1
|
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
|
2
|
Rajanala K, Upadhyay AK. Vaccines for Respiratory Viruses-COVID and Beyond. Vaccines (Basel) 2024; 12:936. [PMID: 39204059 PMCID: PMC11360283 DOI: 10.3390/vaccines12080936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 08/14/2024] [Accepted: 08/20/2024] [Indexed: 09/03/2024] Open
Abstract
The COVID-19 (coronavirus disease 2019) pandemic had an extensive impact on global morbidity and mortality. Several other common respiratory viruses, such as the influenza virus and respiratory syncytial virus (RSV), are endemic or epidemic agents causing acute respiratory infections that are easily transmissible and pose a significant threat to communities due to efficient person-to-person transmission. These viruses can undergo antigenic variation through genetic mutations, resulting in the emergence of novel strains or variants, thereby diminishing the effectiveness of current vaccines, and necessitating ongoing monitoring and adjustment of vaccine antigens. As the virus-specific immunity is maintained only for several weeks or months after the infection, there is an emergent need to develop effective and durable vaccines. Additionally, specific populations, such as elderly or immunocompromised individuals, may exhibit reduced immune responses to respiratory viruses, posing significant challenges to develop vaccines that elicit durable and potent immunity. We present a comprehensive review of the molecular mechanisms underlying the pathogenesis and virulence of common respiratory viruses, such as RSV, influenza virus, and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). We discuss several vaccine approaches that are under development. A thorough understanding of the current strategies and the challenges encountered during the vaccine development process can lead to the advancement of effective next-generation vaccines.
Collapse
|
3
|
Sanchez PL, Andre G, Antipov A, Petrovsky N, Ross TM. Advax-SM™-Adjuvanted COBRA (H1/H3) Hemagglutinin Influenza Vaccines. Vaccines (Basel) 2024; 12:455. [PMID: 38793706 PMCID: PMC11125990 DOI: 10.3390/vaccines12050455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 03/25/2024] [Accepted: 04/22/2024] [Indexed: 05/26/2024] Open
Abstract
Adjuvants enhance immune responses stimulated by vaccines. To date, many seasonal influenza vaccines are not formulated with an adjuvant. In the present study, the adjuvant Advax-SM™ was combined with next generation, broadly reactive influenza hemagglutinin (HA) vaccines that were designed using a computationally optimized broadly reactive antigen (COBRA) methodology. Advax-SM™ is a novel adjuvant comprising inulin polysaccharide and CpG55.2, a TLR9 agonist. COBRA HA vaccines were combined with Advax-SM™ or a comparator squalene emulsion (SE) adjuvant and administered to mice intramuscularly. Mice vaccinated with Advax-SM™ adjuvanted COBRA HA vaccines had increased serum levels of anti-influenza IgG and IgA, high hemagglutination inhibition activity against a panel of H1N1 and H3N2 influenza viruses, and increased anti-influenza antibody secreting cells isolated from spleens. COBRA HA plus Advax-SM™ immunized mice were protected against both morbidity and mortality following viral challenge and, at postmortem, had no detectable lung viral titers or lung inflammation. Overall, the Advax-SM™-adjuvanted COBRA HA formulation provided effective protection against drifted H1N1 and H3N2 influenza viruses.
Collapse
Affiliation(s)
- Pedro L. Sanchez
- Center for Vaccines and Immunology, University of Georgia, Athens, GA 30602, USA;
- Department of Infectious Diseases, University of Georgia, Athens, GA 30602, USA
- Florida Research and Innovation Center, Cleveland Clinic, Port Saint Lucie, FL 34987, USA
| | - Greiciely Andre
- Vaxine Pty Ltd., Adelaide, SA 5046, Australia; (G.A.); (A.A.); (N.P.)
| | - Anna Antipov
- Vaxine Pty Ltd., Adelaide, SA 5046, Australia; (G.A.); (A.A.); (N.P.)
| | - Nikolai Petrovsky
- Vaxine Pty Ltd., Adelaide, SA 5046, Australia; (G.A.); (A.A.); (N.P.)
