1
|
Wu L, Xu W, Jiang H, Yang M, Cun D. Respiratory delivered vaccines: Current status and perspectives in rational formulation design. Acta Pharm Sin B 2024; 14:5132-5160. [PMID: 39807330 PMCID: PMC11725141 DOI: 10.1016/j.apsb.2024.08.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 07/20/2024] [Accepted: 08/18/2024] [Indexed: 01/16/2025] Open
Abstract
The respiratory tract is susceptible to various infections and can be affected by many serious diseases. Vaccination is one of the most promising ways that prevent infectious diseases and treatment of some diseases such as malignancy. Direct delivery of vaccines to the respiratory tract could mimic the natural process of infection and shorten the delivery path, therefore unique mucosal immunity at the first line might be induced and the efficiency of delivery can be high. Despite considerable attempts at the development of respiratory vaccines, the rational formulation design still warrants attention, i.e., how the formulation composition, particle properties, formulation type (liquid or solid), and devices would influence the immune outcome. This article reviews the recent advances in the formulation design and development of respiratory vaccines. The focus is on the state of the art of delivering antigenic compounds through the respiratory tract, overcoming the pulmonary bio-barriers, enhancing delivery efficiencies of respiratory vaccines as well as maintaining the stability of vaccines during storage and use. The choice of devices and the influence of deposition sites on vaccine efficiencies were also reviewed.
Collapse
Affiliation(s)
- Lan Wu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
- Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, Shenyang 110016, China
| | - Wenwen Xu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
- Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, Shenyang 110016, China
| | - Huiyang Jiang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
- Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, Shenyang 110016, China
| | - Mingshi Yang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
- Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, Shenyang 110016, China
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen DK-2100, Denmark
| | - Dongmei Cun
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
- Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, Shenyang 110016, China
- School of Food and Drug, Shenzhen Polytechnic University, China, Shenzhen 518055, China
| |
Collapse
|
2
|
Cowling BJ, Okoli GN. Influenza Vaccine Effectiveness and Progress Towards a Universal Influenza Vaccine. Drugs 2024; 84:1013-1023. [PMID: 39167316 PMCID: PMC11438668 DOI: 10.1007/s40265-024-02083-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/04/2024] [Indexed: 08/23/2024]
Abstract
At various times in recent decades, surges have occurred in optimism about the potential for universal influenza vaccines that provide strong, broad, and long-lasting protection and could substantially reduce the disease burden associated with seasonal influenza epidemics as well as the threat posed by pandemic influenza. Each year more than 500 million doses of seasonal influenza vaccine are administered around the world, with most doses being egg-grown inactivated subunit or split-virion vaccines. These vaccines tend to have moderate effectiveness against medically attended influenza for influenza A(H1N1) and influenza B, and somewhat lower for influenza A(H3N2) where differences between vaccine strains and circulating strains can occur more frequently due to antigenic drift and egg adaptations in the vaccine strains. Several enhanced influenza vaccine platforms have been developed including cell-grown antigen, the inclusion of adjuvants, or higher antigen doses, to improve immunogenicity and protection. During the COVID-19 pandemic there was unprecedented speed in development and roll-out of relatively new vaccine platforms, including mRNA vaccines and viral vector vaccines. These new platforms present opportunities to improve protection for influenza beyond existing products. Other approaches continue to be explored. Incremental improvements in influenza vaccine performance should be achievable in the short to medium term.
Collapse
Affiliation(s)
- Benjamin J Cowling
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China.
- Laboratory of Data Discovery for Health Limited, Hong Kong Science and Technology Park, New Territories, Hong Kong Special Administrative Region, Hong Kong, China.
| | - George N Okoli
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| |
Collapse
|
3
|
Henríquez R, Muñoz-Barroso I. Viral vector- and virus-like particle-based vaccines against infectious diseases: A minireview. Heliyon 2024; 10:e34927. [PMID: 39144987 PMCID: PMC11320483 DOI: 10.1016/j.heliyon.2024.e34927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 06/28/2024] [Accepted: 07/18/2024] [Indexed: 08/16/2024] Open
Abstract
To overcome the limitations of conventional vaccines, new platforms for vaccine design have emerged such as those based on viral vectors and virus-like particles (VLPs). Viral vector vaccines are highly efficient and the onset of protection is quick. Many recombinant vaccine candidates for humans are based on viruses belonging to different families such as Adenoviridae, Retroviridae, Paramyxoviridae, Rhabdoviridae, and Parvoviridae. Also, the first viral vector vaccine licensed for human vaccination was the Japanese encephalitis virus vaccine. Since then, several viral vectors have been approved for vaccination against the viruses of Lassa fever, Ebola, hepatitis B, hepatitis E, SARS-CoV-2, and malaria. VLPs are nanoparticles that mimic viral particles formed from the self-assembly of structural proteins and VLP-based vaccines against hepatitis B and E viruses, human papillomavirus, and malaria have been commercialized. As evidenced by the accelerated production of vaccines against COVID-19, these new approaches are important tools for vaccinology and for generating rapid responses against pathogens and emerging pandemic threats.
Collapse
Affiliation(s)
- Ruth Henríquez
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Edificio Departamental Lab.106. Plaza Doctores de la Reina S/n, 37007, Salamanca, Spain
| | - Isabel Muñoz-Barroso
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Edificio Departamental Lab.106. Plaza Doctores de la Reina S/n, 37007, Salamanca, Spain
| |
Collapse
|
4
|
Santos LC, Fernandes AMS, Alves IA, Serafini MR, Silva LDSE, de Freitas HF, Leite LCC, Santos CC. Trends in Viral Vector-Based Vaccines for Tuberculosis: A Patent Review (2010-2023). Vaccines (Basel) 2024; 12:876. [PMID: 39204002 PMCID: PMC11359462 DOI: 10.3390/vaccines12080876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 07/25/2024] [Accepted: 07/30/2024] [Indexed: 09/03/2024] Open
Abstract
Tuberculosis (TB) is an ancient global public health problem. Several strategies have been applied to develop new and more effective vaccines against TB, from attenuated or inactivated mycobacteria to recombinant subunit or genetic vaccines, including viral vectors. This review aimed to evaluate patents filed between 2010 and 2023 for TB vaccine candidates. It focuses on viral vector-based strategies. A search was carried out in Espacenet, using the descriptors "mycobacterium and tuberculosis" and the classification A61K39. Of the 411 patents preliminarily identified, the majority were related to subunit vaccines, with 10 patents based on viral vector platforms selected in this study. Most of the identified patents belong to the United States or China, with a concentration of patent filings between 2013 and 2023. Adenoviruses were the most explored viral vectors, and the most common immunodominant Mycobacterium tuberculosis (Mtb) antigens were present in all the selected patents. The majority of patents were tested in mouse models by intranasal or subcutaneous route of immunization. In the coming years, an increased use of this platform for prophylactic and/or therapeutic approaches for TB and other diseases is expected. Along with this, expanding knowledge about the safety of this technology is essential to advance its use.
Collapse
Affiliation(s)
- Lana C. Santos
- Serviço de Imunologia das Doenças Infecciosas, Faculdade de Farmácia, Universidade Federal da Bahia, Salvador 40170-115, BA, Brazil; (L.C.S.); (A.M.S.F.); (L.d.S.e.S.)
| | - Antônio Márcio Santana Fernandes
- Serviço de Imunologia das Doenças Infecciosas, Faculdade de Farmácia, Universidade Federal da Bahia, Salvador 40170-115, BA, Brazil; (L.C.S.); (A.M.S.F.); (L.d.S.e.S.)
| | - Izabel Almeida Alves
- Departamento do Medicamento, Faculdade de Farmácia, Universidade Federal da Bahia, Salvador 40170-115, BA, Brazil;
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade do Estado da Bahia, Salvador 41150-000, BA, Brazil
| | - Mairim Russo Serafini
- Departamento de Farmácia, Universidade Federal do Sergipe, São Cristóvão 49100-000, SE, Brazil;
| | - Leandra da Silva e Silva
- Serviço de Imunologia das Doenças Infecciosas, Faculdade de Farmácia, Universidade Federal da Bahia, Salvador 40170-115, BA, Brazil; (L.C.S.); (A.M.S.F.); (L.d.S.e.S.)
| | | | - Luciana C. C. Leite
- Laboratório de Desenvolvimento de Vacinas, Instituto Butantan, São Paulo 05503-900, SP, Brazil;
| | - Carina C. Santos
- Serviço de Imunologia das Doenças Infecciosas, Faculdade de Farmácia, Universidade Federal da Bahia, Salvador 40170-115, BA, Brazil; (L.C.S.); (A.M.S.F.); (L.d.S.e.S.)
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal da Bahia, Salvador 40170-115, BA, Brazil
| |
Collapse
|
5
|
Leong SL, Gras S, Grant EJ. Fighting flu: novel CD8 + T-cell targets are required for future influenza vaccines. Clin Transl Immunology 2024; 13:e1491. [PMID: 38362528 PMCID: PMC10867544 DOI: 10.1002/cti2.1491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 01/29/2024] [Accepted: 01/30/2024] [Indexed: 02/17/2024] Open
Abstract
Seasonal influenza viruses continue to cause severe medical and financial complications annually. Although there are many licenced influenza vaccines, there are billions of cases of influenza infection every year, resulting in the death of over half a million individuals. Furthermore, these figures can rise in the event of a pandemic, as seen throughout history, like the 1918 Spanish influenza pandemic (50 million deaths) and the 1968 Hong Kong influenza pandemic (~4 million deaths). In this review, we have summarised many of the currently licenced influenza vaccines available across the world and current vaccines in clinical trials. We then briefly discuss the important role of CD8+ T cells during influenza infection and why future influenza vaccines should consider targeting CD8+ T cells. Finally, we assess the current landscape of known immunogenic CD8+ T-cell epitopes and highlight the knowledge gaps required to be filled for the design of rational future influenza vaccines that incorporate CD8+ T cells.
