1
|
Niu Y, Yan Y, Hu Y, Yang X, Shi H, Zhou P, Zhu C, Xing M, Zhou D, Wang X. A novel tetravalent influenza vaccine based on one chimpanzee adenoviral vector. Vaccine 2025; 53:126959. [PMID: 40023902 DOI: 10.1016/j.vaccine.2025.126959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 02/20/2025] [Accepted: 02/23/2025] [Indexed: 03/04/2025]
Abstract
Highly effective and broad-spectrum influenza vaccines are urgently required to prevent influenza outbreaks. Hemagglutinin (HA), M2 ectodomain (M2e), and nucleoprotein (NP) are crucial target antigens for the development of universal influenza vaccines. To generate a novel multivalent influenza vaccine, the HA genes of influenza B Yamagata (BY) and Victoria (BV) strains, and the NP gene of H1N1 were cloned into the E1 region of the chimpanzee adenoviral vector, AdC68, and M2e epitopes of H1N1 and H3N2 were fused to the loop region of the AdC68 fiber, resulting in the recombinant adenoviral vector vaccine, AdC-Flu-Tet. The immunoprotective effects of AdC-Flu-Tet were evaluated in the mouse models. The results showed that AdC-Flu-Tet successfully induced robust humoral and cellular immune responses and conferred full protection against H1N1, H3N2, BY, and BV infections. In conclusion, AdC-Flu-Tet is a promising candidate as a novel influenza vaccine with high protective efficacy.
Collapse
Affiliation(s)
- Yixin Niu
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| | - Yao Yan
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| | - Ying Hu
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| | - Xi Yang
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| | - Hongyang Shi
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Ping Zhou
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| | - Caihong Zhu
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| | - Man Xing
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Dongming Zhou
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China; Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Xiang Wang
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China.
| |
Collapse
|
2
|
Zhao Y, Liu J, Peng C, Guo S, Wang B, Chen L, Wang Y, Tang H, Liu L, Pan Q, Li S, Wang J, Yang D, Du E. Cross-protection against homo and heterologous influenza viruses via intranasal administration of an HA chimeric multiepitope nanoparticle vaccine. J Nanobiotechnology 2025; 23:77. [PMID: 39905416 DOI: 10.1186/s12951-025-03122-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 01/13/2025] [Indexed: 02/06/2025] Open
Abstract
BACKGROUND Influenza A viruses (IAVs) cause seasonal influenza epidemics and pose significant threats to public health. However, seasonal influenza vaccines often elicit strain-specific immune responses and confer little protection against mismatched strains. There is an urgent need to develop universal influenza vaccines against emerging and potentially re-emerging influenza virus infections. Multiepitope vaccines combining multiple conserved epitopes can induce more robust and broader immune responses and provide a potential solution. RESULTS Here, we demonstrated that an HA chimeric multiepitope nanoparticle vaccine, delivered intranasally conferred broad protection against challenges with various influenza viruses in mice. The nanoparticle vaccine co-expresses the ectodomain of haemagglutinin (H), three repeated highly conserved ectodomains of matrix protein 2 (M), and the M-cell-targeting ligand Co4B (C) in a baculovirus-insect cell system. These elements (C, H and M) were presented on the surface of self-assembling ferritin (f) in tandem to generate a nanoparticle denoted as CHM-f. Intranasal vaccination with CHM-f nanoparticles elicited robust humoral and cellular immune responses, conferring complete protection against a variety of IAVs, including the A/PR8/34 H1N1 strain, the swine flu H3N2 strain, the avian flu H5N8 strain, and H9N2. When CHM-f nanoparticles adjuvanted with CpG IAMA-002, the weight loss protective effect, cellular immune responses and mucosal IgA responses were significantly augmented. Compared with controls, mice immunized with CHM-f nanoparticles with or without CpG IAMA-002 showed significant reductions in weight loss, lung viral titres and pathological changes. CONCLUSIONS These results suggest that CHM-f nanoparticle with or without CpG IAMA-002 is a promising candidate as a universal influenza vaccine.
Collapse
Affiliation(s)
- Yongqiang Zhao
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Jia Liu
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Chun Peng
- Chengdu NanoVAX Biotechnology Co., Ltd., Chengdu, Sichuan, 610219, China
| | - Shuangshuang Guo
- Yangling Carey Biotechnology Co., Ltd., Yangling, Shaanxi, 712100, China
| | - Bo Wang
- Yangling Carey Biotechnology Co., Ltd., Yangling, Shaanxi, 712100, China
| | - Longping Chen
- Yangling Carey Biotechnology Co., Ltd., Yangling, Shaanxi, 712100, China
| | - Yating Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Haiwen Tang
- Chengdu NanoVAX Biotechnology Co., Ltd., Chengdu, Sichuan, 610219, China
| | - Liming Liu
- Nanjing JSIAMA Biopharmaceuticals Ltd., Nanjing, Jiangsu, 210000, China
| | - Qi Pan
- Nanjing JSIAMA Biopharmaceuticals Ltd., Nanjing, Jiangsu, 210000, China
| | - Shiren Li
- Chengdu NanoVAX Biotechnology Co., Ltd., Chengdu, Sichuan, 610219, China
| | - Jingyu Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Dongni Yang
- Chengdu NanoVAX Biotechnology Co., Ltd., Chengdu, Sichuan, 610219, China.
| | - Enqi Du
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, 712100, China.
- Yangling Carey Biotechnology Co., Ltd., Yangling, Shaanxi, 712100, China.
| |
Collapse
|
3
|
Amarilla-Irusta A, Zenarruzabeitia O, Sevilla A, Sandá V, Lopez-Pardo A, Astarloa-Pando G, Pérez-Garay R, Pérez-Fernández S, Meijide S, Imaz-Ayo N, Arana-Arri E, Amo L, Borrego F. CD151 identifies an NK cell subset that is enriched in COVID-19 patients and correlates with disease severity. J Infect 2024; 89:106304. [PMID: 39374860 DOI: 10.1016/j.jinf.2024.106304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 09/27/2024] [Accepted: 09/29/2024] [Indexed: 10/09/2024]
Abstract
Severe coronavirus disease 2019 (COVID-19) often leads to acute respiratory distress syndrome and multi-organ dysfunction, driven by a dysregulated immune response, including a cytokine storm with elevated proinflammatory cytokine levels. Natural killer (NK) cells are part of the innate immune system with a fundamental role in the defense against viral infections. However, during COVID-19 acute infection, they exhibit an altered phenotype and impaired functionality contributing to the immunopathogenesis of the disease. In this work, we have studied a cohort of patients with COVID-19 (ranging from mild to severe) by analyzing IL-15, TGF-β, PlGF and GDF-15 plasma levels and performing multiparametric flow cytometry studies. Our results revealed that severe COVID-19 patients exhibited high levels of IL-15, PlGF and GDF-15, along with an enrichment of an NK cell subset expressing the CD151 tetraspanin, which correlated with IL-15 plasma levels and disease severity. In patients, these CD151+ NK cells displayed a more activated phenotype characterized by an increased expression of HLA-DR, CD38 and granzyme B, a distinct receptor repertoire, with lower levels of CD160 and CD31 and higher levels of CD55 and, remarkably, a higher expression of tissue-resident markers CD103 and the NK cell decidual marker CD9. Last of all, in individuals with severe disease, we identified an expansion of a CD151brightCD9+ NK cell subset, suggesting that these cells play a specific role in COVID-19. Altogether, our findings suggest that CD151+ NK cells may have a relevant role in COVID-19 immunopathogenesis.
Collapse
Affiliation(s)
| | - Olatz Zenarruzabeitia
- Immunopathology Group, Biobizkaia Health Research Institute, Barakaldo, Spain; Department of Genetics, Physical Anthropology and Animal Physiology, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Arrate Sevilla
- Immunopathology Group, Biobizkaia Health Research Institute, Barakaldo, Spain
| | - Víctor Sandá
- Immunopathology Group, Biobizkaia Health Research Institute, Barakaldo, Spain
| | - Ainara Lopez-Pardo
- Immunopathology Group, Biobizkaia Health Research Institute, Barakaldo, Spain
| | | | - Raquel Pérez-Garay
- Immunopathology Group, Biobizkaia Health Research Institute, Barakaldo, Spain; Clinical Analysis Service, Cruces University Hospital, OSI Ezkerraldea-Enkarterri-Cruces, Barakaldo, Spain
| | - Silvia Pérez-Fernández
- Scientific Coordination Facility, Biobizkaia Health Research Institute, Barakaldo, Spain
| | - Susana Meijide
- Scientific Coordination Facility, Biobizkaia Health Research Institute, Barakaldo, Spain
| | - Natale Imaz-Ayo
- Scientific Coordination Facility, Biobizkaia Health Research Institute, Barakaldo, Spain
| | - Eunate Arana-Arri
- Scientific Coordination Facility, Biobizkaia Health Research Institute, Barakaldo, Spain
| | - Laura Amo
- Immunopathology Group, Biobizkaia Health Research Institute, Barakaldo, Spain; Ikerbasque, Basque Foundation for Science, Bilbao, Spain
| | - Francisco Borrego
- Immunopathology Group, Biobizkaia Health Research Institute, Barakaldo, Spain; Ikerbasque, Basque Foundation for Science, Bilbao, Spain.
| |
Collapse
|
4
|
Calzas C, Alkie TN, Suderman M, Embury-Hyatt C, Khatri V, Le Goffic R, Berhane Y, Bourgault S, Archambault D, Chevalier C. M2e nanovaccines supplemented with recombinant hemagglutinin protect chickens against heterologous HPAI H5N1 challenge. NPJ Vaccines 2024; 9:161. [PMID: 39237609 PMCID: PMC11377767 DOI: 10.1038/s41541-024-00944-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 08/07/2024] [Indexed: 09/07/2024] Open
Abstract
Current poultry vaccines against influenza A viruses target the globular head region of the hemagglutinin (HA1), providing limited protection against antigenically divergent strains. Experimental subunit vaccines based on the conserved ectodomain of the matrix protein 2 (M2e) induce cross-reactive antibody responses, but fail to fully prevent virus shedding after low pathogenic avian influenza (LPAI) virus challenge, and are ineffective against highly pathogenic avian influenza (HPAI) viruses. This study assessed the benefits of combining nanoparticles bearing three tandem M2e repeats (NR-3M2e nanorings or NF-3M2e nanofilaments) with an HA1 subunit vaccine in protecting chickens against a heterologous HPAI H5N1 virus challenge. Chickens vaccinated with the combined formulations developed M2e and HA1-specific antibodies, were fully protected from clinical disease and mortality, and showed no histopathological lesions or virus shedding, unlike those given only HA1, NR-3M2e, or NF-3M2e. Thus, the combined vaccine formulations provided complete cross-protection against HPAI H5N1 virus, and prevented environmental virus shedding, crucial for controlling avian influenza outbreaks.
Collapse
Affiliation(s)
- Cynthia Calzas
- INRAE, UVSQ, UMR892 VIM, Université Paris-Saclay, Jouy-en-Josas, France
| | - Tamiru N Alkie
- National Centre for Foreign Animal Disease, Canadian Food Inspection Agency, Winnipeg, Canada
| | - Matthew Suderman
- Public Health Agency of Canada, National Microbiology Laboratory, Winnipeg, Canada
| | - Carissa Embury-Hyatt
- National Centre for Foreign Animal Disease, Canadian Food Inspection Agency, Winnipeg, Canada
| | - Vinay Khatri
- Chemistry Department, Université du Québec à Montréal, Montréal, QC, Canada
- Department of Biological Sciences, Université du Québec à Montréal, Montréal, QC, Canada
| | - Ronan Le Goffic
- INRAE, UVSQ, UMR892 VIM, Université Paris-Saclay, Jouy-en-Josas, France
| | - Yohannes Berhane
- National Centre for Foreign Animal Disease, Canadian Food Inspection Agency, Winnipeg, Canada
- Department of Veterinary Pathology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Canada
- Department of Animal Science, University of Manitoba, Winnipeg, Canada
| | - Steve Bourgault
- Chemistry Department, Université du Québec à Montréal, Montréal, QC, Canada
| | - Denis Archambault
- Department of Biological Sciences, Université du Québec à Montréal, Montréal, QC, Canada.
| | | |
Collapse
|
5
|
Liang Y, Guo J, Li Z, Liu S, Zhang T, Sun S, Lu F, Zhai Y, Wang W, Ning C, Tan W. A novel method to assess antibody-dependent cell-mediated cytotoxicity against influenza A virus M2 in immunized murine models. BIOSAFETY AND HEALTH 2024; 6:178-185. [PMID: 40078727 PMCID: PMC11895015 DOI: 10.1016/j.bsheal.2024.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 04/13/2024] [Accepted: 04/16/2024] [Indexed: 03/14/2025] Open
Abstract
The matrix protein 2 (M2) is a preferred target for developing a universal vaccine against the influenza A virus (IAV). This study aimed to develop a method for assessing antibody-dependent cell-mediated cytotoxicity (ADCC) associated with M2-based immunization in mice. We first established a stable cell line derived from mouse lymphoma cells (YAC-1) expressing M2 of H3N2. This cell line, designated as YAC-1-M2, was generated using a second-generation lentiviral tricistronic plasmid system to transduce the M2 gene into YAC-1 cells. The ADCC effect induced by polyclonal antibodies targeting matrix protein 2 ectodomain (M2e) was demonstrated by YAC-1-M2 cell lysis by natural killer cells (NK) derived from mice, in the presence of anti-M2 antibodies obtained from mice immunized with an mRNA vaccine based on M2e. This ADCC effect was found to be stronger compared to the effect induced by monoclonal antibodies (14C2) against M2. Moreover, the ADCC effect was enhanced as the effector-to-target ratio of NK to YAC-1-M2 cells increased. In conclusion, we established a novel method to detect ADCC of M2 of IAV, which paves the way for the development of an M2-based universal vaccine against IAV and an in-depth analysis of its mechanism of broad-spectrum immune protection in mice.
