1
|
Grovenstein P, Bhatnagar N, Kim KH, Pal SS, Le CTT, Raha JR, Liu R, Shin CH, Park BR, Du L, Subbiah J, Wang BZ, Kang SM. Influenza 5xM2e mRNA lipid nanoparticle vaccine confers broad immunity and significantly enhances the efficacy of inactivated split vaccination when coadministered. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025; 214:104-114. [PMID: 40073270 PMCID: PMC11844137 DOI: 10.1093/jimmun/vkae013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 10/29/2024] [Indexed: 03/14/2025]
Abstract
Current influenza vaccines are not effective in conferring protection against antigenic variants and pandemics. To improve cross-protection of influenza vaccination, we developed a 5xM2e messenger RNA (mRNA) vaccine encoding the tandem repeat conserved ectodomain (M2e) of ion channel protein M2 derived from human, swine, and avian influenza A viruses. The lipid nanoparticle (LNP)-encapsulated 5xM2e mRNA vaccine was immunogenic, eliciting high levels of M2e-specific IgG antibodies, IFN-γ+ T cells, T follicular helper cells, germinal center phenotypic B cells, and plasma cells. The mice with 5xM2e mRNA vaccination were broadly protected against lethal infection regardless of hemagglutinin (H1, H3, H5) subtypes by preventing severe weight loss. Injection of 5xM2e mRNA LNP vaccine induced acute innate responses recruiting monocytes, macrophages, and diverse subsets of dendritic cells. A single dose of combined 5xM2e mRNA LNP and split vaccines resulted in significantly enhanced and sustainable IgG antibody responses to viral antigens and protection against homologous and heterologous viruses. This study provides a new strategy of combined mRNA and seasonal vaccination, significantly enhancing vaccine protective efficacy.
Collapse
Affiliation(s)
- Phillip Grovenstein
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, United States
| | - Noopur Bhatnagar
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, United States
| | - Ki-Hye Kim
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, United States
| | - Surya Sekhar Pal
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, United States
| | - Chau Thuy Tien Le
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, United States
| | - Jannatul Ruhan Raha
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, United States
| | - Rong Liu
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, United States
| | - Chong Hyun Shin
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, United States
| | - Bo Ryoung Park
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, United States
| | - Lanying Du
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, United States
| | - Jeeva Subbiah
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, United States
| | - Bao-Zhong Wang
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, United States
| | - Sang-Moo Kang
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, United States
| |
Collapse
|
2
|
Di Y, Zhang C, Ren Z, Jiang R, Tang J, Yang S, Wang Z, Yu T, Zhang T, Yu Z, Xu Z, Zhuang X, Jin N, Tian M. The self-assembled nanoparticle-based multi-epitope influenza mRNA vaccine elicits protective immunity against H1N1 and B influenza viruses in mice. Front Immunol 2024; 15:1483720. [PMID: 39445022 PMCID: PMC11497263 DOI: 10.3389/fimmu.2024.1483720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 09/19/2024] [Indexed: 10/25/2024] Open
Abstract
Introduction The influenza virus is recognized as the primary cause of human respiratory diseases, with the current influenza vaccine primarily offering strain-specific immunity and limited protection against drifting strains. Considering this, the development of a broad-spectrum influenza vaccine capable of inducing effective immunity is considered the future direction in combating influenza. Methods The present study proposes a novel mRNA-based multi-epitope influenza vaccine, which combines three conserved antigens derived from the influenza A virus. The antigens consist of M2 ion channel's extracellular domain (M2e), the conserved epitope of located in HA2 of hemagglutinin (H1, H3, B), and HA1 of hemagglutinin. At the same time, trimeric sequences and ferritin were conjugated separately to investigate the immune effects of antigen multivalent presentation. Results Immunization studies conducted on C57BL/6 mice with these vaccines revealed that they can elicit both humoral immunity and CD4+ and CD8+ T cell responses, which collectively contribute to enhancing cross-protective effects. The virus challenge results showed that vaccinated groups had significantly reduced lung damage, lower viral loads in the lungs, nasal turbinates, and trachea, as well as decreased levels of pro-inflammatory cytokines. Conclusion These findings clearly demonstrate the wide range of protective effects provided by these vaccines against H1N1 and B influenza viruses. The present finding highlights the potential of mRNA-based influenza vaccines encoding conserved proteins as a promising strategy for eliciting broad-spectrum protective humoral and cellular immunity against H1N1 and B influenza viruses.
Collapse
Affiliation(s)
- Yaxin Di
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, State Key Laboratory of Pathogen and Biosecurity, Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun, China
| | - Chenchao Zhang
- College of Agriculture, Yanbian University, Yanji, China
| | - Zilin Ren
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, State Key Laboratory of Pathogen and Biosecurity, Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun, China
| | - Renyue Jiang
- College of Agriculture, Yanbian University, Yanji, China
| | - Jiafeng Tang
- College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Songhui Yang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, State Key Laboratory of Pathogen and Biosecurity, Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun, China
| | - Ziliang Wang
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China
| | - Tong Yu
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China
| | - Tong Zhang
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Ziping Yu
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China
| | - Zhiqiang Xu
- College of Agriculture, Yanbian University, Yanji, China
| | - Xinyu Zhuang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, State Key Laboratory of Pathogen and Biosecurity, Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun, China
| | - Ningyi Jin
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, State Key Laboratory of Pathogen and Biosecurity, Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| | - Mingyao Tian
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, State Key Laboratory of Pathogen and Biosecurity, Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun, China
| |
Collapse
|
3
|
Hadj Hassine I, Ben M'hadheb M, Almalki MA, Gharbi J. Virus-like particles as powerful vaccination strategy against human viruses. Rev Med Virol 2024; 34:e2498. [PMID: 38116958 DOI: 10.1002/rmv.2498] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 11/25/2023] [Accepted: 12/08/2023] [Indexed: 12/21/2023]
Abstract
Nowadays, viruses are not only seen as causative agents of viral infectious diseases but also as valuable research materials for various biomedical purposes, including recombinant protein production. When expressed in living or cell-free expression systems, viral structural proteins self-assemble into virus-like particles (VLPs). Mimicking the native form and size of viruses and lacking the genetic material, VLPs are safe and highly immunogenic and thus can be exploited to develop antiviral vaccines. Some vaccines based on VLPs against various infectious pathogens have already been licenced for human use and are available in the commercial market, the latest of which is a VLP-based vaccine to protect against the novel Coronavirus. Despite the success and popularity of VLP subunit vaccines, many more VLPs are still in different stages of design, production, and approval. There are still many challenges that require to be addressed in the future before this surface display system can be widely used as an effective vaccine strategy in combating infectious diseases. In this review, we highlight the use of structural viral proteins to produce VLPs, emphasising their intrinsic properties, structural classification, and main expression host systems. We also compiled the recent scientific literature about VLP-based vaccines to underline the recent advances in their application as a vaccine strategy for preventing and fighting virulent human pathogens. Finally, we presented the key challenges and possible solutions for VLP-based vaccine production.
Collapse
Affiliation(s)
- Ikbel Hadj Hassine
- Virology and Antiviral Strategies Research Unit UR17ES30, Higher Institute of Biotechnology, University of Monastir, Monastir, Tunisia
- USCR-SAG Unit, Higher Institute of Biotechnology, University of Monastirs, Monastir, Tunisia
| | - Manel Ben M'hadheb
- Virology and Antiviral Strategies Research Unit UR17ES30, Higher Institute of Biotechnology, University of Monastir, Monastir, Tunisia
- USCR-SAG Unit, Higher Institute of Biotechnology, University of Monastirs, Monastir, Tunisia
| | - Mohammed A Almalki
- Department of Biological Sciences, College of Science, King Faisal University, Al-Ahsa, Saudi Arabia
| | - Jawhar Gharbi
- Department of Biological Sciences, College of Science, King Faisal University, Al-Ahsa, Saudi Arabia
| |
Collapse
|
4
|
Emami A, Pirbonyeh N, Moattari A, Javanmardi F. A decade genetic diversity in Circulating influenza B virus in Iran (2010-2019): Divergence from WHO-recommended vaccine strains. Vopr Virusol 2023; 68:385-393. [PMID: 38156573 DOI: 10.36233/0507-4088-180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 10/15/2023] [Indexed: 12/30/2023]
Abstract
BACKGROUND Data on the disease burden and circulation patterns of influenza B virus lineages for Iran are limited. OBJECTIVE This review aims to describe the pattern of influenza B occurrence in Iran, comparing it with the proposed vaccine strains and determining the match and mismatch with the prescribed vaccine annually. METHODS Various sources were used to retrieve information of the data; such as information from an online search of databases such as FluNet, GISAID, and NCBI. After extracting protein sequence records in GISAID, sequence alignment with vaccine strain and construction of a phylogenetic tree were performed. Subsequently, categories of the registered circulating strains were evaluated for matching with the vaccine strains. RESULTS Of the total registered influenza-positive samples, 20.21% were related to influenza B virus. The phylogenic tree was designed based on 43 samples registered in the GISAID database; 76.74 and 23.25% sequences were of Yamagata and Victoria lineages, respectively. The most prevalent influenza B virus strains circulating during the study years belonged to the Yamagata lineage. In general, the match of the influenza B virus predominant circulating strains with administrated vaccines was observed in Iran. However, a high level of mismatch between the vaccine strain and Iranian isolates was identified in 2016‒2017. CONCLUSION The review of match and mismatch in influenza vaccine in order to improve the composition of the prescribed vaccine in each region is very important because the vaccine efficacy decreased when the strain included in vaccine did not match the circulating epidemic strain.
Collapse
Affiliation(s)
- A Emami
- Shiraz University of medical sciences
| | | | | | | |
Collapse
|
5
|
Singh RS, Singh A, Masih GD, Batra G, Sharma AR, Joshi R, Prakash A, Suroy B, Sarma P, Prajapat M, Kaur H, Bhattacharyya A, Upadhyay S, Medhi B. A comprehensive insight on the challenges for COVID-19 vaccine: A lesson learnt from other viral vaccines. Heliyon 2023; 9:e16813. [PMID: 37303517 PMCID: PMC10245239 DOI: 10.1016/j.heliyon.2023.e16813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 04/15/2023] [Accepted: 05/29/2023] [Indexed: 06/13/2023] Open
Abstract
The aim of this study is to comprehensively analyze previous viral vaccine programs and identify potential challenges and effective measures for the COVID-19 vaccine program. Previous viral vaccine programs, such as those for HIV, Zika, Influenza, Ebola, Dengue, SARS, and MERS, were evaluated. Paramount challenges were identified, including quasi-species, cross-reactivity, duration of immunity, revaccination, mutation, immunosenescence, and adverse events related to viral vaccines. Although a large population has been vaccinated, mutations in SARS-CoV-2 and adverse events related to vaccines pose significant challenges. Previous vaccine programs have taught us that predicting the final outcome of the current vaccine program for COVID-19 cannot be determined at a given state. Long-term follow-up studies are essential. Validated preclinical studies, long-term follow-up studies, alternative therapeutic approaches, and alternative vaccines are necessary.
Collapse
Affiliation(s)
- Rahul Soloman Singh
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Ashutosh Singh
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Gladson David Masih
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Gitika Batra
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Amit Raj Sharma
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Rupa Joshi
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Ajay Prakash
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Benjamin Suroy
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Phulen Sarma
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Manisha Prajapat
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Hardeep Kaur
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Anusuya Bhattacharyya
- Department of Ophthalmology, Government Medical College & Hospital, Sector-32, Chandigarh, 160030, India
| | - Sujata Upadhyay
- Department of Physiology, Dr. Harvansh Singh Judge Institute of Dental Sciences & Hospital, Panjab University, Chandigarh, 160014, India
| | - Bikash Medhi
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh, 160012, India
| |
Collapse
|
6
|
Tsai HH, Huang PH, Lin LC, Yao BY, Liao WT, Pai CH, Liu YH, Chen HW, Hu CMJ. Lymph Node Follicle-Targeting STING Agonist Nanoshells Enable Single-Shot M2e Vaccination for Broad and Durable Influenza Protection. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023:e2206521. [PMID: 37092580 DOI: 10.1002/advs.202206521] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 03/14/2023] [Indexed: 05/03/2023]
Abstract
The highly conserved matrix protein 2 ectodomain (M2e) of influenza viruses presents a compelling vaccine antigen candidate for stemming the pandemic threat of the mutation-prone pathogen, yet the low immunogenicity of the diminutive M2e peptide renders vaccine development challenging. A highly potent M2e nanoshell vaccine that confers broad and durable influenza protectivity under a single vaccination is shown. Prepared via asymmetric ionic stabilization for nanoscopic curvature formation, polymeric nanoshells co-encapsulating high densities of M2e peptides and stimulator of interferon genes (STING) agonists are prepared. Robust and long-lasting protectivity against heterotypic influenza viruses is achieved with a single administration of the M2e nanoshells in mice. Mechanistically, molecular adjuvancy by the STING agonist and nanoshell-mediated prolongation of M2e antigen exposure in the lymph node follicles synergistically contribute to the heightened anti-M2e humoral responses. STING agonist-triggered T cell helper functions and extended residence of M2e peptides in the follicular dendritic cell network provide a favorable microenvironment that induces Th1-biased antibody production against the diminutive antigen. These findings highlight a versatile nanoparticulate design that leverages innate immune pathways for enhancing the immunogenicity of weak immunogens. The single-shot nanovaccine further provides a translationally viable platform for pandemic preparedness.