Collapse
Affiliation(s)
- Samuel Liwei Leong
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular ScienceLa Trobe UniversityBundooraVICAustralia
| | - Stephanie Gras
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular ScienceLa Trobe UniversityBundooraVICAustralia
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery InstituteMonash UniversityClaytonVICAustralia
| | - Emma J Grant
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular ScienceLa Trobe UniversityBundooraVICAustralia
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery InstituteMonash UniversityClaytonVICAustralia
| |
Collapse
|
6
|
Collatuzzo G, De Palma G, Violante FS, Porru S, Larese Filon F, Fabianova E, Violán C, Vimercati L, Leustean M, Rodriguez-Suarez MM, Sansone E, Sala E, Zunarelli C, Lodi V, Monaco MGL, Spiteri G, Negro C, Beresova J, Carrasco-Ribelles LA, Tafuri S, Asafo SS, Ditano G, Abedini M, Boffetta P. Temporal trends of COVID-19 antibodies in vaccinated healthcare workers undergoing repeated serological sampling: An individual-level analysis within 13 months in the ORCHESTRA cohort. Front Immunol 2023; 13:1079884. [PMID: 36713452 PMCID: PMC9875291 DOI: 10.3389/fimmu.2022.1079884] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 12/13/2022] [Indexed: 01/12/2023] Open
Abstract
Short summary We investigated changes in serologic measurements after COVID-19 vaccination in 19,422 subjects. An individual-level analysis was performed on standardized measurements. Age, infection, vaccine doses, time between doses and serologies, and vaccine type were associated with changes in serologic levels within 13 months. Background Persistence of vaccine immunization is key for COVID-19 prevention. Methods We investigated the difference between two serologic measurements of anti-COVID-19 S1 antibodies in an individual-level analysis on 19,422 vaccinated healthcare workers (HCW) from Italy, Spain, Romania, and Slovakia, tested within 13 months from first dose. Differences in serologic levels were divided by the standard error of the cohort-specific distribution, obtaining standardized measurements. We fitted multivariate linear regression models to identify predictors of difference between two measurements. Results We observed a progressively decreasing difference in serologic levels from <30 days to 210-240 days. Age was associated with an increased difference in serologic levels. There was a greater difference between the two serologic measurements in infected HCW than in HCW who had never been infected; before the first measurement, infected HCW had a relative risk (RR) of 0.81 for one standard deviation in the difference [95% confidence interval (CI) 0.78-0.85]. The RRs for a 30-day increase in time between first dose and first serology, and between the two serologies, were 1.08 (95% CI 1.07-1.10) and 1.04 (95% CI 1.03-1.05), respectively. The first measurement was a strong predictor of subsequent antibody decrease (RR 1.60; 95% CI 1.56-1.64). Compared with Comirnaty, Spikevax (RR 0.83, 95% CI 0.75-0.92) and mixed vaccines (RR 0.61, 95% CI 0.51-0.74) were smaller decrease in serological level (RR 0.46; 95% CI 0.40-0.54). Conclusions Age, COVID-19 infection, number of doses, time between first dose and first serology, time between serologies, and type of vaccine were associated with differences between the two serologic measurements within a 13-month period.
Collapse
Affiliation(s)
- Giulia Collatuzzo
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Giuseppe De Palma
- Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
| | - Francesco S. Violante
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy,Occupational Medicine Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Stefano Porru
- Section of Occupational Medicine, Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | | | - Eleonora Fabianova
- Occupational Health Department, Regional Authority of Public Health, Banská Bystrica, Slovakia
| | - Concepción Violán
- Unitat de Suport a la Recerca Metropolitana Nord, Institut Universitari d’Investigació en Atenció Primària Jordi Gol (IDIAP Jordi Gol), Mataró, Spain,Direcció d’Atenció Primària Metropolitana Nord Institut Català de Salut, Barcelona, Spain,Germans Trias i Pujol Research Institute (IGTP), Badalona, Spain,Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Luigi Vimercati
- Interdisciplinary Department of Medicine, University of Bari, Bari, Italy
| | | | - Marta Maria Rodriguez-Suarez
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA) and Universitario Central de Asturias (HUCA), University of Oviedo, Oviedo, Spain
| | - Emanuele Sansone
- Occupational Medicine Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Emma Sala
- Unit of Occupational Health, Hygiene, Toxicology and Prevention, ASST Ospedali Civili di Brescia, Brescia, Italy
| | - Carlotta Zunarelli
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Vittorio Lodi
- Occupational Medicine Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | | | - Gianluca Spiteri
- Occupational Medicine Unit, University Hospital of Verona, Verona, Italy
| | - Corrado Negro
- Unit of Occupational Medicine, University of Trieste, Trieste, Italy
| | - Jana Beresova
- Occupational Health Department, Regional Authority of Public Health, Banská Bystrica, Slovakia
| | - LucÌa A. Carrasco-Ribelles
- Unitat de Suport a la Recerca Metropolitana Nord, Institut Universitari d’Investigació en Atenció Primària Jordi Gol (IDIAP Jordi Gol), Mataró, Spain
| | - Silvio Tafuri
- Interdisciplinary Department of Medicine, University of Bari, Bari, Italy
| | - Shuffield S. Asafo
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Giorgia Ditano
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Mahsa Abedini
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Paolo Boffetta
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy,Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY, United States,*Correspondence: Paolo Boffetta,
| |
Collapse
|
7
|
Apinda N, Muenthaisong A, Chomjit P, Sangkakam K, Nambooppha B, Rittipornlertrak A, Koonyosying P, Yao Y, Nair V, Sthitmatee N. Simultaneous Protective Immune Responses of Ducks against Duck Plague and Fowl Cholera by Recombinant Duck Enteritis Virus Vector Expressing Pasteurella multocida OmpH Gene. Vaccines (Basel) 2022; 10:vaccines10081358. [PMID: 36016245 PMCID: PMC9415155 DOI: 10.3390/vaccines10081358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 08/17/2022] [Accepted: 08/18/2022] [Indexed: 12/03/2022] Open
Abstract
Duck enteritis virus and Pasteurella multocida are major duck pathogens that induce duck plague and fowl cholera, respectively, in ducks and other waterfowl populations, leading to high levels of morbidity and mortality. Immunization with live attenuated DEV vaccine containing P. multocida outer membrane protein H (OmpH) can provide the most effective protection against these two infectious diseases in ducks. We have recently reported the construction of recombinant DEV expressing P. multocida ompH gene using the CRISPR/Cas9 gene editing strategy with the goal of using it as a bivalent vaccine that can simultaneously protect against both infections. Here we describe the findings of our investigation into the systemic immune responses, potency and clinical protection induced by the two recombinant DEV-ompH vaccine constructs, where one copy each of the ompH gene was inserted into the DEV genome at the UL55-LORF11 and UL44-44.5 intergenic regions, respectively. Our study demonstrated that the insertion of the ompH gene exerted no adverse effect on the DEV parental virus. Moreover, ducklings immunized with the rDEV-ompH-UL55 and rDEV-ompH-UL44 vaccines induced promising levels of P. multocida OmpH-specific as well as DEV-specific antibodies and were completely protected from both diseases. Analysis of the humoral and cellular immunity confirmed the immunogenicity of both recombinant vaccines, which provided strong immune responses against DEV and P. multocida. This study not only provides insights into understanding the immune responses of ducks to recombinant DEV-ompH vaccines but also demonstrates the potential for simultaneous prevention of viral and bacterial infections using viral vectors expressing bacterial immunogens.
Collapse
Affiliation(s)
- Nisachon Apinda
- Department of Veterinary Biosciences and Veterinary Public Health, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai 50100, Thailand
| | - Anucha Muenthaisong
- Department of Veterinary Biosciences and Veterinary Public Health, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai 50100, Thailand
| | - Paweena Chomjit
- Department of Veterinary Biosciences and Veterinary Public Health, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai 50100, Thailand
| | - Kanokwan Sangkakam
- Department of Veterinary Biosciences and Veterinary Public Health, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai 50100, Thailand
| | - Boondarika Nambooppha
- Department of Veterinary Biosciences and Veterinary Public Health, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai 50100, Thailand
| | - Amarin Rittipornlertrak
- Department of Food Animal Clinic, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai 50100, Thailand
| | - Pongpisid Koonyosying
- Department of Veterinary Biosciences and Veterinary Public Health, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai 50100, Thailand
| | - Yongxiu Yao
- The Pirbright Institute, Ash Road, Pirbright, Woking GU24 0NF, UK
| | - Venugopal Nair
- The Pirbright Institute, Ash Road, Pirbright, Woking GU24 0NF, UK
- Jenner Institute, University of Oxford, Oxford OX1 2JD, UK
- Department of Zoology, University of Oxford, 11a Mansfield Road, Oxford OX1 3SZ, UK
| | - Nattawooti Sthitmatee
- Department of Veterinary Biosciences and Veterinary Public Health, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai 50100, Thailand
- Excellence Center in Veterinary Bioscience, Chiang Mai University, Chiang Mai 50100, Thailand
- Correspondence: ; Tel.: +66-53-948-017; Fax: +66-53-948-041
| |
Collapse
|
8
|
Liu Z, Kong Z, Chen M, Shang Y. Design of live-attenuated animal vaccines based on pseudorabies virus platform. ANIMAL DISEASES 2022. [DOI: 10.1186/s44149-022-00044-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
AbstractPseudorabies virus (PRV) is a double-stranded DNA virus with a genome approximating 150 kb in size. PRV contains many non-essential genes that can be replaced with genes encoding heterogenous antigens without affecting viral propagation. With the ability to induce cellular, humoral and mucosal immune responses in the host, PRV is considered to be an ideal and potential live vector for generation of animal vaccines. In this review, we summarize the advances in attenuated recombinant PRVs and design of PRV-based live vaccines as well as the challenge of vaccine application.
Collapse
|
9
|
Andersen TK, Bodin J, Oftung F, Bogen B, Mjaaland S, Grødeland G. Pandemic Preparedness Against Influenza: DNA Vaccine for Rapid Relief. Front Immunol 2021; 12:747032. [PMID: 34691056 PMCID: PMC8531196 DOI: 10.3389/fimmu.2021.747032] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Accepted: 09/20/2021] [Indexed: 01/14/2023] Open
Abstract
The 2009 “swine flu” pandemic outbreak demonstrated the limiting capacity for egg-based vaccines with respect to global vaccine supply within a timely fashion. New vaccine platforms that efficiently can quench pandemic influenza emergences are urgently needed. Since 2009, there has been a profound development of new vaccine platform technologies with respect to prophylactic use in the population, including DNA vaccines. These vaccines are particularly well suited for global pandemic responses as the DNA format is temperature stable and the production process is cheap and rapid. Here, we show that by targeting influenza antigens directly to antigen presenting cells (APC), DNA vaccine efficacy equals that of conventional technologies. A single dose of naked DNA encoding hemagglutinin (HA) from influenza/A/California/2009 (H1N1), linked to a targeting moiety directing the vaccine to major histocompatibility complex class II (MHCII) molecules, raised similar humoral immune responses as the adjuvanted split virion vaccine Pandemrix, widely administered in the 2009 pandemic. Both vaccine formats rapidly induced serum antibodies that could protect mice already 8 days after a single immunization, in contrast to the slower kinetics of a seasonal trivalent inactivated influenza vaccine (TIV). Importantly, the DNA vaccine also elicited cytotoxic T-cell responses that reduced morbidity after vaccination, in contrast to very limited T-cell responses seen after immunization with Pandemrix and TIV. These data demonstrate that DNA vaccines has the potential as a single dose platform vaccine, with rapid protective effects without the need for adjuvant, and confirms the relevance of naked DNA vaccines as candidates for pandemic preparedness.