Collapse
Affiliation(s)
- Yinjie Liang
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning 530021, China
- Key Laboratory of Biosafety, National Health Commission of the People’s Republic of China, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Junjia Guo
- Key Laboratory of Biosafety, National Health Commission of the People’s Republic of China, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
- School of Laboratory Medicine, Wenzhou Medical University, Wenzhou 325035, China
| | - Zhen Li
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning 530021, China
- Key Laboratory of Biosafety, National Health Commission of the People’s Republic of China, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Shiyuan Liu
- Key Laboratory of Biosafety, National Health Commission of the People’s Republic of China, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
- School of Public Health, Baotou Medical College, Baotou 014040, China
| | - Ting Zhang
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning 530021, China
| | - Shucai Sun
- Key Laboratory of Biosafety, National Health Commission of the People’s Republic of China, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
- Department of Nuclear Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, China
| | - Funa Lu
- Key Laboratory of Biosafety, National Health Commission of the People’s Republic of China, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
- Department of Microbiology, School of Basic Medicine, Inner Mongolia Medical University, Hohhot 010110, China
| | - Yuqian Zhai
- Key Laboratory of Biosafety, National Health Commission of the People’s Republic of China, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Wenling Wang
- Key Laboratory of Biosafety, National Health Commission of the People’s Republic of China, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Chuanyi Ning
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning 530021, China
| | - Wenjie Tan
- Key Laboratory of Biosafety, National Health Commission of the People’s Republic of China, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
- School of Laboratory Medicine, Wenzhou Medical University, Wenzhou 325035, China
| |
Collapse
|
6
|
Rijnink WF, Stadlbauer D, Puente-Massaguer E, Okba NMA, Kirkpatrick Roubidoux E, Strohmeier S, Mudd PA, Schmitz A, Ellebedy A, McMahon M, Krammer F. Characterization of non-neutralizing human monoclonal antibodies that target the M1 and NP of influenza A viruses. J Virol 2023; 97:e0164622. [PMID: 37916834 PMCID: PMC10688359 DOI: 10.1128/jvi.01646-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 10/08/2023] [Indexed: 11/03/2023] Open
Abstract
IMPORTANCE Currently, many groups are focusing on isolating both neutralizing and non-neutralizing antibodies to the mutation-prone hemagglutinin as a tool to treat or prevent influenza virus infection. Less is known about the level of protection induced by non-neutralizing antibodies that target conserved internal influenza virus proteins. Such non-neutralizing antibodies could provide an alternative pathway to induce broad cross-reactive protection against multiple influenza virus serotypes and subtypes by partially overcoming influenza virus escape mediated by antigenic drift and shift. Accordingly, more information about the level of protection and potential mechanism(s) of action of non-neutralizing antibodies targeting internal influenza virus proteins could be useful for the design of broadly protective and universal influenza virus vaccines.
Collapse
Affiliation(s)
| | - Daniel Stadlbauer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Eduard Puente-Massaguer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Nisreen M. A. Okba
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Ericka Kirkpatrick Roubidoux
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Shirin Strohmeier
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Philip A. Mudd
- Division of Immunobiology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Aaron Schmitz
- Division of Immunobiology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Ali Ellebedy
- Division of Immunobiology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Meagan McMahon
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Pathology, Molecular and Cell Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
7
|
Ouyang MJ, Ao Z, Olukitibi TA, Lawrynuik P, Shieh C, Kung SKP, Fowke KR, Kobasa D, Yao X. Oral Immunization with rVSV Bivalent Vaccine Elicits Protective Immune Responses, Including ADCC, against Both SARS-CoV-2 and Influenza A Viruses. Vaccines (Basel) 2023; 11:1404. [PMID: 37766083 PMCID: PMC10534613 DOI: 10.3390/vaccines11091404] [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: 07/26/2023] [Revised: 08/14/2023] [Accepted: 08/21/2023] [Indexed: 09/29/2023] Open
Abstract
COVID-19 and influenza both cause enormous disease burdens, and vaccines are the primary measures for their control. Since these viral diseases are transmitted through the mucosal surface of the respiratory tract, developing an effective and convenient mucosal vaccine should be a high priority. We previously reported a recombinant vesicular stomatitis virus (rVSV)-based bivalent vaccine (v-EM2/SPΔC1Delta) that protects animals from both SARS-CoV-2 and influenza viruses via intramuscular and intranasal immunization. Here, we further investigated the immune response induced by oral immunization with this vaccine and its protective efficacy in mice. The results demonstrated that the oral delivery, like the intranasal route, elicited strong and protective systemic immune responses against SARS-CoV-2 and influenza A virus. This included high levels of neutralizing antibodies (NAbs) against SARS-CoV-2, as well as strong anti-SARS-CoV-2 spike protein (SP) antibody-dependent cellular cytotoxicity (ADCC) and anti-influenza M2 ADCC responses in mice sera. Furthermore, it provided efficient protection against challenge with influenza H1N1 virus in a mouse model, with a 100% survival rate and a significantly low lung viral load of influenza virus. All these findings provide substantial evidence for the effectiveness of oral immunization with the rVSV bivalent vaccine.
Collapse
Affiliation(s)
- Maggie Jing Ouyang
- Laboratory of Molecular Human Retrovirology, Department of Medical Microbiology and Infectious Diseases, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, 508-745 Bannatyne Ave, Winnipeg, MB R3E 0J9, Canada; (M.J.O.); (Z.A.); (T.A.O.); (P.L.); (C.S.)
- Department of Medical Microbiology and Infectious Diseases, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, 745 Bannatyne Ave, Winnipeg, MB R3E 0J9, Canada; (K.R.F.); (D.K.)
| | - Zhujun Ao
- Laboratory of Molecular Human Retrovirology, Department of Medical Microbiology and Infectious Diseases, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, 508-745 Bannatyne Ave, Winnipeg, MB R3E 0J9, Canada; (M.J.O.); (Z.A.); (T.A.O.); (P.L.); (C.S.)
- Department of Medical Microbiology and Infectious Diseases, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, 745 Bannatyne Ave, Winnipeg, MB R3E 0J9, Canada; (K.R.F.); (D.K.)
| | - Titus A. Olukitibi
- Laboratory of Molecular Human Retrovirology, Department of Medical Microbiology and Infectious Diseases, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, 508-745 Bannatyne Ave, Winnipeg, MB R3E 0J9, Canada; (M.J.O.); (Z.A.); (T.A.O.); (P.L.); (C.S.)
- Department of Medical Microbiology and Infectious Diseases, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, 745 Bannatyne Ave, Winnipeg, MB R3E 0J9, Canada; (K.R.F.); (D.K.)
| | - Peter Lawrynuik
- Laboratory of Molecular Human Retrovirology, Department of Medical Microbiology and Infectious Diseases, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, 508-745 Bannatyne Ave, Winnipeg, MB R3E 0J9, Canada; (M.J.O.); (Z.A.); (T.A.O.); (P.L.); (C.S.)
| | - Christopher Shieh
- Laboratory of Molecular Human Retrovirology, Department of Medical Microbiology and Infectious Diseases, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, 508-745 Bannatyne Ave, Winnipeg, MB R3E 0J9, Canada; (M.J.O.); (Z.A.); (T.A.O.); (P.L.); (C.S.)
| | - Sam K. P. Kung
- Department of Immunology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0W3, Canada;
| | - Keith R. Fowke
- Department of Medical Microbiology and Infectious Diseases, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, 745 Bannatyne Ave, Winnipeg, MB R3E 0J9, Canada; (K.R.F.); (D.K.)
| | - Darwyn Kobasa
- Department of Medical Microbiology and Infectious Diseases, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, 745 Bannatyne Ave, Winnipeg, MB R3E 0J9, Canada; (K.R.F.); (D.K.)
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3L5, Canada
| | - Xiaojian Yao
- Laboratory of Molecular Human Retrovirology, Department of Medical Microbiology and Infectious Diseases, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, 508-745 Bannatyne Ave, Winnipeg, MB R3E 0J9, Canada; (M.J.O.); (Z.A.); (T.A.O.); (P.L.); (C.S.)
- Department of Medical Microbiology and Infectious Diseases, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, 745 Bannatyne Ave, Winnipeg, MB R3E 0J9, Canada; (K.R.F.); (D.K.)
| |
Collapse
|
8
|
Bricha S, Côté-Cyr M, Tremblay T, Nguyen PT, St-Louis P, Giguère D, Archambault D, Bourgault S. Synthetic Multicomponent Nanovaccines Based on the Molecular Co-assembly of β-Peptides Protect against Influenza A Virus. ACS Infect Dis 2023; 9:1232-1244. [PMID: 37200051 DOI: 10.1021/acsinfecdis.2c00610] [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] [Indexed: 05/19/2023]
Abstract
Peptides with the ability to self-assemble into nanoparticles have emerged as an attractive strategy to design antigen delivery platforms for subunit vaccines. While toll-like receptor (TLR) agonists are promising immunostimulants, their use as soluble agents is limited by their rapid clearance and off-target inflammation. Herein, we harnessed molecular co-assembly to prepare multicomponent cross-β-sheet peptide nanofilaments exposing an antigenic epitope derived from the influenza A virus and a TLR agonist. The TLR7 agonist imiquimod and the TLR9 agonist CpG were respectively functionalized on the assemblies by means of an orthogonal pre- or post-assembly conjugation strategy. The nanofilaments were readily uptaken by dendritic cells, and the TLR agonists retained their activity. Multicomponent nanovaccines induced a robust epitope-specific immune response and completely protected immunized mice from a lethal influenza A virus inoculation. This versatile bottom-up approach is promising for the preparation of synthetic vaccines with customized magnitude and polarization of the immune responses.