Collapse
Affiliation(s)
- Hsiao-Han Tsai
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 115, Taiwan
- Taiwan International Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei, 112, Taiwan
| | - Ping-Han Huang
- Department of Veterinary Medicine, National Taiwan University, Taipei, 10617, Taiwan
| | - Leon Cw Lin
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 115, Taiwan
- Biomedical Translation Research Center, Academia Sinica, Taipei, 115, Taiwan
| | - Bing-Yu Yao
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 115, Taiwan
- Biomedical Translation Research Center, Academia Sinica, Taipei, 115, Taiwan
| | - Wan-Ting Liao
- Department of Veterinary Medicine, National Taiwan University, Taipei, 10617, Taiwan
| | - Chen-Hsueh Pai
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 115, Taiwan
- Biomedical Translation Research Center, Academia Sinica, Taipei, 115, Taiwan
| | - Yu-Han Liu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 115, Taiwan
| | - Hui-Wen Chen
- Department of Veterinary Medicine, National Taiwan University, Taipei, 10617, Taiwan
| | - Che-Ming J Hu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 115, Taiwan
- Taiwan International Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei, 112, Taiwan
- Biomedical Translation Research Center, Academia Sinica, Taipei, 115, Taiwan
- Center of Applied Nanomedicine, National Cheng Kung University, Tainan, 70101, Taiwan
| |
Collapse
|
7
|
Gupta R, Arora K, Roy SS, Joseph A, Rastogi R, Arora NM, Kundu PK. Platforms, advances, and technical challenges in virus-like particles-based vaccines. Front Immunol 2023; 14:1123805. [PMID: 36845125 PMCID: PMC9947793 DOI: 10.3389/fimmu.2023.1123805] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 01/30/2023] [Indexed: 02/11/2023] Open
Abstract
Viral infectious diseases threaten human health and global stability. Several vaccine platforms, such as DNA, mRNA, recombinant viral vectors, and virus-like particle-based vaccines have been developed to counter these viral infectious diseases. Virus-like particles (VLP) are considered real, present, licensed and successful vaccines against prevalent and emergent diseases due to their non-infectious nature, structural similarity with viruses, and high immunogenicity. However, only a few VLP-based vaccines have been commercialized, and the others are either in the clinical or preclinical phases. Notably, despite success in the preclinical phase, many vaccines are still struggling with small-scale fundamental research owing to technical difficulties. Successful production of VLP-based vaccines on a commercial scale requires a suitable platform and culture mode for large-scale production, optimization of transduction-related parameters, upstream and downstream processing, and monitoring of product quality at each step. In this review article, we focus on the advantages and disadvantages of various VLP-producing platforms, recent advances and technical challenges in VLP production, and the current status of VLP-based vaccine candidates at commercial, preclinical, and clinical levels.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Prabuddha K. Kundu
- Department of Research and Development, Premas Biotech Pvt Ltd., Sector IV, Industrial Model Township (IMT), Manesar, Gurgaon, India
| |
Collapse
|
8
|
Trombetta CM, Marchi S, Montomoli E. The baculovirus expression vector system: a modern technology for the future of influenza vaccine manufacturing. Expert Rev Vaccines 2022; 21:1233-1242. [DOI: 10.1080/14760584.2022.2085565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Affiliation(s)
| | - Serena Marchi
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Emanuele Montomoli
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
- VisMederi srl, Siena, Italy
- VisMederi Research srl, Siena, Italy
| |
Collapse
|
9
|
Influenza Vaccine: An Engineering Vision from Virological Importance to Production. BIOTECHNOL BIOPROC E 2022; 27:714-738. [PMID: 36313971 PMCID: PMC9589582 DOI: 10.1007/s12257-022-0115-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Revised: 06/06/2022] [Accepted: 06/12/2022] [Indexed: 01/26/2023]
Abstract
According to data from the World Health Organization (WHO) every year, millions of people are affected by flu. Flu is a disease caused by influenza viruses. For preventing this, seasonal influenza vaccinations are widely considered the most efficient way to protect against the negative effects of the flu. To date, there is no "one-size-fits-all" vaccine that can be effective all over the world to protect against all seasonal or pandemic influenza virus types. Because influenza virus transforms its genetic structure and it can emerges as immunogenically new (antigenic drift) which causes epidemics or new virus subtype (antigenic shift) which causes pandemics. As a result, annual revaccination or new subtype viral vaccine development is required. Currently, three types of vaccines (inactivated, live attenuated, and recombinant) are approved in different countries. These can be named "conventional influenza vaccines" and their production are based on eggs or cell culture. Although, there is good effort to develop new influenza vaccines for broader and longer period of time protection. In this sense these candidate vaccines are called "universal influenza vaccines". In this article, after we mentioned the short history of flu then virus morphology and infection, we explained the diseases caused by the influenza virus in humans. Afterward, we explained in detail the production methods of available influenza vaccines, types of bioreactors used in cell culture based production, conventional and new vaccine types, and development strategies for better vaccines.
Collapse
|
10
|
Baculovirus-derived influenza virus-like particle confers complete protection against lethal H7N9 avian influenza virus challenge in chickens and mice. Vet Microbiol 2022; 264:109306. [DOI: 10.1016/j.vetmic.2021.109306] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 11/29/2021] [Accepted: 12/11/2021] [Indexed: 02/03/2023]
|
11
|
Combination of conserved recombinant proteins (NP & 3M2e) formulated with Alum protected BALB/c mice against influenza A/PR8/H1N1 virus challenge. Biotechnol Lett 2021; 43:2137-2147. [PMID: 34491470 DOI: 10.1007/s10529-021-03174-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Accepted: 08/19/2021] [Indexed: 10/20/2022]
Abstract
PURPOSE Influenza is one of the most important agents of pandemic outbreak causing substantial morbidity and mortality. Vaccination strategies of influenza must be adapted annually due to constant antigenic changes in various strains. Therefore, the present study was conducted to evaluate protective immunity of the conserved influenza proteins. METHODS For this purpose, three tandem repeats of M2e (3M2e) and NP were separately expressed in E. coli and were purified using column chromatography. Female Balb/c mice were injected intradermally with a combination of the purified 3M2e and NP alone or formulated with Alum (AlOH3) adjuvant in three doses. The mice were challenged by intranasal administration of H1N1 (A/PR/8/34) 2 weeks after the last vaccination. RESULTS The results demonstrated that recombinant NP and M2e proteins are immunogenic and could efficiently elicit immune responses in mice compared to non-immunized mice. The combination of 3M2e and NP supplemented with Alum stimulated both NP and M2e-specific antibodies, which were higher than those stimulated by each single antigen plus Alum. In addition, the secretion of IFN-γ and IL-4 as well as the induction of lymphocyte proliferation in mice received the mixture of these proteins with Alum was considerably higher than other groups. Moreover, the highest survival rate (86%) with the least body weight change was observed in the mice immunized with 3M2e and NP supplemented with Alum followed by the mice received NP supplemented with Alum (71%). CONCLUSION Accordingly, this regimen can be considered as an attractive candidate for global vaccination against influenza.
Collapse
|
12
|
Mytle N, Leyrer S, Inglefield JR, Harris AM, Hickey TE, Minang J, Lu H, Ma Z, Andersen H, Grubaugh ND, Guina T, Skiadopoulos MH, Lacy MJ. Influenza Antigens NP and M2 Confer Cross Protection to BALB/c Mice against Lethal Challenge with H1N1, Pandemic H1N1 or H5N1 Influenza A Viruses. Viruses 2021; 13:1708. [PMID: 34578289 PMCID: PMC8473317 DOI: 10.3390/v13091708] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 08/24/2021] [Accepted: 08/26/2021] [Indexed: 02/01/2023] Open
Abstract
Influenza hemagglutinin (HA) is considered a major protective antigen of seasonal influenza vaccine but antigenic drift of HA necessitates annual immunizations using new circulating HA versions. Low variation found within conserved non-HA influenza virus (INFV) antigens may maintain protection with less frequent immunizations. Conserved antigens of influenza A virus (INFV A) that can generate cross protection against multiple INFV strains were evaluated in BALB/c mice using modified Vaccinia virus Ankara (MVA)-vectored vaccines that expressed INFV A antigens hemagglutinin (HA), matrix protein 1 (M1), nucleoprotein (NP), matrix protein 2 (M2), repeats of the external portion of M2 (M2e) or as tandem repeats (METR), and M2e with transmembrane region and cytoplasmic loop (M2eTML). Protection by combinations of non-HA antigens was equivalent to that of subtype-matched HA. Combinations of NP and forms of M2e generated serum antibody responses and protected mice against lethal INFV A challenge using PR8, pandemic H1N1 A/Mexico/4108/2009 (pH1N1) or H5N1 A/Vietnam/1203/2004 (H5N1) viruses, as demonstrated by reduced lung viral burden and protection against weight loss. The highest levels of protection were obtained with NP and M2e antigens delivered as MVA inserts, resulting in broadly protective immunity in mice and enhancement of previous natural immunity to INFV A.
Collapse
Affiliation(s)
- Nutan Mytle
- Emergent BioSolutions, 300 Professional Drive, Gaithersburg, MD 20879, USA; (N.M.); (S.L.); (J.R.I.); (A.M.H.); (T.E.H.); (J.M.); (H.L.); (Z.M.); (N.D.G.); (T.G.); (M.H.S.)
- Biomedical Advanced Research and Development Agency, U.S. Department of Health and Human Services, Washington, DC 20201, USA
| | - Sonja Leyrer
- Emergent BioSolutions, 300 Professional Drive, Gaithersburg, MD 20879, USA; (N.M.); (S.L.); (J.R.I.); (A.M.H.); (T.E.H.); (J.M.); (H.L.); (Z.M.); (N.D.G.); (T.G.); (M.H.S.)
- Roche Diagnostics GmbH, Nonnenwald 2, 82377 Penzberg, Germany
| | - Jon R. Inglefield
- Emergent BioSolutions, 300 Professional Drive, Gaithersburg, MD 20879, USA; (N.M.); (S.L.); (J.R.I.); (A.M.H.); (T.E.H.); (J.M.); (H.L.); (Z.M.); (N.D.G.); (T.G.); (M.H.S.)
- Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Andrea M. Harris
- Emergent BioSolutions, 300 Professional Drive, Gaithersburg, MD 20879, USA; (N.M.); (S.L.); (J.R.I.); (A.M.H.); (T.E.H.); (J.M.); (H.L.); (Z.M.); (N.D.G.); (T.G.); (M.H.S.)
| | - Thomas E. Hickey
- Emergent BioSolutions, 300 Professional Drive, Gaithersburg, MD 20879, USA; (N.M.); (S.L.); (J.R.I.); (A.M.H.); (T.E.H.); (J.M.); (H.L.); (Z.M.); (N.D.G.); (T.G.); (M.H.S.)
- National Cancer Institute, National Institutes of Health, Frederick, MD 20814, USA
| | - Jacob Minang
- Emergent BioSolutions, 300 Professional Drive, Gaithersburg, MD 20879, USA; (N.M.); (S.L.); (J.R.I.); (A.M.H.); (T.E.H.); (J.M.); (H.L.); (Z.M.); (N.D.G.); (T.G.); (M.H.S.)
- Optimal Health Care, 11377 Robinwood Dr, Hagerstown, MD 21742, USA
| | - Hang Lu
- Emergent BioSolutions, 300 Professional Drive, Gaithersburg, MD 20879, USA; (N.M.); (S.L.); (J.R.I.); (A.M.H.); (T.E.H.); (J.M.); (H.L.); (Z.M.); (N.D.G.); (T.G.); (M.H.S.)
| | - Zhidong Ma
- Emergent BioSolutions, 300 Professional Drive, Gaithersburg, MD 20879, USA; (N.M.); (S.L.); (J.R.I.); (A.M.H.); (T.E.H.); (J.M.); (H.L.); (Z.M.); (N.D.G.); (T.G.); (M.H.S.)
| | - Hanné Andersen
- BIOQUAL, Inc., 12301 Parklawn Dr, Rockville, MD 20852, USA;
| | - Nathan D. Grubaugh
- Emergent BioSolutions, 300 Professional Drive, Gaithersburg, MD 20879, USA; (N.M.); (S.L.); (J.R.I.); (A.M.H.); (T.E.H.); (J.M.); (H.L.); (Z.M.); (N.D.G.); (T.G.); (M.H.S.)
- Yale School of Public Health, Yale University, 60 College Street, New Haven, CT 06510, USA
| | - Tina Guina
- Emergent BioSolutions, 300 Professional Drive, Gaithersburg, MD 20879, USA; (N.M.); (S.L.); (J.R.I.); (A.M.H.); (T.E.H.); (J.M.); (H.L.); (Z.M.); (N.D.G.); (T.G.); (M.H.S.)
- AstraZeneca, Gaithersburg, MD 20878, USA
| | - Mario H. Skiadopoulos
- Emergent BioSolutions, 300 Professional Drive, Gaithersburg, MD 20879, USA; (N.M.); (S.L.); (J.R.I.); (A.M.H.); (T.E.H.); (J.M.); (H.L.); (Z.M.); (N.D.G.); (T.G.); (M.H.S.)