Collapse
Affiliation(s)
- Tor Kristian Andersen
- Department of Immunology and Transfusion Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Johanna Bodin
- Division for Infection Control and Environmental Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Fredrik Oftung
- Division for Infection Control and Environmental Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Bjarne Bogen
- Department of Immunology and Transfusion Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Immunology and Transfusion Medicine, Clinic for Laboratory Medicine, Oslo University Hospital, Oslo, Norway
| | - Siri Mjaaland
- Division for Infection Control and Environmental Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Gunnveig Grødeland
- Department of Immunology and Transfusion Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Immunology and Transfusion Medicine, Clinic for Laboratory Medicine, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
10
|
Becker T, Elbahesh H, Reperant LA, Rimmelzwaan GF, Osterhaus ADME. Influenza Vaccines: Successes and Continuing Challenges. J Infect Dis 2021; 224:S405-S419. [PMID: 34590139 PMCID: PMC8482026 DOI: 10.1093/infdis/jiab269] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Influenza vaccines have been available for over 80 years. They have contributed to significant reductions in influenza morbidity and mortality. However, there have been limitations in their effectiveness, in part due to the continuous antigenic evolution of seasonal influenza viruses, but also due to the predominant use of embryonated chicken eggs for their production. The latter furthermore limits their worldwide production timelines and scale. Therefore today, alternative approaches for their design and production are increasingly pursued, with already licensed quadrivalent seasonal influenza vaccines produced in cell cultures, including based on a baculovirus expression system. Next-generation influenza vaccines aim at inducing broader and longer-lasting immune responses to overcome seasonal influenza virus antigenic drift and to timely address the emergence of a new pandemic influenza virus. Tailored approaches target mechanisms to improve vaccine-induced immune responses in individuals with a weakened immune system, in particular older adults.
Collapse
Affiliation(s)
- Tanja Becker
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Husni Elbahesh
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, Hannover, Germany
| | | | - Guus F Rimmelzwaan
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Albert D M E Osterhaus
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, Hannover, Germany
| |
Collapse
|
11
|
Rasouli M, Vakilian F, Ranjbari J. Therapeutic and protective potential of mesenchymal stem cells, pharmaceutical agents and current vaccines against covid-19. Curr Stem Cell Res Ther 2021; 17:166-185. [PMID: 33349221 DOI: 10.2174/1574888x16666201221151853] [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: 03/19/2021] [Revised: 07/07/2021] [Accepted: 07/19/2021] [Indexed: 11/22/2022]
Abstract
It has been almost 18 months since the first outbreak of COVID-19 disease was reported in Wuhan, China. This unexpected devastating phenomenon, raised a great deal of concerns and anxiety among people around the world and imposed a huge economic burden on the nations' health care systems. Accordingly, clinical scientists, pharmacologists and physicians worldwide felt an urgent demand for a safe, effective therapeutic agent, treatment strategy or vaccine in order to prevent or cure the recently-emerged disease. Initially, due to lack of specific pharmacological agents and approved vaccines to combat the COVID-19, the disease control in the confirmed cases was limited to supportive care. Accordingly, repositioning or repurposing current drugs and examining their possible therapeutic efficacy received a great deal of attention. Despite revealing promising results in some clinical trials, the overall results are conflicting. For this reason, there is an urgent to seek and investigate other potential therapeutics. Mesenchymal stem cells (MSC) representing immunomodulatory and regenerative capacity to treat both curable and intractable diseases, have been investigated in COVID-19 clinical trials carried out in different parts of the world. Nevertheless, up to now, none of MSC-based approaches has been approved in controlling COVID-19 infection. Thanks to the fact that the final solution for defeating the pandemic is developing a safe, effective vaccine, enormous efforts and clinical research have been carried out. In this review, we will concisely discuss the safety and efficacy of the most relevant pharmacological agents, MSC-based approaches and candidate vaccines for treating and preventing COVID-19 infection.
Collapse
Affiliation(s)
- Mehdi Rasouli
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran. Iran
| | | | - Javad Ranjbari
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran. Iran
| |
Collapse
|
12
|
Pan C, Yue H, Zhu L, Ma GH, Wang HL. Prophylactic vaccine delivery systems against epidemic infectious diseases. Adv Drug Deliv Rev 2021; 176:113867. [PMID: 34280513 PMCID: PMC8285224 DOI: 10.1016/j.addr.2021.113867] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/20/2021] [Accepted: 07/11/2021] [Indexed: 01/04/2023]
Abstract
Prophylactic vaccines have evolved from traditional whole-cell vaccines to safer subunit vaccines. However, subunit vaccines still face problems, such as poor immunogenicity and low efficiency, while traditional adjuvants are usually unable to meet specific response needs. Advanced delivery vectors are important to overcome these barriers; they have favorable safety and effectiveness, tunable properties, precise location, and immunomodulatory capabilities. Nevertheless, there has been no systematic summary of the delivery systems to cover a wide range of infectious pathogens. We herein summarized and compared the delivery systems for major or epidemic infectious diseases caused by bacteria, viruses, fungi, and parasites. We also included the newly licensed vaccines (e.g., COVID-19 vaccines) and those close to licensure. Furthermore, we highlighted advanced delivery systems with high efficiency, cross-protection, or long-term protection against epidemic pathogens, and we put forward prospects and thoughts on the development of future prophylactic vaccines.
Collapse
Affiliation(s)
- Chao Pan
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing 100071, PR China
| | - Hua Yue
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Li Zhu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing 100071, PR China
| | - Guang-Hui Ma
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China.
| | - Heng-Liang Wang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing 100071, PR China.
| |
Collapse
|
13
|
Chong WC, Chellappan DK, Shukla SD, Peterson GM, Patel RP, Jha NK, Eri RD, Dua K, Tambuwala MM, Shastri MD. An Appraisal of the Current Scenario in Vaccine Research for COVID-19. Viruses 2021; 13:1397. [PMID: 34372603 PMCID: PMC8310376 DOI: 10.3390/v13071397] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/13/2021] [Accepted: 07/16/2021] [Indexed: 12/15/2022] Open
Abstract
The recent coronavirus disease 2019 (COVID-19) outbreak has drawn global attention, affecting millions, disrupting economies and healthcare modalities. With its high infection rate, COVID-19 has caused a colossal health crisis worldwide. While information on the comprehensive nature of this infectious agent, SARS-CoV-2, still remains obscure, ongoing genomic studies have been successful in identifying its genomic sequence and the presenting antigen. These may serve as promising, potential therapeutic targets in the effective management of COVID-19. In an attempt to establish herd immunity, massive efforts have been directed and driven toward developing vaccines against the SARS-CoV-2 pathogen. This review, in this direction, is aimed at providing the current scenario and future perspectives in the development of vaccines against SARS-CoV-2.
Collapse
Affiliation(s)
- Wai Chin Chong
- Department of Molecular and Translational Science, Monash University, Clayton 3168, Australia;
| | - Dinesh K. Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University (IMU), Kuala Lumpur 57000, Malaysia;
| | - Shakti D. Shukla
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney 2007, Australia; (S.D.S.); (K.D.)
| | - Gregory M. Peterson
- School of Pharmacy and Pharmacology, University of Tasmania, Hobart 7005, Australia; (G.M.P.); (R.P.P.)
| | - Rahul P. Patel
- School of Pharmacy and Pharmacology, University of Tasmania, Hobart 7005, Australia; (G.M.P.); (R.P.P.)
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Engineering & Technology (SET), Sharda University, Greater Noida 201310, UP, India;
| | - Rajaraman D. Eri
- School of Health Sciences, College of Health and Medicine, University of Tasmania, Launceston 7248, Australia;
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney 2007, Australia; (S.D.S.); (K.D.)
| | - Murtaza M. Tambuwala
- School of Pharmacy and Pharmaceutical Science, Ulster University, Coleraine BT52 1SA, UK
| | - Madhur D. Shastri
- School of Pharmacy and Pharmacology, University of Tasmania, Hobart 7005, Australia; (G.M.P.); (R.P.P.)
- School of Health Sciences, College of Health and Medicine, University of Tasmania, Launceston 7248, Australia;
| |
Collapse
|
14
|
B Carvalho S, Peixoto C, T Carrondo MJ, S Silva RJ. Downstream processing for influenza vaccines and candidates: An update. Biotechnol Bioeng 2021; 118:2845-2869. [PMID: 33913510 DOI: 10.1002/bit.27803] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 04/10/2021] [Accepted: 04/16/2021] [Indexed: 02/07/2023]
Abstract
Seasonal and pandemic influenza outbreaks present severe health and economic burdens. To overcome limitations on influenza vaccines' availability and effectiveness, researchers chase universal vaccines providing broad, long-lasting protection against multiple influenza subtypes, and including pandemic ones. Novel influenza vaccine designs are under development, in clinical trials, or reaching the market, namely inactivated, or live-attenuated virus, virus-like particles, or recombinant antigens, searching for improved effectiveness; all these bring downstream processing (DSP) new challenges. Having to deal with new influenza strains, including pandemics, requires shorter development time, driving the development of faster bioprocesses. To cope with better upstream processes, new regulatory demands for quality and safety, and cost reduction requirements, new unit operations and integrated processes are increasing DSP efficiency for novel vaccine formats. This review covers recent advances in DSP strategies of different influenza vaccine formats. Focus is given to the improvements on relevant state-of-the-art unit operations, from harvest and clarification to purification steps, ending with sterile filtration and formulation. The development of more efficient unit operations to cope with biophysical properties of the new candidates is discussed: emphasis is given to the design of new stationary phases, 3D printing approaches, and continuous processing tools, such as continuous chromatography. The impact of the production platforms and vaccine designs on the downstream operations for the different influenza vaccine formats approved for this season are highlighted.
Collapse
Affiliation(s)
- Sofia B Carvalho
- Animal Cell Technology Unit, iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal.,Animal Cell Technology Unit, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Cristina Peixoto
- Animal Cell Technology Unit, iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal.,Animal Cell Technology Unit, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Manuel J T Carrondo
- Animal Cell Technology Unit, iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
| | - Ricardo J S Silva
- Animal Cell Technology Unit, iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal.,Animal Cell Technology Unit, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| |
Collapse
|
15
|
Andryukov BG, Besednova NN. Older adults: panoramic view on the COVID-19 vaccination. AIMS Public Health 2021; 8:388-415. [PMID: 34395690 PMCID: PMC8334630 DOI: 10.3934/publichealth.2021030] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Accepted: 05/06/2021] [Indexed: 12/11/2022] Open
Abstract
In December 2020, COVID-19 vaccination started in many countries, with which the world community hopes to stop the further spread of the current pandemic. More than 90% of sick and deceased patients belong to the category of older adults (65 years and older). This category of the population is most vulnerable to infectious diseases, so vaccination is the most effective preventive strategy, the need for which for older adults is indisputable. Here we briefly summarize information about age-related changes in the immune system and present current data on their impact on the formation of the immune response to vaccination. Older age is accompanied by the process of biological aging accompanied by involution of the immune system with increased susceptibility to infections and a decrease in the effect of immunization. Therefore, in the ongoing mass COVID-19 vaccination, the older adults are a growing public health concern. The authors provide an overview of the various types of COVID-19 vaccines approved for mass immunization of the population by the end of 2020, including older adults, as well as an overview of strategies and platforms to improve the effectiveness of vaccination of this population. In the final part, the authors propose for discussion a system for assessing the safety and monitoring the effectiveness of COVID-19 vaccines for the older adults.
