Changbai Road, Beijing 102206, China; (D.W.)
| | - Wenjie Tan
- Key Laboratory of Biosafety, National Health Commissions, National Institute for Viral Disease Control and Prevention, China CDC, 155 Changbai Road, Beijing 102206, China; (D.W.)
- Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, China
- School of Public Health, Xinxiang Medical University, Xinxiang 453003, China
| |
Collapse
|
7
|
Pulkina A, Vasilyev K, Muzhikyan A, Sergeeva M, Romanovskaya-Romanko E, Shurygina AP, Shuklina M, Vasin A, Stukova M, Egorov A. IgGκ Signal Peptide Enhances the Efficacy of an Influenza Vector Vaccine against Respiratory Syncytial Virus Infection in Mice. Int J Mol Sci 2023; 24:11445. [PMID: 37511205 PMCID: PMC10380829 DOI: 10.3390/ijms241411445] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/11/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
Intranasal vaccination using influenza vectors is a promising approach to developing vaccines against respiratory pathogens due to the activation of the mucosa-associated immune response. However, there is no clear evidence of a vector design that could be considered preferable. To find the optimal structure of an influenza vector with a modified NS genomic segment, we constructed four vector expressing identical transgene sequences inherited from the F protein of the respiratory syncytial virus (RSV). Two vectors were designed aiming at transgene accumulation in the cytosol. Another two were supplemented with an IgGκ signal peptide prior to the transgene for its extracellular delivery. Surprisingly, adding the IgGκ substantially enhanced the T-cell immune response to the CD8 epitope of the transgene. Moreover, this strategy allowed us to obtain a better protection of mice from the RSV challenge after a single intranasal immunization. Protection was achieved without antibodies, mediated by a balanced T-cell immune response including the formation of the RSV specific effector CD8+ IFNγ+/IL10+-producing cells and the accumulation of Treg cells preventing immunopathology in the lungs of infected mice. In addition to the presented method for optimizing the influenza vector, our results highlight the possibility of achieving protection against RSV through a respiratory-associated T-cell immune response alone.
Collapse
Affiliation(s)
- Anastasia Pulkina
- Smorodintsev Research Institute of Influenza of the Ministry of Health of the Russian Federation, 197022 St. Petersburg, Russia
| | - Kirill Vasilyev
- Smorodintsev Research Institute of Influenza of the Ministry of Health of the Russian Federation, 197022 St. Petersburg, Russia
| | - Arman Muzhikyan
- Smorodintsev Research Institute of Influenza of the Ministry of Health of the Russian Federation, 197022 St. Petersburg, Russia
| | - Mariia Sergeeva
- Smorodintsev Research Institute of Influenza of the Ministry of Health of the Russian Federation, 197022 St. Petersburg, Russia
| | - Ekaterina Romanovskaya-Romanko
- Smorodintsev Research Institute of Influenza of the Ministry of Health of the Russian Federation, 197022 St. Petersburg, Russia
| | - Anna-Polina Shurygina
- Smorodintsev Research Institute of Influenza of the Ministry of Health of the Russian Federation, 197022 St. Petersburg, Russia
| | - Marina Shuklina
- Smorodintsev Research Institute of Influenza of the Ministry of Health of the Russian Federation, 197022 St. Petersburg, Russia
| | - Andrey Vasin
- Smorodintsev Research Institute of Influenza of the Ministry of Health of the Russian Federation, 197022 St. Petersburg, Russia
| | - Marina Stukova
- Smorodintsev Research Institute of Influenza of the Ministry of Health of the Russian Federation, 197022 St. Petersburg, Russia
| | - Andrej Egorov
- Smorodintsev Research Institute of Influenza of the Ministry of Health of the Russian Federation, 197022 St. Petersburg, Russia
| |
Collapse
|
8
|
Du R, Cui Q, Chen Z, Zhao X, Lin X, Rong L. Revisiting influenza A virus life cycle from a perspective of genome balance. Virol Sin 2023; 38:1-8. [PMID: 36309307 PMCID: PMC10006207 DOI: 10.1016/j.virs.2022.10.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 10/18/2022] [Indexed: 11/06/2022] Open
Abstract
Influenza A virus (IAV) genome comprises eight negative-sense RNA segments, of which the replication is well orchestrated and the delicate balance of multiple segments are dynamically regulated throughout IAV life cycle. However, previous studies seldom discuss these balances except for functional hemagglutinin-neuraminidase balance that is pivotal for both virus entry and release. Therefore, we attempt to revisit IAV life cycle by highlighting the critical role of "genome balance". Moreover, we raise a "balance regression" model of IAV evolution that the virus evolves to rebalance its genome after reassortment or interspecies transmission, and direct a "balance compensation" strategy to rectify the "genome imbalance" as a result of artificial modifications during creation of recombinant IAVs. This review not only improves our understanding of IAV life cycle, but also facilitates both basic and applied research of IAV in future.
Collapse
Affiliation(s)
- Ruikun Du
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China; Qingdao Academy of Chinese Medicinal Sciences, Shandong University of Traditional Chinese Medicine, Qingdao, 266122, China.
| | - Qinghua Cui
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China; Qingdao Academy of Chinese Medicinal Sciences, Shandong University of Traditional Chinese Medicine, Qingdao, 266122, China
| | - Zinuo Chen
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Xiujuan Zhao
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Xiaojing Lin
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Lijun Rong
- Department of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, Chicago, 60612, USA.