Collapse
Affiliation(s)
- Salma Bricha
- Department of Chemistry, Université du Québec à Montréal, C.P.8888, Succursale Centre-Ville, Montréal H3C 3P8, Canada
- Department of Biological Sciences, Université du Québec à Montréal, C.P.8888, Succursale Centre-Ville, Montréal H3C 3P8, Canada
- Quebec Network for Research on Protein Function, Engineering and Applications (PROTEO), Québec H3C 3P8, Canada
- The Swine and Poultry Infectious Diseases Research Centre (CRIPA), Saint-Hyacinthe J2S 2M2, Canada
- The Center of Excellence in Research on Orphan Diseases─Fondation Courtois (CERMO-FC), Montréal H3C 3P8, Canada
| | - Mélanie Côté-Cyr
- Department of Chemistry, Université du Québec à Montréal, C.P.8888, Succursale Centre-Ville, Montréal H3C 3P8, Canada
- Quebec Network for Research on Protein Function, Engineering and Applications (PROTEO), Québec H3C 3P8, Canada
- The Swine and Poultry Infectious Diseases Research Centre (CRIPA), Saint-Hyacinthe J2S 2M2, Canada
- The Center of Excellence in Research on Orphan Diseases─Fondation Courtois (CERMO-FC), Montréal H3C 3P8, Canada
| | - Thomas Tremblay
- Quebec Network for Research on Protein Function, Engineering and Applications (PROTEO), Québec H3C 3P8, Canada
- Department of Chemistry, Université Laval, 1045 Av. De la Médecine, Québec City QC G1V 0A6, Canada
| | - Phuong Trang Nguyen
- Department of Chemistry, Université du Québec à Montréal, C.P.8888, Succursale Centre-Ville, Montréal H3C 3P8, Canada
- Quebec Network for Research on Protein Function, Engineering and Applications (PROTEO), Québec H3C 3P8, Canada
- The Swine and Poultry Infectious Diseases Research Centre (CRIPA), Saint-Hyacinthe J2S 2M2, Canada
- The Center of Excellence in Research on Orphan Diseases─Fondation Courtois (CERMO-FC), Montréal H3C 3P8, Canada
| | - Philippe St-Louis
- Department of Biological Sciences, Université du Québec à Montréal, C.P.8888, Succursale Centre-Ville, Montréal H3C 3P8, Canada
- The Swine and Poultry Infectious Diseases Research Centre (CRIPA), Saint-Hyacinthe J2S 2M2, Canada
- The Center of Excellence in Research on Orphan Diseases─Fondation Courtois (CERMO-FC), Montréal H3C 3P8, Canada
| | - Denis Giguère
- Quebec Network for Research on Protein Function, Engineering and Applications (PROTEO), Québec H3C 3P8, Canada
- Department of Chemistry, Université Laval, 1045 Av. De la Médecine, Québec City QC G1V 0A6, Canada
| | - Denis Archambault
- Department of Biological Sciences, Université du Québec à Montréal, C.P.8888, Succursale Centre-Ville, Montréal H3C 3P8, Canada
- The Swine and Poultry Infectious Diseases Research Centre (CRIPA), Saint-Hyacinthe J2S 2M2, Canada
- The Center of Excellence in Research on Orphan Diseases─Fondation Courtois (CERMO-FC), Montréal H3C 3P8, Canada
| | - Steve Bourgault
- Department of Chemistry, Université du Québec à Montréal, C.P.8888, Succursale Centre-Ville, Montréal H3C 3P8, Canada
- Quebec Network for Research on Protein Function, Engineering and Applications (PROTEO), Québec H3C 3P8, Canada
- The Swine and Poultry Infectious Diseases Research Centre (CRIPA), Saint-Hyacinthe J2S 2M2, Canada
- The Center of Excellence in Research on Orphan Diseases─Fondation Courtois (CERMO-FC), Montréal H3C 3P8, Canada
| |
Collapse
|
9
|
Smith DM, Schafer JR, Tullius B, Witkam L, Paust S. Natural killer cells for antiviral therapy. Sci Transl Med 2023; 15:eabl5278. [PMID: 36599006 DOI: 10.1126/scitranslmed.abl5278] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Natural killer (NK) cell-based immunotherapy is being explored for treating infectious diseases, including viral infections. Here, we discuss evidence of NK cell responses to different viruses, ongoing clinical efforts to treat such infections with NK cell products, and review platforms to generate NK cell products.
Collapse
Affiliation(s)
- Davey M Smith
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | | | | | - Laura Witkam
- Kiadis Pharma, Sanofi, 1105BP Amsterdam, Netherlands
| | - Silke Paust
- Department of Immunology and Microbiology, Scripps Research Institute, La Jolla, CA 92037, USA
| |
Collapse
|
10
|
Pousse L, Korfi K, Medeiros BC, Berrera M, Kumpesa N, Eckmann J, Hutter IK, Griesser V, Karanikas V, Klein C, Amann M. CD25 targeting with the afucosylated human IgG1 antibody RG6292 eliminates regulatory T cells and CD25+ blasts in acute myeloid leukemia. Front Oncol 2023; 13:1150149. [PMID: 37205201 PMCID: PMC10185852 DOI: 10.3389/fonc.2023.1150149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 04/11/2023] [Indexed: 05/21/2023] Open
Abstract
Background Acute Myeloid leukemia is a heterogeneous disease that requires novel targeted treatment options tailored to the patients' specific microenvironment and blast phenotype. Methods We characterized bone marrow and/or blood samples of 37 AML patients and healthy donors by high dimensional flow cytometry and RNA sequencing using computational analysis. In addition, we performed ex vivo ADCC assays using allogeneic NK cells isolated from healthy donors and AML patient material to test the cytotoxic potential of CD25 Mab (also referred to as RG6292 and RO7296682) or isotype control antibody on regulatory T cells and CD25+ AML cells. Results Bone marrow composition, in particular the abundance of regulatory T cells and CD25 expressing AML cells, correlated strongly with that of the blood in patients with time-matched samples. In addition, we observed a strong enrichment in the prevalence of CD25 expressing AML cells in patients bearing a FLT3-ITD mutation or treated with a hypomethylating agent in combination with venetoclax. We adopted a patient-centric approach to study AML clusters with CD25 expression and found it most highly expressed on immature phenotypes. Ex vivo treatment of primary AML patient samples with CD25 Mab, a human CD25 specific glycoengineered IgG1 antibody led to the specific killing of two different cell types, CD25+ AML cells and regulatory T cells, by allogeneic Natural Killer cells. Conclusion The in-depth characterization of patient samples by proteomic and genomic analyses supported the identification of a patient population that may benefit most by harnessing CD25 Mab's dual mode of action. In this pre-selected patient population, CD25 Mab could lead to the specific depletion of regulatory T cells, in addition to leukemic stem cells and progenitor-like AML cells that are responsible for disease progression or relapse.
Collapse
Affiliation(s)
- Laurène Pousse
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Zurich (RICZ), Schlieren, Switzerland
- *Correspondence: Laurène Pousse, ; Maria Amann,
| | - Koorosh Korfi
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Zurich (RICZ), Schlieren, Switzerland
| | - Bruno C. Medeiros
- Genentech, Inc. Hematology Department, South San Francisco, CA, United States
| | - Marco Berrera
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Basel (RICB), Basel, Switzerland
| | - Nadine Kumpesa
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Basel (RICB), Basel, Switzerland
| | - Jan Eckmann
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Münich (RICM), Penzberg, Germany
| | - Idil Karakoc Hutter
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Zurich (RICZ), Schlieren, Switzerland
| | - Vera Griesser
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Basel (RICB), Basel, Switzerland
| | - Vaios Karanikas
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Zurich (RICZ), Schlieren, Switzerland
| | - Christian Klein
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Zurich (RICZ), Schlieren, Switzerland
| | - Maria Amann
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Zurich (RICZ), Schlieren, Switzerland
- *Correspondence: Laurène Pousse, ; Maria Amann,
| |
Collapse
|
11
|
Assessment of Fcγ receptor-dependent binding of influenza hemagglutinin vaccine-induced antibodies in a non-human primate model. iScience 2022; 25:105085. [PMID: 36147947 PMCID: PMC9486051 DOI: 10.1016/j.isci.2022.105085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 08/09/2022] [Accepted: 08/31/2022] [Indexed: 11/23/2022] Open
Abstract
Several cross-protective antibodies that recognize a broad range of influenza A virus (IAV) strains are known to have functions in virus elimination such as Fcγ receptor (FcγR)-effector function and neutralizing activity against the head region. Although few studies have used primary cells as effector cells, the FcγR-effector function was evaluated after isolating each cell subset. Herein, we established an original assay system to evaluate purified FI6 IgG-mediated binding to hemagglutinin (HA)-expressing cells by flow cytometry using peripheral blood mononuclear cells from cynomolgus macaques. In addition, we evaluated the FcγR-effector function of IAV vaccine-induced anti-HA antibodies in cynomolgus macaques after administering the split vaccine. We found several cell types, mainly classical monocytes, bound to HA-expressing target cells in an FcγR-dependent manner, that were dominant in the binding of the cell population. Thus, this assay system could facilitate the development of a universal influenza vaccine.
Collapse
|
12
|
Terrén I, Orrantia A, Astarloa-Pando G, Amarilla-Irusta A, Zenarruzabeitia O, Borrego F. Cytokine-Induced Memory-Like NK Cells: From the Basics to Clinical Applications. Front Immunol 2022; 13:884648. [PMID: 35603208 PMCID: PMC9114299 DOI: 10.3389/fimmu.2022.884648] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Accepted: 04/11/2022] [Indexed: 12/12/2022] Open
Abstract
Natural killer (NK) cells are lymphocytes with a key role in the defense against viral infections and tumor cells. Although NK cells are classified as innate lymphoid cells (ILCs), under certain circumstances they exhibit adaptive and memory-like features. The latter may be achieved, among others, by a brief stimulation with interleukin (IL)-12, IL-15 and IL-18. These cytokine-induced memory-like (CIML) NK cells resemble the trained immunity observed in myeloid cells. CIML NK cells undergo transcriptional, epigenetic and metabolic reprogramming that, along with changes in the expression of cell surface receptors and components of cytotoxic granules, are responsible for their enhanced effector functions after a resting period. In addition, these memory-like NK cells persist for a long time, which make them a good candidate for cancer immunotherapy. Currently, several clinical trials are testing CIML NK cells infusions to treat tumors, mostly hematological malignancies. In relapse/refractory acute myeloid leukemia (AML), the adoptive transfer of CIML NK cells is safe and complete clinical remissions have been observed. In our review, we sought to summarize the current knowledge about the generation and molecular basis of NK cell memory-like responses and the up-to-date results from clinical trials with CIML NK cells.
Collapse
Affiliation(s)
- Iñigo Terrén
- Immunopathology Group, Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
| | - Ane Orrantia
- Immunopathology Group, Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
| | | | | | - Olatz Zenarruzabeitia
- Immunopathology Group, Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
| | - Francisco Borrego
- Immunopathology Group, Biocruces Bizkaia Health Research Institute, Barakaldo, Spain.,Ikerbasque, Basque Foundation for Science, Bilbao, Spain
| |
Collapse
|
13
|
van Vloten JP, Matuszewska K, Minow MAA, Minott JA, Santry LA, Pereira M, Stegelmeier AA, McAusland TM, Klafuric EM, Karimi K, Colasanti J, McFadden DG, Petrik JJ, Bridle BW, Wootton SK. Oncolytic Orf virus licenses NK cells via cDC1 to activate innate and adaptive antitumor mechanisms and extends survival in a murine model of late-stage ovarian cancer. J Immunother Cancer 2022; 10:jitc-2021-004335. [PMID: 35296558 PMCID: PMC8928368 DOI: 10.1136/jitc-2021-004335] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/12/2022] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Novel therapies are needed to improve outcomes for women diagnosed with ovarian cancer. Oncolytic viruses are multifunctional immunotherapeutic biologics that preferentially infect cancer cells and stimulate inflammation with the potential to generate antitumor immunity. Herein we describe Parapoxvirus ovis (Orf virus (OrfV)), an oncolytic poxvirus, as a viral immunotherapy for ovarian cancer. METHODS The immunotherapeutic potential of OrfV was tested in the ID8 orthotopic mouse model of end-stage epithelial ovarian carcinoma. Immune cell profiling, impact on secondary lesion development and survival were evaluated in OrfV-treated mice as well as in Batf3 knockout, mice depleted of specific immune cell subsets and in mice where the primary tumor was removed. Finally, we interrogated gene expression datasets from primary human ovarian tumors from the International Cancer Genome Consortium database to determine whether the interplay we observed between natural killer (NK) cells, classical type 1 dendritic cells (cDC1s) and T cells exists and influences outcomes in human ovarian cancer. RESULTS OrfV was an effective monotherapy in a murine model of advanced-stage epithelial ovarian cancer. OrfV intervention relied on NK cells, which when depleted abrogated antitumor CD8+ T-cell responses. OrfV therapy was shown to require cDC1s in experiments with BATF3 knockout mice, which do not have mature cDC1s. Furthermore, cDC1s governed antitumor NK and T-cell responses to mediate antitumor efficacy following OrfV. Primary tumor removal, a common treatment option in human patients, was effectively combined with OrfV for optimal therapeutic outcome. Analysis of human RNA sequencing datasets revealed that cDC1s correlate with NK cells in human ovarian cancer and that intratumoral NK cells correlate positively with survival. CONCLUSIONS The data herein support the translational potential of OrfV as an NK stimulating immunotherapeutic for the treatment of advanced-stage ovarian cancer.