Collapse
Affiliation(s)
- Boris G Andryukov
- G.P. Somov Institute of Epidemiology and Microbiology, Russian Federal Service for Surveillance on Consumer Rights Protection and Human Wellbeing, 690087, Vladivostok, Russia
- Far Eastern Federal University (FEFU), 690091, Vladivostok, Russia
| | - Natalya N Besednova
- G.P. Somov Institute of Epidemiology and Microbiology, Russian Federal Service for Surveillance on Consumer Rights Protection and Human Wellbeing, 690087, Vladivostok, Russia
| |
Collapse
|
16
|
Bezbaruah R, Borah P, Kakoti BB, Al-Shar’I NA, Chandrasekaran B, Jaradat DMM, Al-Zeer MA, Abu-Romman S. Developmental Landscape of Potential Vaccine Candidates Based on Viral Vector for Prophylaxis of COVID-19. Front Mol Biosci 2021; 8:635337. [PMID: 33937326 PMCID: PMC8082173 DOI: 10.3389/fmolb.2021.635337] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 02/05/2021] [Indexed: 12/18/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2, SARS-CoV-2, arose at the end of 2019 as a zoonotic virus, which is the causative agent of the novel coronavirus outbreak COVID-19. Without any clear indications of abatement, the disease has become a major healthcare threat across the globe, owing to prolonged incubation period, high prevalence, and absence of existing drugs or vaccines. Development of COVID-19 vaccine is being considered as the most efficient strategy to curtail the ongoing pandemic. Following publication of genetic sequence of SARS-CoV-2, globally extensive research and development work has been in progress to develop a vaccine against the disease. The use of genetic engineering, recombinant technologies, and other computational tools has led to the expansion of several promising vaccine candidates. The range of technology platforms being evaluated, including virus-like particles, peptides, nucleic acid (DNA and RNA), recombinant proteins, inactivated virus, live attenuated viruses, and viral vectors (replicating and non-replicating) approaches, are striking features of the vaccine development strategies. Viral vectors, the next-generation vaccine platforms, provide a convenient method for delivering vaccine antigens into the host cell to induce antigenic proteins which can be tailored to arouse an assortment of immune responses, as evident from the success of smallpox vaccine and Ervebo vaccine against Ebola virus. As per the World Health Organization, till January 22, 2021, 14 viral vector vaccine candidates are under clinical development including 10 nonreplicating and four replicating types. Moreover, another 39 candidates based on viral vector platform are under preclinical evaluation. This review will outline the current developmental landscape and discuss issues that remain critical to the success or failure of viral vector vaccine candidates against COVID-19.
Collapse
Affiliation(s)
- Rajashri Bezbaruah
- Department of Pharmaceutical Sciences, Faculty of Science and Engineering, Dibrugarh University, Dibrugarh, India
| | - Pobitra Borah
- School of Pharmacy, Graphic Era Hill University, Dehradun, India
| | - Bibhuti Bhushan Kakoti
- Department of Pharmaceutical Sciences, Faculty of Science and Engineering, Dibrugarh University, Dibrugarh, India
| | - Nizar A. Al-Shar’I
- Department of Medicinal Chemistry and Pharmacognosy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| | | | - Da’san M. M. Jaradat
- Department of Chemistry, Faculty of Science, Al-Balqa Applied University, Al-Salt, Jordan
| | - Munir A. Al-Zeer
- Department of Applied Biochemistry, Institute of Biotechnology, Technical University of Berlin, Berlin, Germany
| | - Saeid Abu-Romman
- Department of Biotechnology, Faculty of Agricultural Technology, Al-Balqa Applied University, Al-Salt, Jordan
| |
Collapse
|
17
|
Tournier JN, Kononchik J. Virus Eradication and Synthetic Biology: Changes with SARS-CoV-2? Viruses 2021; 13:569. [PMID: 33800626 PMCID: PMC8066276 DOI: 10.3390/v13040569] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 03/25/2021] [Accepted: 03/26/2021] [Indexed: 12/24/2022] Open
Abstract
The eradication of infectious diseases has been achieved only once in history, in 1980, with smallpox. Since 1988, significant effort has been made to eliminate poliomyelitis viruses, but eradication is still just out of reach. As the goal of viral disease eradication approaches, the ability to recreate historically eradicated viruses using synthetic biology has the potential to jeopardize the long-term sustainability of eradication. However, the emergence of the severe acute respiratory syndrome-coronavirus (SARS-CoV)-2 pandemic has highlighted our ability to swiftly and resolutely respond to a potential outbreak. This virus has been synthetized faster than any other in the past and is resulting in vaccines before most attenuated candidates reach clinical trials. Here, synthetic biology has the opportunity to demonstrate its truest potential to the public and solidify a footing in the world of vaccines.
Collapse
Affiliation(s)
- Jean-Nicolas Tournier
- Microbiology and Infectious Diseases Department, Institut de Recherche Biomédicale des Armées, 91220 Brétigny-sur-Orge, France;
- CNRS UMR-3569, Innovative Vaccine Laboratory, Virology Department, Institut Pasteur, 75015 Paris, France
- Ecole du Val-de-Grâce, 75005 Paris, France
| | - Joseph Kononchik
- Microbiology and Infectious Diseases Department, Institut de Recherche Biomédicale des Armées, 91220 Brétigny-sur-Orge, France;
- US Army Medical Research Institute of Chemical Defense (USAMRICD), 8350 Ricketts Point Rd., Aberdeen Proving Ground, MD 21010, USA
- Toxicology and Chemical Risk Department, Institut de Recherche Biomédicale des Armées, 91220 Brétigny-sur-Orge, France
| |
Collapse
|
18
|
Lin PH, Liang CY, Yao BY, Chen HW, Pan CF, Wu LL, Lin YH, Hsu YS, Liu YH, Chen PJ, Hu CMJ, Yang HC. Robust induction of T RMs by combinatorial nanoshells confers cross-strain sterilizing immunity against lethal influenza viruses. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2021; 21:299-314. [PMID: 33898629 PMCID: PMC8047433 DOI: 10.1016/j.omtm.2021.03.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 03/17/2021] [Indexed: 01/01/2023]
Abstract
Antigen-specific lung-resident memory T cells (TRMs) constitute the first line of defense that mediates rapid protection against respiratory pathogens and inspires novel vaccine designs against infectious pandemic threats, yet effective means of inducing TRMs, particularly via non-viral vectors, remain challenging. Here, we demonstrate safe and potent induction of lung-resident TRMs using a biodegradable polymeric nanoshell that co-encapsulates antigenic peptides and TLR9 agonist CpG-oligodeoxynucleotide (CpG-ODN) in a virus-mimicking structure. Through subcutaneous priming and intranasal boosting, the combinatorial nanoshell vaccine elicits prominent lung-resident CD4+ and CD8+ T cells that surprisingly show better durability than live viral infections. In particular, nanoshells containing CpG-ODN and a pair of conserved class I and II major histocompatibility complex-restricted influenza nucleoprotein-derived antigenic peptides are demonstrated to induce near-sterilizing immunity against lethal infections with influenza A viruses of different strains and subtypes in mice, resulting in rapid elimination of replicating viruses. We further examine the pulmonary transport dynamic and optimal composition of the nanoshell vaccine conducive for robust TRM induction as well as the benefit of subcutaneous priming on TRM replenishment. The study presents a practical vaccination strategy for inducing protective TRM-mediated immunity, offering a compelling platform and critical insights in the ongoing quest toward a broadly protective vaccine against universal influenza as well as other respiratory pathogens.
Collapse
Affiliation(s)
- Pin-Hung Lin
- Graduate Institute of Microbiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chieh-Yu Liang
- Graduate Institute of Microbiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Bing-Yu Yao
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Hui-Wen Chen
- Department of Veterinary Medicine, National Taiwan University, Taipei, Taiwan
| | - Ching-Fu Pan
- Graduate Institute of Microbiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Li-Ling Wu
- Institute of Physiology, National Yang-Ming Chiao-Tung University, Taipei City, Taiwan
| | - Yi-Hsuan Lin
- Graduate Institute of Microbiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yu-Sung Hsu
- Graduate Institute of Microbiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yu-Han Liu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Pei-Jer Chen
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan.,Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan.,Hepatitis Research Center, National Taiwan University Hospital, Taipei, Taiwan
| | - Che-Ming Jack Hu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Hung-Chih Yang
- Graduate Institute of Microbiology, College of Medicine, National Taiwan University, Taipei, Taiwan.,Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| |
Collapse
|
19
|
Junter GA, Lebrun L. Polysaccharide-based chromatographic adsorbents for virus purification and viral clearance. J Pharm Anal 2020; 10:291-312. [PMID: 32292625 PMCID: PMC7104128 DOI: 10.1016/j.jpha.2020.01.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 01/10/2020] [Accepted: 01/11/2020] [Indexed: 12/20/2022] Open
Abstract
Viruses still pose a significant threat to human and animal health worldwide. In the fight against viral infections, high-purity viral stocks are needed for manufacture of safer vaccines. It is also a priority to ensure the viral safety of biopharmaceuticals such as blood products. Chromatography techniques are widely implemented at both academic and industrial levels in the purification of viral particles, whole viruses and virus-like particles to remove viral contaminants from biopharmaceutical products. This paper focuses on polysaccharide adsorbents, particulate resins and membrane adsorbers, used in virus purification/removal chromatography processes. Different chromatographic modes are surveyed, with particular attention to ion exchange and affinity/pseudo-affinity adsorbents among which commercially available agarose-based resins (Sepharose®) and cellulose-based membrane adsorbers (Sartobind®) occupy a dominant position. Mainly built on the development of new ligands coupled to conventional agarose/cellulose matrices, the development perspectives of polysaccharide-based chromatography media in this antiviral area are stressed in the conclusive part.
Collapse
Affiliation(s)
- Guy-Alain Junter
- Normandie Univ, UNIROUEN, INSA Rouen, CNRS, PBS, 76000, Rouen, France
| | - Laurent Lebrun
- Normandie Univ, UNIROUEN, INSA Rouen, CNRS, PBS, 76000, Rouen, France
| |
Collapse
|
20
|
Prospects and Challenges in the Development of Universal Influenza Vaccines. Vaccines (Basel) 2020; 8:vaccines8030361. [PMID: 32640619 PMCID: PMC7563311 DOI: 10.3390/vaccines8030361] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 06/29/2020] [Accepted: 06/30/2020] [Indexed: 01/19/2023] Open
Abstract
Current influenza vaccines offer suboptimal protection and depend on annual reformulation and yearly administration. Vaccine technology has rapidly advanced during the last decade, facilitating development of next-generation influenza vaccines that can target a broader range of influenza viruses. The development and licensure of a universal influenza vaccine could provide a game changing option for the control of influenza by protecting against all influenza A and B viruses. Here we review important findings and considerations regarding the development of universal influenza vaccines and what we can learn from this moving forward with a SARS-CoV-2 vaccine design.