- U.S. Department of Health and Human Services, National Institutes of Health, Bethesda, MD 20892, USA
| | - Michael J. Lacy
- Emergent BioSolutions, 300 Professional Drive, Gaithersburg, MD 20879, USA; (N.M.); (S.L.); (J.R.I.); (A.M.H.); (T.E.H.); (J.M.); (H.L.); (Z.M.); (N.D.G.); (T.G.); (M.H.S.)
| |
Collapse
|
13
|
Sadeghi S, Bandehpour M, Haji Molla Hoseini M, Sharifnia Z. Intranasal administration of immunogenic poly-epitope from influenza H1N1 and H3N2 viruses adjuvanted with chitin and chitosan microparticles in BALB/c mice. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2021; 24:1126-1137. [PMID: 34804430 PMCID: PMC8591766 DOI: 10.22038/ijbms.2021.58087.12909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 07/19/2021] [Indexed: 11/06/2022]
Abstract
OBJECTIVES Prevalence of influenza virus, creates the need to achieve an efficient vaccine against it. We examined whether the predicted antigenic epitopes of HA, NP, and M2 proteins of the influenza H1N1 and H3N2 viruses accompanied by chitin and chitosan biopolymers might be relevant to the induction of effective proper mucosal responses. MATERIALS AND METHODS The construct was prepared using B and T cell predicted epitopes of HA, NP, and M2 proteins from the influenza H1N1 and H3N2 viruses by considering haplotype "d" as a dominant allele in the BALB/c mice. Intranasal immunization with purified LPS free recombinant protein together with chitin and chitosan microparticles as adjuvants was administered at an interval of 2 weeks in thirty-five BALB/c female mice which were divided into seven groups. Ten days after the last immunization, humoral and cellular immune responses were examined. RESULTS Elevated systemic IgG2a, IgA, and mucosal IgA revealed a humoral response to the construct. An increase in the number of IFN-γ-producing cells in re-stimulation of splenocytes in the culture medium by poly-tope as well as rise in the concentrations of IL-6, IL-17, and TNF-α along with the regulatory response of IL-10, presented the capacity of the designed protein to provoke significant immune responses. The neutralization test ultimately confirmed the high efficacy of the protein in inhibiting the virus. CONCLUSION The results support the fact that immunogenic poly-tope protein in the presence of chitin and chitosan microparticles as mucosal adjuvants is able to induce humoral and cell-mediated responses in BALB/c mice.
Collapse
Affiliation(s)
- Sahar Sadeghi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mojgan Bandehpour
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Cellular and Molecular Biology Research Center, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mostafa Haji Molla Hoseini
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Medical Nanotechnology and Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zarin Sharifnia
- Cellular and Molecular Biology Research Center, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
14
|
El-Saadony MT, Zabermawi NM, Zabermawi NM, Burollus MA, Shafi ME, Alagawany M, Yehia N, Askar AM, Alsafy SA, Noreldin AE, Khafaga AF, Dhama K, Elnesr SS, Elwan HAM, Cerbo AD, El-Tarabily KA, Abd El-Hack ME. Nutritional Aspects and Health Benefits of Bioactive Plant Compounds against Infectious Diseases: A Review. FOOD REVIEWS INTERNATIONAL 2021. [DOI: 10.1080/87559129.2021.1944183] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Mohamed T. El-Saadony
- Department of Agricultural Microbiology, Faculty of Agriculture, Zagazig University, Zagazig, Egypt
| | - Nidal M. Zabermawi
- Department of Biological Sciences, Microbiology, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Nehal M. Zabermawi
- Laboratory Department, King Abdulaziz Hospital (KAAH), Ministry of Health, Jeddah, Saudi Arabia
| | - Maryam A. Burollus
- Laboratory Department, King Abdulaziz Hospital (KAAH), Ministry of Health, Jeddah, Saudi Arabia
| | - Manal E. Shafi
- Department of Biological Sciences, Microbiology, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mahmoud Alagawany
- Department of Poultry, Faculty of Agriculture, Zagazig University, Zagazig, Egypt
| | - Nahed Yehia
- Reference Laboratory for Veterinary Quality Control on Poultry Production, Animal Health Research Institute, Agricultural Research Center, Cairo, Egypt
| | - Ahmed M. Askar
- Faculty of Veterinary Medicine, Damanhour University, Damanhour, Egypt
| | - Sara A. Alsafy
- Faculty of Veterinary Medicine, Damanhour University, Damanhour, Egypt
| | - Ahmed E. Noreldin
- Department of Histology and Cytology, Faculty of Veterinary Medicine, Damanhour University, Damanhour, Egypt
| | - Asmaa F. Khafaga
- Department of Pathology, Faculty of Veterinary Medicine, Alexandria University, Edfina, Egypt
| | - Kuldeep Dhama
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, India
| | - Shaaban S. Elnesr
- Poultry Production Department, Faculty of Agriculture, Fayoum University, Fayoum, Egypt
| | - Hamada A. M. Elwan
- Animal and Poultry Production Department, Faculty of Agriculture, Minia University, El-Minya, Egypt
| | - Alessandro Di Cerbo
- School of Biosciences and Veterinary Medicine, University of Camerino, Matelica, Italy
| | - Khaled A. El-Tarabily
- Department of Biology, College of Science, United Arab Emirates University, Al-Ain, United Arab Emirates
- Biosecurity and One health Research Centre, Harry Butler Institute, Murdoch University, Murdoch, Western Australia, Australia
| | | |
Collapse
|
15
|
Animal Models Utilized for the Development of Influenza Virus Vaccines. Vaccines (Basel) 2021; 9:vaccines9070787. [PMID: 34358203 PMCID: PMC8310120 DOI: 10.3390/vaccines9070787] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 07/08/2021] [Accepted: 07/10/2021] [Indexed: 12/25/2022] Open
Abstract
Animal models have been an important tool for the development of influenza virus vaccines since the 1940s. Over the past 80 years, influenza virus vaccines have evolved into more complex formulations, including trivalent and quadrivalent inactivated vaccines, live-attenuated vaccines, and subunit vaccines. However, annual effectiveness data shows that current vaccines have varying levels of protection that range between 40–60% and must be reformulated every few years to combat antigenic drift. To address these issues, novel influenza virus vaccines are currently in development. These vaccines rely heavily on animal models to determine efficacy and immunogenicity. In this review, we describe seasonal and novel influenza virus vaccines and highlight important animal models used to develop them.
Collapse
|
16
|
Lopez CE, Legge KL. Influenza A Virus Vaccination: Immunity, Protection, and Recent Advances Toward A Universal Vaccine. Vaccines (Basel) 2020; 8:E434. [PMID: 32756443 PMCID: PMC7565301 DOI: 10.3390/vaccines8030434] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 07/28/2020] [Accepted: 07/31/2020] [Indexed: 12/19/2022] Open
Abstract
Influenza virus infections represent a serious public health threat and account for significant morbidity and mortality worldwide due to seasonal epidemics and periodic pandemics. Despite being an important countermeasure to combat influenza virus and being highly efficacious when matched to circulating influenza viruses, current preventative strategies of vaccination against influenza virus often provide incomplete protection due the continuous antigenic drift/shift of circulating strains of influenza virus. Prevention and control of influenza virus infection with vaccines is dependent on the host immune response induced by vaccination and the various vaccine platforms induce different components of the local and systemic immune response. This review focuses on the immune basis of current (inactivated influenza vaccines (IIV) and live attenuated influenza vaccines (LAIV)) as well as novel vaccine platforms against influenza virus. Particular emphasis will be placed on how each platform induces cross-protection against heterologous influenza viruses, as well as how this immunity compares to and contrasts from the "gold standard" of immunity generated by natural influenza virus infection.
Collapse
Affiliation(s)
- Christopher E. Lopez
- Department of Microbiology and Immunology University of Iowa, Iowa City, IA 52242, USA;
- Department of Pathology, University of Iowa, Iowa City, IA 52242, USA
| | - Kevin L. Legge
- Department of Microbiology and Immunology University of Iowa, Iowa City, IA 52242, USA;
- Department of Pathology, University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
17
|
Baloxavir Marboxil Single-dose Treatment in Influenza-infected Children: A Randomized, Double-blind, Active Controlled Phase 3 Safety and Efficacy Trial (miniSTONE-2). Pediatr Infect Dis J 2020; 39:700-705. [PMID: 32516282 PMCID: PMC7360097 DOI: 10.1097/inf.0000000000002747] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
BACKGROUND Baloxavir marboxil (baloxavir) is a novel, cap-dependent endonuclease inhibitor that has previously demonstrated efficacy in the treatment of influenza in adults and adolescents. We assessed the safety and efficacy of baloxavir in otherwise healthy children with acute influenza. METHODS MiniSTONE-2 (Clinicaltrials.gov: NCT03629184) was a double-blind, randomized, active controlled trial enrolling children 1-<12 years old with a clinical diagnosis of influenza. Children were randomized 2:1 to receive either a single dose of oral baloxavir or oral oseltamivir twice daily for 5 days. The primary endpoint was incidence, severity and timing of adverse events (AEs); efficacy was a secondary endpoint. RESULTS In total, 173 children were randomized and dosed, 115 to the baloxavir group and 58 to the oseltamivir group. Characteristics of participants were similar between treatment groups. Overall, 122 AEs were reported in 84 (48.6%) children. Incidence of AEs was similar between baloxavir and oseltamivir groups (46.1% vs. 53.4%, respectively). The most common AEs were gastrointestinal (vomiting/diarrhea) in both groups [baloxavir: 12 children (10.4%); oseltamivir: 10 children (17.2%)]. No deaths, serious AEs or hospitalizations were reported. Median time (95% confidence interval) to alleviation of signs and symptoms of influenza was similar between groups: 138.1 (116.6-163.2) hours with baloxavir versus 150.0 (115.0-165.7) hours with oseltamivir. CONCLUSIONS Oral baloxavir is well tolerated and effective at alleviating symptoms in otherwise healthy children with acute influenza. Baloxavir provides a new therapeutic option with a simple oral dosing regimen.
Collapse
|
18
|
Abd Raman HS, Tan S, August JT, Khan AM. Dynamics of Influenza A (H5N1) virus protein sequence diversity. PeerJ 2020; 7:e7954. [PMID: 32518710 PMCID: PMC7261124 DOI: 10.7717/peerj.7954] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 09/26/2019] [Indexed: 11/20/2022] Open
Abstract
Background Influenza A (H5N1) virus is a global concern with potential as a pandemic threat. High sequence variability of influenza A viruses is a major challenge for effective vaccine design. A continuing goal towards this is a greater understanding of influenza A (H5N1) proteome sequence diversity in the context of the immune system (antigenic diversity), the dynamics of mutation, and effective strategies to overcome the diversity for vaccine design. Methods Herein, we report a comprehensive study of the dynamics of H5N1 mutations by analysis of the aligned overlapping nonamer positions (1–9, 2–10, etc.) of more than 13,000 protein sequences of avian and human influenza A (H5N1) viruses, reported over at least 50 years. Entropy calculations were performed on 9,408 overlapping nonamer position of the proteome to study the diversity in the context of immune system. The nonamers represent the predominant length of the binding cores for peptides recognized by the cellular immune system. To further dissect the sequence diversity, each overlapping nonamer position was quantitatively analyzed for four patterns of sequence diversity motifs: index, major, minor and unique. Results Almost all of the aligned overlapping nonamer positions of each viral proteome exhibited variants (major, minor, and unique) to the predominant index sequence. Each variant motif displayed a characteristic pattern of incidence change in relation to increased total variants. The major variant exhibited a restrictive pyramidal incidence pattern, with peak incidence at 50% total variants. Post this peak incidence, the minor variants became the predominant motif for majority of the positions. Unique variants, each sequence observed only once, were present at nearly all of the nonamer positions. The diversity motifs (index and variants) demonstrated complex inter-relationships, with motif switching being a common phenomenon. Additionally, 25 highly conserved sequences were identified to be shared across viruses of both hosts, with half conserved to several other influenza A subtypes. Discussion The presence of distinct sequences (nonatypes) at nearly all nonamer positions represents a large repertoire of reported viral variants in the proteome, which influence the variability dynamics of the viral population. This work elucidated and provided important insights on the components that make up the viral diversity, delineating inherent patterns in the organization of sequence changes that function in the viral fitness-selection. Additionally, it provides a catalogue of all the mutational changes involved in the dynamics of H5N1 viral diversity for both avian and human host populations. This work provides data relevant for the design of prophylactics and therapeutics that overcome the diversity of the virus, and can aid in the surveillance of existing and future strains of influenza viruses.
Collapse
Affiliation(s)
| | - Swan Tan
- School of Data Sciences, Perdana University, Serdang, Selangor, Malaysia.,Institute for Immunology and Informatics, University of Rhode Island, Providence, RI, United States of America
| | - Joseph Thomas August
- School of Medicine, Johns Hopkins University, Baltimore, MD, United States of America
| | - Asif M Khan
- School of Data Sciences, Perdana University, Serdang, Selangor, Malaysia.,School of Medicine, Johns Hopkins University, Baltimore, MD, United States of America.,Beykoz Institute of Life Sciences and Biotechnology, Bezmialem Vakif University, Beykoz, Istanbul, Turkey
| |
Collapse
|
19
|
Gao R, Sheng Z, Sreenivasan CC, Wang D, Li F. Influenza A Virus Antibodies with Antibody-Dependent Cellular Cytotoxicity Function. Viruses 2020; 12:v12030276. [PMID: 32121563 PMCID: PMC7150983 DOI: 10.3390/v12030276] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 02/26/2020] [Accepted: 02/26/2020] [Indexed: 12/11/2022] Open
Abstract
Influenza causes millions of cases of hospitalizations annually and remains a public health concern on a global scale. Vaccines are developed and have proven to be the most effective countermeasures against influenza infection. Their efficacy has been largely evaluated by hemagglutinin inhibition (HI) titers exhibited by vaccine-induced neutralizing antibodies, which correlate fairly well with vaccine-conferred protection. Contrarily, non-neutralizing antibodies and their therapeutic potential are less well defined, yet, recent advances in anti-influenza antibody research indicate that non-neutralizing Fc-effector activities, especially antibody-dependent cellular cytotoxicity (ADCC), also serve as a critical mechanism in antibody-mediated anti-influenza host response. Monoclonal antibodies (mAbs) with Fc-effector activities have the potential for prophylactic and therapeutic treatment of influenza infection. Inducing mAbs mediated Fc-effector functions could be a complementary or alternative approach to the existing neutralizing antibody-based prevention and therapy. This review mainly discusses recent advances in Fc-effector functions, especially ADCC and their potential role in influenza countermeasures. Considering the complexity of anti-influenza approaches, future vaccines may need a cocktail of immunogens in order to elicit antibodies with broad-spectrum protection via multiple protective mechanisms.