Collapse
Affiliation(s)
- Jacob P van Vloten
- Department of Pathobiology, University of Guelph, Guelph, Ontario, Canada
| | - Kathy Matuszewska
- Department of Biomedical Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Mark A A Minow
- Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| | - Jessica A Minott
- Department of Pathobiology, University of Guelph, Guelph, Ontario, Canada
| | - Lisa A Santry
- Department of Pathobiology, University of Guelph, Guelph, Ontario, Canada
| | - Madison Pereira
- Department of Biomedical Sciences, University of Guelph, Guelph, Ontario, Canada
| | | | - Thomas M McAusland
- Department of Pathobiology, University of Guelph, Guelph, Ontario, Canada
| | - Elaine M Klafuric
- Department of Pathobiology, University of Guelph, Guelph, Ontario, Canada
| | - Khalil Karimi
- Department of Pathobiology, University of Guelph, Guelph, Ontario, Canada
| | - Joseph Colasanti
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| | - D Grant McFadden
- Biodesign Institute, Arizona State University, Tempe, Arizona, USA
| | - James J Petrik
- Department of Biomedical Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Byram W Bridle
- Department of Pathobiology, University of Guelph, Guelph, Ontario, Canada
| | - Sarah K Wootton
- Department of Pathobiology, University of Guelph, Guelph, Ontario, Canada
| |
Collapse
|
14
|
Vanderven HA, Esterbauer R, Jegaskanda S, Tan HX, Wheatley AK, Kent SJ. Poor protective potential of influenza nucleoprotein antibodies despite wide prevalence. Immunol Cell Biol 2021; 100:49-60. [PMID: 34687553 DOI: 10.1111/imcb.12508] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 10/20/2021] [Accepted: 10/22/2021] [Indexed: 11/28/2022]
Abstract
Humans are exposed to influenza virus through periodic infections. Due to these repeated exposures, human populations commonly have elevated antibody titers targeting the conserved internal influenza virus nucleoprotein (NP). Despite the presence of anti-NP antibodies, humans are acutely susceptible to drifted influenza viruses with antigenically different surface proteins and the protective potential of human NP antibodies is unclear. In this study, high levels of anti-NP antibody and NP-specific B cells were detected in both adult humans and influenza-infected mice, confirming that NP is a major target of humoral immunity. Through sorting single B cells from influenza-exposed human adults, we generated a panel of 11 anti-NP monoclonal antibodies (mAbs). The majority of anti-NP human mAbs generated were capable of engaging cellular Fc receptors and bound NP on the surface of influenza-infected cell lines in vitro, suggesting that anti-NP mAbs have the potential to mediate downstream Fc effector functions such as antibody-dependent cellular cytotoxicity and antibody-dependent phagocytosis. However, human anti-NP mAbs were not protective in vivo when passively transferred into a murine influenza challenge model. Future in vivo studies examining the synergistic effect of anti-NP mAbs infused with other influenza-specific mAbs are warranted.
Collapse
Affiliation(s)
- Hillary A Vanderven
- Biomedicine, College of Public Health, Medical and Veterinary Sciences, James Cook University, Douglas, QLD, Australia.,Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC, Australia
| | - Robyn Esterbauer
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC, Australia
| | - Sinthujan Jegaskanda
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC, Australia
| | - Hyon-Xhi Tan
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC, Australia
| | - Adam K Wheatley
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC, Australia
| | - Stephen J Kent
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC, Australia.,Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, University of Melbourne, Parkville, VIC, Australia.,Melbourne Sexual Health Centre, Department of Infectious Diseases, Alfred Health, Central Clinical School, Monash University, Carlton, VIC, Australia
| |
Collapse
|
15
|
Mikelez-Alonso I, Magadán S, González-Fernández Á, Borrego F. Natural killer (NK) cell-based immunotherapies and the many faces of NK cell memory: A look into how nanoparticles enhance NK cell activity. Adv Drug Deliv Rev 2021; 176:113860. [PMID: 34237404 DOI: 10.1016/j.addr.2021.113860] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 06/21/2021] [Accepted: 07/01/2021] [Indexed: 12/16/2022]
Abstract
Natural killer (NK) cells are lymphocytes able to exert potent antitumor and antiviral functions by different means. Besides their classification as innate lymphoid cells (ILCs), NK cells exhibit memory-like and memory responses after cytokine preactivation, viral infections and hapten exposure. Multiple NK cell-based immunotherapies have been developed and are currently being tested, including the possibility to translate the NK cell memory responses into the clinic. Nevertheless, still there is a need to improve these therapies, especially for the treatment of solid tumors, and nanotechnology represents an attractive option to increase NK cell effector functions against transformed cells. In this article, we review the basis of NK cell activity, the diversity of the NK cell memory responses and the current NK cell-based immunotherapies that are being used in the clinic. Furthermore, we take a look into nanotechnology-based strategies targeting NK cells to modulate their responses for effective immunotherapy.
Collapse
Affiliation(s)
- Idoia Mikelez-Alonso
- Biocruces Bizkaia Health Research Institute, Immunopathology Group, Barakaldo, Spain; Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Donostia - San Sebastián, Spain
| | - Susana Magadán
- CINBIO, Universidade de Vigo, Immunology Group, Vigo, Spain; Galicia Sur Health Research Institute (IIS-GS), Hospital Alvaro Cunqueiro, Vigo, Spain
| | - África González-Fernández
- CINBIO, Universidade de Vigo, Immunology Group, Vigo, Spain; Galicia Sur Health Research Institute (IIS-GS), Hospital Alvaro Cunqueiro, Vigo, Spain
| | - Francisco Borrego
- Biocruces Bizkaia Health Research Institute, Immunopathology Group, Barakaldo, Spain; Ikerbasque, Basque Foundation for Science, Bilbao, Spain.
| |
Collapse
|
16
|
Pedrina J, Stambas J. Targeting the Host Response: Can We Manipulate Extracellular Matrix Metalloproteinase Activity to Improve Influenza Virus Infection Outcomes? Front Mol Biosci 2021; 8:703456. [PMID: 34291090 PMCID: PMC8287203 DOI: 10.3389/fmolb.2021.703456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 06/08/2021] [Indexed: 11/25/2022] Open
Abstract
Each year, hundreds of thousands of individuals succumb to influenza virus infection and its associated complications. Several preventative and therapeutic options may be applied in order to preserve life. These traditional approaches include administration of seasonal influenza vaccines, pharmacological interventions in the form of antiviral drug therapy and supportive clinical approaches including mechanical ventilation and extracorporeal membrane oxygenation. While these measures have shown varying degrees of success, antiviral therapies and vaccination are constrained due to ongoing antigenic drift. Moreover, clinical approaches can also be associated with complications and drawbacks. These factors have led to the exploration and development of more sophisticated and nuanced therapeutic approaches involving host proteins. Advances in immunotherapy in the cancer field or administration of steroids following virus infection have highlighted the therapeutic potential of targeting host immune responses. We have now reached a point where we can consider the contribution of other “non-traditional” host components such as the extracellular matrix in immunity. Herein, we will review current, established therapeutic interventions and consider novel therapeutic approaches involving the extracellular matrix.
Collapse
Affiliation(s)
- Jess Pedrina
- Faculty of Health, School of Medicine, Deakin University, Geelong, VIC, Australia
| | - John Stambas
- Faculty of Health, School of Medicine, Deakin University, Geelong, VIC, Australia
| |
Collapse
|
17
|
Tan MP, Tan WS, Mohamed Alitheen NB, Yap WB. M2e-Based Influenza Vaccines with Nucleoprotein: A Review. Vaccines (Basel) 2021; 9:739. [PMID: 34358155 PMCID: PMC8310010 DOI: 10.3390/vaccines9070739] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 06/29/2021] [Accepted: 07/01/2021] [Indexed: 11/29/2022] Open
Abstract
Discovery of conserved antigens for universal influenza vaccines warrants solutions to a number of concerns pertinent to the currently licensed influenza vaccines, such as annual reformulation and mismatching with the circulating subtypes. The latter causes low vaccine efficacies, and hence leads to severe disease complications and high hospitalization rates among susceptible and immunocompromised individuals. A universal influenza vaccine ensures cross-protection against all influenza subtypes due to the presence of conserved epitopes that are found in the majority of, if not all, influenza types and subtypes, e.g., influenza matrix protein 2 ectodomain (M2e) and nucleoprotein (NP). Despite its relatively low immunogenicity, influenza M2e has been proven to induce humoral responses in human recipients. Influenza NP, on the other hand, promotes remarkable anti-influenza T-cell responses. Additionally, NP subunits are able to assemble into particles which can be further exploited as an adjuvant carrier for M2e peptide. Practically, the T-cell immunodominance of NP can be transferred to M2e when it is fused and expressed as a chimeric protein in heterologous hosts such as Escherichia coli without compromising the antigenicity. Given the ability of NP-M2e fusion protein in inducing cross-protective anti-influenza cell-mediated and humoral immunity, its potential as a universal influenza vaccine is therefore worth further exploration.
Collapse
Affiliation(s)
- Mei Peng Tan
- Department of Cell and Molecular Biology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang 43400, Malaysia; (M.P.T.); (N.B.M.A.)
- Center for Toxicology and Health Risk Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Malaysia
| | - Wen Siang Tan
- Department of Microbiology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang 43400, Malaysia;
- Laboratory of Vaccine and Biomolecules, Institute of Bioscience, Universiti Putra Malaysia, Serdang 43400, Malaysia
| | - Noorjahan Banu Mohamed Alitheen
- Department of Cell and Molecular Biology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang 43400, Malaysia; (M.P.T.); (N.B.M.A.)
| | - Wei Boon Yap
- Center for Toxicology and Health Risk Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Malaysia
- Biomedical Science Program, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Malaysia
| |
Collapse
|
18
|
Jung J, Mundle ST, Ustyugova IV, Horton AP, Boutz DR, Pougatcheva S, Prabakaran P, McDaniel JR, King GR, Park D, Person MD, Ye C, Tan B, Tanno Y, Kim JE, Curtis NC, DiNapoli J, Delagrave S, Ross TM, Ippolito GC, Kleanthous H, Lee J, Georgiou G. Influenza vaccination in the elderly boosts antibodies against conserved viral proteins and egg-produced glycans. J Clin Invest 2021; 131:148763. [PMID: 34196304 PMCID: PMC8245176 DOI: 10.1172/jci148763] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 05/19/2021] [Indexed: 12/25/2022] Open
Abstract
Seasonal influenza vaccination elicits a diminished adaptive immune response in the elderly, and the mechanisms of immunosenescence are not fully understood. Using Ig-Seq, we found a marked increase with age in the prevalence of cross-reactive (CR) serum antibodies that recognize both the H1N1 (vaccine-H1) and H3N2 (vaccine-H3) components of an egg-produced split influenza vaccine. CR antibodies accounted for 73% ± 18% of the serum vaccine responses in a cohort of elderly donors, 65% ± 15% in late middle-aged donors, and only 13% ± 5% in persons under 35 years of age. The antibody response to non-HA antigens was boosted by vaccination. Recombinant expression of 19 vaccine-H1+H3 CR serum monoclonal antibodies (s-mAbs) revealed that they predominantly bound to non-HA influenza proteins. A sizable fraction of vaccine-H1+H3 CR s-mAbs recognized with high affinity the sulfated glycans, in particular sulfated type 2 N-acetyllactosamine (Galβ1-4GalNAcβ), which is found on egg-produced proteins and thus unlikely to contribute to protection against influenza infection in humans. Antibodies against sulfated glycans in egg-produced vaccine had been identified in animals but were not previously characterized in humans. Collectively, our results provide a quantitative basis for how repeated exposure to split influenza vaccine correlates with unintended focusing of serum antibody responses to non-HA antigens that may result in suboptimal immunity against influenza.
Collapse
Affiliation(s)
- Jiwon Jung
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas, USA
| | - Sophia T. Mundle
- Sanofi Pasteur Inc., Research North America, Cambridge, Massachusetts, USA
| | - Irina V. Ustyugova
- Sanofi Pasteur Inc., Research North America, Cambridge, Massachusetts, USA
| | | | | | | | - Ponraj Prabakaran
- Sanofi Pasteur Inc., Research North America, Cambridge, Massachusetts, USA
| | | | | | - Daechan Park
- Institute for Cellular and Molecular Biology, and
| | - Maria D. Person
- Biological Mass Spectrometry Facility, The University of Texas at Austin, Austin, Texas, USA
| | - Congxi Ye
- Department of Molecular Biosciences
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire, USA
| | - Bing Tan
- Department of Chemical Engineering
| | | | - Jin Eyun Kim
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas, USA
| | - Nicholas C. Curtis
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire, USA
| | - Joshua DiNapoli
- Sanofi Pasteur Inc., Research North America, Cambridge, Massachusetts, USA
| | - Simon Delagrave
- Sanofi Pasteur Inc., Research North America, Cambridge, Massachusetts, USA
| | - Ted M. Ross
- Center for Vaccines and Immunology, University of Georgia, Athens, Georgia, USA
| | - Gregory C. Ippolito
- Department of Molecular Biosciences
- Department of Oncology, Dell Medical School, The University of Texas at Austin, Austin, Texas, USA
| | - Harry Kleanthous
- Sanofi Pasteur Inc., Research North America, Cambridge, Massachusetts, USA
| | - Jiwon Lee
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire, USA
| | - George Georgiou
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas, USA
- Department of Chemical Engineering
- Department of Molecular Biosciences
- Institute for Cellular and Molecular Biology, and
- Department of Oncology, Dell Medical School, The University of Texas at Austin, Austin, Texas, USA
| |
Collapse
|
19
|
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
|
20
|
Guthmiller JJ, Utset HA, Wilson PC. B Cell Responses against Influenza Viruses: Short-Lived Humoral Immunity against a Life-Long Threat. Viruses 2021; 13:965. [PMID: 34067435 PMCID: PMC8224597 DOI: 10.3390/v13060965] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 05/17/2021] [Accepted: 05/19/2021] [Indexed: 12/25/2022] Open
Abstract
Antibodies are critical for providing protection against influenza virus infections. However, protective humoral immunity against influenza viruses is limited by the antigenic drift and shift of the major surface glycoproteins, hemagglutinin and neuraminidase. Importantly, people are exposed to influenza viruses throughout their life and tend to reuse memory B cells from prior exposure to generate antibodies against new variants. Despite this, people tend to recall memory B cells against constantly evolving variable epitopes or non-protective antigens, as opposed to recalling them against broadly neutralizing epitopes of hemagglutinin. In this review, we discuss the factors that impact the generation and recall of memory B cells against distinct viral antigens, as well as the immunological limitations preventing broadly neutralizing antibody responses. Lastly, we discuss how next-generation vaccine platforms can potentially overcome these obstacles to generate robust and long-lived protection against influenza A viruses.