Collapse
|
21
|
Meilleur CE, Memarnejadian A, Shivji AN, Benoit JM, Tuffs SW, Mele TS, Singh B, Dikeakos JD, Topham DJ, Mu HH, Bennink JR, McCormick JK, Haeryfar SMM. Discordant rearrangement of primary and anamnestic CD8+ T cell responses to influenza A viral epitopes upon exposure to bacterial superantigens: Implications for prophylactic vaccination, heterosubtypic immunity and superinfections. PLoS Pathog 2020; 16:e1008393. [PMID: 32433711 PMCID: PMC7239382 DOI: 10.1371/journal.ppat.1008393] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 02/10/2020] [Indexed: 12/21/2022] Open
Abstract
Infection with (SAg)-producing bacteria may precede or follow infection with or vaccination against influenza A viruses (IAVs). However, how SAgs alter the breadth of IAV-specific CD8+ T cell (TCD8) responses is unknown. Moreover, whether recall responses mediating heterosubtypic immunity to IAVs are manipulated by SAgs remains unexplored. We employed wild-type (WT) and mutant bacterial SAgs, SAg-sufficient/deficient Staphylococcus aureus strains, and WT, mouse-adapted and reassortant IAV strains in multiple in vivo settings to address the above questions. Contrary to the popular view that SAgs delete or anergize T cells, systemic administration of staphylococcal enterotoxin B (SEB) or Mycoplasma arthritidis mitogen before intraperitoneal IAV immunization enlarged the clonal size of ‘select’ IAV-specific TCD8 and reshuffled the hierarchical pattern of primary TCD8 responses. This was mechanistically linked to the TCR Vβ makeup of the impacted clones rather than their immunodominance status. Importantly, SAg-expanded TCD8 retained their IFN-γ production and cognate cytolytic capacities. The enhancing effect of SEB on immunodominant TCD8 was also evident in primary responses to vaccination with heat-inactivated and live attenuated IAV strains administered intramuscularly and intranasally, respectively. Interestingly, in prime-boost immunization settings, the outcome of SEB administration depended strictly upon the time point at which this SAg was introduced. Accordingly, SEB injection before priming raised CD127highKLRG1low memory precursor frequencies and augmented the anamnestic responses of SEB-binding TCD8. By comparison, introducing SEB before boosting diminished recall responses to IAV-derived epitopes drastically and indiscriminately. This was accompanied by lower Ki67 and higher Fas, LAG-3 and PD-1 levels consistent with a pro-apoptotic and/or exhausted phenotype. Therefore, SAgs can have contrasting impacts on anti-IAV immunity depending on the naïve/memory status and the TCR composition of exposed TCD8. Finally, local administration of SEB or infection with SEB-producing S. aureus enhanced pulmonary TCD8 responses to IAV. Our findings have clear implications for superinfections and prophylactic vaccination. Exposure to bacterial superantigens (SAgs) is often a consequence of infection with common Gram-positive bacteria causing septic and toxic shock or food poisoning. How SAgs affect the magnitude, breadth and quality of infection/vaccine-elicited CD8+ T cell (TCD8) responses to respiratory viral pathogens, including influenza A viruses (IAVs), is far from clear. Also importantly, superinfections with IAVs and SAg-producing bacteria are serious clinical occurrences during seasonal and pandemic flu and require urgent attention. We demonstrate that two structurally distinct SAgs, including staphylococcal enterotoxin B (SEB), unexpectedly enhance primary TCD8 responses to ‘select’ IAV-derived epitopes depending on the TCR makeup of the responding clones. Intriguingly, the timing of exposure to SEB dictates the outcome of prime-boost immunization. Seeing a SAg before priming raises memory precursor frequencies and augments anamnestic TCD8 responses. Conversely, a SAg encounter before boosting renders TCD8 prone to death or exhaustion and impedes recall responses, thus likely compromising heterosubtypic immunity to IAVs. Finally, local exposure to SEB increases the pulmonary response of immunodominant IAV-specific TCD8. These findings shed new light on how bacterial infections and SAgs influence the effectiveness of anti-IAV TCD8 responses, and have, as such, wide-ranging implications for preventative vaccination and infection control.
Collapse
Affiliation(s)
- Courtney E. Meilleur
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
| | - Arash Memarnejadian
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
| | - Adil N. Shivji
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
| | - Jenna M. Benoit
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
| | - Stephen W. Tuffs
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
| | - Tina S. Mele
- Division of General Surgery, Department of Surgery, Western University, London, Ontario, Canada
- Division of Critical Care Medicine, Department of Medicine, Western University, London, Ontario, Canada
| | - Bhagirath Singh
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
- Lawson Health Research Institute, London, Ontario, Canada
- Centre for Human Immunology, Western University, London, Ontario, Canada
| | - Jimmy D. Dikeakos
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
| | - David J. Topham
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Hong-Hua Mu
- Division of Rheumatology, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, Utah, United States of America
| | - Jack R. Bennink
- Viral Immunology Section, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - John K. McCormick
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
- Lawson Health Research Institute, London, Ontario, Canada
- Centre for Human Immunology, Western University, London, Ontario, Canada
| | - S. M. Mansour Haeryfar
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
- Division of General Surgery, Department of Surgery, Western University, London, Ontario, Canada
- Lawson Health Research Institute, London, Ontario, Canada
- Centre for Human Immunology, Western University, London, Ontario, Canada
- Division of Clinical Immunology & Allergy, Department of Medicine, Western University, London, Ontario, Canada
- * E-mail:
| |
Collapse
|
22
|
Wagner A, Weinberger B. Vaccines to Prevent Infectious Diseases in the Older Population: Immunological Challenges and Future Perspectives. Front Immunol 2020; 11:717. [PMID: 32391017 PMCID: PMC7190794 DOI: 10.3389/fimmu.2020.00717] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 03/30/2020] [Indexed: 12/15/2022] Open
Abstract
Infectious diseases are a major cause for morbidity and mortality in the older population. Demographic changes will lead to increasing numbers of older persons over the next decades. Prevention of infections becomes increasingly important to ensure healthy aging for the individual, and to alleviate the socio-economic burden for societies. Undoubtedly, vaccines are the most efficient health care measure to prevent infections. Age-associated changes of the immune system are responsible for decreased immunogenicity and clinical efficacy of most currently used vaccines in older age. Efficacy of standard influenza vaccines is only 30-50% in the older population. Several approaches, such as higher antigen dose, use of MF59 as adjuvant and intradermal administration have been implemented in order to specifically target the aged immune system. The use of a 23-valent polysaccharide vaccine against Streptococcus pneumoniae has been amended by a 13-valent conjugated pneumococcal vaccine originally developed for young children several years ago to overcome at least some of the limitations of the T cell-independent polysaccharide antigens, but still is only approximately 50% protective against pneumonia. A live-attenuated vaccine against herpes zoster, which has been available for several years, demonstrated efficacy of 51% against herpes zoster and 67% against post-herpetic neuralgia. Protection was lower in the very old and decreased several years after vaccination. Recently, a recombinant vaccine containing the viral glycoprotein gE and the novel adjuvant AS01B has been licensed. Phase III studies demonstrated efficacy against herpes zoster of approx. 90% even in the oldest age groups after administration of two doses and many countries now recommend the preferential use of this vaccine. There are still many infectious diseases causing substantial morbidity in the older population, for which no vaccines are available so far. Extensive research is ongoing to develop vaccines against novel targets with several vaccine candidates already being clinically tested, which have the potential to substantially reduce health care costs and to save many lives. In addition to the development of novel and improved vaccines, which specifically target the aged immune system, it is also important to improve uptake of the existing vaccines in order to protect the vulnerable, older population.
Collapse
Affiliation(s)
- Angelika Wagner
- Department of Pathophysiology, Infectiology, and Immunology, Institute of Specific Prophylaxis and Tropical Medicine, Medical University of Vienna, Vienna, Austria
| | - Birgit Weinberger
- Institute for Biomedical Aging Research, Universität Innsbruck, Innsbruck, Austria
| |
Collapse
|
23
|
Demminger DE, Walz L, Dietert K, Hoffmann H, Planz O, Gruber AD, von Messling V, Wolff T. Adeno-associated virus-vectored influenza vaccine elicits neutralizing and Fcγ receptor-activating antibodies. EMBO Mol Med 2020; 12:e10938. [PMID: 32163240 PMCID: PMC7207162 DOI: 10.15252/emmm.201910938] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 02/11/2020] [Accepted: 02/12/2020] [Indexed: 12/14/2022] Open
Abstract
The current seasonal inactivated influenza vaccine protects only against a narrow range of virus strains as it triggers a dominant antibody response toward the hypervariable hemagglutinin (HA) head region. The discovery of rare broadly protective antibodies against conserved regions in influenza virus proteins has propelled research on distinct antigens and delivery methods to efficiently induce broad immunity toward drifted or shifted virus strains. Here, we report that adeno‐associated virus (AAV) vectors expressing influenza virus HA or chimeric HA protected mice against homologous and heterologous virus challenges. Unexpectedly, immunization even with wild‐type HA induced antibodies recognizing the HA‐stalk and activating FcγR‐dependent responses indicating that AAV‐vectored expression balances HA head‐ and HA stalk‐specific humoral responses. Immunization with AAV‐HA partially protected also ferrets against a harsh virus challenge. Results from this study provide a rationale for further clinical development of AAV vectors as influenza vaccine platform, which could benefit from their approved use in human gene therapy.
Collapse
Affiliation(s)
- Daniel E Demminger
- Unit 17-Influenza and Other Respiratory Viruses, Robert Koch Institute, Berlin, Germany
| | - Lisa Walz
- Veterinary Medicine Division, Paul-Ehrlich-Institute, Langen, Germany
| | - Kristina Dietert
- Department of Veterinary Medicine, Institute of Veterinary Pathology, Berlin, Germany
| | - Helen Hoffmann
- Department of Immunology, Interfaculty Institute for Cell Biology, Eberhard Karls University, Tübingen, Germany
| | - Oliver Planz
- Department of Immunology, Interfaculty Institute for Cell Biology, Eberhard Karls University, Tübingen, Germany
| | - Achim D Gruber
- Department of Veterinary Medicine, Institute of Veterinary Pathology, Berlin, Germany
| | | | - Thorsten Wolff
- Unit 17-Influenza and Other Respiratory Viruses, Robert Koch Institute, Berlin, Germany
| |
Collapse
|
24
|
Calzas C, Chevalier C. Innovative Mucosal Vaccine Formulations Against Influenza A Virus Infections. Front Immunol 2019; 10:1605. [PMID: 31379823 PMCID: PMC6650573 DOI: 10.3389/fimmu.2019.01605] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 06/27/2019] [Indexed: 12/11/2022] Open
Abstract
Despite efforts made to develop efficient preventive strategies, infections with influenza A viruses (IAV) continue to cause serious clinical and economic problems. Current licensed human vaccines are mainly inactivated whole virus particles or split-virion administered via the parenteral route. These vaccines provide incomplete protection against IAV in high-risk groups and are poorly/not effective against the constant antigenic drift/shift occurring in circulating strains. Advances in mucosal vaccinology and in the understanding of the protective anti-influenza immune mechanisms suggest that intranasal immunization is a promising strategy to fight against IAV. To date, human mucosal anti-influenza vaccines consist of live attenuated strains administered intranasally, which elicit higher local humoral and cellular immune responses than conventional parenteral vaccines. However, because of inconsistent protective efficacy and safety concerns regarding the use of live viral strains, new vaccine candidates are urgently needed. To prime and induce potent and long-lived protective immune responses, mucosal vaccine formulations need to ensure the immunoavailability and the immunostimulating capacity of the vaccine antigen(s) at the mucosal surfaces, while being minimally reactogenic/toxic. The purpose of this review is to compile innovative delivery/adjuvant systems tested for intranasal administration of inactivated influenza vaccines, including micro/nanosized particulate carriers such as lipid-based particles, virus-like particles and polymers associated or not with immunopotentiatory molecules including microorganism-derived toxins, Toll-like receptor ligands and cytokines. The capacity of these vaccines to trigger specific mucosal and systemic humoral and cellular responses against IAV and their (cross)-protective potential are considered.
