1
|
Stepanova E, Isakova-Sivak I, Matyushenko V, Mezhenskaya D, Kudryavtsev I, Kostromitina A, Chistiakova A, Rak A, Bazhenova E, Prokopenko P, Kotomina T, Donina S, Novitskaya V, Sivak K, Karal-Ogly D, Rudenko L. Safety and Immunogenicity Study of a Bivalent Vaccine for Combined Prophylaxis of COVID-19 and Influenza in Non-Human Primates. Vaccines (Basel) 2024; 12:1099. [PMID: 39460266 PMCID: PMC11511058 DOI: 10.3390/vaccines12101099] [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: 08/19/2024] [Revised: 09/23/2024] [Accepted: 09/24/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND Influenza and SARS-CoV-2 viruses are two highly variable pathogens. We have developed a candidate bivalent live vaccine based on the strain of licensed A/Leningrad/17-based cold-adapted live attenuated influenza vaccine (LAIV) of H3N2 subtype, which expressed SARS-CoV-2 immunogenic T-cell epitopes. A cassette encoding fragments of S and N proteins of SARS-CoV-2 was inserted into the influenza NA gene using the P2A autocleavage site. In this study, we present the results of preclinical evaluation of the developed bivalent vaccine in a non-human primate model. METHODS Rhesus macaques (Macaca mulatta) (n = 3 per group) were immunized intranasally with 7.5 lg EID50 of the LAIV/CoV-2 bivalent vaccine, a control non-modified H3N2 LAIV or a placebo (chorioallantoic fluid) using a sprayer device, twice, with a 28-day interval. The blood samples were collected at days 0, 3, 28 and 35 for hematological and biochemical assessment. Safety was also assessed by monitoring body weight, body temperature and clinical signs of the disease. Immune responses to influenza virus were assessed both by determining serum antibody titers in hemagglutination inhibition assay, microneutralization assay and IgG ELISA. T-cell responses were measured both to influenza and SARS-CoV-2 antigens using ELISPOT and flow cytometry. Three weeks after the second immunization, animals were challenged with 105 PFU of Delta SARS-CoV-2. The body temperature, weight and challenge virus shedding were monitored for 5 days post-challenge. In addition, virus titers in various organs and histopathology were evaluated on day 6 after SARS-CoV-2 infection. RESULTS There was no toxic effect of the immunizations on the hematological and coagulation hemostasis of animals. No difference in the dynamics of the average weight and thermometry results were found between the groups of animals. Both LAIV and LAIV/CoV-2 variants poorly replicated in the upper respiratory tract of rhesus macaques. Nevertheless, despite this low level of virus shedding, influenza-specific serum IgG responses were detected in the group of monkeys immunized with the LAIV/CoV-2 bivalent but not in the LAIV group. Furthermore, T-cell responses to both influenza and SARS-CoV-2 viruses were detected in the LAIV/CoV-2 vaccine group only. The animals were generally resistant to SARS-CoV-2 challenge, with minimal virus shedding in the placebo and LAIV groups. Histopathological changes in vaccinated animals were decreased compared to the PBS group, suggesting a protective effect of the chimeric vaccine candidate. CONCLUSIONS The candidate bivalent vaccine was safe and immunogenic for non-human primates and warrants its further evaluation in clinical trials.