Collapse
Affiliation(s)
- Jenna J. Guthmiller
- Section of Rheumatology, Department of Medicine, University of Chicago, Chicago, IL 60637, USA; (H.A.U.); (P.C.W.)
| | - Henry A. Utset
- Section of Rheumatology, Department of Medicine, University of Chicago, Chicago, IL 60637, USA; (H.A.U.); (P.C.W.)
| | - Patrick C. Wilson
- Section of Rheumatology, Department of Medicine, University of Chicago, Chicago, IL 60637, USA; (H.A.U.); (P.C.W.)
- Committee on Immunology, University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
21
|
Matrix Protein 2 Extracellular Domain-Specific Monoclonal Antibodies Are an Effective and Potentially Universal Treatment for Influenza A. J Virol 2021; 95:JVI.01027-20. [PMID: 33268521 PMCID: PMC8092830 DOI: 10.1128/jvi.01027-20] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Influenza virus infection causes significant morbidity and mortality worldwide. Humans fail to make a universally protective memory immune response to influenza A. Hemagglutinin and Neuraminidase undergo antigenic drift and shift, resulting in new influenza A strains to which humans are naive. Seasonal vaccines are often ineffective and escape mutants have been reported to all treatments for influenza A. In the absence of a universal influenza A vaccine or treatment, influenza A will remain a significant threat to human health. The extracellular domain of the M2-ion channel (M2e) is an ideal antigenic target for a universal therapeutic agent, as it is highly conserved across influenza A serotypes, has a low mutation rate, and is essential for viral entry and replication. Previous M2e-specific monoclonal antibodies (M2e-MAbs) show protective potential against influenza A, however, they are either strain specific or have limited efficacy. We generated seven murine M2e-MAbs and utilized in vitro and in vivo assays to validate the specificity of our novel M2e-MAbs and to explore the universality of their protective potential. Our data shows our M2e-MAbs bind to M2e peptide, HEK cells expressing the M2 channel, as well as, influenza virions and MDCK-ATL cells infected with influenza viruses of multiple serotypes. Our antibodies significantly protect highly influenza A virus susceptible BALB/c mice from lethal challenge with H1N1 A/PR/8/34, pH1N1 A/CA/07/2009, H5N1 A/Vietnam/1203/2004, and H7N9 A/Anhui/1/2013 by improving survival rates and weight loss. Based on these results, at least four of our seven M2e-MAbs show strong potential as universal influenza A treatments.IMPORTANCE Despite a seasonal vaccine and multiple therapeutic treatments, Influenza A remains a significant threat to human health. The biggest obstacle is producing a vaccine or treatment for influenza A is their universality or efficacy against not only seasonal variances in the influenza virus, but also against all human, avian, and swine serotypes and, therefore, potential pandemic strains. M2e has huge potential as a target for a vaccine or treatment against influenza A. It is the most conserved external protein on the virus. Antibodies against M2e have made it to clinical trials, but not succeeded. Here, we describe novel M2e antibodies produced in mice that are not only protective at low doses, but that we extensively test to determine their universality and found to be cross protective against all strains tested. Additionally, our work begins to elucidate the critical role of isotype for an influenza A monoclonal antibody therapeutic.
Collapse
|
22
|
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: 8] [Impact Index Per Article: 2.0] [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
|
23
|
Zottig X, Al-Halifa S, Côté-Cyr M, Calzas C, Le Goffic R, Chevalier C, Archambault D, Bourgault S. Self-assembled peptide nanorod vaccine confers protection against influenza A virus. Biomaterials 2021; 269:120672. [PMID: 33476893 DOI: 10.1016/j.biomaterials.2021.120672] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 12/29/2020] [Accepted: 01/06/2021] [Indexed: 12/23/2022]
Abstract
Proteinaceous nanostructures have emerged as a promising strategy to develop safe and efficient subunit vaccines. The ability of synthetic β-sheet self-assembling peptides to stabilize antigenic determinants and to potentiate the epitope-specific immune responses have highlighted their potential as an immunostimulating platform for antigen delivery. Nonetheless, the intrinsic polymorphism of the resulting cross-β fibrils, their length in the microscale and their close structural similarity with pathological amyloids could limit their usage in vaccinology. In this study, we harnessed electrostatic capping motifs to control the self-assembly of a chimeric peptide comprising a 10-mer β-sheet sequence and a highly conserved epitope derived from the influenza A virus (M2e). Self-assembly led to the formation of 100-200 nm long uniform nanorods (NRs) displaying the M2e epitope on their surface. These cross-β assemblies differed from prototypical amyloid fibrils owing to low polydispersity, short length, non-binding to thioflavin T and Congo Red dyes, and incapacity to seed homologous amyloid assembly. M2e-NRs were efficiently uptaken by antigen presenting cells and the cross-β quaternary architecture activated the Toll-like receptor 2 and stimulated dendritic cells. Mice subcutaneous immunization revealed a robust M2e-specific IgG response, which was dependent on self-assembly into NRs. Upon intranasal immunization in combination with the polymeric adjuvant montanide gel, M2e-NRs conferred complete protection with absence of clinical signs against a lethal experimental infection with the H1N1 influenza A virus. These findings indicate that by acting as an immunostimulator and delivery system, synthetic peptide-based NRs constitute a versatile self-adjuvanted nanoplatform for the delivery of subunit vaccines.
Collapse
Affiliation(s)
- Ximena Zottig
- Chemistry Department, Université du Québec à Montréal, Montreal, Canada; Quebec Network for Research on Protein Function, Engineering and Applications (PROTEO), Quebec, Canada; Department of Biological Sciences, Université du Québec à Montréal, Montreal, Canada; The Swine and Poultry Infectious Diseases Research Centre (CRIPA), Sainte-Hyacinthe, Canada
| | - Soultan Al-Halifa
- Chemistry Department, Université du Québec à Montréal, Montreal, Canada; Quebec Network for Research on Protein Function, Engineering and Applications (PROTEO), Quebec, Canada; Department of Biological Sciences, Université du Québec à Montréal, Montreal, Canada; The Swine and Poultry Infectious Diseases Research Centre (CRIPA), Sainte-Hyacinthe, Canada
| | - Mélanie Côté-Cyr
- Chemistry Department, Université du Québec à Montréal, Montreal, Canada; Quebec Network for Research on Protein Function, Engineering and Applications (PROTEO), Quebec, Canada; Department of Biological Sciences, Université du Québec à Montréal, Montreal, Canada; The Swine and Poultry Infectious Diseases Research Centre (CRIPA), Sainte-Hyacinthe, Canada
| | - Cynthia Calzas
- UR892 VIM, Equipe Virus Influenza, Université Paris-Saclay, INRAE, Jouy-en-Josas, France
| | - Ronan Le Goffic
- UR892 VIM, Equipe Virus Influenza, Université Paris-Saclay, INRAE, Jouy-en-Josas, France
| | - Christophe Chevalier
- UR892 VIM, Equipe Virus Influenza, Université Paris-Saclay, INRAE, Jouy-en-Josas, France
| | - Denis Archambault
- Department of Biological Sciences, Université du Québec à Montréal, Montreal, Canada; The Swine and Poultry Infectious Diseases Research Centre (CRIPA), Sainte-Hyacinthe, Canada.
| | - Steve Bourgault
- Chemistry Department, Université du Québec à Montréal, Montreal, Canada; Quebec Network for Research on Protein Function, Engineering and Applications (PROTEO), Quebec, Canada; The Swine and Poultry Infectious Diseases Research Centre (CRIPA), Sainte-Hyacinthe, Canada.
| |
Collapse
|
24
|
Frank K, Paust S. Dynamic Natural Killer Cell and T Cell Responses to Influenza Infection. Front Cell Infect Microbiol 2020; 10:425. [PMID: 32974217 PMCID: PMC7461885 DOI: 10.3389/fcimb.2020.00425] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 07/10/2020] [Indexed: 12/12/2022] Open
Abstract
Influenza viruses have perplexed scientists for over a hundred years. Yearly vaccines limit their spread, but they do not prevent all infections. Therapeutic treatments for those experiencing severe infection are limited; further advances are held back by insufficient understanding of the fundamental immune mechanisms responsible for immunopathology. NK cells and T cells are essential in host responses to influenza infection. They produce immunomodulatory cytokines and mediate the cytotoxic response to infection. An imbalance in NK and T cell responses can lead to two outcomes: excessive inflammation and tissue damage or insufficient anti-viral functions and uncontrolled infection. The main cause of death in influenza patients is the former, mediated by hyperinflammatory responses termed “cytokine storm.” NK cells and T cells contribute to cytokine storm, but they are also required for viral clearance. Many studies have attempted to distinguish protective and pathogenic components of the NK cell and T cell influenza response, but it has become clear that they are dynamic and integrated processes. This review will analyze how NK cell and T cell effector functions during influenza infection affect the host response and correlate with morbidity and mortality outcomes.
Collapse
Affiliation(s)
- Kayla Frank
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, United States.,The Skaggs Graduate Program in Chemical and Biological Sciences, The Scripps Research Institute, La Jolla, CA, United States
| | - Silke Paust
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, United States.,The Skaggs Graduate Program in Chemical and Biological Sciences, The Scripps Research Institute, La Jolla, CA, United States
| |
Collapse
|
25
|
Prospects and Challenges in the Development of Universal Influenza Vaccines. Vaccines (Basel) 2020; 8:vaccines8030361. [PMID: 32640619 PMCID: PMC7563311 DOI: 10.3390/vaccines8030361] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 06/29/2020] [Accepted: 06/30/2020] [Indexed: 01/19/2023] Open
Abstract
Current influenza vaccines offer suboptimal protection and depend on annual reformulation and yearly administration. Vaccine technology has rapidly advanced during the last decade, facilitating development of next-generation influenza vaccines that can target a broader range of influenza viruses. The development and licensure of a universal influenza vaccine could provide a game changing option for the control of influenza by protecting against all influenza A and B viruses. Here we review important findings and considerations regarding the development of universal influenza vaccines and what we can learn from this moving forward with a SARS-CoV-2 vaccine design.
Collapse
|
26
|
Vanderven HA, Kent SJ. The protective potential of Fc-mediated antibody functions against influenza virus and other viral pathogens. Immunol Cell Biol 2020; 98:253-263. [PMID: 31914207 DOI: 10.1111/imcb.12312] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 12/20/2019] [Accepted: 12/23/2019] [Indexed: 01/08/2023]
Abstract
In recent years, there has been a renewed interest in utilizing antibody fragment crystallizable (Fc) functions to prevent and control viral infections. The protective and therapeutic potential of Fc-mediated antibody functions have been assessed for some clinically important human viruses, including HIV, hemorrhagic fever viruses and influenza virus. There is mounting evidence that influenza-specific antibodies with Fc-mediated functions, such as antibody-dependent cellular cytotoxicity and antibody-dependent phagocytosis, can aid in the clearance of influenza virus infection. Recent influenza challenge studies and intravenous immunoglobulin G therapy studies in humans suggest a protective role for Fc effector functions in vivo. Broadly reactive influenza antibodies with Fc-mediated functions are prevalent in the human population and could inform the development of a universally protective influenza vaccine or therapy. In this review, we explore the utility of antibodies with Fc-mediated effector functions against viral infections with a focus on influenza virus.