Collapse
Affiliation(s)
- Cynthia Calzas
- VIM, UR892, Equipe Virus Influenza, INRA, University PARIS-SACLAY, Jouy-en-Josas, France
| | - Christophe Chevalier
- VIM, UR892, Equipe Virus Influenza, INRA, University PARIS-SACLAY, Jouy-en-Josas, France
| |
Collapse
|
25
|
Khanna M, Manocha N, Himanshi, Joshi G, Saxena L, Saini S. Role of retroviral vector-based interventions in combating virus infections. Future Virol 2019. [DOI: 10.2217/fvl-2018-0151] [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
The deployment of viruses as vaccine-vectors has witnessed recent developments owing to a better understanding of viral genomes and mechanism of interaction with the immune system. Vaccine delivery by viral vectors offers various advantages over traditional approaches. Viral vector vaccines are one of the best candidates for activating the cellular arm of the immune system, coupled with the induction of significant humoral responses. Hence, there is a broad scope for the development of effective vaccines against many diseases using viruses as vectors. Further studies are required before an ideal vaccine-vector is developed and licensed for use in humans. In this article, we have outlined the use of retroviral vectors in developing vaccines against various viral diseases.
Collapse
Affiliation(s)
- Madhu Khanna
- Virology Unit, Vallabhbhai Patel Chest Institute, University of Delhi, Delhi 110 007, India
| | - Nilanshu Manocha
- Virology Unit, Vallabhbhai Patel Chest Institute, University of Delhi, Delhi 110 007, India
| | - Himanshi
- Virology Unit, Vallabhbhai Patel Chest Institute, University of Delhi, Delhi 110 007, India
| | - Garima Joshi
- Virology Unit, Vallabhbhai Patel Chest Institute, University of Delhi, Delhi 110 007, India
| | - Latika Saxena
- Virology Unit, Vallabhbhai Patel Chest Institute, University of Delhi, Delhi 110 007, India
| | - Sanjesh Saini
- Virology Unit, Vallabhbhai Patel Chest Institute, University of Delhi, Delhi 110 007, India
| |
Collapse
|
26
|
Keshwara R, Hagen KR, Abreu-Mota T, Papaneri AB, Liu D, Wirblich C, Johnson RF, Schnell MJ. A Recombinant Rabies Virus Expressing the Marburg Virus Glycoprotein Is Dependent upon Antibody-Mediated Cellular Cytotoxicity for Protection against Marburg Virus Disease in a Murine Model. J Virol 2019; 93:e01865-18. [PMID: 30567978 PMCID: PMC6401435 DOI: 10.1128/jvi.01865-18] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 12/10/2018] [Indexed: 12/14/2022] Open
Abstract
Marburg virus (MARV) is a filovirus related to Ebola virus (EBOV) associated with human hemorrhagic disease. Outbreaks are sporadic and severe, with a reported case mortality rate of upward of 88%. There is currently no antiviral or vaccine available. Given the sporadic nature of outbreaks, vaccines provide the best approach for long-term control of MARV in regions of endemicity. We have developed an inactivated rabies virus-vectored MARV vaccine (FILORAB3) to protect against Marburg virus disease. Immunogenicity studies in our labs have shown that a Th1-biased seroconversion to both rabies virus and MARV glycoproteins (GPs) is beneficial for protection in a preclinical murine model. As such, we adjuvanted FILORAB3 with glucopyranosyl lipid adjuvant (GLA), a Toll-like receptor 4 agonist, in a squalene-in-water emulsion. Across two different BALB/c mouse challenge models, we achieved 92% protection against murine-adapted Marburg virus (ma-MARV). Although our vaccine elicited strong MARV GP antibodies, it did not strongly induce neutralizing antibodies. Through both in vitro and in vivo approaches, we elucidated a critical role for NK cell-dependent antibody-mediated cellular cytotoxicity (ADCC) in vaccine-induced protection. Overall, these findings demonstrate that FILORAB3 is a promising vaccine candidate for Marburg virus disease.IMPORTANCE Marburg virus (MARV) is a virus similar to Ebola virus and also causes a hemorrhagic disease which is highly lethal. In contrast to EBOV, only a few vaccines have been developed against MARV, and researchers do not understand what kind of immune responses are required to protect from MARV. Here we show that antibodies directed against MARV after application of our vaccine protect in an animal system but fail to neutralize the virus in a widely used virus neutralization assay against MARV. This newly discovered activity needs to be considered more when analyzing MARV vaccines or infections.
Collapse
Affiliation(s)
- Rohan Keshwara
- Department of Microbiology and Immunology, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Katie R Hagen
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Maryland, USA
| | - Tiago Abreu-Mota
- Department of Microbiology and Immunology, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, Pennsylvania, USA
- Life and Health Sciences Research Institute (ICVS) School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's, PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Amy B Papaneri
- Emerging Viral Pathogens Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - David Liu
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Maryland, USA
| | - Christoph Wirblich
- Department of Microbiology and Immunology, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Reed F Johnson
- Emerging Viral Pathogens Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Matthias J Schnell
- Department of Microbiology and Immunology, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, Pennsylvania, USA
- Jefferson Vaccine Center, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
27
|
Gerlach T, Elbahesh H, Saletti G, Rimmelzwaan GF. Recombinant influenza A viruses as vaccine vectors. Expert Rev Vaccines 2019; 18:379-392. [PMID: 30777467 DOI: 10.1080/14760584.2019.1582338] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
INTRODUCTION Various viruses, including poxviruses, adenoviruses and vesicular stomatitis virus, have been considered as vaccine vectors for the delivery of antigens of interest in the development of vaccines against newly emerging pathogens. AREAS COVERED Here, we review results that have been obtained with influenza A viruses (IAV) as vaccine vectors. With the advent of reverse genetics technology, IAV-based recombinant vaccine candidates have been constructed that induce protective immunity to a variety of different pathogens of interest, including West Nile virus, Plasmodium falciparum and respiratory syncytial virus. The various cloning strategies to produce effective and attenuated, safe to use IAV-based viral vectors are discussed. EXPERT COMMENTARY It was concluded that IAV-based vector system has several advantages and holds promise for further development.
Collapse
Affiliation(s)
- Thomas Gerlach
- a Research Center for Emerging Infections and Zoonoses (RIZ) , University of Veterinary Medicine Hannover (TiHo) , Hannover , Germany
| | - Husni Elbahesh
- a Research Center for Emerging Infections and Zoonoses (RIZ) , University of Veterinary Medicine Hannover (TiHo) , Hannover , Germany
| | - Giulietta Saletti
- a Research Center for Emerging Infections and Zoonoses (RIZ) , University of Veterinary Medicine Hannover (TiHo) , Hannover , Germany
| | - Guus F Rimmelzwaan
- a Research Center for Emerging Infections and Zoonoses (RIZ) , University of Veterinary Medicine Hannover (TiHo) , Hannover , Germany
| |
Collapse
|
28
|
Elbahesh H, Saletti G, Gerlach T, Rimmelzwaan GF. Broadly protective influenza vaccines: design and production platforms. Curr Opin Virol 2019; 34:1-9. [DOI: 10.1016/j.coviro.2018.11.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 11/07/2018] [Indexed: 01/04/2023]
|
29
|
Magnusson SE, Altenburg AF, Bengtsson KL, Bosman F, de Vries RD, Rimmelzwaan GF, Stertman L. Matrix-M™ adjuvant enhances immunogenicity of both protein- and modified vaccinia virus Ankara-based influenza vaccines in mice. Immunol Res 2019; 66:224-233. [PMID: 29594879 PMCID: PMC5899102 DOI: 10.1007/s12026-018-8991-x] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Influenza viruses continuously circulate in the human population and escape recognition by virus neutralizing antibodies induced by prior infection or vaccination through accumulation of mutations in the surface proteins hemagglutinin (HA) and neuraminidase (NA). Various strategies to develop a vaccine that provides broad protection against different influenza A viruses are under investigation, including use of recombinant (r) viral vectors and adjuvants. The replication-deficient modified vaccinia virus Ankara (MVA) is a promising vaccine vector that efficiently induces B and T cell responses specific for the antigen of interest. It is assumed that live vaccine vectors do not require an adjuvant to be immunogenic as the vector already mediates recruitment and activation of immune cells. To address this topic, BALB/c mice were vaccinated with either protein- or rMVA-based HA influenza vaccines, formulated with or without the saponin-based Matrix-M™ adjuvant. Co-formulation with Matrix-M significantly increased HA vaccine immunogenicity, resulting in antigen-specific humoral and cellular immune responses comparable to those induced by unadjuvanted rMVA-HA. Of special interest, rMVA-HA immunogenicity was also enhanced by addition of Matrix-M, demonstrated by enhanced HA inhibition antibody titres and cellular immune responses. Matrix-M added to either protein- or rMVA-based HA vaccines mediated recruitment and activation of antigen-presenting cells and lymphocytes to the draining lymph node 24 and 48 h post-vaccination. Taken together, these results suggest that adjuvants can be used not only with protein-based vaccines but also in combination with rMVA to increase vaccine immunogenicity, which may be a step forward to generate new and more effective influenza vaccines.
Collapse
Affiliation(s)
| | | | | | - Fons Bosman
- Amatsigroup NV, Biologicals Unit, Ghent, Belgium
| | - Rory D de Vries
- Department of Viroscience, Erasmus MC, Rotterdam, The Netherlands
| | - Guus F Rimmelzwaan
- Department of Viroscience, Erasmus MC, Rotterdam, The Netherlands
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine, Hannover, Germany
| | | |
Collapse
|
30
|
Abstract
Various adenovirus (AdV) vector systems have proven to be lucrative options for gene delivery. They can serve as potential vaccine candidates for prevention of several common infectious diseases and hold the promise for gene therapy, especially for cancer. Several AdV vector-based therapies are currently at various stages of clinical trials worldwide, which make an immense interest of both the clinicians and researchers. Since these vectors are easy to manipulate, have broad tropism, and have the capability to yield high titers, this delivery system has a wide range of applications for different clinical settings. This chapter emphasizes on some of the current usages of AdV vectors and their production methods.