Collapse
Affiliation(s)
- Ekaterina Stepanova
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Irina Isakova-Sivak
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Victoria Matyushenko
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Daria Mezhenskaya
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Igor Kudryavtsev
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Arina Kostromitina
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Anna Chistiakova
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Alexandra Rak
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Ekaterina Bazhenova
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Polina Prokopenko
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Tatiana Kotomina
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Svetlana Donina
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Vlada Novitskaya
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Konstantin Sivak
- Smorodintsev Research Institute of Influenza, Saint-Petersburg 197376, Russia;
| | - Dzhina Karal-Ogly
- Center of Preclinical Research, Research Institute of Medical Primatology, Sochi 354376, Russia;
| | - Larisa Rudenko
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| |
Collapse
|
2
|
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
|
3
|
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
|
4
|
Stepanova E, Isakova-Sivak I, Mezhenskaya D, Niskanen S, Matyushenko V, Bazhenova E, Rak A, Wong PF, Prokopenko P, Kotomina T, Krutikova E, Legotskiy S, Neterebskii B, Ostroukhova T, Sivak K, Orshanskaya Y, Yakovlev K, Rudenko L. Expression of the SARS-CoV-2 receptor-binding domain by live attenuated influenza vaccine virus as a strategy for designing a bivalent vaccine against COVID-19 and influenza. Virol J 2024; 21:82. [PMID: 38589848 PMCID: PMC11003101 DOI: 10.1186/s12985-024-02350-w] [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: 01/09/2024] [Accepted: 03/22/2024] [Indexed: 04/10/2024] Open
Abstract
Influenza and SARS-CoV-2 are two major respiratory pathogens that cocirculate in humans and cause serious illness with the potential to exacerbate disease in the event of co-infection. To develop a bivalent vaccine, capable of protecting against both infections, we inserted the receptor-binding domain (RBD) of the SARS-CoV-2 spike protein into hemagglutinin (HA) molecule or into the open reading frame of the truncated nonstructural protein 1 (NS1) of live attenuated influenza vaccine (LAIV) virus and assessed phenotypic characteristics of the rescued LAIV-RBD viruses, as well as their immunogenicity in mouse and Syrian hamster animal models. A panel of 9 recombinant LAIV-RBD viruses was rescued using the A/Leningrad/17 backbone. Notably, only two variants with RBD insertions into the HA molecule could express sufficient quantities of RBD protein in infected MDCK cells. Intranasal immunization of mice induced high levels of anti-influenza antibody responses in all chimeric LAIV-RBD viruses, which was comparable to the LAIV virus vector. The RBD-specific antibody responses were most pronounced in the variant expressing RBD194 fragment as a chimeric HA protein. This candidate was further tested in Syrian hamsters and was shown to be immunogenic and capable of protecting animals against both infections.
Collapse
Affiliation(s)
| | | | - Daria Mezhenskaya
- Institute of Experimental Medicine, Saint Petersburg, 197022, Russia
| | - Sergei Niskanen
- Joint-Stock Company «BIOCAD» (JSC «BIOCAD») Saint Petersburg, Intracity Municipality the Settlement of Strelna, the Settlement of Strelna, ul. Svyazi, d. 38, str. 1, pomeshch. 89, Saint Petersburg, 198515, Russia
| | | | | | - Alexandra Rak
- Institute of Experimental Medicine, Saint Petersburg, 197022, Russia
| | - Pei Fong Wong
- Institute of Experimental Medicine, Saint Petersburg, 197022, Russia
| | - Polina Prokopenko
- Institute of Experimental Medicine, Saint Petersburg, 197022, Russia
| | - Tatiana Kotomina
- Institute of Experimental Medicine, Saint Petersburg, 197022, Russia
| | - Elena Krutikova
- Institute of Experimental Medicine, Saint Petersburg, 197022, Russia
| | - Sergei Legotskiy
- Joint-Stock Company «BIOCAD» (JSC «BIOCAD») Saint Petersburg, Intracity Municipality the Settlement of Strelna, the Settlement of Strelna, ul. Svyazi, d. 38, str. 1, pomeshch. 89, Saint Petersburg, 198515, Russia
| | - Bogdan Neterebskii
- Joint-Stock Company «BIOCAD» (JSC «BIOCAD») Saint Petersburg, Intracity Municipality the Settlement of Strelna, the Settlement of Strelna, ul. Svyazi, d. 38, str. 1, pomeshch. 89, Saint Petersburg, 198515, Russia
| | - Tatiana Ostroukhova
- Joint-Stock Company «BIOCAD» (JSC «BIOCAD») Saint Petersburg, Intracity Municipality the Settlement of Strelna, the Settlement of Strelna, ul. Svyazi, d. 38, str. 1, pomeshch. 89, Saint Petersburg, 198515, Russia
| | - Konstantin Sivak
- Smorodintsev Research Institute of Influenza, Saint Petersburg, 197376, Russia
| | - Yana Orshanskaya