Collapse
Affiliation(s)
- Hillary A Vanderven
- Discipline of Biomedicine, College of Public Health, Medical and Veterinary Sciences, James Cook University, Douglas, QLD, Australia.,Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC, Australia
| | - Stephen J Kent
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC, Australia.,Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, University of Melbourne, Parkville, VIC, Australia.,Melbourne Sexual Health Centre, Department of Infectious Diseases, Alfred Health, Central Clinical School, Monash University, Clayton, VIC, Australia
| |
Collapse
|
27
|
Terrén I, Orrantia A, Mikelez-Alonso I, Vitallé J, Zenarruzabeitia O, Borrego F. NK Cell-Based Immunotherapy in Renal Cell Carcinoma. Cancers (Basel) 2020; 12:cancers12020316. [PMID: 32013092 PMCID: PMC7072691 DOI: 10.3390/cancers12020316] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 01/14/2020] [Accepted: 01/23/2020] [Indexed: 02/06/2023] Open
Abstract
Natural killer (NK) cells are cytotoxic lymphocytes that are able to kill tumor cells without prior sensitization. It has been shown that NK cells play a pivotal role in a variety of cancers, highlighting their relevance in tumor immunosurveillance. NK cell infiltration has been reported in renal cell carcinoma (RCC), the most frequent kidney cancer in adults, and their presence has been associated with patients’ survival. However, the role of NK cells in this disease is not yet fully understood. In this review, we summarize the biology of NK cells and the mechanisms through which they are able to recognize and kill tumor cells. Furthermore, we discuss the role that NK cells play in renal cell carcinoma, and review current strategies that are being used to boost and exploit their cytotoxic capabilities.
Collapse
Affiliation(s)
- Iñigo Terrén
- Immunopathology Group, Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain; (I.T.); (A.O.); (I.M.-A.); (J.V.); (O.Z.)
| | - Ane Orrantia
- Immunopathology Group, Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain; (I.T.); (A.O.); (I.M.-A.); (J.V.); (O.Z.)
| | - Idoia Mikelez-Alonso
- Immunopathology Group, Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain; (I.T.); (A.O.); (I.M.-A.); (J.V.); (O.Z.)
- CIC biomaGUNE, 20014 Donostia-San Sebastián, Spain
| | - Joana Vitallé
- Immunopathology Group, Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain; (I.T.); (A.O.); (I.M.-A.); (J.V.); (O.Z.)
| | - Olatz Zenarruzabeitia
- Immunopathology Group, Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain; (I.T.); (A.O.); (I.M.-A.); (J.V.); (O.Z.)
| | - Francisco Borrego
- Immunopathology Group, Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain; (I.T.); (A.O.); (I.M.-A.); (J.V.); (O.Z.)
- Ikerbasque, Basque Foundation for Science, 48013 Bilbao, Spain
- Correspondence: ; Tel.: +34-94-600-6000 (ext. 7079)
| |
Collapse
|
28
|
Mezhenskaya D, Isakova-Sivak I, Rudenko L. M2e-based universal influenza vaccines: a historical overview and new approaches to development. J Biomed Sci 2019; 26:76. [PMID: 31629405 PMCID: PMC6800501 DOI: 10.1186/s12929-019-0572-3] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 10/01/2019] [Indexed: 01/04/2023] Open
Abstract
The influenza A virus was isolated for the first time in 1931, and the first attempts to develop a vaccine against the virus began soon afterwards. In addition to causing seasonal epidemics, influenza viruses can cause pandemics at random intervals, which are very hard to predict. Vaccination is the most effective way of preventing the spread of influenza infection. However, seasonal vaccination is ineffective against pandemic influenza viruses because of antigenic differences, and it takes approximately six months from isolation of a new virus to develop an effective vaccine. One of the possible ways to fight the emergence of pandemics may be by using a new type of vaccine, with a long and broad spectrum of action. The extracellular domain of the M2 protein (M2e) of influenza A virus is a conservative region, and an attractive target for a universal influenza vaccine. This review gives a historical overview of the study of M2 protein, and summarizes the latest developments in the preparation of M2e-based universal influenza vaccines.
Collapse
Affiliation(s)
- Daria Mezhenskaya
- Department of Virology, Institute of Experimental Medicine, 12 Acad. Pavlov Street, St. Petersburg, 197376, Russia
| | - Irina Isakova-Sivak
- Department of Virology, Institute of Experimental Medicine, 12 Acad. Pavlov Street, St. Petersburg, 197376, Russia.
| | - Larisa Rudenko
- Department of Virology, Institute of Experimental Medicine, 12 Acad. Pavlov Street, St. Petersburg, 197376, Russia
| |
Collapse
|
29
|
Padilla-Quirarte HO, Lopez-Guerrero DV, Gutierrez-Xicotencatl L, Esquivel-Guadarrama F. Protective Antibodies Against Influenza Proteins. Front Immunol 2019; 10:1677. [PMID: 31379866 PMCID: PMC6657620 DOI: 10.3389/fimmu.2019.01677] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2019] [Accepted: 07/04/2019] [Indexed: 12/21/2022] Open
Abstract
The influenza A virus infection continues to be a threat to the human population. The seasonal variation of the virus and the likelihood of periodical pandemics caused by completely new virus strains make it difficult to produce vaccines that efficiently protect against this infection. Antibodies (Abs) are very important in preventing the infection and in blocking virus propagation once the infection has taken place. However, the precise protection mechanism provided by these Abs still needs to be established. Furthermore, most research has focused on Abs directed to the globular head domain of hemagglutinin (HA). However, other domains of HA (like the stem) and other proteins are also able to elicit protective Ab responses. In this article, we review the current knowledge about the role of both neutralizing and non-neutralizing anti-influenza proteins Abs that play a protective role during infection or vaccination.
Collapse
Affiliation(s)
- Herbey O Padilla-Quirarte
- LIV, Facultad de Medicina, Universidad Autonoma del Estado de Morelos, Cuernavaca, Mexico.,Instituto de Biotecnologia, Universidad Nacional Autonoma de Mexico, Cuernavaca, Mexico
| | | | | | | |
Collapse
|
30
|
Targeting M2e to DEC-205 induces an enhanced serum antibody-dependent heterosubtypic protection against influenza A virus infection. Vaccine 2019; 37:2624-2633. [DOI: 10.1016/j.vaccine.2019.02.050] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 01/23/2019] [Accepted: 02/15/2019] [Indexed: 01/07/2023]
|
31
|
Estrada LD, Schultz-Cherry S. Development of a Universal Influenza Vaccine. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2019; 202:392-398. [PMID: 30617121 PMCID: PMC6327971 DOI: 10.4049/jimmunol.1801054] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 08/30/2018] [Indexed: 12/17/2022]
Abstract
The severity of the 2017-18 influenza season, combined with the low efficacy for some vaccine components, highlights the need to improve our current seasonal influenza vaccine. Thus, the National Institute of Allergy and Infectious Diseases recently announced a strategic plan to improve current influenza vaccines and eventually develop a "universal" influenza vaccine. This review will highlight the many different strategies being undertaken in pursuit of this goal and the exciting advances made by the influenza community. There is no doubt that an improved influenza vaccine is on the horizon.
Collapse
Affiliation(s)
- Leonardo D Estrada
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN 38105
| | - Stacey Schultz-Cherry
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN 38105
| |
Collapse
|
32
|
Sicca F, Neppelenbroek S, Huckriede A. Effector mechanisms of influenza-specific antibodies: neutralization and beyond. Expert Rev Vaccines 2018; 17:785-795. [PMID: 30145912 DOI: 10.1080/14760584.2018.1516553] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Antibodies directed against influenza virus execute their protective function by exploiting a variety of effector mechanisms. Neutralizing antibodies have been thoroughly studied because of their pivotal role in preventing influenza virus infection and their presence in host serum is correlated with protection. Influenza antibodies can also exploit non-neutralizing effector mechanisms, which until recently have been largely overlooked. AREAS COVERED Here, we discuss the antibody response to influenza virus in its entire breadth. Neutralizing antibodies mostly target variable epitopes on influenza surface proteins and interfere with virus binding, fusion, or egress. Non-neutralizing antibodies instead usually target conserved epitopes which can be located on surface as well as internal proteins. They drive viral clearance via interaction of their Fc region with components of the innate immune system such as immune effector cells (e.g. NK cells, macrophages) or the complement system. EXPERT COMMENTARY Recent research has unraveled that influenza-specific antibodies target multiple proteins and make use of diverse effector mechanisms. Often these antibodies are cross-reactive among virus strains of the same subtype or even between subtypes. As such they are induced early in life and are boosted by regular encounters with virus or vaccine. Designing strategies to optimally exploit these pre-existing antibodies may represent the key for the development of new broadly protective influenza vaccines.
Collapse
Affiliation(s)
- Federica Sicca
- a Department of Medical Microbiology , University of Groningen, University Medical Center Groningen , Groningen , The Netherlands
| | - Sam Neppelenbroek
- a Department of Medical Microbiology , University of Groningen, University Medical Center Groningen , Groningen , The Netherlands
| | - Anke Huckriede
- a Department of Medical Microbiology , University of Groningen, University Medical Center Groningen , Groningen , The Netherlands
| |
Collapse
|
33
|
Tsybalova LM, Stepanova LA, Shuklina MA, Mardanova ES, Kotlyarov RY, Potapchuk MV, Petrov SA, Blokhina EA, Ravin NV. Combination of M2e peptide with stalk HA epitopes of influenza A virus enhances protective properties of recombinant vaccine. PLoS One 2018; 13:e0201429. [PMID: 30138320 PMCID: PMC6107133 DOI: 10.1371/journal.pone.0201429] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 07/16/2018] [Indexed: 12/14/2022] Open
Abstract
Background Influenza infection could be more effectively controlled if a multi-purpose vaccine with the ability to induce responses against most, or all, influenza A subtypes could be generated. Conserved viral proteins are a promising basis for the creation of a broadly protective vaccine. In the present study, the immunogenicity and protective properties of three recombinant proteins (vaccine candidates), comprising conserved viral proteins fused with bacterial flagellin, were compared. Methods Balb/c mice were immunized intranasally with recombinant proteins comprising either one viral protein (the ectodomain of the M2 protein, ‘M2e’) or two viral proteins (M2e and the hemagglutinin second subunit ‘HA2’ epitope) genetically fused with flagellin. Further, two different consensus variants of HA2 were used. Therefore, three experimental positives were used in addition to the negative control (Flg-his). The mucosal, humoral, and T-cell immune responses to these constructs were evaluated. Result We have demonstrated that insertion of the HA2 consensus polypeptide (aa 76–130), derived from either the first (HA2-1) or second (HA2-2) virus phylogenetic group, into the recombinant Flg4M2e protein significantly enhanced its immunogenicity and protective properties. Intranasal administration of the vaccine candidates (Flg-HA2-2-4M2e or Flg-HA2-1-4M2e) induced considerable mucosal and systemic responses directed at both the M2e-protein and, in general, the influenza A virus. However, the immune response elicited by the Flg-HA2-1-4M2e protein was weaker than the one generated by Flg-HA2-2-4M2e. These recombinant proteins containing both viral peptides provide complete protection from lethal challenge with various influenza viruses: A/H3N2; A/H2N2; and A/H5N1. Conclusion This study demonstrates that the intranasal administration of Flg-HA2-2-4M2e recombinant protein induces a strong immune response which provides broad protection against various influenza viruses. This construct is therefore a strong candidate for development as a universal vaccine.
Collapse
Affiliation(s)
- Liudmila M. Tsybalova
- Department of Vaccinology, Smorodintsev Research Institute of Influenza, Ministry of Health of the Russian Federation, St. Petersburg, Russia
- * E-mail:
| | - Liudmila A. Stepanova
- Department of Vaccinology, Smorodintsev Research Institute of Influenza, Ministry of Health of the Russian Federation, St. Petersburg, Russia
| | - Marina A. Shuklina
- Department of Vaccinology, Smorodintsev Research Institute of Influenza, Ministry of Health of the Russian Federation, St. Petersburg, Russia
| | - Eugenia S. Mardanova
- Institute of Bioengineering, Research Center of Biotechnology, Russian Academy of Sciences, Moscow, Russia
| | - Roman Y. Kotlyarov
- Institute of Bioengineering, Research Center of Biotechnology, Russian Academy of Sciences, Moscow, Russia
| | - Marina V. Potapchuk
- Department of Vaccinology, Smorodintsev Research Institute of Influenza, Ministry of Health of the Russian Federation, St. Petersburg, Russia
| | - Sergei A. Petrov
- Department of Vaccinology, Smorodintsev Research Institute of Influenza, Ministry of Health of the Russian Federation, St. Petersburg, Russia
| | - Elena A. Blokhina
- Institute of Bioengineering, Research Center of Biotechnology, Russian Academy of Sciences, Moscow, Russia
| | - Nikolai V. Ravin
- Institute of Bioengineering, Research Center of Biotechnology, Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
34
|
The Potential Role of Fc-Receptor Functions in the Development of a Universal Influenza Vaccine. Vaccines (Basel) 2018; 6:vaccines6020027. [PMID: 29772781 PMCID: PMC6027188 DOI: 10.3390/vaccines6020027] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 05/08/2018] [Accepted: 05/10/2018] [Indexed: 01/02/2023] Open
Abstract
Despite global vaccination efforts, influenza virus continues to cause yearly epidemics and periodic pandemics throughout most of the world. Many of us consider the generation of broader, potent and long-lasting immunity against influenza viruses as critical in curtailing the global health and economic impact that influenza currently plays. To date, classical vaccinology has relied on the generation of neutralizing antibodies as the benchmark to measure vaccine effectiveness. However, recent developments in numerous related fields of biomedical research including, HIV, HSV and DENV have emphasized the importance of Fc-mediate effector functions in pathogenesis and immunity. The concept of Fc effector functions in contributing to protection from illness is not a new concept and has been investigated in the field for over four decades. However, in recent years the application and study of Fc effector functions has become revitalized with new knowledge and technologies to characterize their potential importance in immunity. In this perspective, we describe the current state of the field of Influenza Fc effector functions and discuss its potential utility in universal vaccine design in the future.