Collapse
Affiliation(s)
- Ekramy E Sayedahmed
- Department of Comparative Pathobiology, Purdue Institute for Inflammation, Immunology, and Infectious Disease, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA
| | - Rashmi Kumari
- Department of Comparative Pathobiology, Purdue Institute for Inflammation, Immunology, and Infectious Disease, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA
| | - Suresh K Mittal
- Department of Comparative Pathobiology, Purdue Institute for Inflammation, Immunology, and Infectious Disease, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA.
| |
Collapse
|
31
|
Kochhar S, Excler JL, Bok K, Gurwith M, McNeil MM, Seligman SJ, Khuri-Bulos N, Klug B, Laderoute M, Robertson JS, Singh V, Chen RT. Defining the interval for monitoring potential adverse events following immunization (AEFIs) after receipt of live viral vectored vaccines. Vaccine 2018; 37:5796-5802. [PMID: 30497831 PMCID: PMC6535369 DOI: 10.1016/j.vaccine.2018.08.085] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 08/27/2018] [Indexed: 12/13/2022]
Abstract
Live viral vectors that express heterologous antigens of the target pathogen are being investigated in the development of novel vaccines against serious infectious agents like HIV and Ebola. As some live recombinant vectored vaccines may be replication-competent, a key challenge is defining the length of time for monitoring potential adverse events following immunization (AEFI) in clinical trials and epidemiologic studies. This time period must be chosen with care and based on considerations of pre-clinical and clinical trials data, biological plausibility and practical feasibility. The available options include: (1) adapting from the current relevant regulatory guidelines; (2) convening a panel of experts to review the evidence from a systematic literature search to narrow down a list of likely potential or known AEFI and establish the optimal risk window(s); and (3) conducting "near real-time" prospective monitoring for unknown clustering's of AEFI in validated large linked vaccine safety databases using Rapid Cycle Analysis for pre-specified adverse events of special interest (AESI) and Treescan to identify previously unsuspected outcomes. The risk window established by any of these options could be used along with (4) establishing a registry of clinically validated pre-specified AESI to include in case-control studies. Depending on the infrastructure, human resources and databases available in different countries, the appropriate option or combination of options can be determined by regulatory agencies and investigators.
Collapse
Affiliation(s)
- Sonali Kochhar
- Global Healthcare Consulting, New Delhi, India; Erasmus MC, University Medical Center, Rotterdam, the Netherlands; University of Washington, Seattle, USA
| | | | - Karin Bok
- National Vaccine Program Office, Office of the Assistant Secretary for Health, US Department of Health and Human Services, Washington DC, USA
| | | | - Michael M McNeil
- Immunization Safety Office, Centers for Disease Control and Prevention (CDC), Atlanta, GA, USA
| | - Stephen J Seligman
- Department of Microbiology and Immunology, New York Medical College, NY, USA; St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller University, New York, NY, USA
| | - Najwa Khuri-Bulos
- Division of Infectious Disease, Jordan University Hospital, Amman, Jordan
| | - Bettina Klug
- Division Immunology, Paul-Ehrlich-Institut, Langen, Germany
| | | | - James S Robertson
- Independent Adviser (formerly of National Institute for Biological Standards and Control), Potters Bar, UK
| | - Vidisha Singh
- Division of HIV/AIDS Prevention, National Center for HIV/AIDS, Viral Hepatitis, STD, and TB Prevention (NCHHSTP), USA
| | - Robert T Chen
- Division of HIV/AIDS Prevention, National Center for HIV/AIDS, Viral Hepatitis, STD, and TB Prevention (NCHHSTP), USA; Centers for Disease Control and Prevention (CDC), Atlanta, GA, USA.
| | | |
Collapse
|
32
|
Avian Influenza A Virus Pandemic Preparedness and Vaccine Development. Vaccines (Basel) 2018; 6:vaccines6030046. [PMID: 30044370 PMCID: PMC6161001 DOI: 10.3390/vaccines6030046] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 07/17/2018] [Accepted: 07/21/2018] [Indexed: 12/24/2022] Open
Abstract
Influenza A viruses can infect a wide range of hosts, creating opportunities for zoonotic transmission, i.e., transmission from animals to humans, and placing the human population at constant risk of potential pandemics. In the last hundred years, four influenza A virus pandemics have had a devastating effect, especially the 1918 influenza pandemic that took the lives of at least 40 million people. There is a constant risk that currently circulating avian influenza A viruses (e.g., H5N1, H7N9) will cause a new pandemic. Vaccines are the cornerstone in preparing for and combating potential pandemics. Despite exceptional advances in the design and development of (pre-)pandemic vaccines, there are still serious challenges to overcome, mainly caused by intrinsic characteristics of influenza A viruses: Rapid evolution and a broad host range combined with maintenance in animal reservoirs, making it near impossible to predict the nature and source of the next pandemic virus. Here, recent advances in the development of vaccination strategies to prepare against a pandemic virus coming from the avian reservoir will be discussed. Furthermore, remaining challenges will be addressed, setting the agenda for future research in the development of new vaccination strategies against potentially pandemic influenza A viruses.
Collapse
|
33
|
de Vries RD, Altenburg AF, Nieuwkoop NJ, de Bruin E, van Trierum SE, Pronk MR, Lamers MM, Richard M, Nieuwenhuijse DF, Koopmans MPG, Kreijtz JHCM, Fouchier RAM, Osterhaus ADME, Sutter G, Rimmelzwaan GF. Induction of Cross-Clade Antibody and T-Cell Responses by a Modified Vaccinia Virus Ankara-Based Influenza A(H5N1) Vaccine in a Randomized Phase 1/2a Clinical Trial. J Infect Dis 2018; 218:614-623. [PMID: 29912453 PMCID: PMC6047453 DOI: 10.1093/infdis/jiy214] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 04/15/2018] [Indexed: 11/14/2022] Open
Abstract
Background High-pathogenicity avian influenza viruses continue to circulate in poultry and wild birds and occasionally infect humans, sometimes with fatal outcomes. Development of vaccines is a priority to prepare for potential pandemics but is complicated by antigenic variation of the surface glycoprotein hemagglutinin. We report the immunological profile induced by human immunization with modified vaccinia virus Ankara (MVA) expressing the hemagglutinin gene of influenza A(H5N1) virus A/Vietnam/1194/04 (rMVA-H5). Methods In a double-blinded phase 1/2a clinical trial, 79 individuals received 1 or 2 injections of rMVA-H5 or vector control. Twenty-seven study subjects received a booster immunization after 1 year. The breadth, magnitude, and properties of vaccine-induced antibody and T-cell responses were characterized. Results rMVA-H5 induced broadly reactive antibody responses, demonstrated by protein microarray, hemagglutination inhibition, virus neutralization, and antibody-dependent cellular cytotoxicity assays. Antibodies cross-reacted with antigenically distinct H5 viruses, including the recently emerged subtypes H5N6 and H5N8 and the currently circulating subtype H5N1. In addition, the induction of T cells specific for H5 viruses of 2 different clades was demonstrated. Conclusions rMVA-H5 induced immune responses that cross-reacted with H5 viruses of various clades. These findings validate rMVA-H5 as vaccine candidate against antigenically distinct H5 viruses. Clinical Trials Registration NTR3401.
Collapse
Affiliation(s)
- Rory D de Vries
- Department of Viroscience, Postgraduate School of Molecular Medicine, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Arwen F Altenburg
- Department of Viroscience, Postgraduate School of Molecular Medicine, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Nella J Nieuwkoop
- Department of Viroscience, Postgraduate School of Molecular Medicine, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Erwin de Bruin
- Department of Viroscience, Postgraduate School of Molecular Medicine, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Stella E van Trierum
- Department of Viroscience, Postgraduate School of Molecular Medicine, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Mark R Pronk
- Department of Viroscience, Postgraduate School of Molecular Medicine, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Mart M Lamers
- Department of Viroscience, Postgraduate School of Molecular Medicine, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Mathilde Richard
- Department of Viroscience, Postgraduate School of Molecular Medicine, Erasmus Medical Center, Rotterdam, the Netherlands
| | - David F Nieuwenhuijse
- Department of Viroscience, Postgraduate School of Molecular Medicine, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Marion P G Koopmans
- Department of Viroscience, Postgraduate School of Molecular Medicine, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Joost H C M Kreijtz
- Department of Viroscience, Postgraduate School of Molecular Medicine, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Ron A M Fouchier
- Department of Viroscience, Postgraduate School of Molecular Medicine, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Albert D M E Osterhaus
- Department of Viroscience, Postgraduate School of Molecular Medicine, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Gerd Sutter
- Institute for Infectious Diseases and Zoonoses, Ludwig Maximilian University of Munich, Munich
- German Center for Infection Research, Hannover, Germany
| | - Guus F Rimmelzwaan
- Department of Viroscience, Postgraduate School of Molecular Medicine, Erasmus Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
34
|
Sebastian S, Lambe T. Clinical Advances in Viral-Vectored Influenza Vaccines. Vaccines (Basel) 2018; 6:E29. [PMID: 29794983 PMCID: PMC6027524 DOI: 10.3390/vaccines6020029] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 05/21/2018] [Accepted: 05/21/2018] [Indexed: 12/27/2022] Open
Abstract
Influenza-virus-mediated disease can be associated with high levels of morbidity and mortality, particularly in younger children and older adults. Vaccination is the primary intervention used to curb influenza virus infection, and the WHO recommends immunization for at-risk individuals to mitigate disease. Unfortunately, influenza vaccine composition needs to be updated annually due to antigenic shift and drift in the viral immunogen hemagglutinin (HA). There are a number of alternate vaccination strategies in current development which may circumvent the need for annual re-vaccination, including new platform technologies such as viral-vectored vaccines. We discuss the different vectored vaccines that have been or are currently in clinical trials, with a forward-looking focus on immunogens that may be protective against seasonal and pandemic influenza infection, in the context of viral-vectored vaccines. We also discuss future perspectives and limitations in the field that will need to be addressed before new vaccines can significantly impact disease levels.
Collapse
Affiliation(s)
- Sarah Sebastian
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Headington, Oxford OX3 DQ, UK.
| | - Teresa Lambe
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Headington, Oxford OX3 DQ, UK.
| |
Collapse
|
35
|
Gu H, Gao Y, Zhou S, Sun F, Zhao Z, Wang K, Zhao L, Zhang P, Wang Z, Zhang S, Wang X, Yang P. Bivalent vaccine platform based on ca influenza virus vaccine elicits protective immunity against human adenoviruses. Antiviral Res 2018; 153:78-84. [DOI: 10.1016/j.antiviral.2018.02.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 02/07/2018] [Accepted: 02/20/2018] [Indexed: 11/28/2022]
|
36
|
Altenburg AF, van Trierum SE, de Bruin E, de Meulder D, van de Sandt CE, van der Klis FRM, Fouchier RAM, Koopmans MPG, Rimmelzwaan GF, de Vries RD. Effects of pre-existing orthopoxvirus-specific immunity on the performance of Modified Vaccinia virus Ankara-based influenza vaccines. Sci Rep 2018; 8:6474. [PMID: 29692427 PMCID: PMC5915537 DOI: 10.1038/s41598-018-24820-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 04/10/2018] [Indexed: 11/26/2022] Open
Abstract
The replication-deficient orthopoxvirus modified vaccinia virus Ankara (MVA) is a promising vaccine vector against various pathogens and has an excellent safety record. However, pre-existing vector-specific immunity is frequently suggested to be a drawback of MVA-based vaccines. To address this issue, mice were vaccinated with MVA-based influenza vaccines in the presence or absence of orthopoxvirus-specific immunity. Importantly, protective efficacy of an MVA-based influenza vaccine against a homologous challenge was not impaired in the presence of orthopoxvirus-specific pre-existing immunity. Nonetheless, orthopoxvirus-specific pre-existing immunity reduced the induction of antigen-specific antibodies under specific conditions and completely prevented induction of antigen-specific T cell responses by rMVA-based vaccination. Notably, antibodies induced by vaccinia virus vaccination, both in mice and humans, were not capable of neutralizing MVA. Thus, when using rMVA-based vaccines it is important to consider the main correlate of protection induced by the vaccine, the vaccine dose and the orthopoxvirus immune status of vaccine recipients.