| | - Ted M. Ross
- Center for Vaccines and Immunology, University of Georgia, Athens, GA 30602, USA;
- Department of Infectious Diseases, University of Georgia, Athens, GA 30602, USA
- Florida Research and Innovation Center, Cleveland Clinic, Port Saint Lucie, FL 34987, USA
- Department of Infection Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| |
Collapse
|
4
|
Stepanova E, Isakova-Sivak I, Mezhenskaya D, Niskanen S, Matyushenko V, Bazhenova E, Rak A, Wong PF, Prokopenko P, Kotomina T, Krutikova E, Legotskiy S, Neterebskii B, Ostroukhova T, Sivak K, Orshanskaya Y, Yakovlev K, Rudenko L. Expression of the SARS-CoV-2 receptor-binding domain by live attenuated influenza vaccine virus as a strategy for designing a bivalent vaccine against COVID-19 and influenza. Virol J 2024; 21:82. [PMID: 38589848 PMCID: PMC11003101 DOI: 10.1186/s12985-024-02350-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 03/22/2024] [Indexed: 04/10/2024] Open
Abstract
Influenza and SARS-CoV-2 are two major respiratory pathogens that cocirculate in humans and cause serious illness with the potential to exacerbate disease in the event of co-infection. To develop a bivalent vaccine, capable of protecting against both infections, we inserted the receptor-binding domain (RBD) of the SARS-CoV-2 spike protein into hemagglutinin (HA) molecule or into the open reading frame of the truncated nonstructural protein 1 (NS1) of live attenuated influenza vaccine (LAIV) virus and assessed phenotypic characteristics of the rescued LAIV-RBD viruses, as well as their immunogenicity in mouse and Syrian hamster animal models. A panel of 9 recombinant LAIV-RBD viruses was rescued using the A/Leningrad/17 backbone. Notably, only two variants with RBD insertions into the HA molecule could express sufficient quantities of RBD protein in infected MDCK cells. Intranasal immunization of mice induced high levels of anti-influenza antibody responses in all chimeric LAIV-RBD viruses, which was comparable to the LAIV virus vector. The RBD-specific antibody responses were most pronounced in the variant expressing RBD194 fragment as a chimeric HA protein. This candidate was further tested in Syrian hamsters and was shown to be immunogenic and capable of protecting animals against both infections.
Collapse
Affiliation(s)
| | | | - Daria Mezhenskaya
- Institute of Experimental Medicine, Saint Petersburg, 197022, Russia
| | - Sergei Niskanen
- Joint-Stock Company «BIOCAD» (JSC «BIOCAD») Saint Petersburg, Intracity Municipality the Settlement of Strelna, the Settlement of Strelna, ul. Svyazi, d. 38, str. 1, pomeshch. 89, Saint Petersburg, 198515, Russia
| | | | | | - Alexandra Rak
- Institute of Experimental Medicine, Saint Petersburg, 197022, Russia
| | - Pei Fong Wong
- Institute of Experimental Medicine, Saint Petersburg, 197022, Russia
| | - Polina Prokopenko
- Institute of Experimental Medicine, Saint Petersburg, 197022, Russia
| | - Tatiana Kotomina
- Institute of Experimental Medicine, Saint Petersburg, 197022, Russia
| | - Elena Krutikova
- Institute of Experimental Medicine, Saint Petersburg, 197022, Russia
| | - Sergei Legotskiy
- Joint-Stock Company «BIOCAD» (JSC «BIOCAD») Saint Petersburg, Intracity Municipality the Settlement of Strelna, the Settlement of Strelna, ul. Svyazi, d. 38, str. 1, pomeshch. 89, Saint Petersburg, 198515, Russia
| | - Bogdan Neterebskii
- Joint-Stock Company «BIOCAD» (JSC «BIOCAD») Saint Petersburg, Intracity Municipality the Settlement of Strelna, the Settlement of Strelna, ul. Svyazi, d. 38, str. 1, pomeshch. 89, Saint Petersburg, 198515, Russia
| | - Tatiana Ostroukhova
- Joint-Stock Company «BIOCAD» (JSC «BIOCAD») Saint Petersburg, Intracity Municipality the Settlement of Strelna, the Settlement of Strelna, ul. Svyazi, d. 38, str. 1, pomeshch. 89, Saint Petersburg, 198515, Russia
| | - Konstantin Sivak
- Smorodintsev Research Institute of Influenza, Saint Petersburg, 197376, Russia
| | - Yana Orshanskaya
- Smorodintsev Research Institute of Influenza, Saint Petersburg, 197376, Russia
| | - Kirill Yakovlev
- Smorodintsev Research Institute of Influenza, Saint Petersburg, 197376, Russia
| | - Larisa Rudenko
- Institute of Experimental Medicine, Saint Petersburg, 197022, Russia
| |