Collapse
|
35
|
Saletti G, Gerlach T, Rimmelzwaan GF. Influenza vaccines: 'tailor-made' or 'one fits all'. Curr Opin Immunol 2018; 53:102-110. [PMID: 29734023 DOI: 10.1016/j.coi.2018.04.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 04/12/2018] [Accepted: 04/16/2018] [Indexed: 02/02/2023]
Abstract
Currently used inactivated influenza vaccines aim at the induction of virus-neutralizing antibodies directed to the variable head domain of the viral hemagglutinin. Although these vaccines are effective against antigenically matching virus strains, they offer little protection against antigenically distinct drift variants or potentially pandemic viruses of alternative subtypes. In the last decades, the threat of novel influenza pandemics has sparked research efforts to develop vaccines that induce more broadly protective immunity. Here, we discuss the immune responses induced by conventional 'tailor-made' inactivated and live influenza vaccines and novel 'one fits all' candidate vaccines able to induce cross-reactive virus-specific antibody and T cell responses and to afford protection to a wider range of influenza viruses.
Collapse
Affiliation(s)
- Giulietta Saletti
- University of Veterinary Medicine (TiHo), Research Center for Emerging Infections and Zoonoses (RIZ), Bünteweg 17, 30559 Hannover, Germany
| | - Thomas Gerlach
- University of Veterinary Medicine (TiHo), Research Center for Emerging Infections and Zoonoses (RIZ), Bünteweg 17, 30559 Hannover, Germany
| | - Guus F Rimmelzwaan
- University of Veterinary Medicine (TiHo), Research Center for Emerging Infections and Zoonoses (RIZ), Bünteweg 17, 30559 Hannover, Germany.
| |
Collapse
|
36
|
Terrén I, Mikelez I, Odriozola I, Gredilla A, González J, Orrantia A, Vitallé J, Zenarruzabeitia O, Borrego F. Implication of Interleukin-12/15/18 and Ruxolitinib in the Phenotype, Proliferation, and Polyfunctionality of Human Cytokine-Preactivated Natural Killer Cells. Front Immunol 2018; 9:737. [PMID: 29713323 PMCID: PMC5911648 DOI: 10.3389/fimmu.2018.00737] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 03/26/2018] [Indexed: 12/30/2022] Open
Abstract
A brief in vitro stimulation of natural killer (NK) cells with interleukin (IL)-12, IL-15, and IL-18 endow them a memory-like behavior, characterized by higher effector responses when they are restimulated after a resting period of time. These preactivated NK cells, also known as cytokine-induced memory-like (CIML) NK cells, have several properties that make them a promising tool in cancer immunotherapy. In the present study, we have described the effect that different combinations of IL-12, IL-15, and IL-18 have on the generation of human CIML NK cells. Our data points to a major contribution of IL-15 to CIML NK cell-mediated cytotoxicity against target cells. However, the synergistic effect of the three cytokines grant them the best polyfunctional profile, that is, cells that simultaneously degranulate (CD107a) and produce multiple cytokines and chemokines such as interferon γ, tumor necrosis factor α, and C-C motif chemokine ligand 3. We have also analyzed the involvement of each cytokine and their combinations in the expression of homing receptors CXCR4 and CD62L, as well as the expression of CD25 and IL-2-induced proliferation. Furthermore, we have tested the effects of the Jak1/2 inhibitor ruxolitinib in the generation of CIML NK cells. We found that ruxolitinib-treated CIML NK cells expressed lower levels of CD25 than non-treated CIML NK cells, but exhibited similar proliferation in response to IL-2. In addition, we have also found that ruxolitinib-treated NK cells displayed reduced effector functions after the preactivation, which can be recovered after a 4 days expansion phase in the presence of low doses of IL-2. Altogether, our results describe the impact that each cytokine and the Jak1/2 pathway have in the phenotype, IL-2-induced proliferation, and effector functions of human CIML NK cells.
Collapse
Affiliation(s)
- Iñigo Terrén
- Immunopathology Group, BioCruces Health Research Institute, Barakaldo, Spain
| | - Idoia Mikelez
- Immunopathology Group, BioCruces Health Research Institute, Barakaldo, Spain
- CIC biomaGUNE, Donostia-San Sebastián, Spain
| | - Irati Odriozola
- Immunopathology Group, BioCruces Health Research Institute, Barakaldo, Spain
| | - Andrea Gredilla
- Immunopathology Group, BioCruces Health Research Institute, Barakaldo, Spain
| | - Javier González
- Immunopathology Group, BioCruces Health Research Institute, Barakaldo, Spain
| | - Ane Orrantia
- Immunopathology Group, BioCruces Health Research Institute, Barakaldo, Spain
| | - Joana Vitallé
- Immunopathology Group, BioCruces Health Research Institute, Barakaldo, Spain
| | | | - Francisco Borrego
- Immunopathology Group, BioCruces Health Research Institute, Barakaldo, Spain
- Ikerbasque, Basque Foundation for Science, Bilbao, Spain
- Basque Center for Transfusion and Human Tissues, Galdakao, Spain
| |
Collapse
|
37
|
Sautto GA, Kirchenbaum GA, Ross TM. Towards a universal influenza vaccine: different approaches for one goal. Virol J 2018; 15:17. [PMID: 29370862 PMCID: PMC5785881 DOI: 10.1186/s12985-017-0918-y] [Citation(s) in RCA: 132] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 12/21/2017] [Indexed: 12/14/2022] Open
Abstract
Influenza virus infection is an ongoing health and economic burden causing epidemics with pandemic potential, affecting 5–30% of the global population annually, and is responsible for millions of hospitalizations and thousands of deaths each year. Annual influenza vaccination is the primary prophylactic countermeasure aimed at limiting influenza burden. However, the effectiveness of current influenza vaccines are limited because they only confer protective immunity when there is antigenic similarity between the selected vaccine strains and circulating influenza isolates. The major targets of the antibody response against influenza virus are the surface glycoprotein antigens hemagglutinin (HA) and neuraminidase (NA). Hypervariability of the amino acid sequences encoding HA and NA is largely responsible for epidemic and pandemic influenza outbreaks, and are the consequence of antigenic drift or shift, respectively. For this reason, if an antigenic mismatch exists between the current vaccine and circulating influenza isolates, vaccinated people may not be afforded complete protection. There is currently an unmet need to develop an effective “broadly-reactive” or “universal” influenza vaccine capable of conferring protection against both seasonal and newly emerging pre-pandemic strains. A number of novel influenza vaccine approaches are currently under evaluation. One approach is the elicitation of an immune response against the “Achille’s heel” of the virus, i.e. conserved viral proteins or protein regions shared amongst seasonal and pre-pandemic strains. Alternatively, other approaches aim toward eliciting a broader immune response capable of conferring protection against the diversity of currently circulating seasonal influenza strains. In this review, the most promising under-development universal vaccine approaches are discussed with an emphasis on those targeting the HA glycoprotein. In particular, their strengths and potential short-comings are discussed. Ultimately, the upcoming clinical evaluation of these universal vaccine approaches will be fundamental to determine their effectiveness against preventing influenza virus infection and/or reducing transmission and disease severity.
Collapse
Affiliation(s)
- Giuseppe A Sautto
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, USA
| | - Greg A Kirchenbaum
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, USA
| | - Ted M Ross
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, USA. .,Department of Infectious Diseases, University of Georgia, Athens, GA, USA.
| |
Collapse
|
38
|
Morrison BJ, Roman JA, Luke TC, Nagabhushana N, Raviprakash K, Williams M, Sun P. Antibody-dependent NK cell degranulation as a marker for assessing antibody-dependent cytotoxicity against pandemic 2009 influenza A(H1N1) infection in human plasma and influenza-vaccinated transchromosomic bovine intravenous immunoglobulin therapy. J Virol Methods 2017. [PMID: 28624584 PMCID: PMC7113754 DOI: 10.1016/j.jviromet.2017.06.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Assay that assesses influenza antibodies capable of NK cell degranulation. Description of NK cell degranulation titer determination by CD107a expression. Positive correlation between influenza HAI titers and NK cell degranulation titers. Transchromosomic bovine intravenous immunoglobulin therapy has high NK cell titer.
This study describes an antibody-dependent NK cell degranulation assay, as a biomarker to assess antibody-dependent cellular cytotoxicity (ADCC) response in influenza plasma and for antibody therapies against influenza infection. The concentration of neutralizing antibodies (NAbs) against the hemagglutinin receptor of influenza viruses is a current determinant in protection against infection, particularly following receipt of the seasonal influenza vaccine. However, this is a limited assessment of protection, because: (i) NAb titers that incur full protection vary; and (ii) NAb titers do not account for the entire breadth of antibody responses against viral infection. Previous reports have indicated that antibodies that prime ADCC play a vital role in controlling influenza infections, and thus should be quantified for assessing protection against influenza. This report demonstrates a non-radioactive assay that assesses NK cell activation as a marker of ADCC, in which NK cells interact with opsonized viral antigen expressed on the surface of infected Raji target cells resulting in effector cell degranulation (surrogate CD107a expression). A positive correlation was determined between HAI titers and sustained NK cell activation, although NK cell activation was seen in plasma samples with HAI titers below 40 and varied amongst samples with high HAI titers. Furthermore, sustained NK cell degranulation was determined for influenza-vaccinated transchromosomic bovine intravenous immunoglobulin, indicating the potential utility of this therapy for influenza treatment. We conclude that this assay is reproducible and relevant.
Collapse
Affiliation(s)
- Brian J Morrison
- Viral and Rickettsial Diseases Department, Infectious Diseases Directorate, Naval Medical Research Center, Silver Spring, MD, USA.
| | - Jessica A Roman
- Viral and Rickettsial Diseases Department, Infectious Diseases Directorate, Naval Medical Research Center, Silver Spring, MD, USA
| | - Thomas C Luke
- Viral and Rickettsial Diseases Department, Infectious Diseases Directorate, Naval Medical Research Center, Silver Spring, MD, USA
| | - Nishith Nagabhushana
- Viral and Rickettsial Diseases Department, Infectious Diseases Directorate, Naval Medical Research Center, Silver Spring, MD, USA
| | - Kanakatte Raviprakash
- Viral and Rickettsial Diseases Department, Infectious Diseases Directorate, Naval Medical Research Center, Silver Spring, MD, USA
| | - Maya Williams
- Viral and Rickettsial Diseases Department, Infectious Diseases Directorate, Naval Medical Research Center, Silver Spring, MD, USA
| | - Peifang Sun
- Viral and Rickettsial Diseases Department, Infectious Diseases Directorate, Naval Medical Research Center, Silver Spring, MD, USA
| |
Collapse
|
39
|
Jegaskanda S, Vanderven HA, Wheatley AK, Kent SJ. Fc or not Fc; that is the question: Antibody Fc-receptor interactions are key to universal influenza vaccine design. Hum Vaccin Immunother 2017; 13:1-9. [PMID: 28332900 DOI: 10.1080/21645515.2017.1290018] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
A universal vaccine that provides long-lasting protection from both epidemic and pandemic influenza viruses remains the "holy grail" of influenza vaccine research. Though virus neutralization assays are the current benchmark of measuring vaccine effectiveness, it is clear that Fc-receptor functions can drastically improve the effectiveness of antibodies and vaccines in vivo. Antibodies that kill virus-infected cells and/or elicit an antiviral environment, termed antibody-dependent cellular cytotoxicity (ADCC)-mediating antibodies, provide a link between the innate and adaptive immune response. New technologies allowing the rapid isolation and characterization of monoclonal antibodies (mAb) have yielded a plethora of mAbs which target conserved regions of influenza virus, such as the hemagglutinin (HA) stem region. Many such mAbs have been used to gain a better understanding of Fc-receptor functions in vivo. In parallel, several studies have characterized the induction of polyclonal ADCC following influenza vaccination and infection in humans. Taken together, these studies suggest that ADCC-mediating antibodies (ADCC-Abs) significantly contribute to host immunity against influenza virus and may be a mechanism to exploit for rational vaccine and therapeutic design. We discuss recent research on influenza-specific ADCC and potential future avenues to extend our understanding.