| |
Collapse
|
9
|
Losa-Martin O, Frisuelos-Garcia A, Delgado-Iribarren A, Martin-deCabo MR, Martin-Segarra O, Vegas-Serrano A, Hervas-Gomez R, Moreno-Nuñez L, Velasco-Arribas M, Losa-Garcia JE. Infección por virus respiratorio sincitial en adultos: diferencias con la gripe. Enferm Infecc Microbiol Clin 2022. [DOI: 10.1016/j.eimc.2022.07.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
10
|
Zhao X, Lin X, Li P, Chen Z, Zhang C, Manicassamy B, Rong L, Cui Q, Du R. Expanding the tolerance of segmented Influenza A Virus genome using a balance compensation strategy. PLoS Pathog 2022; 18:e1010756. [PMID: 35926068 PMCID: PMC9380948 DOI: 10.1371/journal.ppat.1010756] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 08/16/2022] [Accepted: 07/21/2022] [Indexed: 12/17/2022] Open
Abstract
Reporter viruses provide powerful tools for both basic and applied virology studies, however, the creation and exploitation of reporter influenza A viruses (IAVs) have been hindered by the limited tolerance of the segmented genome to exogenous modifications. Interestingly, our previous study has demonstrated the underlying mechanism that foreign insertions reduce the replication/transcription capacity of the modified segment, impairing the delicate balance among the multiple segments during IAV infection. In the present study, we developed a “balance compensation” strategy by incorporating additional compensatory mutations during initial construction of recombinant IAVs to expand the tolerance of IAV genome. As a proof of concept, promoter-enhancing mutations were introduced within the modified segment to rectify the segments imbalance of a reporter influenza PR8-NS-Gluc virus, while directed optimization of the recombinant IAV was successfully achieved. Further, we generated recombinant IAVs expressing a much larger firefly luciferase (Fluc) by coupling with a much stronger compensatory enhancement, and established robust Fluc-based live-imaging mouse models of IAV infection. Our strategy feasibly expands the tolerance for foreign gene insertions in the segmented IAV genome, which opens up better opportunities to develop more versatile reporter IAVs as well as live attenuated influenza virus-based vaccines for other important human pathogens.
Collapse
Affiliation(s)
- Xiujuan Zhao
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiaojing Lin
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Ping Li
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zinuo Chen
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Chengcheng Zhang
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Balaji Manicassamy
- Department of Microbiology and Immunology, University of Iowa, Iowa, United States of America
| | - Lijun Rong
- Department of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, Chicago, United States of America
- * E-mail: (LR); (QC); (RD)
| | - Qinghua Cui
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
- Qingdao Academy of Chinese Medicinal Sciences, Shandong University of Traditional Chinese Medicine, Qingdao, China
- * E-mail: (LR); (QC); (RD)
| | - Ruikun Du
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
- Qingdao Academy of Chinese Medicinal Sciences, Shandong University of Traditional Chinese Medicine, Qingdao, China
- * E-mail: (LR); (QC); (RD)
| |
Collapse
|
11
|
Development of a T Cell-Based COVID-19 Vaccine Using a Live Attenuated Influenza Vaccine Viral Vector. Vaccines (Basel) 2022; 10:vaccines10071142. [PMID: 35891306 PMCID: PMC9318028 DOI: 10.3390/vaccines10071142] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/06/2022] [Accepted: 07/14/2022] [Indexed: 12/12/2022] Open
Abstract
The COVID-19 pandemic emerged in 2020 and has caused an unprecedented burden to all countries in the world. SARS-CoV-2 continues to circulate and antigenically evolve, enabling multiple reinfections. To address the issue of the virus antigenic variability, T cell-based vaccines are being developed, which are directed to more conserved viral epitopes. We used live attenuated influenza vaccine (LAIV) virus vector to generate recombinant influenza viruses expressing various T-cell epitopes of SARS-CoV-2 from either neuraminidase (NA) or non-structural (NS1) genes, via the P2A self-cleavage site. Intranasal immunization of human leukocyte antigen-A*0201 (HLA-A2.1) transgenic mice with these recombinant viruses did not result in significant SARS-CoV-2-specific T-cell responses, due to the immunodominance of NP366 influenza T-cell epitope. However, side-by-side stimulation of peripheral blood mononuclear cells (PBMCs) of COVID-19 convalescents with recombinant viruses and LAIV vector demonstrated activation of memory T cells in samples stimulated with LAIV/SARS-CoV-2, but not LAIV alone. Hamsters immunized with a selected LAIV/SARS-CoV-2 prototype were protected against challenge with influenza virus and a high dose of SARS-CoV-2 of Wuhan and Delta lineages, which was confirmed by reduced weight loss, milder clinical symptoms and less pronounced histopathological signs of SARS-CoV-2 infection in the lungs, compared to LAIV- and mock-immunized animals. Overall, LAIV is a promising platform for the development of a bivalent vaccine against influenza and SARS-CoV-2.
Collapse
|
12
|
Venkataraman S, Hefferon K, Makhzoum A, Abouhaidar M. Combating Human Viral Diseases: Will Plant-Based Vaccines Be the Answer? Vaccines (Basel) 2021; 9:vaccines9070761. [PMID: 34358177 PMCID: PMC8310141 DOI: 10.3390/vaccines9070761] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 07/02/2021] [Accepted: 07/04/2021] [Indexed: 12/28/2022] Open
Abstract
Molecular pharming or the technology of application of plants and plant cell culture to manufacture high-value recombinant proteins has progressed a long way over the last three decades. Whether generated in transgenic plants by stable expression or in plant virus-based transient expression systems, biopharmaceuticals have been produced to combat several human viral diseases that have impacted the world in pandemic proportions. Plants have been variously employed in expressing a host of viral antigens as well as monoclonal antibodies. Many of these biopharmaceuticals have shown great promise in animal models and several of them have performed successfully in clinical trials. The current review elaborates the strategies and successes achieved in generating plant-derived vaccines to target several virus-induced health concerns including highly communicable infectious viral diseases. Importantly, plant-made biopharmaceuticals against hepatitis B virus (HBV), hepatitis C virus (HCV), the cancer-causing virus human papillomavirus (HPV), human immunodeficiency virus (HIV), influenza virus, zika virus, and the emerging respiratory virus, severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) have been discussed. The use of plant virus-derived nanoparticles (VNPs) and virus-like particles (VLPs) in generating plant-based vaccines are extensively addressed. The review closes with a critical look at the caveats of plant-based molecular pharming and future prospects towards further advancements in this technology. The use of biopharmed viral vaccines in human medicine and as part of emergency response vaccines and therapeutics in humans looks promising for the near future.