Collapse
MESH Headings
- Adaptive Immunity
- Animals
- Antibodies, Neutralizing/chemistry
- Antibodies, Neutralizing/immunology
- Antibodies, Viral/chemistry
- Antibodies, Viral/immunology
- Antibody-Dependent Cell Cytotoxicity
- Hemagglutinin Glycoproteins, Influenza Virus/chemistry
- Hemagglutinin Glycoproteins, Influenza Virus/immunology
- Host-Pathogen Interactions/immunology
- Humans
- Immunity, Innate
- Influenza A virus/immunology
- Influenza Vaccines/immunology
- Influenza, Human/immunology
- Influenza, Human/prevention & control
- Influenza, Human/virology
- Structure-Activity Relationship
Collapse
Affiliation(s)
- Rongyuan Gao
- Department of Biology and Microbiology, South Dakota State University, Brookings, SD 57007, USA; (R.G.); (C.C.S.)
| | - Zizhang Sheng
- Zuckerman Institute, Columbia University, New York, NY 10027, USA;
| | - Chithra C. Sreenivasan
- Department of Biology and Microbiology, South Dakota State University, Brookings, SD 57007, USA; (R.G.); (C.C.S.)
| | - Dan Wang
- Department of Biology and Microbiology, South Dakota State University, Brookings, SD 57007, USA; (R.G.); (C.C.S.)
- Correspondence: (D.W.); (F.L.)
| | - Feng Li
- Department of Biology and Microbiology, South Dakota State University, Brookings, SD 57007, USA; (R.G.); (C.C.S.)
- BioSNTR, Brookings, SD 57007, USA
- Correspondence: (D.W.); (F.L.)
| |
Collapse
|
20
|
Quan FS, Basak S, Chu KB, Kim SS, Kang SM. Progress in the development of virus-like particle vaccines against respiratory viruses. Expert Rev Vaccines 2020; 19:11-24. [PMID: 31903811 PMCID: PMC7103727 DOI: 10.1080/14760584.2020.1711053] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Introduction: Influenza virus, human respiratory syncytial virus (RSV), and human metapneumovirus (HMPV) are important human respiratory pathogens. Recombinant virus-like particle (VLP) vaccines are suggested to be potential promising platforms to protect against these respiratory viruses. This review updates important progress in the development of VLP vaccines against respiratory viruses.Areas Covered: This review summarizes progress in developing VLP and nanoparticle-based vaccines against influenza virus, RSV, and HMPV. The PubMed was mainly used to search for important research articles published since 2010 although earlier key articles were also referenced. The research area covered includes VLP and nanoparticle platform vaccines against seasonal, pandemic, and avian influenza viruses as well as RSV and HMPV respiratory viruses. The production methods, immunogenic properties, and vaccine efficacy of respiratory VLP vaccines in preclinical animal models and clinical studies were reviewed in this article.Expert opinion: Previous and current preclinical and clinical studies suggest that recombinant VLP and nanoparticle vaccines are expected to be developed as promising alternative platforms against respiratory viruses in future. Therefore, continued research efforts are warranted.
Collapse
Affiliation(s)
- Fu-Shi Quan
- Department of Medical Zoology, Kyung Hee University School of Medicine, Seoul, Republic of Korea.,Medical Research Center for Bioreaction to Reactive Oxygen Species and Biomedical Science Institute, School of Medicine, Graduate school, Kyung Hee University, Seoul, Republic of Korea
| | - Swarnendu Basak
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - Ki-Back Chu
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - Sung Soo Kim
- Medical Research Center for Bioreaction to Reactive Oxygen Species and Biomedical Science Institute, School of Medicine, Graduate school, Kyung Hee University, Seoul, Republic of Korea.,Department of Biochemistry and Molecular Biology, Kyung Hee University School of Medicine, Seoul, Republic of Korea
| | - Sang-Moo Kang
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| |
Collapse
|
21
|
Durous L, Rosa-Calatrava M, Petiot E. Advances in influenza virus-like particles bioprocesses. Expert Rev Vaccines 2019; 18:1285-1300. [DOI: 10.1080/14760584.2019.1704262] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Laurent Durous
- Virologie et Pathologie Humaine - VirPath team - Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, CNRS UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
| | - Manuel Rosa-Calatrava
- Virologie et Pathologie Humaine - VirPath team - Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, CNRS UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
- VirNext, Faculté de Médecine RTH Laennec, Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
| | - Emma Petiot
- Virologie et Pathologie Humaine - VirPath team - Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, CNRS UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
| |
Collapse
|
22
|
Saleh M, Nowroozi J, Fotouhi F, Farahmand B. Physicochemical study of the influenza A virus M2 protein and aluminum salt adjuvant interaction as a vaccine candidate model. Future Virol 2019. [DOI: 10.2217/fvl-2019-0019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Aim: The present study evaluated the structural changes resulting from the interaction between a recombinant influenza A virus M2 protein and aluminum hydroxide adjuvant to investigate the antigen for further immunological studies. Materials & methods: Membrane protein II was produced from the H1N1 subtype of human influenza A virus. The interaction between M2 protein and alum inum hydroxide adjuvant was evaluated by physicochemical techniques including scanning electron microscope, UV-Vis spectra, Fourier-transform infrared spectroscopy and circular dichroism spectroscopy. Results: Physicochemical methods showed high-level protein adsorption and accessibility to the effective parts of the protein. Conclusion: It was concluded that M2 protein secondary structural perturbations, including the α-helix-to-β-sheet transition, enhanced its mechanical properties toward adsorption.
Collapse
Affiliation(s)
- Maryam Saleh
- Department of Microbiology, Faculty of Biological Science, Islamic Azad University, North Tehran Branch, Tehran, Iran
| | - Jamileh Nowroozi
- Department of Microbiology, Faculty of Biological Science, Islamic Azad University, North Tehran Branch, Tehran, Iran
| | - Fatemeh Fotouhi
- Department of Influenza & Respiratory viruses, Pasteur Institute of Iran, Tehran, Iran
| | - Behrokh Farahmand
- Department of Influenza & Respiratory viruses, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
23
|
Trombetta CM, Marchi S, Manini I, Lazzeri G, Montomoli E. Challenges in the development of egg-independent vaccines for influenza. Expert Rev Vaccines 2019; 18:737-750. [DOI: 10.1080/14760584.2019.1639503] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
| | - Serena Marchi
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Ilaria Manini
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Giacomo Lazzeri
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Emanuele Montomoli
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
- VisMederi srl, Siena, Italy
| |
Collapse
|
24
|
Senapati S, Darling RJ, Loh D, Schneider IC, Wannemuehler MJ, Narasimhan B, Mallapragada SK. Pentablock Copolymer Micelle Nanoadjuvants Enhance Cytosolic Delivery of Antigen and Improve Vaccine Efficacy while Inducing Low Inflammation. ACS Biomater Sci Eng 2019; 5:1332-1342. [PMID: 33405651 PMCID: PMC8627116 DOI: 10.1021/acsbiomaterials.8b01591] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
As the focus has shifted from traditional killed or live, attenuated vaccines toward subunit vaccines, improvements in vaccine safety have been confronted with low immunogenicity of protein antigens. This issue has been addressed by synthesizing and designing a wide variety of antigen carriers and adjuvants, such as Toll-like receptor agonists (e.g., MPLA, CpG). Studies have focused on optimizing adjuvants for improved cellular trafficking, cytosolic availability, and improved antigen presentation. In this work, we describe the design of novel amphiphilic pentablock copolymer (PBC) adjuvants that exhibit high biocompatibility and reversible pH- and temperature-sensitive micelle formation. We demonstrate improved humoral immunity in mice in response to single-dose immunization with PBC micelle adjuvants compared with soluble antigen alone. With the motive of exploring the mechanism of action of these PBC micelles, we studied intracellular trafficking of these PBC micelles with a model antigen and demonstrated that the PBC micelles associate with the antigen and enhance its cytosolic delivery to antigen-presenting cells. We posit that these PBC micelles operate via immune-enhancing mechanisms that are different from that of traditional Toll-like receptor activating adjuvants. The metabolic profile of antigen-presenting cells stimulated with traditional adjuvants and the PBC micelles also suggests distinct mechanisms of action. A key finding from this study is the low production of nitric oxide and reactive oxygen species by antigen-presenting cells when stimulated by PBC micelle adjuvants in sharp contrast to TLR adjuvants. Together, these studies provide a basis for rationally developing novel vaccine adjuvants that are safe, that induce low inflammation, and that can efficiently deliver antigen to the cytosol.
Collapse
Affiliation(s)
- Sujata Senapati
- Department of Chemical and Biological Engineering, Iowa State University, Ames, Iowa 50011, United States
- Nanovaccine Institute, Iowa State University, Ames, Iowa 50011, United States
| | - Ross J. Darling
- Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, Iowa 50011, United States
- Nanovaccine Institute, Iowa State University, Ames, Iowa 50011, United States
| | - Darren Loh
- Department of Chemical and Biological Engineering, Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Ian C. Schneider
- Department of Chemical and Biological Engineering, Iowa State University, Ames, Iowa 50011, United States
- Nanovaccine Institute, Iowa State University, Ames, Iowa 50011, United States
| | - Michael J. Wannemuehler
- Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, Iowa 50011, United States
- Nanovaccine Institute, Iowa State University, Ames, Iowa 50011, United States
| | - Balaji Narasimhan
- Department of Chemical and Biological Engineering, Iowa State University, Ames, Iowa 50011, United States
- Nanovaccine Institute, Iowa State University, Ames, Iowa 50011, United States
| | - Surya K. Mallapragada
- Department of Chemical and Biological Engineering, Iowa State University, Ames, Iowa 50011, United States
- Nanovaccine Institute, Iowa State University, Ames, Iowa 50011, United States
| |
Collapse
|
25
|
Abstract
BACKGROUND The influenza virus is a pathogenic virus responsible for large numbers of deaths and long-term disabilities worldwide. Although the very young, the very old, and immunocompromised individuals are most susceptible, the effects of the influenza virus can be observed across the entire spectrum of individuals. DISCUSSION Infection with the influenza virus induces a substantial inflammatory and immunologic response and induces marked pulmonary inflammation. Many aspects of influenza affect surgical patients directly. Vaccines are one of the most effective measures aimed at reducing the prevalence and severity of many infectious diseases, including the influenza virus. Vaccination programs remain one of the highest priorities across the spectrum of countries, research institutions such as the National Institutes of Health, international health agencies such as the World Health Organization (WHO), and major non-profit organizations. CONCLUSION This review addresses aspects of the immune and inflammatory response to influenza, with a focus on the elderly population and healthcare providers who may act as reservoirs for virus transmission to the vulnerable surgical population.
Collapse
Affiliation(s)
- Daithi S Heffernan
- Division of Surgical Research, Department of Surgery, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, Rhode Island
| |
Collapse
|
26
|
Highly conserved hemagglutinin peptides of H1N1 influenza virus elicit immune response. 3 Biotech 2018; 8:492. [PMID: 30498665 DOI: 10.1007/s13205-018-1509-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2017] [Accepted: 11/09/2018] [Indexed: 01/24/2023] Open
Abstract
In the current study, two highly conserved (> 90%) H1N1 hemagglutinin peptides STDTVDTVLEKNVTVTHSVNL (H1) and KVNSVIEKMNTQFTAVGKEF (H2) containing multiple T-cell epitopes have been assessed for their immunogenic potential in vitro, subjecting peripheral blood mononuclear cells from healthy volunteers to repetitive stimulation of chemically synthesised H1 and H2 peptides, and measuring their interferon (IFN)-γ level (ELISA) and proliferation (MTT assay). Further, these peptides were analysed for their binding affinity with 18 different human leukocyte antigen (HLA) class I and II by means of molecular docking. All seven samples tested for H1- and H2-induced IFN-γ secretion were found to have enhanced IFN-γ production. Six (H1) and five (H2) samples have shown proliferative response compared to unstimulated cells. Peptide-induced IFN-γ secretion and proliferation in healthy samples represent the immunogenic potential of these peptides. Further, molecular docking results reveal that the peptides have comparable binding energy to that of native bound peptide for both HLA classes which indicates that these peptides have the capability to be presented by different HLA molecules required for T-cell response. Hence, these conserved immunogenic hemagglutinin peptides are potential candidates for influenza vaccine development.