Collapse
|
5
|
Wong PF, Isakova-Sivak I, Stepanova E, Krutikova E, Bazhenova E, Rekstin A, Rudenko L. Development of Cross-Reactive Live Attenuated Influenza Vaccine Candidates against Both Lineages of Influenza B Virus. Vaccines (Basel) 2024; 12:95. [PMID: 38250908 PMCID: PMC10821225 DOI: 10.3390/vaccines12010095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 12/30/2023] [Accepted: 01/16/2024] [Indexed: 01/23/2024] Open
Abstract
BACKGROUND Influenza viruses continue to cause a significant social and economic burden globally. Vaccination is recognized as the most effective measure to control influenza. Live attenuated influenza vaccines (LAIVs) are an effective means of preventing influenza, especially among children. A reverse genetics (RG) system is required to rapidly update the antigenic composition of vaccines, as well as to design LAIVs with a broader spectrum of protection. Such a system has been developed for the Russian LAIVs only for type A strains, but not for influenza B viruses (IBV). METHODS All genes of the B/USSR/60/69 master donor virus (B60) were cloned into RG plasmids, and the engineered B60, as well as a panel of IBV LAIV reassortants were rescued from plasmid DNAs encoding all viral genes. The engineered viruses were evaluated in vitro and in a mouse model. RESULTS The B60 RG system was successfully developed, which made it possible to rescue LAIV reassortants with the desired antigenic composition, including hybrid strains with hemagglutinin and neuraminidase genes belonging to the viruses from different IBV lineages. The LAIV candidate carrying the HA of the B/Victoria-lineage virus and NA from the B/Yamagata-lineage virus demonstrated optimal characteristics in terms of safety, immunogenicity and cross-protection, prompting its further assessment as a broadly protective component of trivalent LAIV. CONCLUSIONS The new RG system for B60 MDV allowed the rapid generation of type B LAIV reassortants with desired genome compositions. The generation of hybrid LAIV reassortants with HA and NA genes belonging to the opposite IBV lineages is a promising approach for the development of IBV vaccines with broad cross-protection.
Collapse
Affiliation(s)
| | - Irina Isakova-Sivak
- Department of Virology, Institute of Experimental Medicine, 197022 St. Petersburg, Russia; (P.-F.W.); (E.S.); (E.K.); (E.B.); (A.R.); (L.R.)
| | | | | | | | | | | |
Collapse
|
6
|
Zhang X, Zhang J, Chen S, He Q, Bai Y, Liu J, Wang Z, Liang Z, Chen L, Mao Q, Xu M. Progress and challenges in the clinical evaluation of immune responses to respiratory mucosal vaccines. Expert Rev Vaccines 2024; 23:362-370. [PMID: 38444382 DOI: 10.1080/14760584.2024.2326094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 02/28/2024] [Indexed: 03/07/2024]
Abstract
INTRODUCTION Following the coronavirus disease pandemic, respiratory mucosal vaccines that elicit both mucosal and systemic immune responses have garnered increasing attention. However, human physiological characteristics pose significant challenges in the evaluation of mucosal immunity, which directly impedes the development and application of respiratory mucosal vaccines. AREAS COVERED This study summarizes the characteristics of immune responses in the respiratory mucosa and reviews the current status and challenges in evaluating immune response to respiratory mucosal vaccines. EXPERT OPINION Secretory Immunoglobulin A (S-IgA) is a major effector molecule at mucosal sites and a commonly used indicator for evaluating respiratory mucosal vaccines. However, the unique physiological structure of the respiratory tract pose significant challenges for the clinical collection and detection of S-IgA. Therefore, it is imperative to develop a sampling method with high collection efficiency and acceptance, a sensitive detection method, reference materials for mucosal antibodies, and to establish a threshold for S-IgA that correlates with clinical protection. Sample collection is even more challenging when evaluating mucosal cell immunity. Therefore, a mucosal cell sampling method with high operability and high tolerance should be established. Targets of the circulatory system capable of reflecting mucosal cellular immunity should also be explored.