Collapse
Affiliation(s)
- Srividhya Venkataraman
- Virology Laboratory, Department of Cell & Systems Biology, University of Toronto, Toronto, ON M5S 3B2, Canada; (K.H.); (M.A.)
- Correspondence:
| | - Kathleen Hefferon
- Virology Laboratory, Department of Cell & Systems Biology, University of Toronto, Toronto, ON M5S 3B2, Canada; (K.H.); (M.A.)
| | - Abdullah Makhzoum
- Department of Biological Sciences & Biotechnology, Botswana International University of Science & Technology, Palapye, Botswana;
| | - Mounir Abouhaidar
- Virology Laboratory, Department of Cell & Systems Biology, University of Toronto, Toronto, ON M5S 3B2, Canada; (K.H.); (M.A.)
| |
Collapse
|
13
|
Universal Live-Attenuated Influenza Vaccine Candidates Expressing Multiple M2e Epitopes Protect Ferrets against a High-Dose Heterologous Virus Challenge. Viruses 2021; 13:v13071280. [PMID: 34209093 PMCID: PMC8310119 DOI: 10.3390/v13071280] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 06/28/2021] [Accepted: 06/29/2021] [Indexed: 12/14/2022] Open
Abstract
The development of an influenza vaccine with broad protection and durability remains an attractive idea due to the high mutation rate of the influenza virus. An extracellular domain of Matrix 2 protein (M2e) is among the most attractive target for the universal influenza vaccine owing to its high conservancy rate. Here, we generated two recombinant live attenuated influenza vaccine (LAIV) candidates encoding four M2e epitopes representing consensus sequences of human, avian and swine influenza viruses, and studied them in a preclinical ferret model. Both LAIV+4M2e viruses induced higher levels of M2e-specific antibodies compared to the control LAIV strain, with the LAIV/HA+4M2e candidate being significantly more immunogenic than the LAIV/NS+4M2e counterpart. A high-dose heterosubtypic influenza virus challenge revealed the highest degree of protection after immunization with LAIV/HA+4M2e strain, followed by the NS-modified LAIV and the classical LAIV virus. Furthermore, only the immune sera from the LAIV/HA+4M2e-immunized ferrets protected mice from a panel of lethal influenza viruses encoding M genes of various origins. These data suggest that the improved cross-protection of the LAIV/HA+4M2e universal influenza vaccine candidate was mediated by the M2e-targeted antibodies. Taking into account the safety profile and improved cross-protective potential, the LAIV/HA+4M2e vaccine warrants its further evaluation in a phase I clinical trial.
Collapse
|
14
|
Kumar M, Kumari N, Thakur N, Bhatia SK, Saratale GD, Ghodake G, Mistry BM, Alavilli H, Kishor DS, Du X, Chung SM. A Comprehensive Overview on the Production of Vaccines in Plant-Based Expression Systems and the Scope of Plant Biotechnology to Combat against SARS-CoV-2 Virus Pandemics. PLANTS (BASEL, SWITZERLAND) 2021; 10:1213. [PMID: 34203729 PMCID: PMC8232254 DOI: 10.3390/plants10061213] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/28/2021] [Accepted: 06/12/2021] [Indexed: 12/23/2022]
Abstract
Many pathogenic viral pandemics have caused threats to global health; the COVID-19 pandemic is the latest. Its transmission is growing exponentially all around the globe, putting constraints on the health system worldwide. A novel coronavirus, severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), causes this pandemic. Many candidate vaccines are available at this time for COVID-19, and there is a massive international race underway to procure as many vaccines as possible for each country. However, due to heavy global demand, there are strains in global vaccine production. The use of a plant biotechnology-based expression system for vaccine production also represents one part of this international effort, which is to develop plant-based heterologous expression systems, virus-like particles (VLPs)-vaccines, antiviral drugs, and a rapid supply of antigen-antibodies for detecting kits and plant origin bioactive compounds that boost the immunity and provide tolerance to fight against the virus infection. This review will look at the plant biotechnology platform that can provide the best fight against this global pandemic.
Collapse
Affiliation(s)
- Manu Kumar
- Department of Life Science, College of Life Science and Biotechnology, Dongguk University, Seoul 10326, Korea; (M.K.); (D.S.K.); (X.D.)
| | - Nisha Kumari
- Department of Radiology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul 03080, Korea;
| | - Nishant Thakur
- Department of Hospital Pathology, Yeouido St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, 10, 63-ro, Yeongdeungpo-gu, Seoul 07345, Korea;
| | - Shashi Kant Bhatia
- Department of Biological Engineering, College of Engineering, Konkuk University, 1 Hwayang-dong, Gwangjin-gu, Seoul 05029, Korea;
| | - Ganesh Dattatraya Saratale
- Department of Food Science and Biotechnology, Dongguk University, Seoul 10326, Korea; (G.D.S.); (B.M.M.)
| | - Gajanan Ghodake
- Department of Biological and Environmental Science, Dongguk University, Seoul 10326, Korea;
| | - Bhupendra M. Mistry
- Department of Food Science and Biotechnology, Dongguk University, Seoul 10326, Korea; (G.D.S.); (B.M.M.)