- Smorodintsev Research Institute of Influenza, Saint Petersburg, 197376, Russia
| | - Kirill Yakovlev
- Smorodintsev Research Institute of Influenza, Saint Petersburg, 197376, Russia
| | - Larisa Rudenko
- Institute of Experimental Medicine, Saint Petersburg, 197022, Russia
| |
Collapse
|
5
|
Raman SNT, Zetner A, Hashem AM, Patel D, Wu J, Gravel C, Gao J, Zhang W, Pfeifle A, Tamming L, Parikh K, Cao J, Tam R, Safronetz D, Chen W, Johnston MJ, Wang L, Sauve S, Rosu-Myles M, Domselaar GV, Li X. Bivalent vaccines effectively protect mice against influenza A and respiratory syncytial viruses. Emerg Microbes Infect 2023; 12:2192821. [PMID: 36927227 PMCID: PMC10171128 DOI: 10.1080/22221751.2023.2192821] [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: 01/19/2023] [Accepted: 03/14/2023] [Indexed: 03/17/2023]
Abstract
Influenza and Respiratory Syncytial virus (RSV) infections together contribute significantly to the burden of acute lower respiratory tract infections. Despite the disease burden, no approved RSV vaccine is available. While approved vaccines are available for influenza, seasonal vaccination is required to maintain protection. In addition to both being respiratory viruses, they follow a common seasonality, which warrants the necessity for a concerted vaccination approach. Here, we designed bivalent vaccines by utilizing highly conserved sequences, targeting both influenza A and RSV, as either a chimeric antigen or individual antigens separated by a ribosome skipping sequence. These vaccines were found to be effective in protecting the animals from challenge by either virus, with mechanisms of protection being substantially interrogated in this communication.
Collapse
Affiliation(s)
- Sathya N. Thulasi Raman
- Centre for Oncology and Regulatory Research, Biologic and Radiopharmaceutical Drugs Directorate, Health Products and Food Branch, Health Canada and WHO Collaborating Center for Standardization and Evaluation of Biologicals, Ottawa, Canada
| | - Adrian Zetner
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Canada
| | - Anwar M. Hashem
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Microbiology and Parasitology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Devina Patel
- Centre for Oncology and Regulatory Research, Biologic and Radiopharmaceutical Drugs Directorate, Health Products and Food Branch, Health Canada and WHO Collaborating Center for Standardization and Evaluation of Biologicals, Ottawa, Canada
| | - Jianguo Wu
- Centre for Oncology and Regulatory Research, Biologic and Radiopharmaceutical Drugs Directorate, Health Products and Food Branch, Health Canada and WHO Collaborating Center for Standardization and Evaluation of Biologicals, Ottawa, Canada
| | - Caroline Gravel
- Centre for Oncology and Regulatory Research, Biologic and Radiopharmaceutical Drugs Directorate, Health Products and Food Branch, Health Canada and WHO Collaborating Center for Standardization and Evaluation of Biologicals, Ottawa, Canada
| | - Jun Gao
- Centre for Vaccines Clinical Trials and Biostatistics, Biologic and Radiopharmaceutical Drugs Directorate, Health Products and Food Branch, Health Canada, Ottawa, Canada
| | - Wanyue Zhang
- Centre for Oncology and Regulatory Research, Biologic and Radiopharmaceutical Drugs Directorate, Health Products and Food Branch, Health Canada and WHO Collaborating Center for Standardization and Evaluation of Biologicals, Ottawa, Canada
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Annabelle Pfeifle
- Centre for Oncology and Regulatory Research, Biologic and Radiopharmaceutical Drugs Directorate, Health Products and Food Branch, Health Canada and WHO Collaborating Center for Standardization and Evaluation of Biologicals, Ottawa, Canada
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Levi Tamming
- Centre for Oncology and Regulatory Research, Biologic and Radiopharmaceutical Drugs Directorate, Health Products and Food Branch, Health Canada and WHO Collaborating Center for Standardization and Evaluation of Biologicals, Ottawa, Canada
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Karan Parikh
- Centre for Oncology and Regulatory Research, Biologic and Radiopharmaceutical Drugs Directorate, Health Products and Food Branch, Health Canada and WHO Collaborating Center for Standardization and Evaluation of Biologicals, Ottawa, Canada
| | - Jingxin Cao
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Canada
| | - Roger Tam
- Centre for Oncology and Regulatory Research, Biologic and Radiopharmaceutical Drugs Directorate, Health Products and Food Branch, Health Canada and WHO Collaborating Center for Standardization and Evaluation of Biologicals, Ottawa, Canada
| | - David Safronetz
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Canada
| | - Wangxue Chen
- Human Health Therapeutics Research Center, National Research Council of Canada, Ottawa, Canada
| | - Michael J.W. Johnston