Collapse
Affiliation(s)
- Arwen F Altenburg
- Department of Viroscience, Postgraduate School of Molecular Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Stella E van Trierum
- Department of Viroscience, Postgraduate School of Molecular Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Erwin de Bruin
- Department of Viroscience, Postgraduate School of Molecular Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Dennis de Meulder
- Department of Viroscience, Postgraduate School of Molecular Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Carolien E van de Sandt
- Department of Viroscience, Postgraduate School of Molecular Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Fiona R M van der Klis
- Centre for Infectious Disease Control (Cib), National Institute of Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Ron A M Fouchier
- Department of Viroscience, Postgraduate School of Molecular Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Marion P G Koopmans
- Department of Viroscience, Postgraduate School of Molecular Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Guus F Rimmelzwaan
- Department of Viroscience, Postgraduate School of Molecular Medicine, Erasmus MC, Rotterdam, The Netherlands.,Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine, Hannover, Germany
| | - Rory D de Vries
- Department of Viroscience, Postgraduate School of Molecular Medicine, Erasmus MC, Rotterdam, The Netherlands.
| |
Collapse
|
37
|
Meseda CA, Atukorale V, Soto J, Eichelberger MC, Gao J, Wang W, Weiss CD, Weir JP. Immunogenicity and Protection Against Influenza H7N3 in Mice by Modified Vaccinia Virus Ankara Vectors Expressing Influenza Virus Hemagglutinin or Neuraminidase. Sci Rep 2018; 8:5364. [PMID: 29599502 PMCID: PMC5876369 DOI: 10.1038/s41598-018-23712-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 03/14/2018] [Indexed: 12/18/2022] Open
Abstract
Influenza subtypes such as H7 have pandemic potential since they are able to infect humans with severe consequences, as evidenced by the ongoing H7N9 infections in China that began in 2013. The diversity of H7 viruses calls for a broadly cross-protective vaccine for protection. We describe the construction of recombinant modified vaccinia virus Ankara (MVA) vectors expressing the hemagglutinin (HA) or neuraminidase (NA) from three H7 viruses representing both Eurasian and North American H7 lineages – A/mallard/Netherlands/12/2000 (H7N3), A/Canada/rv444/2004 (H7N3), and A/Shanghai/02/2013 (H7N9). These vectors were evaluated for immunogenicity and protective efficacy against H7N3 virus in a murine model of intranasal challenge. High levels of H7-, N3-, and N9-specific antibodies, including neutralizing antibodies, were induced by the MVA-HA and MVA-NA vectors. Mice vaccinated with MVA vectors expressing any of the H7 antigens were protected, suggesting cross-protection among H7 viruses. In addition, MVA vectors expressing N3 but not N9 elicited protection against H7N3 virus challenge. Similar outcomes were obtained when immune sera from MVA vector-immunized mice were passively transferred to naïve mice prior to challenge with the H7N3 virus. The results support the further development of an MVA vector platform as a candidate vaccine for influenza strains with pandemic potential.
Collapse
Affiliation(s)
- Clement A Meseda
- Laboratory of DNA Viruses, Center for Biologics Evaluations and Research, Food and Drug Administration, 10903 New Hampshire Ave, Silver Spring, MD, 20993, USA
| | - Vajini Atukorale
- Laboratory of DNA Viruses, Center for Biologics Evaluations and Research, Food and Drug Administration, 10903 New Hampshire Ave, Silver Spring, MD, 20993, USA
| | - Jackeline Soto
- Laboratory of DNA Viruses, Center for Biologics Evaluations and Research, Food and Drug Administration, 10903 New Hampshire Ave, Silver Spring, MD, 20993, USA
| | - Maryna C Eichelberger
- Laboratory of Respiratory Viral Diseases, Center for Biologics Evaluations and Research, Food and Drug Administration, 10903 New Hampshire Ave, Silver Spring, MD, 20993, USA
| | - Jin Gao
- Laboratory of Respiratory Viral Diseases, Center for Biologics Evaluations and Research, Food and Drug Administration, 10903 New Hampshire Ave, Silver Spring, MD, 20993, USA
| | - Wei Wang
- Laboratory of Immunoregulation, Division of Viral Products, Center for Biologics Evaluations and Research, Food and Drug Administration, 10903 New Hampshire Ave, Silver Spring, MD, 20993, USA
| | - Carol D Weiss
- Laboratory of Immunoregulation, Division of Viral Products, Center for Biologics Evaluations and Research, Food and Drug Administration, 10903 New Hampshire Ave, Silver Spring, MD, 20993, USA
| | - Jerry P Weir
- Laboratory of DNA Viruses, Center for Biologics Evaluations and Research, Food and Drug Administration, 10903 New Hampshire Ave, Silver Spring, MD, 20993, USA.
| |
Collapse
|
38
|
Modified Vaccinia Virus Ankara Preferentially Targets Antigen Presenting Cells In Vitro, Ex Vivo and In Vivo. Sci Rep 2017; 7:8580. [PMID: 28819261 PMCID: PMC5561217 DOI: 10.1038/s41598-017-08719-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 07/13/2017] [Indexed: 11/09/2022] Open
Abstract
Modified Vaccinia virus Ankara (MVA) is a promising vaccine vector with an excellent safety profile. However, despite extensive pre-clinical and clinical testing, surprisingly little is known about the cellular tropism of MVA, especially in relevant animal species. Here, we performed in vitro, ex vivo and in vivo experiments with recombinant MVA expressing green fluorescent protein (rMVA-GFP). In both human peripheral blood mononuclear cells and mouse lung explants, rMVA-GFP predominantly infected antigen presenting cells. Subsequent in vivo experiments performed in mice, ferrets and non-human primates indicated that preferential targeting of dendritic cells and alveolar macrophages was observed after respiratory administration, although subtle differences were observed between the respective animal species. Following intramuscular injection, rMVA-GFP was detected in interdigitating cells between myocytes, but also in myocytes themselves. These data are important in advancing our understanding of the basis for the immunogenicity of MVA-based vaccines and aid rational vaccine design and delivery strategies.
Collapse
|
39
|
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
|
40
|
Altenburg AF, Magnusson SE, Bosman F, Stertman L, de Vries RD, Rimmelzwaan GF. Protein and modified vaccinia virus Ankara-based influenza virus nucleoprotein vaccines are differentially immunogenic in BALB/c mice. Clin Exp Immunol 2017; 190:19-28. [PMID: 28665497 DOI: 10.1111/cei.13004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/13/2017] [Indexed: 02/06/2023] Open
Abstract
Because of the high variability of seasonal influenza viruses and the eminent threat of influenza viruses with pandemic potential, there is great interest in the development of vaccines that induce broadly protective immunity. Most probably, broadly protective influenza vaccines are based on conserved proteins, such as nucleoprotein (NP). NP is a vaccine target of interest as it has been shown to induce cross-reactive antibody and T cell responses. Here we tested and compared various NP-based vaccine preparations for their capacity to induce humoral and cellular immune responses to influenza virus NP. The immunogenicity of protein-based vaccine preparations with Matrix-M™ adjuvant as well as recombinant viral vaccine vector modified Vaccinia virus Ankara (MVA) expressing the influenza virus NP gene, with or without modifications that aim at optimization of CD8+ T cell responses, was addressed in BALB/c mice. Addition of Matrix-M™ adjuvant to NP wild-type protein-based vaccines significantly improved T cell responses. Furthermore, recombinant MVA expressing the influenza virus NP induced strong antibody and CD8+ T cell responses, which could not be improved further by modifications of NP to increase antigen processing and presentation.
Collapse
Affiliation(s)
- A F Altenburg
- Department of Viroscience, Postgraduate School of Molecular Medicine, Erasmus MC, Rotterdam, the Netherlands
| | | | - F Bosman
- AmatsiQ-Biologicals, Ghent, Belgium
| | | | - R D de Vries
- Department of Viroscience, Postgraduate School of Molecular Medicine, Erasmus MC, Rotterdam, the Netherlands
| | - G F Rimmelzwaan
- Department of Viroscience, Postgraduate School of Molecular Medicine, Erasmus MC, Rotterdam, the Netherlands
| |
Collapse
|
41
|
Karlsson I, Borggren M, Nielsen J, Christensen D, Williams J, Fomsgaard A. Increased humoral immunity by DNA vaccination using an α-tocopherol-based adjuvant. Hum Vaccin Immunother 2017; 13:1823-1830. [PMID: 28613978 DOI: 10.1080/21645515.2017.1321183] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
DNA vaccines induce broad immunity, which involves both humoral and strong cellular immunity, and can be rapidly designed for novel or evolving pathogens such as influenza. However, the humoral immunogenicity in humans and higher animals has been suboptimal compared with that of traditional vaccine approaches. We tested whether the emulsion-based and α-tocopherol containing adjuvant Diluvac Forte® has the ability to enhance the immunogenicity of a naked DNA vaccine (i.e., plasmid DNA). As a model vaccine, we used plasmids encoding both a surface-exposed viral glycoprotein (hemagglutinin) and an internal non-glycosylated nucleoprotein in the Th1/Th2 balanced CB6F1 mouse model. The naked DNA (50 µg) was premixed at a 1:1 volume/volume ratio with Diluvac Forte®, an emulsion containing different concentrations of α-tocopherol, the emulsion alone or endotoxin-free phosphate-buffered saline (PBS). The animals received 2 intracutaneous immunizations spaced 3 weeks apart. When combined with Diluvac Forte® or the emulsion containing α-tocopherol, the DNA vaccine induced a more potent and balanced immunoglobulin G (IgG)1 and IgG2c response, and both IgG subclass responses were significantly enhanced by the adjuvant. The DNA vaccine also induced CD4+ and CD8+ vaccine-specific T cells; however, the adjuvant did not exert a significant impact. We concluded that the emulsion-based adjuvant Diluvac Forte® enhanced the immunogenicity of a naked DNA vaccine encoding influenza proteins and that the adjuvant constituent α-tocopherol plays an important role in this immunogenicity. This induction of a potent and balanced humoral response without impairment of cellular immunity constitutes an important advancement toward effective DNA vaccines.
Collapse
Affiliation(s)
- Ingrid Karlsson
- a Virus Research and Development Laboratory, Department of Virus and Microbiological Special Diagnostics , Statens Serum Institut , Copenhagen , Denmark
| | - Marie Borggren
- a Virus Research and Development Laboratory, Department of Virus and Microbiological Special Diagnostics , Statens Serum Institut , Copenhagen , Denmark
| | - Jens Nielsen
- a Virus Research and Development Laboratory, Department of Virus and Microbiological Special Diagnostics , Statens Serum Institut , Copenhagen , Denmark
| | - Dennis Christensen
- b Department of Infectious Disease Immunology, Vaccine Adjuvant Research , Statens Serum Institut , Copenhagen , Denmark
| | - Jim Williams
- c Nature Technology Corporation , Lincoln , NE , USA
| | - Anders Fomsgaard
- a Virus Research and Development Laboratory, Department of Virus and Microbiological Special Diagnostics , Statens Serum Institut , Copenhagen , Denmark.,d Infectious Disease Research Unit, Clinical Institute , University of Southern Denmark , Odense , Denmark
| |
Collapse
|