| | - Hemasundar Alavilli
- Department of Biochemistry and Molecular Biology, College of Medicine, Korea University, Seoul 02841, Korea;
| | - D. S. Kishor
- Department of Life Science, College of Life Science and Biotechnology, Dongguk University, Seoul 10326, Korea; (M.K.); (D.S.K.); (X.D.)
| | - Xueshi Du
- Department of Life Science, College of Life Science and Biotechnology, Dongguk University, Seoul 10326, Korea; (M.K.); (D.S.K.); (X.D.)
| | - Sang-Min Chung
- Department of Life Science, College of Life Science and Biotechnology, Dongguk University, Seoul 10326, Korea; (M.K.); (D.S.K.); (X.D.)
| |
Collapse
|
15
|
Park BR, Kim KH, Kotomina T, Kim MC, Kwon YM, Jeeva S, Jung YJ, Bhatnagar N, Isakova-Sivak I, Mezhenskaya D, Rudenko L, Wang BZ, Kang SM. Broad cross protection by recombinant live attenuated influenza H3N2 seasonal virus expressing conserved M2 extracellular domain in a chimeric hemagglutinin. Sci Rep 2021; 11:4151. [PMID: 33603072 PMCID: PMC7893060 DOI: 10.1038/s41598-021-83704-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 02/03/2021] [Indexed: 12/24/2022] Open
Abstract
Hemagglutinin (HA)-based current vaccines provide suboptimum cross protection. Influenza A virus contains an ion channel protein M2 conserved extracellular domain (M2e), a target for developing universal vaccines. Here we generated reassortant influenza virus rgH3N2 4xM2e virus (HA and NA from A/Switzerland/9715293/2013/(H3N2)) expressing chimeric 4xM2e-HA fusion proteins with 4xM2e epitopes inserted into the H3 HA N-terminus. Recombinant rgH3N2 4xM2e virus was found to retain equivalent growth kinetics as rgH3N2 in egg substrates. Intranasal single inoculation of mice with live rgH3N2 4xM2e virus was effective in priming the induction of M2e specific IgG antibody responses in mucosal and systemic sites as well as T cell responses. The rgH3N2 4xM2e primed mice were protected against a broad range of different influenza A virus subtypes including H1N1, H3N2, H5N1, H7N9, and H9N2. The findings support a new approach to improve the efficacy of current vaccine platforms by recombinant influenza virus inducing immunity to HA and cross protective M2e antigens.
Collapse
Affiliation(s)
- Bo Ryoung Park
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA
| | - Ki-Hye Kim
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA
| | - Tatiana Kotomina
- Department of Virology, Institute of Experimental Medicine, St Petersburg, Russia
| | - Min-Chul Kim
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA
- CARESIDE Co., Ltd., Seongnam, Gyeonggi-do, Republic of Korea
| | - Young-Man Kwon
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA
| | - Subbiah Jeeva
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA
| | - Yu-Jin Jung
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA
| | - Noopur Bhatnagar
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA
| | - Irina Isakova-Sivak
- Department of Virology, Institute of Experimental Medicine, St Petersburg, Russia
| | - Daria Mezhenskaya
- Department of Virology, Institute of Experimental Medicine, St Petersburg, Russia
| | - Larisa Rudenko
- Department of Virology, Institute of Experimental Medicine, St Petersburg, Russia
| | - Bao-Zhong Wang
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA
| | - Sang-Moo Kang
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA.
| |
Collapse
|
16
|
Mezhenskaya D, Isakova-Sivak I, Kotomina T, Matyushenko V, Kim MC, Bhatnagar N, Kim KH, Kang SM, Rudenko L. A Strategy to Elicit M2e-Specific Antibodies Using a Recombinant H7N9 Live Attenuated Influenza Vaccine Expressing Multiple M2e Tandem Repeats. Biomedicines 2021; 9:biomedicines9020133. [PMID: 33535408 PMCID: PMC7912525 DOI: 10.3390/biomedicines9020133] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 01/26/2021] [Accepted: 01/27/2021] [Indexed: 12/21/2022] Open
Abstract
Influenza viruses remain a serious public health problem. Vaccination is the most effective way to prevent the disease; however, seasonal influenza vaccines demonstrate low or no effectiveness against antigenically drifted and newly emerged influenza viruses. Different strategies of eliciting immune responses against conserved parts of various influenza virus proteins are being developed worldwide. We constructed a universal live attenuated influenza vaccine (LAIV) candidate with enhanced breadth of protection by modifying H7N9 LAIV by incorporating four epitopes of M2 protein extracellular part into its hemagglutinin molecule. The new recombinant H7N9+4M2e vaccine induced anti-M2e antibody responses and demonstrated increased protection against heterosubtypic challenge viruses in direct and serum passive protection studies, compared to the classical H7N9 LAIV. The results of our study suggest that the H7N9+4M2e warrants further investigation in pre-clinical and phase 1 clinical trials.
Collapse
Affiliation(s)
- Daria Mezhenskaya
- Department of Virology, Institute of Experimental Medicine, 197376 Saint Petersburg, Russia; (D.M.); (T.K.); (V.M.); (L.R.)
| | - Irina Isakova-Sivak
- Department of Virology, Institute of Experimental Medicine, 197376 Saint Petersburg, Russia; (D.M.); (T.K.); (V.M.); (L.R.)