Collapse
|
27
|
Keshavarz M, Mirzaei H, Salemi M, Momeni F, Mousavi MJ, Sadeghalvad M, Arjeini Y, Solaymani-Mohammadi F, Sadri Nahand J, Namdari H, Mokhtari-Azad T, Rezaei F. Influenza vaccine: Where are we and where do we go? Rev Med Virol 2018; 29:e2014. [PMID: 30408280 DOI: 10.1002/rmv.2014] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 09/22/2018] [Accepted: 09/25/2018] [Indexed: 12/11/2022]
Abstract
The alarming rise of morbidity and mortality caused by influenza pandemics and epidemics has drawn attention worldwide since the last few decades. This life-threatening problem necessitates the development of a safe and effective vaccine to protect against incoming pandemics. The currently available flu vaccines rely on inactivated viral particles, M2e-based vaccine, live attenuated influenza vaccine (LAIV) and virus like particle (VLP). While inactivated vaccines can only induce systemic humoral responses, LAIV and VLP vaccines stimulate both humoral and cellular immune responses. Yet, these vaccines have limited protection against newly emerging viral strains. These strains, however, can be targeted by universal vaccines consisting of conserved viral proteins such as M2e and capable of inducing cross-reactive immune response. The lack of viral genome in VLP and M2e-based vaccines addresses safety concern associated with existing attenuated vaccines. With the emergence of new recombinant viral strains each year, additional effort towards developing improved universal vaccine is warranted. Besides various types of vaccines, microRNA and exosome-based vaccines have been emerged as new types of influenza vaccines which are associated with new and effective properties. Hence, development of a new generation of vaccines could contribute to better treatment of influenza.
Collapse
Affiliation(s)
- Mohsen Keshavarz
- Department of Medical Virology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
| | - Maryam Salemi
- Department of Genomics and Genetic Engineering, Razi Vaccine and Serum Research Institute (RVSRI), Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| | - Fatemeh Momeni
- Thalassemia and Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohammad Javad Mousavi
- Department of Immunology and Allergy, Faculty of Medicine, Bushehr University of Medical Sciences, Bushehr, Iran.,Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mona Sadeghalvad
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Yaser Arjeini
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Farid Solaymani-Mohammadi
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Javid Sadri Nahand
- Department of Medical Virology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Haideh Namdari
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Talat Mokhtari-Azad
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Farhad Rezaei
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
28
|
Design of a Universal Influenza A Vaccine Candidate Based on M2e.FliC; Immunoinformatics Analysis, Protein Modeling, and Its Expression in Escherichia coli. Jundishapur J Microbiol 2018. [DOI: 10.5812/jjm.66592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
29
|
Dhakal S, Cheng X, Salcido J, Renu S, Bondra K, Lakshmanappa YS, Misch C, Ghimire S, Feliciano-Ruiz N, Hogshead B, Krakowka S, Carson K, McDonough J, Lee CW, Renukaradhya GJ. Liposomal nanoparticle-based conserved peptide influenza vaccine and monosodium urate crystal adjuvant elicit protective immune response in pigs. Int J Nanomedicine 2018; 13:6699-6715. [PMID: 30425484 PMCID: PMC6205527 DOI: 10.2147/ijn.s178809] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Background Influenza (flu) is a constant threat to humans and animals, and vaccination is one of the most effective ways to mitigate the disease. Due to incomplete protection induced by current flu vaccines, development of novel flu vaccine candidates is warranted to achieve greater efficacy against constantly evolving flu viruses. Methods In the present study, we used liposome nanoparticle (<200 nm diameter)-based subunit flu vaccine containing ten encapsulated highly conserved B and T cell epitope peptides to induce protective immune response against a zoonotic swine influenza A virus (SwIAV) H1N1 challenge infection in a pig model. Furthermore, we used monosodium urate (MSU) crystals as an adjuvant and co-administered the vaccine formulation as an intranasal mist to flu-free nursery pigs, twice at 3-week intervals. Results Liposome peptides flu vaccine delivered with MSU adjuvant improved the hemagglutination inhibition antibody titer and mucosal IgA response against the SwIAV challenge and also against two other highly genetically variant IAVs. Liposomal vaccines also enhanced the frequency of peptides and virus-specific T-helper/memory cells and IFN-γ response. The improved specific cellular and mucosal humoral immune responses in adjuvanted liposomal peptides flu vaccine partially protected pigs from flu-induced fever and pneumonic lesions, and reduced the nasal virus shedding and viral load in the lungs. Conclusion Overall, our study shows great promise for using liposome and MSU adjuvant- based subunit flu vaccine through the intranasal route, and provides scope for future, pre-clinical investigations in a pig model for developing potent human intranasal subunit flu vaccines.
Collapse
Affiliation(s)
- Santosh Dhakal
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, Wooster, OH 44691, USA, .,Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA,
| | - Xingguo Cheng
- Pharmaceuticals and Bioengineering Department, Chemistry and Chemical Engineering Division, Southwest Research Institute, San Antonio, TX 78238-0510, USA,
| | - John Salcido
- Pharmaceuticals and Bioengineering Department, Chemistry and Chemical Engineering Division, Southwest Research Institute, San Antonio, TX 78238-0510, USA,
| | - Sankar Renu
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, Wooster, OH 44691, USA, .,Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA,
| | - Kathy Bondra
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, Wooster, OH 44691, USA, .,Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA,
| | - Yashavantha Shaan Lakshmanappa
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, Wooster, OH 44691, USA, .,Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA,
| | - Christina Misch
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, Wooster, OH 44691, USA, .,Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA,
| | - Shristi Ghimire
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, Wooster, OH 44691, USA, .,Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA,
| | - Ninoshkaly Feliciano-Ruiz
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, Wooster, OH 44691, USA, .,Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA,
| | - Bradley Hogshead
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, Wooster, OH 44691, USA, .,Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA,
| | - Steven Krakowka
- The Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
| | - Kenneth Carson
- Pharmaceuticals and Bioengineering Department, Chemistry and Chemical Engineering Division, Southwest Research Institute, San Antonio, TX 78238-0510, USA,
| | - Joseph McDonough
- Pharmaceuticals and Bioengineering Department, Chemistry and Chemical Engineering Division, Southwest Research Institute, San Antonio, TX 78238-0510, USA,
| | - Chang Won Lee
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, Wooster, OH 44691, USA, .,Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA,
| | - Gourapura J Renukaradhya
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, Wooster, OH 44691, USA, .,Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA,
| |
Collapse
|
30
|
Lee DK, Lee EY, Kim RH, Kwak HW, Kim JY, Kim H, Kang KW, Lee SM, Park JH, Chang J, Nam JH. Effect of apoptosis-associated speck-like protein containing a caspase recruitment domain on vaccine efficacy: Overcoming the effects of its deficiency with aluminum hydroxide adjuvant. Microbiol Immunol 2018; 62:176-186. [PMID: 29315762 DOI: 10.1111/1348-0421.12569] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 12/08/2017] [Accepted: 12/22/2017] [Indexed: 11/30/2022]
Abstract
Host factors such as nutritional status and immune cell state are important for vaccine efficacy. Inflammasome activation may be important for triggering vaccine-induced humoral and cell-mediated immune responses. Formulations with alum as a typical adjuvant to overcome the effects of host factors have recently been shown to induce inflammasome activation, which augments vaccine efficacy. Apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC) is one of the main components of inflammasomes, but it is not clear whether ASC affects the vaccine-induced immune response. Herein, we used two types of vaccines: inactivated influenza vaccine not formulated with alum, and HPV vaccine formulated with alum. We gave the vaccines to ASC knockout (ASC-/- ) mice to investigate the role of ASC in vaccine efficacy. Influenza vaccine-immunized ASC-/- mice did not show antibody titers in week 2 after the first vaccination. After boosting, the antibody titer in ASC-/- mice was about half that in wild type (WT) mice. Furthermore, a cytotoxic T-lymphocyte response against influenza vaccine was not induced in ASC-/- mice. Therefore, vaccinated ASC-/- mice did not show effective protection against viral challenge. ASC-/- mice immunized with alum-formulated HPV vaccine showed similar antibody titers and T-cell proliferation compared with immunized WT mice. However, the HPV vaccine without alum induced up to threefold lower titers of HPV-specific antibody titers in ASC-/- mice compared with those in WT mice. These findings suggest that alum in vaccine can overcome the ASC-deficient condition.
Collapse
Affiliation(s)
- Deuk-Ki Lee
- Department of Biotechnology, The Catholic University of Korea, Bucheon, 14662, South Korea
| | - Eun-Young Lee
- Department of Biotechnology, The Catholic University of Korea, Bucheon, 14662, South Korea
| | - Ryoon-Ho Kim
- Department of Biotechnology, The Catholic University of Korea, Bucheon, 14662, South Korea
| | - Hye-Won Kwak
- Department of Biotechnology, The Catholic University of Korea, Bucheon, 14662, South Korea
| | - Joo Young Kim
- Division of Life & Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, South Korea
| | - Hun Kim
- SK Chemical, Sampyeong-dong, Bundang-gu, Seongnam-si, Gyeonggi-do, 13493, South Korea
| | - Kyung-Won Kang
- Department of Biotechnology, Chonbuk National University, Iksan, 570-752, South Korea
| | - Sang-Myeong Lee
- Department of Biotechnology, Chonbuk National University, Iksan, 570-752, South Korea
| | - Jong-Hwan Park
- Laboratory Animal Medicine, College of Veterinary Medicine, Chonnam National University, Gwangju, 61186, South Korea
| | - Jun Chang
- Division of Life & Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, South Korea
| | - Jae-Hwan Nam
- Department of Biotechnology, The Catholic University of Korea, Bucheon, 14662, South Korea
| |
Collapse
|
31
|
Dehghan A, Shahsavandi S, Jabalameli L. Improvement Efficacy of Influenza Nanovaccine in Combination with Hemokinin-1 Molecular Adjuvant. Avicenna J Med Biotechnol 2018; 10:208-213. [PMID: 30555652 PMCID: PMC6252024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
BACKGROUND H9N2 avian influenza viruses have the potential to become the next human pandemic threat and next generation vaccine technologies are needed. Current studies introduce nanoparticles as a proper vaccine delivery vehicle for induction of protective immunity. In this study, the efficacy of chitosan nanoparticle-based H9N2 influenza vaccine with and without hemokinin-1 (HK-1) as a molecular adjuvant to induce protective immunity against the virus was examined. METHODS The H9N2 antigen was prepared in MDCK cells and inactivated with formalin. The inactivated antigen alone and in combination with HK-1 was encapsulated into chitosan nanoparticles. Groups of BALB/c mice received chitosan nanoparticle-based H9N2 antigen alone or in combination with HK-1 in a prime/boost platform via eye drop method. To evaluate the efficacy of the adjuvanted-nanovaccine candidate, systemic antibody responses were compared among the groups of animals. RESULTS Serological analysis indicated that mice receiving the HK-1/H9N2 nanoparticles formulation induced higher antibody titers that were sustained until the end of experiment. However, in the immunized mice, influenza specific antibody titers were comparable to that in the animals which were immunized either with inactivated antigen alone or the H9N2 nanoparticles without HK-1 adjuvant. CONCLUSION The data demonstrate the synergy between HK-1 as an adjuvant and chitosan nanoparticles as a delivery antigen/adjuvant carrier in the improvement of influenza immune responses.
Collapse
Affiliation(s)
- Atefeh Dehghan
- Department of Microbiology, Karaj Branch, Islamic Azad University, Karaj, Iran
| | - Shahla Shahsavandi
- Razi Vaccine and Serum Research Institute, Agricultural Research Education and Extension Organization, Karaj, Iran,Corresponding author: Shahla Shahsavandi, Ph.D., Razi Vaccine and Serum Research Institute, Agricultural Research Education and Extension Organization, Karaj, Iran, Tel: +98 263 4570038, Fax: +98 263 4552194, E-mail:
| | - Leila Jabalameli
- Department of Microbiology, Karaj Branch, Islamic Azad University, Karaj, Iran
| |
Collapse
|
32
|
Anderson AJ, Snelling TL, Moore HC, Blyth CC. Advances in Vaccines to Prevent Viral Respiratory Illnesses in Children. Paediatr Drugs 2017; 19:523-531. [PMID: 28808938 DOI: 10.1007/s40272-017-0257-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Childhood vaccination has played a critical role in the reduction of morbidity and mortality from communicable diseases, including specific respiratory pathogens. Acute lower respiratory infection (ALRI) of both bacterial and viral aetiology continues to impact global child health. Key bacterial pathogens including Streptococcus pneumoniae and Haemophilus influenza type b are specifically targeted with current vaccination programmes, while at present there are less effective strategies for the prevention of viral disease. Influenza vaccines, including both live attenuated intranasal vaccines and inactivated influenza vaccines, are limited by seasonal strain variation and unsustained immunity. Research into the development of a universal influenza vaccine is ongoing; potential targets are the conserved regions of the virus such as the M2e antigen and hemagglutinin stalk. Respiratory syncytial virus (RSV) and parainfluenza virus 3 (PIV3) are the viral pathogens most commonly causing ALRI in children, particularly the infant population. Currently, no vaccine exists for either virus. Over the last decade, promising advances have been made. Protection of neonates via maternal RSV immunisation is being assessed in a phase III clinical trial, with many other candidates for RSV and PIV3 at less advanced stages of development.
Collapse
Affiliation(s)
- Aleisha J Anderson
- Department of Infectious Diseases, Princess Margaret Hospital, Roberts Rd, Subiaco, WA, Australia
| | - Tom L Snelling
- Department of Infectious Diseases, Princess Margaret Hospital, Roberts Rd, Subiaco, WA, Australia
- Wesfarmers Centre for Vaccines and Infectious Diseases, Telethon Kids Institute, Roberts Rd, Subiaco, WA, Australia
| | - Hannah C Moore
- Wesfarmers Centre for Vaccines and Infectious Diseases, Telethon Kids Institute, Roberts Rd, Subiaco, WA, Australia
| | - Christopher C Blyth
- Department of Infectious Diseases, Princess Margaret Hospital, Roberts Rd, Subiaco, WA, Australia.