- Centre for Oncology and Regulatory Research, Biologic and Radiopharmaceutical Drugs Directorate, Health Products and Food Branch, Health Canada and WHO Collaborating Center for Standardization and Evaluation of Biologicals, Ottawa, Canada
- Department of Chemistry, Carleton University, Ottawa, Canada
| | - Lisheng Wang
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Simon Sauve
- Centre for Oncology and Regulatory Research, Biologic and Radiopharmaceutical Drugs Directorate, Health Products and Food Branch, Health Canada and WHO Collaborating Center for Standardization and Evaluation of Biologicals, Ottawa, Canada
| | - Michael Rosu-Myles
- Centre for Oncology and Regulatory Research, Biologic and Radiopharmaceutical Drugs Directorate, Health Products and Food Branch, Health Canada and WHO Collaborating Center for Standardization and Evaluation of Biologicals, Ottawa, Canada
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Gary Van Domselaar
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Canada
| | - Xuguang Li
- Centre for Oncology and Regulatory Research, Biologic and Radiopharmaceutical Drugs Directorate, Health Products and Food Branch, Health Canada and WHO Collaborating Center for Standardization and Evaluation of Biologicals, Ottawa, Canada
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Canada
| |
Collapse
|
6
|
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: 1.0] [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
|
7
|
Isticato R. Bacterial Spore-Based Delivery System: 20 Years of a Versatile Approach for Innovative Vaccines. Biomolecules 2023; 13:947. [PMID: 37371527 DOI: 10.3390/biom13060947] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 05/25/2023] [Accepted: 06/03/2023] [Indexed: 06/29/2023] Open
Abstract
Mucosal vaccines offer several advantages over injectable conventional vaccines, such as the induction of adaptive immunity, with secretory IgA production at the entry site of most pathogens, and needle-less vaccinations. Despite their potential, only a few mucosal vaccines are currently used. Developing new effective mucosal vaccines strongly relies on identifying innovative antigens, efficient adjuvants, and delivery systems. Several approaches based on phages, bacteria, or nanoparticles have been proposed to deliver antigens to mucosal surfaces. Bacterial spores have also been considered antigen vehicles, and various antigens have been successfully exposed on their surface. Due to their peculiar structure, spores conjugate the advantages of live microorganisms with synthetic nanoparticles. When mucosally administered, spores expressing antigens have been shown to induce antigen-specific, protective immune responses. This review accounts for recent progress in the formulation of spore-based mucosal vaccines, describing a spore's structure, specifically the spore surface, and the diverse approaches developed to improve its efficiency as a vehicle for heterologous antigen presentation.
Collapse
Affiliation(s)
- Rachele Isticato
- Department of Biology, University of Naples Federico II, Complesso Universitario Monte S. Angelo, Via Cinthia 4, 80126 Naples, Italy
- Interuniversity Center for Studies on Bioinspired Agro-Environmental Technology (BAT Center), 80055 Naples, Italy
| |
Collapse
|
8
|
Wang SY, Liu WQ, Li YQ, Li JX, Zhu FC. A China-developed adenovirus vector-based COVID-19 vaccine: review of the development and application of Ad5-nCov. Expert Rev Vaccines 2023; 22:704-713. [PMID: 37501516 DOI: 10.1080/14760584.2023.2242528] [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: 01/21/2023] [Accepted: 07/26/2023] [Indexed: 07/29/2023]
Abstract
INTRODUCTION The global spread of COVID-19 has prompted the development of vaccines. A recombinant adenovirus type-5 vectored COVID-19 vaccine (Ad5-nCoV) developed by Chinese scientists has been authorized for use as a prime and booster dose in China and several other countries. AREAS COVERED We searched published articles as of 4 May 2023, on PubMed using keywords related to Adenovirus vector, vaccine, and SARS-CoV-2. We reported the progress and outcomes of Ad5-nCov, including vaccine efficacy, safety, immunogenicity based on pre-clinical trials, clinical trials, and real-world studies for primary and booster doses. EXPERT OPINION Ad5-nCoV is a significant advancement in Chinese vaccine development technology. Evidence from clinical trials and real-world studies has demonstrated well-tolerated, highly immunogenic, and efficacy of Ad5-nCoV in preventing severe/critical COVID-19. Aerosolized Ad5-nCoV, given via a novel route, could elicit mucosal immunity and improve the vaccine efficacy, enhance the production capacity and availability, and reduce the potential negative impact of preexisting antibodies. However, additional research is necessary to evaluate the long-term safety and immunogenicity of Ad5-nCoV, its efficacy against emerging variants, its effectiveness in a real-world context of hybrid immunity, and its cost-effectiveness, particularly with respect to aerosolized Ad5-nCoV.