- Correspondence:
| | - Tatiana Kotomina
- Department of Virology, Institute of Experimental Medicine, 197376 Saint Petersburg, Russia; (D.M.); (T.K.); (V.M.); (L.R.)
| | - Victoria Matyushenko
- Department of Virology, Institute of Experimental Medicine, 197376 Saint Petersburg, Russia; (D.M.); (T.K.); (V.M.); (L.R.)
| | - Min-Chul Kim
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA; (M.-C.K.); (N.B.); (K.-H.K.); (S.-M.K.)
| | - Noopur Bhatnagar
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA; (M.-C.K.); (N.B.); (K.-H.K.); (S.-M.K.)
| | - Ki-Hye Kim
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA; (M.-C.K.); (N.B.); (K.-H.K.); (S.-M.K.)
| | - Sang-Moo Kang
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA; (M.-C.K.); (N.B.); (K.-H.K.); (S.-M.K.)
| | - Larisa Rudenko
- Department of Virology, Institute of Experimental Medicine, 197376 Saint Petersburg, Russia; (D.M.); (T.K.); (V.M.); (L.R.)
| |
Collapse
|
17
|
Jung YJ, Lee YN, Kim KH, Lee Y, Jeeva S, Park BR, Kang SM. Recombinant Live Attenuated Influenza Virus Expressing Conserved G-Protein Domain in a Chimeric Hemagglutinin Molecule Induces G-Specific Antibodies and Confers Protection against Respiratory Syncytial Virus. Vaccines (Basel) 2020; 8:vaccines8040716. [PMID: 33271920 PMCID: PMC7711863 DOI: 10.3390/vaccines8040716] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 11/04/2020] [Accepted: 11/13/2020] [Indexed: 01/13/2023] Open
Abstract
Respiratory syncytial virus (RSV) is one of the most important pathogens causing significant morbidity and mortality in infants and the elderly. Live attenuated influenza vaccine (LAIV) is a licensed vaccine platform in humans and it is known to induce broader immune responses. RSV G attachment proteins mediate virus binding to the target cells and they contain a conserved central domain with neutralizing epitopes. Here, we generated recombinant LAIV based on the attenuated A/Puerto Rico/8/1934 virus backbone, expressing an RSV conserved G-domain in a chimeric hemagglutinin (HA) fusion molecule (HA-G). The attenuated phenotypes of chimeric HA-G LAIV were evident by restricted replication in the upper respiratory tract and low temperature growth characteristics. The immunization of mice with chimeric HA-G LAIV induced significant increases in G-protein specific IgG2a (T helper type 1) and IgG antibody-secreting cell responses in lung, bronchioalveolar fluid, bone marrow, and spleens after RSV challenge. Vaccine-enhanced disease that is typically caused by inactivated-RSV vaccination was not observed in chimeric HA-G LAIV as analyzed by lung histopathology. These results in this study suggest a new approach of developing an RSV vaccine candidate while using recombinant LAIV, potentially conferring protection against influenza virus and RSV.
Collapse
Affiliation(s)
- Yu-Jin Jung
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA; (Y.-J.J.); (Y.-N.L.); (K.-H.K.); (Y.L.); (S.J.); (B.R.P.)
| | - Yu-Na Lee
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA; (Y.-J.J.); (Y.-N.L.); (K.-H.K.); (Y.L.); (S.J.); (B.R.P.)
- Animal and Plant Quarantine Agency, Gimcheon, Gyeongsangbukdo 39660, Korea
| | - Ki-Hye Kim
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA; (Y.-J.J.); (Y.-N.L.); (K.-H.K.); (Y.L.); (S.J.); (B.R.P.)
| | - Youri Lee
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA; (Y.-J.J.); (Y.-N.L.); (K.-H.K.); (Y.L.); (S.J.); (B.R.P.)
| | - Subbiah Jeeva
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA; (Y.-J.J.); (Y.-N.L.); (K.-H.K.); (Y.L.); (S.J.); (B.R.P.)
| | - Bo Ryoung Park
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA; (Y.-J.J.); (Y.-N.L.); (K.-H.K.); (Y.L.); (S.J.); (B.R.P.)
| | - Sang-Moo Kang
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA; (Y.-J.J.); (Y.-N.L.); (K.-H.K.); (Y.L.); (S.J.); (B.R.P.)
- Correspondence:
| |
Collapse
|
18
|
Hwang HS, Chang M, Kim YA. Influenza-Host Interplay and Strategies for Universal Vaccine Development. Vaccines (Basel) 2020; 8:vaccines8030548. [PMID: 32962304 PMCID: PMC7564814 DOI: 10.3390/vaccines8030548] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 09/11/2020] [Accepted: 09/18/2020] [Indexed: 12/24/2022] Open
Abstract
Influenza is an annual epidemic and an occasional pandemic caused by pathogens that are responsible for infectious respiratory disease. Humans are highly susceptible to the infection mediated by influenza A viruses (IAV). The entry of the virus is mediated by the influenza virus hemagglutinin (HA) glycoprotein that binds to the cellular sialic acid receptors and facilitates the fusion of the viral membrane with the endosomal membrane. During IAV infection, virus-derived pathogen-associated molecular patterns (PAMPs) are recognized by host intracellular specific sensors including toll-like receptors (TLRs), C-type lectin receptors, retinoic acid-inducible gene-I (RIG-I)-like receptors (RLRs), and nucleotide-binding oligomerization domain (NOD)-like receptors (NLRs) either on the cell surface or intracellularly in endosomes. Herein, we comprehensively review the current knowledge available on the entry of the influenza virus into host cells and the molecular details of the influenza virus–host interface. We also highlight certain strategies for the development of universal influenza vaccines.