- Wesfarmers Centre for Vaccines and Infectious Diseases, Telethon Kids Institute, Roberts Rd, Subiaco, WA, Australia.
- Discipline of Paediatrics, School of Medicine, Princess Margaret Hospital, University of Western Australia, Roberts Rd, Subiaco, WA, Australia.
- PathWest Laboratory Medicine WA, Department of Microbiology, QEII Medical Centre, Nedlands, WA, Australia.
| |
Collapse
|
33
|
Bahadoran A, Ebrahimi M, Yeap SK, Safi N, Moeini H, Hair-Bejo M, Hussein MZ, Omar AR. Induction of a robust immune response against avian influenza virus following transdermal inoculation with H5-DNA vaccine formulated in modified dendrimer-based delivery system in mouse model. Int J Nanomedicine 2017; 12:8573-8585. [PMID: 29270010 PMCID: PMC5729183 DOI: 10.2147/ijn.s139126] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
This study was aimed to evaluate the immunogenicity of recombinant plasmid deoxyribonucleic acid (DNA), pBud-H5-green fluorescent protein (GFP)-interferon-regulatory factor (IRF)3 following delivery using polyamidoamine (PAMAM) dendrimer and transactivator of transcription (TAT)-conjugated PAMAM dendrimer as well as the effect of IRF3 as the genetic adjuvant. BALB/c mice were vaccinated transdermally with pBud-H5-GFP, PAMAM/pBud-H5-GFP, TAT-PAMAM/pBud-H5-GFP, and TAT-PAMAM/pBud-H5-GFP-IRF3. The expression analysis of H5 gene from the blood by using quantitative real-time reverse transcriptase polymerase chain reaction confirmed the ability of PAMAM dendrimer as a carrier for gene delivery, as well as the ability of TAT peptide to enhance the delivery efficiency of PAMAM dendrimer. Mice immunized with modified PAMAM by TAT peptide showed higher hemagglutination inhibition titer, and larger CD3+/CD4+ T cells and CD3+/CD8+ T cells population, as well as the production of cytokines, namely, interferon (IFN)-γ, interleukin (IL)-2, IL-15, IL-12, IL-6, and tumor necrosis factor-α compared with those immunized with native PAMAM. These results suggest that the function of TAT peptide as a cell-penetrating peptide is able to enhance the gene delivery, which results in rapid distribution of H5 in the tissues of the immunized mice. Furthermore, pBud-H5-GFP co-expressing IRF3 as a genetic adjuvant demonstrated the highest hemagglutination inhibition titer besides larger CD3+/CD4+ and CD3+/CD8+ T cells population, and strong Th1-like cytokine responses among all the systems tested. In conclusion, TAT-PAMAM dendrimer-based delivery system with IRF3 as a genetic adjuvant is an attractive transdermal DNA vaccine delivery system utilized to evaluate the efficacy of the developed DNA vaccine in inducing protection during challenge with virulent H5N1 virus.
Collapse
Affiliation(s)
- Azadeh Bahadoran
- Institute of Bioscience, Universiti Putra Malaysia, UPM, Serdang.,Department of Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur
| | - Mehdi Ebrahimi
- Department of Veterinary Preclinical Sciences, Universiti Putra Malaysia, UPM, Serdang, Malaysia
| | - Swee Keong Yeap
- Institute of Bioscience, Universiti Putra Malaysia, UPM, Serdang
| | - Nikoo Safi
- Institute of Bioscience, Universiti Putra Malaysia, UPM, Serdang
| | | | - Mohd Hair-Bejo
- Institute of Bioscience, Universiti Putra Malaysia, UPM, Serdang.,Department of Veterinary Pathology and Microbiology, Universiti Putra Malaysia, UPM
| | - Mohd Zobir Hussein
- Advanced Technology Institute, Universiti Putra Malaysia, UPM, Serdang, Malaysia
| | - Abdul Rahman Omar
- Institute of Bioscience, Universiti Putra Malaysia, UPM, Serdang.,Department of Veterinary Pathology and Microbiology, Universiti Putra Malaysia, UPM
| |
Collapse
|
34
|
Development of a novel dual-domain nanoparticle antigen construct for universal influenza vaccine. Vaccine 2017; 35:7026-7032. [PMID: 29102171 DOI: 10.1016/j.vaccine.2017.10.051] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 08/07/2017] [Accepted: 10/16/2017] [Indexed: 12/25/2022]
Abstract
A highly effective antigen construct for presenting conserved antigen domains is essential to the development of a universal influenza vaccine. We have developed a novel dual-domain nanoparticle fusion protein (DDNFP) which allows independent presentation of two conserved domains. The conserved domains used were from two separate viral surface proteins, M2e of M2 and fusion peptide (FP) or long alpha helix (CD) of HA2. The carrier is a novel nanoparticle protein - the dodecameric DNA binding protein from starved cells (Dps) of bacteria or archaea. Dps was found to be uniquely capable of simultaneous fusion and surface presentation at both N- and C-termini while retaining the ability to form nanoparticles. Thus, DDNFPs with M2e and FP or CD fused at N- and C-termini of Dps from E. coli (EcDps) or other bacteria were first constructed based on the H1 subtype sequences along with corresponding single-domain nanoparticle fusion proteins (SDNFPs). They were expressed at high levels in bacteria and found to form nanoparticles of the expected size (∼9 nm). They were stable against treatment at high temperatures. The DDNFPs (M2e-EcDps-FP and M2e-EcDps-CD) induced strong antibody responses against individual antigen domains and provided full protection against lethal challenge with PR8 virus (H1N1). Importantly, the protection by DDNFPs was synergistically enhanced as compared to SDNFPs. The M2e-EcDps-CD provided an even stronger protection than M2e-EcDps-FP and therefore appeared to be the superior construct. Together, with novel domain combination, enhanced protection and ease of production, this M2e/CD DDNFP could potentially be a highly effective antigen construct for the universal influenza vaccine.
Collapse
|
35
|
Lohia N, Baranwal M. Immune responses to highly conserved influenza A virus matrix 1 peptides. Microbiol Immunol 2017; 61:225-231. [PMID: 28429374 DOI: 10.1111/1348-0421.12485] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 04/12/2017] [Accepted: 04/18/2017] [Indexed: 01/07/2023]
Abstract
Influenza vaccine development is considered to be complicated and challenging. Constantly evolving influenza viruses require continuous global monitoring and reformulation of the vaccine strains. Peptides that are conserved among different strains and subtypes of influenza A virus are strongly considered to be attractive targets for development of cross protective influenza vaccines that stimulate cellular responses. In this study, three highly conserved (>90%) matrix 1 peptides that contain multiple T cell epitopes, ILGFVFTLTVPSERGLQRRRF (PM 1), LIRHENRMVLASTTAKA (PM 2) and LQAYQKRMGVQMQR (PM 3), were assessed for their immunogenic potential in vitro by subjecting peripheral blood mononuclear cells from healthy volunteers to repetitive stimulation with these chemically synthesised peptides and measuring their IFN-γ concentrations, proliferation by ELISA, and 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay, respectively. Seven samples were screened for immunogenicity of PM 1 and PM 2, and six for that of PM 3. All six samples had positive responses (IFN-γ secretion) to PM 3 stimulation, as did five and three for PM 2 and PM 1 respectively. In contrast, seven (PM 1 and PM 2) and four (PM 3) samples showed proliferative response as compared with unstimulated cells. The encouraging immunogenic response generated by these highly conserved matrix 1 peptides indicates they are prospective candidates for development of broadly reactive influenza vaccines.
Collapse
Affiliation(s)
- Neha Lohia
- Department of Biotechnology, Thapar University, Patiala147004, India
| | - Manoj Baranwal
- Department of Biotechnology, Thapar University, Patiala147004, India
| |
Collapse
|
36
|
Kim YJ, Ko EJ, Kim MC, Lee YN, Kim KH, Jung YJ, Kang SM. Roles of antibodies to influenza A virus hemagglutinin, neuraminidase, and M2e in conferring cross protection. Biochem Biophys Res Commun 2017; 493:393-398. [PMID: 28887040 DOI: 10.1016/j.bbrc.2017.09.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Accepted: 09/04/2017] [Indexed: 12/20/2022]
Abstract
Although neuraminidase (NA) is the second major viral glycoprotein of influenza virus, its immune mechanism as a vaccine target has been less considered. Here we compared the properties of antibodies and the efficacy of cross protection by N1 and N2 NA proteins, inactivated split influenza vaccines (split), and tandem repeat extracellular domain M2 on virus-like particles (M2e5x VLP). Anti-NA immune sera could confer better cross-protection against multiple heterologous influenza viruses correlating with NA inhibition activity compared to split vaccine immune sera. Whereas split vaccine was superior to NA in conferring homologous protection. NA and M2e immune sera each showed comparable survival protection. Protective efficacy by NA immune sera was lower in Fc receptor common γ-chain deficient mice but comparable in C3 complement deficient mice compared to that in wild type mice, suggesting a role of Fc receptor in NA immunity.
Collapse
Affiliation(s)
- Yu-Jin Kim
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Eun-Ju Ko
- 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
| | - Yu-Na Lee
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Ki-Hye Kim
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Yu-Jin Jung
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Sang-Moo Kang
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA.
| |
Collapse
|
37
|
Riese P, Guzmán CA. Roads to advanced vaccines: influenza case study. Microb Biotechnol 2017; 10:1036-1040. [PMID: 28809451 PMCID: PMC5609253 DOI: 10.1111/1751-7915.12835] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 07/20/2017] [Indexed: 11/30/2022] Open
Abstract
Vaccines represent a cornerstone to ensure healthy lives and promote well‐being for all at all ages. However, there are many diseases for which vaccines are not available, are relatively ineffective or need to be adapted periodically. Advances in microbial biotechnology will contribute to overcoming these roadblocks by laying the groundwork for improving and creating new approaches for developing better vaccines, as illustrated here in the case of influenza.
Collapse
Affiliation(s)
- Peggy Riese
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, D-38124, Braunschweig, Germany
| | - Carlos A Guzmán
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, D-38124, Braunschweig, Germany
| |
Collapse
|
38
|
Abstract
Influenza is an acute respiratory illness, caused by influenza A, B, and C viruses, that occurs in local outbreaks or seasonal epidemics. Clinical illness follows a short incubation period and presentation ranges from asymptomatic to fulminant, depending on the characteristics of both the virus and the individual host. Influenza A viruses can also cause sporadic infections or spread worldwide in a pandemic when novel strains emerge in the human population from an animal host. New approaches to influenza prevention and treatment for management of both seasonal influenza epidemics and pandemics are desirable. In this Seminar, we discuss the clinical presentation, transmission, diagnosis, management, and prevention of seasonal influenza infection. We also review the animal-human interface of influenza, with a focus on current pandemic threats.
Collapse
Affiliation(s)
- Catharine Paules
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Kanta Subbarao
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
39
|
Choi WS, Lloren KKS, Baek YH, Song MS. The significance of avian influenza virus mouse-adaptation and its application in characterizing the efficacy of new vaccines and therapeutic agents. Clin Exp Vaccine Res 2017; 6:83-94. [PMID: 28775972 PMCID: PMC5540968 DOI: 10.7774/cevr.2017.6.2.83] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 05/03/2017] [Accepted: 05/12/2017] [Indexed: 11/26/2022] Open
Abstract
Due to the increased frequency of interspecies transmission of avian influenza viruses, studies designed to identify the molecular determinants that could lead to an expansion of the host range have been increased. A variety of mouse-based mammalian-adaptation studies of avian influenza viruses have provided insight into the genetic alterations of various avian influenza subtypes that may contribute to the generation of a pandemic virus. To date, the studies have focused on avian influenza subtypes H5, H6, H7, H9, and H10 which have recently caused human infection. Although mice cannot fully reflect the course of human infection with avian influenza, these mouse studies can be a useful method for investigating potential mammalian adaptive markers against newly emerging avian influenza viruses. In addition, due to the lack of appropriate vaccines against the diverse emerging influenza viruses, the generation of mouse-adapted lethal variants could contribute to the development of effective vaccines or therapeutic agents. Within this review, we will summarize studies that have demonstrated adaptations of avian influenza viruses that result in an altered pathogenicity in mice which may suggest the potential application of mouse-lethal strains in the development of influenza vaccines and/or therapeutics in preclinical studies.
Collapse
Affiliation(s)
- Won-Suk Choi
- Department of Microbiology, College of Medicine and Medical Research Institute, Chungbuk National University, Cheongju, Korea
| | - Khristine Kaith S Lloren
- Department of Microbiology, College of Medicine and Medical Research Institute, Chungbuk National University, Cheongju, Korea
| | - Yun Hee Baek
- Department of Microbiology, College of Medicine and Medical Research Institute, Chungbuk National University, Cheongju, Korea
| | - Min-Suk Song
- Department of Microbiology, College of Medicine and Medical Research Institute, Chungbuk National University, Cheongju, Korea
| |
Collapse
|
40
|
van de Wakker SI, Fischer MJ, Oosting RS. New drug-strategies to tackle viral-host interactions for the treatment of influenza virus infections. Eur J Pharmacol 2017; 809:178-190. [DOI: 10.1016/j.ejphar.2017.05.038] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Revised: 04/24/2017] [Accepted: 05/19/2017] [Indexed: 12/13/2022]
|
41
|
Wang C, Yan F, Zheng X, Wang H, Jin H, Wang C, Zhao Y, Feng N, Wang T, Gao Y, Yang S, Xia X. Porcine epidemic diarrhea virus virus-like particles produced in insect cells induce specific immune responses in mice. Virus Genes 2017; 53:548-554. [PMID: 28357676 PMCID: PMC7088547 DOI: 10.1007/s11262-017-1450-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 03/21/2017] [Indexed: 02/06/2023]
Abstract
Porcine epidemic diarrhea virus (PEDV), which causes 80-100% mortality in neonatal piglets, is one of the most devastating viral diseases affecting swine worldwide. To date, the lack of effective vaccines and drugs is the main problem preventing control of the global spread of PEDV. In this study, we produced PEDV virus-like particles (VLPs) composed of S, M, and E proteins with a baculovirus expression system and tested them via indirect immunofluorescence assay (IFA)and Western blot analysis. Electron microscopy showed that the morphological structure of the PEDV VLPs was similar to that of the protovirus. Microneutralization assays and ELISpot analysis demonstrated that PEDV VLPs induced highly specific antibody responses and Th2-mediated humoral immunity. As a result, the PEDV VLPs displayed excellent immunogenicity in mice. Therefore, a VLP-based vaccine has the potential to prevent PEDV infection.