Collapse
Affiliation(s)
- Shen-Yu Wang
- Department of Immunization Programe, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, China
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Wen-Qing Liu
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yu-Qing Li
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jing-Xin Li
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
- NHC Key Laboratory of Enteric Pathogenic Microbiology (Jiangsu Provincial Center for Disease Control and Prevention), Nanjing, China
- Institute of Global Public Health and Emergency Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Feng-Cai Zhu
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
- NHC Key Laboratory of Enteric Pathogenic Microbiology (Jiangsu Provincial Center for Disease Control and Prevention), Nanjing, China
- Institute of Global Public Health and Emergency Pharmacy, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
9
|
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: 4.5] [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
|
10
|
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: 3.5] [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
|
11
|
Zhang R, Hung IFN. Approaches in broadening the neutralizing antibody response of the influenza vaccine. Expert Rev Vaccines 2021; 20:1539-1547. [PMID: 34549677 DOI: 10.1080/14760584.2021.1984887] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
INTRODUCTION Influenza vaccine is the mainstay for influenza prevention and elicits immune response and antigen-specific neutralizing antibodies against influenza virus. However, antigenic drift and shift can confer influenza virus to escape from the immune response induced by vaccine, and then reduce the vaccine effectiveness. AREAS COVERED To improve effect and neutralizing antibody response of vaccine for heterologous influenza virus, a literature review of preclinical and clinical studies published before August 2021 and searched in PubMed, which evaluated vaccine effectiveness improved by adjuvants and administration route. EXPERT OPINION The review showed that adjuvant, including imiquimod, GLA, MF59, and AS03, can improve the effectiveness of influenza vaccines by regulating immune system. Subjects receiving influenza vaccine combined with these adjuvants showed enhanced antibody response against homologous and heterologous virus strains compared to those vaccinated without adjuvant. This review also discussed the role of intradermal vaccination. In contrast to intramuscular vaccination, intradermal vaccination elicited a robust and prolonged antibody response against vaccine strains and drifted virus than intramuscular vaccination.
Collapse
Affiliation(s)
- Ruiqi Zhang
- Department of Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Ivan Fan-Ngai Hung
- Department of Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| |
Collapse
|
12
|
Oh N, Park S, Kim JW, Park JH. Photothermal Transfection for Effective Nonviral Genome Editing. ACS APPLIED BIO MATERIALS 2021; 4:5678-5685. [PMID: 35006736 DOI: 10.1021/acsabm.1c00465] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The efficient nonviral delivery of nucleic acids into the cytoplasm is needed to fully realize the potential of gene therapy. Although cationic lipids and nanoparticles have been widely used to improve the intracellular delivery of nucleic acids, they suffer from cytotoxicity and poor endosomal escape, thus limiting the transfection efficacy. Here, we developed a photothermal transfection platform for efficient and biosafe intracellular delivery of nucleic acids. Photothermal transfection was carried out by irradiation of cells co-treated with Lipofectamine-plasmid DNA complexes and PEGylated gold nanorods (GNRs) using an NIR laser for 30 min and subsequent incubation of the cells for 30 min without laser irradiation. Compared to conventional Lipofectamine-based transfection, our photothermal transfection platform significantly improved the transfection efficiency in difficult-to-transfect human primary cells including human dermal fibroblasts while maintaining the cell viability. The photothermal heating did not leave the GNRs inside the cell, thereby minimizing the cellular damage. Furthermore, the photothermal transfection platform showed superior genome editing abilities (both gene cleavage and insertion) in human dermal fibroblasts than conventional Lipofectamine-based transfection.