Collapse
Affiliation(s)
- Hye Suk Hwang
- Alan G. MacDiarmid Energy Research Institute, Chonnam National University, Gwangju 61186, Korea;
| | - Mincheol Chang
- Alan G. MacDiarmid Energy Research Institute, Chonnam National University, Gwangju 61186, Korea;
- Department of Polymer Engineering, Graduate School, Chonnam National University, Gwangju 61186, Korea
- School of Polymer Science and Engineering, Chonnam National University, Gwangju 61186, Korea
- Correspondence: (M.C.); (Y.A.K.); Tel.: +82-62-530-1771 (M.C.); +82-62-530-1871 (Y.A.K.)
| | - Yoong Ahm Kim
- Alan G. MacDiarmid Energy Research Institute, Chonnam National University, Gwangju 61186, Korea;
- Department of Polymer Engineering, Graduate School, Chonnam National University, Gwangju 61186, Korea
- School of Polymer Science and Engineering, Chonnam National University, Gwangju 61186, Korea
- Correspondence: (M.C.); (Y.A.K.); Tel.: +82-62-530-1771 (M.C.); +82-62-530-1871 (Y.A.K.)
| |
Collapse
|
19
|
Kotomina T, Isakova-Sivak I, Stepanova E, Mezhenskaya D, Matyushenko V, Prokopenko P, Sivak K, Kiseleva I, Rudenko L. Neutralizing epitope of the Fusion Protein of Respiratory Syncytial Virus Embedded in the HA Molecule of LAIV Virus is not Sufficient to Prevent RS Virus Pulmonary Replication but Ameliorates Lung Pathology following RSV Infection in Mice. Open Microbiol J 2020. [DOI: 10.2174/1874285802014010147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Aims:
To develop experimental bivalent vaccines against influenza and RSV using a cold-adapted LAIV backbone.
Background:
Respiratory syncytial virus (RSV) is a causative agent of bronchiolitis and pneumonia in young children, elderly and immunocompromised adults. No vaccine against RSV has been licensed to date for various reasons. One of the promising platforms for designing RSV vaccine is the use of live attenuated influenza vaccine (LAIV) viruses to deliver RSV epitopes to the respiratory mucosa.
Objective:
To generate recombinant LAIV viruses encoding a neutralizing epitope of the RSV fusion protein and assess their protective potential against both influenza and RSV infections in a mouse model.
Methods:
Reverse genetics methods were used to rescue recombinant LAIV+HA/RSV viruses expressing chimeric hemagglutinins encoding the RSV-F epitope at its N-terminus using two different flexible linkers. BALB/c mice were intranasally immunized with two doses of the recombinant viruses and then challenged with the influenza virus or RSV. The LAIV viral vector and formalin-inactivated RSV (FI-RSV) were included as control vaccines. Protection was assessed by the reduction of virus pulmonary titers. In addition, RSV-induced lung pathology was evaluated by histopathology studies.
Results:
Two rescued chimeric LAIV+HA/RSV viruses were identical to the LAIV vector in terms of replication capacity in vitro and in vivo. The RSV-F neutralizing epitope was successfully expressed only if inserted into the HA molecule via G-linker, but not A-linker. Both chimeric viruses induced high influenza-specific antibody levels and fully protected mice against a lethal influenza challenge virus. However, they induced weak anti-RSV antibody responses which did not prevent RS virus replication upon challenge, and only LAIV-HA+G-RSV variant protected mice against RSV-induced lung pathology.
Conclusion:
Although the designed LAIV-RSV chimeric viruses were unable to neutralize the RS virus pulmonary replication, the LAIV-HA+G-RSV reduced RSV-induced lung pathology and can be considered a promising bivalent vaccine against influenza and RSV infections and warrants its further development.
Collapse
|
20
|
Matyushenko V, Kotomina T, Kudryavtsev I, Mezhenskaya D, Prokopenko P, Matushkina A, Sivak K, Muzhikyan A, Rudenko L, Isakova-Sivak I. Conserved T-cell epitopes of respiratory syncytial virus (RSV) delivered by recombinant live attenuated influenza vaccine viruses efficiently induce RSV-specific lung-localized memory T cells and augment influenza-specific resident memory T-cell responses. Antiviral Res 2020; 182:104864. [PMID: 32585323 PMCID: PMC7313889 DOI: 10.1016/j.antiviral.2020.104864] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 05/30/2020] [Accepted: 06/16/2020] [Indexed: 12/14/2022]
Abstract
Respiratory syncytial virus (RSV) can cause recurrent infection in people because it does not stimulate a long-lived immunological memory. There is an urgent need to develop a safe and efficacious vaccine against RSV that would induce immunological memory without causing immunopathology following natural RSV infection. We have previously generated two recombinant live attenuated influenza vaccine (LAIV) viruses that encode immunodominant T-cell epitopes of RSV M2 protein in the neuraminidase or NS1 genes. These chimeric vaccines afforded protection against influenza and RSV infection in mice, without causing pulmonary eosinophilia or inflammatory RSV disease. The current study assessed the formation of influenza-specific and RSV-specific CD4 and CD8 T-cell responses in the lungs of mice, with special attention to the lung tissue-resident memory T cell subsets (TRM). The RSV epitopes did not affect influenza-specific CD4 effector memory T cell (Tem) levels in the lungs. The majority of these cells formed by LAIV or LAIV-RSV viruses had CD69+CD103- phenotype. Both LAIV+NA/RSV and LAIV+NS/RSV recombinant viruses induced significant levels of RSV M282 epitope-specific lung-localized CD8 Tem cells expressing both CD69 and CD103 TRM markers. Surprisingly, the CD69+CD103+ influenza-specific CD8 Tem responses were augmented by the addition of RSV epitopes, possibly as a result of the local microenvironment formed by the RSV-specific memory T cells differentiating to TRM in the lungs of mice immunized with LAIV-RSV chimeric viruses. This study provides evidence that LAIV vector-based vaccination can induce robust lung-localized T-cell immunity to the inserted T-cell epitope of a foreign pathogen, without altering the immunogenicity of the viral vector itself. Two LAIV-RSV vaccine viruses induced RSV M282-specific effector memory CD8 T cells producing both IFNγ and TNFα cytokines. The inserted RSV epitopes did not affect influenza-specific CD4 Tem levels in the lungs of immunized mice. LAIV-RSV viruses induced RSV M282-specific lung-localized CD8 Tem cells expressing both CD69 and CD103 TRM markers. The magnitude of RSV M282-specific CD8 Tem responses correlates with protection against RSV-induced lung pathology. The addition of RSV epitopes into the LAIV strain augmented CD69+CD103+ influenza-specific CD8 Tem responses in the lungs.