Collapse
Affiliation(s)
- Cuiling Wang
- College of Animal Science and Technology, Shihezi University, Shihezi, China
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, China
| | - Feihu Yan
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, China
| | - Xuexing Zheng
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, China
- School of Public Health, Shandong University, Jinan, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Hualei Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Hongli Jin
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, China
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Chong Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, China
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Yongkun Zhao
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Na Feng
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Tiecheng Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Yuwei Gao
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Songtao Yang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, China.
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China.
| | - Xianzhu Xia
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, China.
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China.
| |
Collapse
|
42
|
Zhu W, Wang C, Wang BZ. From Variation of Influenza Viral Proteins to Vaccine Development. Int J Mol Sci 2017; 18:ijms18071554. [PMID: 28718801 PMCID: PMC5536042 DOI: 10.3390/ijms18071554] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 07/10/2017] [Accepted: 07/14/2017] [Indexed: 11/19/2022] Open
Abstract
Recurrent influenza epidemics and occasional pandemics are one of the most important global public health concerns and are major causes of human morbidity and mortality. Influenza viruses can evolve through antigen drift and shift to overcome the barriers of human immunity, leading to host adaption and transmission. Mechanisms underlying this viral evolution are gradually being elucidated. Vaccination is an effective method for the prevention of influenza virus infection. However, the emergence of novel viruses, including the 2009 pandemic influenza A (H1N1), the avian influenza A virus (H7N9), and the highly pathogenic avian influenza A virus (HPAI H5N1), that have infected human populations frequently in recent years reveals the tremendous challenges to the current influenza vaccine strategy. A better vaccine that provides protection against a wide spectrum of various influenza viruses and long-lasting immunity is urgently required. Here, we review the evolutionary changes of several important influenza proteins and the influence of these changes on viral antigenicity, host adaption, and viral pathogenicity. Furthermore, we discuss the development of a potent universal influenza vaccine based on this knowledge.
Collapse
Affiliation(s)
- Wandi Zhu
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, Atlanta, GA 30303, USA.
| | - Chao Wang
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, Atlanta, GA 30303, USA.
| | - Bao-Zhong Wang
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, Atlanta, GA 30303, USA.
| |
Collapse
|
43
|
Puth S, Hong SH, Park MJ, Lee HH, Lee YS, Jeong K, Kang IC, Koh JT, Moon B, Park SC, Rhee JH, Lee SE. Mucosal immunization with a flagellin-adjuvanted Hgp44 vaccine enhances protective immune responses in a murine Porphyromonas gingivalis infection model. Hum Vaccin Immunother 2017; 13:2794-2803. [PMID: 28604268 PMCID: PMC5718812 DOI: 10.1080/21645515.2017.1327109] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Chronic periodontitis is caused by interactions between the oral polymicrobial community and host factors. Periodontal diseases are associated with dysbiotic shift in oral microbiota. Vaccination against periodontopathic bacteria could be a fundamental therapeutic to modulate polymicrobial biofilms. Because oral cavity is the site of periodontopathic bacterial colonization, mucosal vaccines should provide better protection than vaccines administered systemically. We previously reported that bacterial flagellin is an excellent mucosal adjuvant. In this study, we investigated whether mucosal immunization with a flagellin-adjuvanted polypeptide vaccine induces protective immune responses using a Porphyromonas gingivalis infection model. We used the Hgp44 domain polypeptide of Arg-gingipain A (RgpA) as a mucosal antigen. Intranasal (IN) immunization induced a significantly higher Hgp44-specific IgG titer in the serum of mice than sublingual (SL) administration. The co-administration of flagellin potentiated serum IgG responses for both the IN and SL vaccinations. On the other hand, the anti-Hgp44-specific IgA titer in the saliva was comparable between IN and SL vaccinations, suggesting SL administration as more compliant vaccination route for periodontal vaccines. The co-administration of flagellin significantly potentiated the secretory IgA response in saliva also. Furthermore, mice administered a mixture of Hgp44 and flagellin via the IN and SL routes exhibited significant reductions in alveolar bone loss induced by live P. gingivalis infections. An intranasally administered Hgp44-flagellin fusion protein induced a comparable level of Hgp44-specific antibody responses to the mixture of Hgp44 and flagellin. Overall, a flagellin-adjuvanted Hgp44 antigen would serve an important component for a multivalent mucosal vaccine against polymicrobial periodontitis.
Collapse
Affiliation(s)
- Sao Puth
- a Clinical Vaccine R&D Center , Chonnam National University , Hwasun-gun , Jeonnam , Republic of Korea.,b Department of Microbiology , Chonnam National University Medical School , Hwasun-gun , Jeonnam , Republic of Korea
| | - Seol Hee Hong
- a Clinical Vaccine R&D Center , Chonnam National University , Hwasun-gun , Jeonnam , Republic of Korea.,b Department of Microbiology , Chonnam National University Medical School , Hwasun-gun , Jeonnam , Republic of Korea
| | - Mi Jin Park
- a Clinical Vaccine R&D Center , Chonnam National University , Hwasun-gun , Jeonnam , Republic of Korea.,b Department of Microbiology , Chonnam National University Medical School , Hwasun-gun , Jeonnam , Republic of Korea
| | - Hye Hwa Lee
- a Clinical Vaccine R&D Center , Chonnam National University , Hwasun-gun , Jeonnam , Republic of Korea.,c Department of Pharmacology and Dental Therapeutics, School of Dentistry , Chonnam National University , Gwangju , Republic of Korea ; Department of Microbiology , Chonnam National University Medical School , Hwasun-gun , Jeonnam , Republic of Korea
| | - Youn Suhk Lee
- a Clinical Vaccine R&D Center , Chonnam National University , Hwasun-gun , Jeonnam , Republic of Korea.,c Department of Pharmacology and Dental Therapeutics, School of Dentistry , Chonnam National University , Gwangju , Republic of Korea ; Department of Microbiology , Chonnam National University Medical School , Hwasun-gun , Jeonnam , Republic of Korea
| | - Kwangjoon Jeong
- a Clinical Vaccine R&D Center , Chonnam National University , Hwasun-gun , Jeonnam , Republic of Korea.,b Department of Microbiology , Chonnam National University Medical School , Hwasun-gun , Jeonnam , Republic of Korea
| | - In-Chol Kang
- d Department of Oral Microbiology, School of Dentistry , Chonnam National University , Gwangju , Republic of Korea
| | - Jeong Tae Koh
- c Department of Pharmacology and Dental Therapeutics, School of Dentistry , Chonnam National University , Gwangju , Republic of Korea ; Department of Microbiology , Chonnam National University Medical School , Hwasun-gun , Jeonnam , Republic of Korea
| | - Byounggon Moon
- e Well Aging Research Center, Samsung Adv. Inst. of Technology (SAIT) , Samsung Electronics Co., Ltd. Suwon-si , Gyeonggi-do , Republic of Korea
| | - Sang Chul Park
- e Well Aging Research Center, Samsung Adv. Inst. of Technology (SAIT) , Samsung Electronics Co., Ltd. Suwon-si , Gyeonggi-do , Republic of Korea
| | - Joon Haeng Rhee
- a Clinical Vaccine R&D Center , Chonnam National University , Hwasun-gun , Jeonnam , Republic of Korea.,b Department of Microbiology , Chonnam National University Medical School , Hwasun-gun , Jeonnam , Republic of Korea
| | - Shee Eun Lee
- a Clinical Vaccine R&D Center , Chonnam National University , Hwasun-gun , Jeonnam , Republic of Korea.,c Department of Pharmacology and Dental Therapeutics, School of Dentistry , Chonnam National University , Gwangju , Republic of Korea ; Department of Microbiology , Chonnam National University Medical School , Hwasun-gun , Jeonnam , Republic of Korea
| |
Collapse
|
44
|
Pendzialek J, Roose K, Smet A, Schepens B, Kufer P, Raum T, Baeuerle PA, Muenz M, Saelens X, Fiers W. Bispecific T cell engaging antibody constructs targeting a universally conserved part of the viral M2 ectodomain cure and prevent influenza A virus infection. Antiviral Res 2017; 141:155-164. [PMID: 28257797 DOI: 10.1016/j.antiviral.2017.02.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 02/08/2017] [Accepted: 02/27/2017] [Indexed: 11/25/2022]
Abstract
The ectodomain of the influenza A matrix protein 2 (M2e) is highly conserved amongst all influenza virus A subtypes. M2e is present on the surface of influenza A virus-infected cells, and therefore a suitable target for broadly protective therapies. We designed bispecific T cell engaging (BiTE®) antibody constructs specific for M2e by genetically fusing a single chain variable fragment (scFv) derived from an M2e-specific murine monoclonal antibody with a CD3ɛ-specific scFv. These so-called FLU BiTE® antibody constructs selectively mediate T cell dependent lysis of M2-expressing and influenza A virus infected cells and protect BALB/c mice against challenge with different influenza A virus subtypes. By humanizing the M2e-binding scFv, we generated human-like FLU BiTE® antibody constructs, with increased in vitro cytotoxic activity and in vivo protective capacity against influenza A virus infection. FLU BiTE® antibody constructs represent a promising new curative and prophylactic treatment option for influenza disease.
Collapse
Affiliation(s)
| | - Kenny Roose
- Medical Biotechnology Center, VIB, Technologiepark 927, 9052, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Technologiepark 927, 9052, Ghent, Belgium
| | - Anouk Smet
- Medical Biotechnology Center, VIB, Technologiepark 927, 9052, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Technologiepark 927, 9052, Ghent, Belgium
| | - Bert Schepens
- Medical Biotechnology Center, VIB, Technologiepark 927, 9052, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Technologiepark 927, 9052, Ghent, Belgium
| | - Peter Kufer
- ARM, Amgen Research (Munich) GmbH, Munich, Germany
| | - Tobias Raum
- ARM, Amgen Research (Munich) GmbH, Munich, Germany
| | | | - Markus Muenz
- ARM, Amgen Research (Munich) GmbH, Munich, Germany
| | - Xavier Saelens
- Medical Biotechnology Center, VIB, Technologiepark 927, 9052, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Technologiepark 927, 9052, Ghent, Belgium
| | - Walter Fiers
- Medical Biotechnology Center, VIB, Technologiepark 927, 9052, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Technologiepark 927, 9052, Ghent, Belgium.
| |
Collapse
|
45
|
Chowdhury MYE, Kim TH, Uddin MB, Kim JH, Hewawaduge CY, Ferdowshi Z, Sung MH, Kim CJ, Lee JS. Mucosal vaccination of conserved sM2, HA2 and cholera toxin subunit A1 (CTA1) fusion protein with poly gamma-glutamate/chitosan nanoparticles (PC NPs) induces protection against divergent influenza subtypes. Vet Microbiol 2017; 201:240-251. [PMID: 28284616 DOI: 10.1016/j.vetmic.2017.01.020] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 01/18/2017] [Accepted: 01/18/2017] [Indexed: 01/13/2023]
Abstract
To develop a safe and effective mucosal vaccine that broad cross protection against seasonal or emerging influenza A viruses, we generated a mucosal influenza vaccine system combining the highly conserved matrix protein-2 (sM2), fusion peptide of hemagglutinin (HA2), the well-known mucosal adjuvant cholera toxin subunit A1 (CTA1) and poly-γ-glutamic acid (γ-PGA)-chitosan nanoparticles (PC NPs), which are safe, natural materials that are able to target the mucosal membrane as a mucosal adjuvant. The mucosal administration of sM2HA2CTA1/PC NPs could induce a high degree of systemic immunity (IgG and IgA) at the site of inoculation as well as at remote locations and also significantly increase the levels of sM2- or HA2-specific cell-mediated immune response. In challenge tests in BALB/c mice with 10 MLD50 of A/EM/Korea/W149/06(H5N1), A/Puerto Rico/8/34(H1N1), A/Aquatic bird/Korea/W81/2005(H5N2), A/Aquatic bird/Korea/W44/2005 (H7N3) or A/Chicken/Korea/116/2004(H9N2) viruses, the recombinant sM2HA2CTA1/PC NPs provided cross protection against divergent lethal influenza subtypes and also the protection was maintained up to six months after vaccination. Thus, sM2HA2CTA1/PC NPs could be a promising strategy for a universal influenza vaccine.