Collapse
Affiliation(s)
- Nuri Oh
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Sooyeon Park
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Jin Woo Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Ji-Ho Park
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea.,KAIST Institute for Health Science and Technology, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| |
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: 1.7] [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
|
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: 1.3] [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
|
15
|
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
|
16
|
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: 4.0] [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
|
17
|
Lemoine C, Thakur A, Krajišnik D, Guyon R, Longet S, Razim A, Górska S, Pantelić I, Ilić T, Nikolić I, Lavelle EC, Gamian A, Savić S, Milicic A. Technological Approaches for Improving Vaccination Compliance and Coverage. Vaccines (Basel) 2020; 8:E304. [PMID: 32560088 PMCID: PMC7350210 DOI: 10.3390/vaccines8020304] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 06/13/2020] [Accepted: 06/14/2020] [Indexed: 12/18/2022] Open
Abstract
Vaccination has been well recognised as a critically important tool in preventing infectious disease, yet incomplete immunisation coverage remains a major obstacle to achieving disease control and eradication. As medical products for global access, vaccines need to be safe, effective and inexpensive. In line with these goals, continuous improvements of vaccine delivery strategies are necessary to achieve the full potential of immunisation. Novel technologies related to vaccine delivery and route of administration, use of advanced adjuvants and controlled antigen release (single-dose immunisation) approaches are expected to contribute to improved coverage and patient compliance. This review discusses the application of micro- and nano-technologies in the alternative routes of vaccine administration (mucosal and cutaneous vaccination), oral vaccine delivery as well as vaccine encapsulation with the aim of controlled antigen release for single-dose vaccination.
Collapse
Affiliation(s)
- Céline Lemoine
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Rue Michel-Servet 1, 1221 Geneva, Switzerland;
- Vaccine Formulation Institute, Chemin des Aulx 14, 1228 Plan-les-Ouates, Switzerland
| | - Aneesh Thakur
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen Ø, Denmark;
| | - Danina Krajišnik
- Department of Pharmaceutical Technology and Cosmetology, University of Belgrade-Faculty of Pharmacy, Vojvode Stepe 450, 11221 Belgrade, Serbia; (D.K.); (I.P.); (T.I.); (I.N.); (S.S.)
| | - Romain Guyon
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, UK;
| | - Stephanie Longet
- Virology & Pathogenesis Group, Public Health England, Manor Farm Road, Porton Down, Salisbury SP4 0JG, UK;
| | - Agnieszka Razim
- Department of Microbiology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, ul. Rudolfa Weigla 12, 53-114 Wroclaw, Poland; (A.R.); (S.G.)
| | - Sabina Górska
- Department of Microbiology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, ul. Rudolfa Weigla 12, 53-114 Wroclaw, Poland; (A.R.); (S.G.)
| | - Ivana Pantelić
- Department of Pharmaceutical Technology and Cosmetology, University of Belgrade-Faculty of Pharmacy, Vojvode Stepe 450, 11221 Belgrade, Serbia; (D.K.); (I.P.); (T.I.); (I.N.); (S.S.)
| | - Tanja Ilić
- Department of Pharmaceutical Technology and Cosmetology, University of Belgrade-Faculty of Pharmacy, Vojvode Stepe 450, 11221 Belgrade, Serbia; (D.K.); (I.P.); (T.I.); (I.N.); (S.S.)
| | - Ines Nikolić
- Department of Pharmaceutical Technology and Cosmetology, University of Belgrade-Faculty of Pharmacy, Vojvode Stepe 450, 11221 Belgrade, Serbia; (D.K.); (I.P.); (T.I.); (I.N.); (S.S.)
| | - Ed C. Lavelle
- The Adjuvant Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, DO2R590 Dublin, Ireland;
| | - Andrzej Gamian
- Department of Immunology of Infectious Diseases, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, ul. Rudolfa Weigla 12, 53-114 Wroclaw, Poland;
| | - Snežana Savić
- Department of Pharmaceutical Technology and Cosmetology, University of Belgrade-Faculty of Pharmacy, Vojvode Stepe 450, 11221 Belgrade, Serbia; (D.K.); (I.P.); (T.I.); (I.N.); (S.S.)