Collapse
Affiliation(s)
- Victoria Matyushenko
- Department of Virology, Institute of Experimental Medicine, Saint Petersburg, Russia
| | - Tatiana Kotomina
- Department of Virology, Institute of Experimental Medicine, Saint Petersburg, Russia
| | - Igor Kudryavtsev
- Department of Immunology, Institute of Experimental Medicine, Saint Petersburg, Russia
| | - Daria Mezhenskaya
- Department of Virology, Institute of Experimental Medicine, Saint Petersburg, Russia
| | - Polina Prokopenko
- Department of Virology, Institute of Experimental Medicine, Saint Petersburg, Russia
| | - Anastasia Matushkina
- Department of Virology, Institute of Experimental Medicine, Saint Petersburg, Russia
| | - Konstantin Sivak
- Smorodintsev Research Institute of Influenza, Saint Petersburg, Russia
| | - Arman Muzhikyan
- Smorodintsev Research Institute of Influenza, Saint Petersburg, Russia
| | - Larisa Rudenko
- Department of Virology, Institute of Experimental Medicine, Saint Petersburg, Russia
| | - Irina Isakova-Sivak
- Department of Virology, Institute of Experimental Medicine, Saint Petersburg, Russia.
| |
Collapse
|
21
|
Prospects of and Barriers to the Development of Epitope-Based Vaccines against Human Metapneumovirus. Pathogens 2020; 9:pathogens9060481. [PMID: 32570728 PMCID: PMC7350342 DOI: 10.3390/pathogens9060481] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 06/10/2020] [Accepted: 06/16/2020] [Indexed: 12/21/2022] Open
Abstract
Human metapneumovirus (HMPV) is a major cause of respiratory illnesses in children, the elderly and immunocompromised patients. Although this pathogen was only discovered in 2001, an enormous amount of research has been conducted in order to develop safe and effective vaccines to prevent people from contracting the disease. In this review, we summarize current knowledge about the most promising experimental B- and T-cell epitopes of human metapneumovirus for the rational design of HMPV vaccines using vector delivery systems, paying special attention to the conservation of these epitopes among different lineages/genotypes of HMPV. The prospects of the successful development of an epitope-based HMPV vaccine are discussed in the context of recent findings regarding HMPV’s ability to modulate host immunity. In particular, we discuss the lack of data on experimental human CD4 T-cell epitopes for HMPV despite the role of CD4 lymphocytes in both the induction of higher neutralizing antibody titers and the establishment of CD8 memory T-cell responses. We conclude that current research should be focused on searching for human CD4 T-cell epitopes of HMPV that can help us to design a safe and cross-protective epitope-based HMPV vaccine.
Collapse
|
22
|
Isakova-Sivak I, Matyushenko V, Stepanova E, Matushkina A, Kotomina T, Mezhenskaya D, Prokopenko P, Kudryavtsev I, Kopeykin P, Sivak K, Rudenko L. Recombinant Live Attenuated Influenza Vaccine Viruses Carrying Conserved T-cell Epitopes of Human Adenoviruses Induce Functional Cytotoxic T-Cell Responses and Protect Mice against Both Infections. Vaccines (Basel) 2020; 8:E196. [PMID: 32344618 PMCID: PMC7349758 DOI: 10.3390/vaccines8020196] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 04/17/2020] [Accepted: 04/22/2020] [Indexed: 01/01/2023] Open
Abstract
Human adenoviruses (AdVs) are one of the most common causes of acute respiratory viral infections worldwide. Multiple AdV serotypes with low cross-reactivity circulate in the human population, making the development of an effective vaccine very challenging. In the current study, we designed a cross-reactive AdV vaccine based on the T-cell epitopes conserved among various AdV serotypes, which were inserted into the genome of a licensed cold-adapted live attenuated influenza vaccine (LAIV) backbone. We rescued two recombinant LAIV-AdV vaccines by inserting the selected AdV T-cell epitopes into the open reading frame of full-length NA and truncated the NS1 proteins of the H7N9 LAIV virus. We then tested the bivalent vaccines for their efficacy against influenza and human AdV5 in a mouse model. The vaccine viruses were attenuated in C57BL/6J mice and induced a strong influenza-specific antibody and cell-mediated immunity, fully protecting the mice against virulent influenza virus infection. The CD8 T-cell responses induced by both LAIV-AdV candidates were functional and efficiently killed the target cells loaded either with influenza NP366 or AdV DBP418 peptides. In addition, high levels of recall memory T cells targeted to an immunodominant H2b-restricted CD8 T-cell epitope were detected in the immunized mice after the AdV5 challenge, and the magnitude of these responses correlated with the level of protection against pulmonary pathology caused by the AdV5 infection. Our findings suggest that the developed recombinant vaccines can be used for combined protection against influenza and human adenoviruses and warrant further evaluation on humanized animal models and subsequent human trials.