Collapse
Affiliation(s)
- Mohammed Y E Chowdhury
- College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea; Faculty of Veterinary Medicine, Chittagong Veterinary and Animal Sciences University, Chittagong, Bangladesh
| | - Tae-Hwan Kim
- College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Md Bashir Uddin
- College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea; Faculty of Veterinary & Animal Science, Sylhet Agricultural University, Sylhet -3100, Bangladesh
| | - Jae-Hoon Kim
- College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - C Y Hewawaduge
- College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Zannatul Ferdowshi
- College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea; Department of Biochemistry and Biotechnology, University of Science and Technology Chittagong, Bangladesh
| | | | - Chul-Joong Kim
- College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea.
| | - Jong-Soo Lee
- College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea.
| |
Collapse
|
46
|
Soleimani S, Shahsavandi S, Maddadgar O. Improvement influenza HA2 DNA vaccine cellular and humoral immune responses with Mx bio adjuvant. Biologicals 2016; 46:6-10. [PMID: 28027847 DOI: 10.1016/j.biologicals.2016.11.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 11/29/2016] [Accepted: 11/29/2016] [Indexed: 11/19/2022] Open
Abstract
Immunization with DNA vaccines as a novel alternative to conventional vaccination strategy requires adjuvant for improving vaccine efficacy. The conserved immunogenic HA2 subunit, which harbors neutralizing epitopes is a promising vaccine candidate against influenza viruses. In this study, for the first time we explore the idea of using host interferon inducible Mx protein to increase the immunogenicity of HA2 H9N2 influenza DNA vaccine. The potency and safety of the Mx adjuvanted-HA2 vaccine was evaluated in BALB/c mice by different prime-boost strategies. To assess the effect of the vaccination on the virus clearance rate, mice were challenged with homologous influenza virus. Administration of the adjuvanted vaccine and boosting with the same regimen could effectively enhance both humoral and cellular immune responses in treated mice. These data demonstrated that Mx as host defense peptide can be potentiated for improving influenza vaccine efficacy.
Collapse
MESH Headings
- Animals
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- Chemotherapy, Adjuvant/methods
- Enzyme-Linked Immunosorbent Assay
- Female
- Immunity, Cellular/drug effects
- Immunity, Cellular/immunology
- Immunity, Humoral/drug effects
- Immunity, Humoral/immunology
- Immunization, Secondary/methods
- Influenza A Virus, H9N2 Subtype/immunology
- Influenza Vaccines/administration & dosage
- Influenza Vaccines/immunology
- Mice, Inbred BALB C
- Myxovirus Resistance Proteins/administration & dosage
- Myxovirus Resistance Proteins/immunology
- Orthomyxoviridae Infections/immunology
- Orthomyxoviridae Infections/prevention & control
- Orthomyxoviridae Infections/virology
- Treatment Outcome
- Vaccination/methods
- Vaccines, DNA/administration & dosage
- Vaccines, DNA/immunology
Collapse
Affiliation(s)
- Sina Soleimani
- Faculty of Veterinary Medicine, Tehran University, P.O. Box 14155-6453, Tehran, Iran; Razi Vaccine & Serum Research Institute, Agricultural Research Education and Extension Organization, P.O. Box 31975-148, Karaj, Iran
| | - Shahla Shahsavandi
- Razi Vaccine & Serum Research Institute, Agricultural Research Education and Extension Organization, P.O. Box 31975-148, Karaj, Iran.
| | - Omid Maddadgar
- Faculty of Veterinary Medicine, Tehran University, P.O. Box 14155-6453, Tehran, Iran
| |
Collapse
|
47
|
Gasper DJ, Neldner B, Plisch EH, Rustom H, Carrow E, Imai H, Kawaoka Y, Suresh M. Effective Respiratory CD8 T-Cell Immunity to Influenza Virus Induced by Intranasal Carbomer-Lecithin-Adjuvanted Non-replicating Vaccines. PLoS Pathog 2016; 12:e1006064. [PMID: 27997610 PMCID: PMC5173246 DOI: 10.1371/journal.ppat.1006064] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 11/14/2016] [Indexed: 01/31/2023] Open
Abstract
CD8+ cytotoxic T lymphocytes (CTLs) are critical for clearing many viral infections, and protective CTL memory can be induced by vaccination with attenuated viruses and vectors. Non-replicating vaccines are typically potentiated by the addition of adjuvants that enhance humoral responses, however few are capable of generating CTL responses. Adjuplex is a carbomer-lecithin-based adjuvant demonstrated to elicit robust humoral immunity to non-replicating antigens. We report that mice immunized with non-replicating Adjuplex-adjuvanted vaccines generated robust antigen-specific CTL responses. Vaccination by the subcutaneous or the intranasal route stimulated systemic and mucosal CTL memory respectively. However, only CTL memory induced by intranasal vaccination was protective against influenza viral challenge, and correlated with an enhancement of memory CTLs in the airways and CD103+ CD69+ CXCR3+ resident memory-like CTLs in the lungs. Mechanistically, Myd88-deficient mice mounted primary CTL responses to Adjuplex vaccines that were similar in magnitude to wild-type mice, but exhibited altered differentiation of effector cell subsets. Immune potentiating effects of Adjuplex entailed alterations in the frequency of antigen-presenting-cell subsets in vaccine draining lymph nodes, and in the lungs and airways following intranasal vaccination. Further, Adjuplex enhanced the ability of dendritic cells to promote antigen-induced proliferation of naïve CD8 T cells by modulating antigen uptake, its intracellular localization, and rate of processing. Taken together, we have identified an adjuvant that elicits both systemic and mucosal CTL memory to non-replicating antigens, and engenders protective CTL-based heterosubtypic immunity to influenza A virus in the respiratory tract. Further, findings presented in this manuscript have provided key insights into the mechanisms and factors that govern the induction and programming of systemic and protective memory CTLs in the respiratory tract. Current respiratory-virus vaccines typically employ non-replicating antigens and rely solely on the generation of humoral responses for protection. Viruses such as influenza can mutate and escape these responses, thereby limiting immunity and necessitating revaccination. Cell-mediated immunity (CMI) could provide broader protection by targeting viral components that infrequently mutate, however non-replicating vaccines capable of inducing CMI are not available. Impediments to vaccine development include an incomplete understanding of the nature of protective respiratory CMI and a lack of vaccine adjuvants capable of eliciting CMI to non-replicating antigens. Using a mouse model, we characterized the protective immunity afforded by CMI responses to non-replicating vaccines formulated with the adjuvant Adjuplex. We found that vaccination via either the subcutaneous or intranasal route was capable of inducing potent CMI responses. However, only intranasal vaccination protected against challenge with heterosubtypic influenza viruses. This protection correlated with enhancement of T cells with a resident-memory phenotype in the lungs. Additionally, mechanistic studies showed that Adjuplex affects antigen-presenting cells via activation and alteration of antigen uptake, processing, and presentation. The current studies: (1) identified an adjuvant that elicits protective CMI to respiratory viral pathogens; (2) suggested that stimulation of protective CMI in the respiratory tract requires intranasal vaccine delivery.
Collapse
Affiliation(s)
- David J Gasper
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America.,Comparative Biomedical Sciences Graduate Program, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Brandon Neldner
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Erin H Plisch
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Hani Rustom
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Emily Carrow
- Advanced Bioadjuvants, Omaha, Nebraska, United States of America
| | - Hirotaka Imai
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Yoshihiro Kawaoka
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - M Suresh
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| |
Collapse
|
48
|
An avian influenza H5N1 virus vaccine candidate based on the extracellular domain produced in yeast system as subviral particles protects chickens from lethal challenge. Antiviral Res 2016; 133:242-9. [PMID: 27498036 DOI: 10.1016/j.antiviral.2016.08.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 08/01/2016] [Indexed: 11/22/2022]
Abstract
Highly pathogenic avian influenza is an on-going problem in poultry and a potential human pandemic threat. Pandemics occur suddenly and vaccine production must be fast and effective to be of value in controlling the spread of the virus. In this study we evaluated the potential of a recombinant protein from the extracellular domain of an H5 hemagglutinin protein produced in a yeast expression system to act as an effective vaccine. Protein production was efficient, with up to 200 mg purified from 1 L of culture medium. We showed that the deletion of the multibasic cleavage site from the protein improves oligomerization and, consequentially, its immunogenicity. We also showed that immunization with this deleted protein protected chickens from challenge with a highly pathogenic avian influenza H5N1 virus. Our results suggest that this recombinant protein produced in yeast may be an effective vaccine against H5N1 virus in poultry.
Collapse
|
49
|
Rosendahl Huber SK, Luimstra JJ, van Beek J, Hoppes R, Jacobi RHJ, Hendriks M, Kapteijn K, Ouwerkerk C, Rodenko B, Ovaa H, de Jonge J. Chemical Modification of Influenza CD8+ T-Cell Epitopes Enhances Their Immunogenicity Regardless of Immunodominance. PLoS One 2016; 11:e0156462. [PMID: 27333291 PMCID: PMC4917206 DOI: 10.1371/journal.pone.0156462] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 05/13/2016] [Indexed: 11/19/2022] Open
Abstract
T cells are essential players in the defense against infection. By targeting the MHC class I antigen-presenting pathway with peptide-based vaccines, antigen-specific T cells can be induced. However, low immunogenicity of peptides poses a challenge. Here, we set out to increase immunogenicity of influenza-specific CD8+ T cell epitopes. By substituting amino acids in wild type sequences with non-proteogenic amino acids, affinity for MHC can be increased, which may ultimately enhance cytotoxic CD8+ T cell responses. Since preventive vaccines against viruses should induce a broad immune response, we used this method to optimize influenza-specific epitopes of varying dominance. For this purpose, HLA-A*0201 epitopes GILGFVFTL, FMYSDFHFI and NMLSTVLGV were selected in order of decreasing MHC-affinity and dominance. For all epitopes, we designed chemically enhanced altered peptide ligands (CPLs) that exhibited greater binding affinity than their WT counterparts; even binding scores of the high affinity GILGFVFTL epitope could be improved. When HLA-A*0201 transgenic mice were vaccinated with selected CPLs, at least 2 out of 4 CPLs of each epitope showed an increase in IFN-γ responses of splenocytes. Moreover, modification of the low affinity epitope NMLSTVLGV led to an increase in the number of mice that responded. By optimizing three additional influenza epitopes specific for HLA-A*0301, we show that this strategy can be extended to other alleles. Thus, enhancing binding affinity of peptides provides a valuable tool to improve the immunogenicity and range of preventive T cell-targeted peptide vaccines.
Collapse
Affiliation(s)
- Sietske K. Rosendahl Huber
- Centre for Infectious Disease Control (Cib), National Institute for Public Health and the Environment, Bilthoven, the Netherlands
| | - Jolien J. Luimstra
- Division of Cell Biology, Netherlands Cancer Institute, Amsterdam, the Netherlands
- Institute for Chemical Immunology (ICI), Utrecht, the Netherlands
| | - Josine van Beek
- Centre for Infectious Disease Control (Cib), National Institute for Public Health and the Environment, Bilthoven, the Netherlands
| | - Rieuwert Hoppes
- Division of Cell Biology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Ronald H. J. Jacobi
- Centre for Infectious Disease Control (Cib), National Institute for Public Health and the Environment, Bilthoven, the Netherlands
| | - Marion Hendriks
- Centre for Infectious Disease Control (Cib), National Institute for Public Health and the Environment, Bilthoven, the Netherlands
| | - Kim Kapteijn
- Division of Cell Biology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Casper Ouwerkerk
- Division of Cell Biology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Boris Rodenko
- Division of Cell Biology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Huib Ovaa
- Division of Cell Biology, Netherlands Cancer Institute, Amsterdam, the Netherlands
- Institute for Chemical Immunology (ICI), Utrecht, the Netherlands
| | - Jørgen de Jonge
- Centre for Infectious Disease Control (Cib), National Institute for Public Health and the Environment, Bilthoven, the Netherlands
- * E-mail:
| |
Collapse
|
50
|
Immunogenicity of Virus Like Particle Forming Baculoviral DNA Vaccine against Pandemic Influenza H1N1. PLoS One 2016; 11:e0154824. [PMID: 27149064 PMCID: PMC4858234 DOI: 10.1371/journal.pone.0154824] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 04/19/2016] [Indexed: 12/29/2022] Open
Abstract
An outbreak of influenza H1N1 in 2009, representing the first influenza pandemic of the 21st century, was transmitted to over a million individuals and claimed 18,449 lives. The current status in many countries is to prepare influenza vaccine using cell-based or egg-based killed vaccine. However, traditional influenza vaccine platforms have several limitations. To overcome these limitations, many researchers have tried various approaches to develop alternative production platforms. One of the alternative approach, we reported the efficacy of influenza HA vaccination using a baculoviral DNA vaccine (AcHERV-HA). However, the immune response elicited by the AcHERV-HA vaccine, which only targets the HA antigen, was lower than that of the commercial killed vaccine. To overcome the limitations of this previous vaccine, we constructed a human endogenous retrovirus (HERV) envelope-coated, baculovirus-based, virus-like-particle (VLP)–forming DNA vaccine (termed AcHERV-VLP) against pandemic influenza A/California/04/2009 (pH1N1). BALB/c mice immunized with AcHERV-VLP (1×107 FFU AcHERV-VLP, i.m.) and compared with mice immunized with the killed vaccine or mice immunized with AcHERV-HA. As a result, AcHERV-VLP immunization produced a greater humoral immune response and exhibited neutralizing activity with an intrasubgroup H1 strain (PR8), elicited neutralizing antibody production, a high level of interferon-γ secretion in splenocytes, and diminished virus shedding in the lung after challenge with a lethal dose of influenza virus. In conclusion, VLP-forming baculovirus DNA vaccine could be a potential vaccine candidate capable of efficiently delivering DNA to the vaccinee and VLP forming DNA eliciting stronger immunogenicity than egg-based killed vaccines.
Collapse
|