Collapse
Affiliation(s)
- Sinthujan Jegaskanda
- a Department of Microbiology and Immunology , University of Melbourne, Peter Doherty Institute for Infection and Immunity , Melbourne , Victoria , Australia
| | - Hillary A Vanderven
- a Department of Microbiology and Immunology , University of Melbourne, Peter Doherty Institute for Infection and Immunity , Melbourne , Victoria , Australia
| | - Adam K Wheatley
- a Department of Microbiology and Immunology , University of Melbourne, Peter Doherty Institute for Infection and Immunity , Melbourne , Victoria , Australia
| | - Stephen J Kent
- a Department of Microbiology and Immunology , University of Melbourne, Peter Doherty Institute for Infection and Immunity , Melbourne , Victoria , Australia.,b ARC Centre for Excellence in Convergent Bio-Nano Science and Technology , University of Melbourne , Melbourne , Australia.,c Melbourne Sexual Health Centre, Department of Infectious Diseases, Alfred Health, Central Clinical School , Monash University , Melbourne , Australia
| |
Collapse
|
40
|
Ye ZW, Yuan S, Poon KM, Wen L, Yang D, Sun Z, Li C, Hu M, Shuai H, Zhou J, Zhang MY, Zheng BJ, Chu H, Yuen KY. Antibody-Dependent Cell-Mediated Cytotoxicity Epitopes on the Hemagglutinin Head Region of Pandemic H1N1 Influenza Virus Play Detrimental Roles in H1N1-Infected Mice. Front Immunol 2017; 8:317. [PMID: 28377769 PMCID: PMC5359280 DOI: 10.3389/fimmu.2017.00317] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2017] [Accepted: 03/06/2017] [Indexed: 01/07/2023] Open
Abstract
Engaging the antibody-dependent cell-mediated cytotoxicity (ADCC) for killing of virus-infected cells and secretion of antiviral cytokines and chemokines was incorporated as one of the important features in the design of universal influenza vaccines. However, investigation of the ADCC epitopes on the highly immunogenic influenza hemagglutinin (HA) head region has been rarely reported. In this study, we determined the ADCC and antiviral activities of two putative ADCC epitopes, designated E1 and E2, on the HA head of a pandemic H1N1 influenza virus in vitro and in a lethal mouse model. Our data demonstrated that sera from the E1-vaccinated mice could induce high ADCC activities. Importantly, the induction of ADCC response modestly decreased viral load in the lungs of H1N1-infected mice. However, the elevated ADCC significantly increased mouse alveolar damage and mortality than that of the PBS-vaccinated group (P < 0.0001). The phenotype was potentially due to an exaggerated inflammatory cell infiltration triggered by ADCC, as an upregulated release of cytotoxic granules (perforin) was observed in the lung tissue of E1-vaccinated mice after H1N1 influenza virus challenge. Overall, our data suggested that ADCC elicited by certain domains of HA head region might have a detrimental rather than protective effect during influenza virus infection. Thus, future design of universal influenza vaccine shall strike a balance between the induction of protective immunity and potential side effects of ADCC.
Collapse
Affiliation(s)
- Zi-Wei Ye
- Department of Microbiology, The University of Hong Kong , Hong Kong
| | - Shuofeng Yuan
- Department of Microbiology, The University of Hong Kong , Hong Kong
| | - Kwok-Man Poon
- Department of Microbiology, The University of Hong Kong , Hong Kong
| | - Lei Wen
- Department of Microbiology, The University of Hong Kong , Hong Kong
| | - Dong Yang
- Department of Microbiology, The University of Hong Kong , Hong Kong
| | - Zehua Sun
- Department of Microbiology, The University of Hong Kong , Hong Kong
| | - Cun Li
- Department of Microbiology, The University of Hong Kong , Hong Kong
| | - Meng Hu
- Department of Microbiology, The University of Hong Kong , Hong Kong
| | - Huiping Shuai
- Department of Microbiology, The University of Hong Kong , Hong Kong
| | - Jie Zhou
- Department of Microbiology, The University of Hong Kong, Hong Kong; State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong
| | - Mei-Yun Zhang
- Department of Microbiology, The University of Hong Kong , Hong Kong
| | - Bo-Jian Zheng
- Department of Microbiology, The University of Hong Kong , Hong Kong
| | - Hin Chu
- Department of Microbiology, The University of Hong Kong, Hong Kong; State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong
| | - Kwok-Yung Yuen
- Department of Microbiology, The University of Hong Kong, Hong Kong; State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong; Research Centre of Infection and Immunology, The University of Hong Kong, Hong Kong; Carol Yu Centre for Infection, The University of Hong Kong, Hong Kong
| |
Collapse
|
41
|
Vanderven HA, Jegaskanda S, Wheatley AK, Kent SJ. Antibody-dependent cellular cytotoxicity and influenza virus. Curr Opin Virol 2017; 22:89-96. [PMID: 28088123 DOI: 10.1016/j.coviro.2016.12.002] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Revised: 12/11/2016] [Accepted: 12/19/2016] [Indexed: 10/20/2022]
Abstract
Antibodies are a key defence against influenza infection and disease, but neutralizing antibodies are often strain-specific and of limited utility against divergent or pandemic viruses. There is now considerable evidence that influenza-specific antibodies with Fc-mediated effector functions, such as antibody-dependent cellular cytotoxicity (ADCC), can assist in the clearance of influenza infection in vitro and in animal models. Further, ADCC-mediating antibodies that recognize a broad array of influenza strains are common in humans, likely as a result of being regularly exposed to influenza infections. The concept that influenza-specific ADCC can assist in the partial control of influenza infections in humans is gaining momentum. This review examines the utility of influenza-specific ADCC antibodies.
Collapse
Affiliation(s)
- Hillary A Vanderven
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
| | - Sinthujan Jegaskanda
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
| | - Adam K Wheatley
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
| | - Stephen J Kent
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia; Melbourne Sexual Health Clinic and Infectious Diseases Department, Alfred Hospital, Monash University Central Clinical School, Carlton, Victoria, Australia; ARC Centre for Excellence in Convergent Bio-Nano Science and Technology, University of Melbourne, Melbourne, Australia.
| |
Collapse
|
42
|
Kolpe A, Schepens B, Fiers W, Saelens X. M2-based influenza vaccines: recent advances and clinical potential. Expert Rev Vaccines 2016; 16:123-136. [DOI: 10.1080/14760584.2017.1240041] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- Annasaheb Kolpe
- Medical Biotechnology Center, VIB, Ghent, B-9052, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Bert Schepens
- Medical Biotechnology Center, VIB, Ghent, B-9052, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Walter Fiers
- Medical Biotechnology Center, VIB, Ghent, B-9052, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Xavier Saelens
- Medical Biotechnology Center, VIB, Ghent, B-9052, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| |
Collapse
|
43
|
Recombinant production and characterization of human anti-influenza virus monoclonal antibodies identified from hybridomas fused with human lymphocytes. Biologicals 2016; 44:394-402. [PMID: 27464991 DOI: 10.1016/j.biologicals.2016.05.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Revised: 05/10/2016] [Accepted: 05/14/2016] [Indexed: 01/09/2023] Open
Abstract
In previous studies, hybridomas producing human immunoglobulin G, the antibodies 5E4 and 5A7 against influenza A and B virus were established using a novel human lymphocyte fusion partner, SPYMEG. In the present study, we succeeded in achieving the recombinant production and secretion of 5E4 and 5A7 in Chinese hamster ovary cells. Our N-glycan analysis by intact-mass detection and liquid chromatography mass spectrometry showed that recombinant 5E4 and 5A7 have one N-glycan and the typical mammalian-type N-glycan structures similar to those in hybridomas. However, the glycan distribution was slightly different among these antibodies. The amount of high-mannose-type structures was under 10% of the total N-glycans of recombinant 5E4 and 5A7, compared to 20% of the 5E4 and 5A7 produced in hybridomas. The amount of galactosylated N-glycans was increased in recombinants. Approximately 80% of the N-glycans of all antibodies was fucosylated, and no sialylated N-glycan was found. Recombinant 5E4 and 5A7 neutralized pandemic influenza A virus specifically, and influenza B virus broadly, quite similar to the 5E4 and 5A7 produced in hybridomas, respectively. Here we demonstrated that recombinants of antibodies identified from hybridomas fused with SPYMEG have normal N-glycans and that their neutralizing activities bear comparison with those of the original antibodies.
Collapse
|
44
|
Jegaskanda S, Luke C, Hickman HD, Sangster MY, Wieland-Alter WF, McBride JM, Yewdell JW, Wright PF, Treanor J, Rosenberger CM, Subbarao K. Generation and Protective Ability of Influenza Virus-Specific Antibody-Dependent Cellular Cytotoxicity in Humans Elicited by Vaccination, Natural Infection, and Experimental Challenge. J Infect Dis 2016; 214:945-52. [PMID: 27354365 DOI: 10.1093/infdis/jiw262] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 06/17/2016] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Nonneutralizing antibodies (Abs) involved in antibody-dependent cellular cytotoxicity (ADCC) may provide some protection from influenza virus infection. The ability of influenza vaccines to induce ADCC-mediating Abs (ADCC-Abs) in adults and children is unclear. METHODS We quantified ADCC-Abs in serum samples from adults who received a dose of inactivated subunit vaccine (ISV) targeting monovalent 2009 pandemic influenza A(H1N1) virus or live-attenuated influenza vaccine (LAIV) or who had laboratory-confirmed influenza A(H1N1) virus infection. We also measured ADCC-Abs in children who either received a dose of trivalent seasonal ISV followed by trivalent seasonal LAIV or 2 doses of LAIV. Finally, we assessed the ability of low and high ADCC-Ab titers to protect adults from experimental challenge with influenza A/Wisconsin/67/131/2005(H3N2) virus. RESULTS Adults and children who received a dose of ISV had a robust increase in ADCC-Ab titers to both recombinant hemagglutinin (rHA) protein and homologous virus-infected cells. There was no detectable increase in titers of ADCC-Abs to rHA or virus-infected cells in adults and children who received LAIV. Higher titers (≥320) of preexisting ADCC-Abs were associated with lower virus replication and a significant reduction in total symptom scores in experimentally infected adults. CONCLUSIONS ADCC-Ab titers increased following experimental influenza virus infection in adults and after ISV administration in both children and adults.
Collapse
Affiliation(s)
- Sinthujan Jegaskanda
- Laboratory of Infectious Diseases Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | | | - Heather D Hickman
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Mark Y Sangster
- Department of Medicine, University of Rochester Medical Center, New York
| | - Wendy F Wieland-Alter
- Department of Pediatrics, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | | | - Jon W Yewdell
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Peter F Wright
- Department of Pediatrics, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | - John Treanor
- Department of Medicine, University of Rochester Medical Center, New York
| | | | | |
Collapse
|
45
|
Lee YN, Kim MC, Lee YT, Kim YJ, Kang SM. Mechanisms of Cross-protection by Influenza Virus M2-based Vaccines. Immune Netw 2015; 15:213-21. [PMID: 26557805 PMCID: PMC4637342 DOI: 10.4110/in.2015.15.5.213] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2015] [Revised: 08/25/2015] [Accepted: 09/10/2015] [Indexed: 01/06/2023] Open
Abstract
Current influenza virus vaccines are based on strain-specific surface glycoprotein hemagglutinin (HA) antigens and effective only when the predicted vaccine strains and circulating viruses are well-matched. The current strategy of influenza vaccination does not prevent the pandemic outbreaks and protection efficacy is reduced or ineffective if mutant strains emerge. It is of high priority to develop effective vaccines and vaccination strategies conferring a broad range of cross protection. The extracellular domain of M2 (M2e) is highly conserved among human influenza A viruses and has been utilized to develop new vaccines inducing cross protection against different subtypes of influenza A virus. However, immune mechanisms of cross protection by M2e-based vaccines still remain to be fully elucidated. Here, we review immune correlates and mechanisms conferring cross protection by M2e-based vaccines. Molecular and cellular immune components that are known to be involved in M2 immune-mediated protection include antibodies, B cells, T cells, alveolar macrophages, Fc receptors, complements, and natural killer cells. Better understanding of protective mechanisms by immune responses induced by M2e vaccination will help facilitate development of broadly cross protective vaccines against influenza A virus.
Collapse
Affiliation(s)
- Yu-Na Lee
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Min-Chul Kim
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA. ; Animal and Plant Quarantine Agency, Anyang 14089, Korea
| | - Young-Tae Lee
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Yu-Jin Kim
- 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
| |
Collapse
|