Collapse
Affiliation(s)
- Irina Isakova-Sivak
- Institute of Experimental Medicine, 197376 Saint Petersburg, Russia; (V.M.); (E.S.); (A.M.); (T.K.); (D.M.); (P.P.); (I.K.); (P.K.); (L.R.)
| | - Victoria Matyushenko
- Institute of Experimental Medicine, 197376 Saint Petersburg, Russia; (V.M.); (E.S.); (A.M.); (T.K.); (D.M.); (P.P.); (I.K.); (P.K.); (L.R.)
| | - Ekaterina Stepanova
- Institute of Experimental Medicine, 197376 Saint Petersburg, Russia; (V.M.); (E.S.); (A.M.); (T.K.); (D.M.); (P.P.); (I.K.); (P.K.); (L.R.)
| | - Anastasia Matushkina
- Institute of Experimental Medicine, 197376 Saint Petersburg, Russia; (V.M.); (E.S.); (A.M.); (T.K.); (D.M.); (P.P.); (I.K.); (P.K.); (L.R.)
| | - Tatiana Kotomina
- Institute of Experimental Medicine, 197376 Saint Petersburg, Russia; (V.M.); (E.S.); (A.M.); (T.K.); (D.M.); (P.P.); (I.K.); (P.K.); (L.R.)
| | - Daria Mezhenskaya
- Institute of Experimental Medicine, 197376 Saint Petersburg, Russia; (V.M.); (E.S.); (A.M.); (T.K.); (D.M.); (P.P.); (I.K.); (P.K.); (L.R.)
| | - Polina Prokopenko
- Institute of Experimental Medicine, 197376 Saint Petersburg, Russia; (V.M.); (E.S.); (A.M.); (T.K.); (D.M.); (P.P.); (I.K.); (P.K.); (L.R.)
| | - Igor Kudryavtsev
- Institute of Experimental Medicine, 197376 Saint Petersburg, Russia; (V.M.); (E.S.); (A.M.); (T.K.); (D.M.); (P.P.); (I.K.); (P.K.); (L.R.)
| | - Pavel Kopeykin
- Institute of Experimental Medicine, 197376 Saint Petersburg, Russia; (V.M.); (E.S.); (A.M.); (T.K.); (D.M.); (P.P.); (I.K.); (P.K.); (L.R.)
| | - Konstantin Sivak
- Smorodintsev Research Institute of Influenza, 197376 Saint Petersburg, Russia;
| | - Larisa Rudenko
- Institute of Experimental Medicine, 197376 Saint Petersburg, Russia; (V.M.); (E.S.); (A.M.); (T.K.); (D.M.); (P.P.); (I.K.); (P.K.); (L.R.)
| |
Collapse
|
23
|
Rosales-Mendoza S, Márquez-Escobar VA, González-Ortega O, Nieto-Gómez R, Arévalo-Villalobos JI. What Does Plant-Based Vaccine Technology Offer to the Fight against COVID-19? Vaccines (Basel) 2020; 8:E183. [PMID: 32295153 PMCID: PMC7349371 DOI: 10.3390/vaccines8020183] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 04/08/2020] [Accepted: 04/09/2020] [Indexed: 12/28/2022] Open
Abstract
The emergence of new pathogenic viral strains is a constant threat to global health, with the new coronavirus strain COVID-19 as the latest example. COVID-19, caused by the SARS-CoV-2 virus has quickly spread around the globe. This pandemic demands rapid development of drugs and vaccines. Plant-based vaccines are a technology with proven viability, which have led to promising results for candidates evaluated at the clinical level, meaning this technology could contribute towards the fight against COVID-19. Herein, a perspective in how plant-based vaccines can be developed against COVID-19 is presented. Injectable vaccines could be generated by using transient expression systems, which offer the highest protein yields and are already adopted at the industrial level to produce VLPs-vaccines and other biopharmaceuticals under GMPC-processes. Stably-transformed plants are another option, but this approach requires more time for the development of antigen-producing lines. Nonetheless, this approach offers the possibility of developing oral vaccines in which the plant cell could act as the antigen delivery agent. Therefore, this is the most attractive approach in terms of cost, easy delivery, and mucosal immunity induction. The development of multiepitope, rationally-designed vaccines is also discussed regarding the experience gained in expression of chimeric immunogenic proteins in plant systems.
Collapse
Affiliation(s)
- Sergio Rosales-Mendoza
- Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, Av. Dr. Manuel Nava 6, San Luis Potosí 78210, Mexico; (V.A.M.-E.); (O.G.-O.); (R.N.-G.); (J.I.A.-V.)
- Sección de Biotecnología, Centro de Investigación en Ciencias de la Salud y Biomedicina, Universidad Autónoma de San Luis Potosí, Av. Sierra Leona 550, Lomas 2ª Sección, San Luis Potosí 78210, Mexico
| | - Verónica A. Márquez-Escobar
- Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, Av. Dr. Manuel Nava 6, San Luis Potosí 78210, Mexico; (V.A.M.-E.); (O.G.-O.); (R.N.-G.); (J.I.A.-V.)
- Sección de Biotecnología, Centro de Investigación en Ciencias de la Salud y Biomedicina, Universidad Autónoma de San Luis Potosí, Av. Sierra Leona 550, Lomas 2ª Sección, San Luis Potosí 78210, Mexico
| | - Omar González-Ortega
- Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, Av. Dr. Manuel Nava 6, San Luis Potosí 78210, Mexico; (V.A.M.-E.); (O.G.-O.); (R.N.-G.); (J.I.A.-V.)
| | - Ricardo Nieto-Gómez
- Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, Av. Dr. Manuel Nava 6, San Luis Potosí 78210, Mexico; (V.A.M.-E.); (O.G.-O.); (R.N.-G.); (J.I.A.-V.)
- Sección de Biotecnología, Centro de Investigación en Ciencias de la Salud y Biomedicina, Universidad Autónoma de San Luis Potosí, Av. Sierra Leona 550, Lomas 2ª Sección, San Luis Potosí 78210, Mexico
| | - Jaime I. Arévalo-Villalobos
- Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, Av. Dr. Manuel Nava 6, San Luis Potosí 78210, Mexico; (V.A.M.-E.); (O.G.-O.); (R.N.-G.); (J.I.A.-V.)
- Sección de Biotecnología, Centro de Investigación en Ciencias de la Salud y Biomedicina, Universidad Autónoma de San Luis Potosí, Av. Sierra Leona 550, Lomas 2ª Sección, San Luis Potosí 78210, Mexico
| |
Collapse
|