| | - Anita Milicic
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, UK;
| |
Collapse
|
18
|
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: 2.3] [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
|
19
|
A Recombinant Influenza A/H1N1 Carrying A Short Immunogenic Peptide of MERS-CoV as Bivalent Vaccine in BALB/c Mice. Pathogens 2019; 8:pathogens8040281. [PMID: 31810359 PMCID: PMC6963271 DOI: 10.3390/pathogens8040281] [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: 10/22/2019] [Revised: 11/21/2019] [Accepted: 11/30/2019] [Indexed: 01/26/2023] Open
Abstract
Middle East Respiratory Syndrome Coronavirus (MERS-CoV) became a global human health threat since its first documentation in humans in 2012. An efficient vaccine for the prophylaxis of humans in hotspots of the infection (e.g., Saudi Arabia) is necessary but no commercial vaccines are yet approved. In this study, a chimeric DNA construct was designed to encode an influenza A/H1N1 NA protein which is flanking immunogenic amino acids (aa) 736–761 of MERS-CoV spike protein. Using the generated chimeric construct, a novel recombinant vaccine strain against pandemic influenza A virus (H1N1pdm09) and MERS-CoV was generated (chimeric bivalent 5 + 3). The chimeric bivalent 5 + 3 vaccine strain comprises a recombinant PR8-based vaccine, expressing the PB1, HA, and chimeric NA of pandemic 2009 H1N1. Interestingly, an increase in replication efficiency of the generated vaccine strain was observed when compared to the PR8-based 5 + 3 H1N1pdm09 vaccine strain that lacks the MERS-CoV spike peptide insert. In BALB/c mice, the inactivated chimeric bivalent vaccine induced potent and specific neutralizing antibodies against MERS-CoV and H1N1pdm09. This novel approach succeeded in developing a recombinant influenza virus with potential use as a bivalent vaccine against H1N1pdm09 and MERS-CoV. This approach provides a basis for the future development of chimeric influenza-based vaccines against MERS-CoV and other viruses.
Collapse
|
20
|
Kotomina T, Isakova-Sivak I, Matyushenko V, Kim KH, Lee Y, Jung YJ, Kang SM, Rudenko L. Recombinant live attenuated influenza vaccine viruses carrying CD8 T-cell epitopes of respiratory syncytial virus protect mice against both pathogens without inflammatory disease. Antiviral Res 2019; 168:9-17. [PMID: 31075351 PMCID: PMC6620116 DOI: 10.1016/j.antiviral.2019.05.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 05/01/2019] [Accepted: 05/02/2019] [Indexed: 12/29/2022]
Abstract
Respiratory syncytial virus (RSV) is the most common cause of lower respiratory disease in young children, elderly and immunocompromised adults. There is no licensed vaccine against RSV although development of an effective and safe RSV vaccine has been a high priority for several decades. Among the various vaccine platforms, the viral-vectored RSV vaccines based on licensed cold-adapted live attenuated influenza vaccine (LAIV) might offer an advantage of inducing adequate mucosal CD8 T cell immunity at the infection site of respiratory pathogens. We constructed two recombinant LAIV viruses expressing immunodominant T-cell epitopes of RSV M2-1 protein. The results in this study provide evidence that RSV CD8 T cell epitopes delivered by LAIV viral vector could confer protection against RSV infection without causing pulmonary eosinophilia and inflammatory RSV disease in mice. In addition, these chimeric LAIV-RSV vaccines retained their attenuated phenotype and ability to protect against virulent influenza virus, thus providing a unique approach to fight against two dangerous respiratory viral pathogens using a single vaccine preparation.
Collapse
Affiliation(s)
- Tatiana Kotomina
- Department of Virology, Institute of Experimental Medicine, 12 Acad. Pavlov Street, 197376, Russia
| | - Irina Isakova-Sivak
- Department of Virology, Institute of Experimental Medicine, 12 Acad. Pavlov Street, 197376, Russia.
| | - Victoria Matyushenko
- Department of Virology, Institute of Experimental Medicine, 12 Acad. Pavlov Street, 197376, Russia
| | - Ki-Hye Kim
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA
| | - Youri Lee
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA
| | - Yu-Jin Jung
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA
| | - Sang-Moo Kang
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA
| | - Larisa Rudenko
- Department of Virology, Institute of Experimental Medicine, 12 Acad. Pavlov Street, 197376, Russia
| |
Collapse
|