Collapse
Affiliation(s)
- Xuanxuan Zhang
- State Key Laboratory of Drug Regulatory Science, Institute of Biological Products, National Institutes for Food and Drug Control, Beijing, China
| | - Jialu Zhang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Si Chen
- Drug and Vaccine Research Center, Guangzhou National Laboratory, Guangzhou, China
| | - Qian He
- State Key Laboratory of Drug Regulatory Science, Institute of Biological Products, National Institutes for Food and Drug Control, Beijing, China
| | - Yu Bai
- State Key Laboratory of Drug Regulatory Science, Institute of Biological Products, National Institutes for Food and Drug Control, Beijing, China
| | - Jianyang Liu
- State Key Laboratory of Drug Regulatory Science, Institute of Biological Products, National Institutes for Food and Drug Control, Beijing, China
| | - Zhongfang Wang
- Drug and Vaccine Research Center, Guangzhou National Laboratory, Guangzhou, China
| | - Zhenglun Liang
- State Key Laboratory of Drug Regulatory Science, Institute of Biological Products, National Institutes for Food and Drug Control, Beijing, China
| | - Ling Chen
- Drug and Vaccine Research Center, Guangzhou National Laboratory, Guangzhou, China
- State Key Laboratory of Respiratory Disease, Guangdong Laboratory of Computational Biomedicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Qunying Mao
- State Key Laboratory of Drug Regulatory Science, Institute of Biological Products, National Institutes for Food and Drug Control, Beijing, China
| | - Miao Xu
- State Key Laboratory of Drug Regulatory Science, Institute of Biological Products, National Institutes for Food and Drug Control, Beijing, China
| |
Collapse
|
7
|
Ivanov PA, Lyashko AV, Kost VY, Lomakina NF, Rtishchev AA, Bunkova NI, Timofeeva TA, Balanova MA, Ionov SA, Gorikov DV, Markushin SG. Determination of cold-adapted influenza virus (Orthomyxoviridae: Alphainfluenzavirus) polymerase activity by the minigenome method with a fluorescent protein. Vopr Virusol 2023; 68:526-535. [PMID: 38156568 DOI: 10.36233/0507-4088-203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Indexed: 12/30/2023]
Abstract
INTRODUCTION Polymerase proteins PB1 and PB2 determine the cold-adapted phenotype of the influenza virus A/Krasnodar/101/35/59 (H2N2), as was shown earlier. OBJECTIVE The development of the reporter construct to determine the activity of viral polymerase at 33 and 37 °C using the minigenome method. MATERIALS AND METHODS Co-transfection of Cos-1 cells with pHW2000 plasmids expressing viral polymerase proteins PB1, PB2, PA, NP (minigenome) and reporter construct. RESULTS Based on segment 8, two reporter constructs were created that contain a direct or inverted NS1-GFP-NS2 sequence for the expression of NS2 and NS1 proteins translationally fused with green fluorescent protein (GFP), which allowed the evaluation the transcriptional and/or replicative activity of viral polymerase. CONCLUSION Polymerase of virus A/Krasnodar/101/35/59 (H2N2) has higher replicative and transcriptional activity at 33 °C than at 37 °C. Its transcriptional activity is more temperature-dependent than its replicative activity. The replicative and transcriptional activity of polymerase A/Puerto Rico/8/34 virus (H1N1, Mount Sinai variant) have no significant differences and do not depend on temperature.
Collapse
Affiliation(s)
- P A Ivanov
- N.F. Gamaleya National Research Centre for Epidemiology and Microbiology, the Russian Ministry of Health
| | - A V Lyashko
- N.F. Gamaleya National Research Centre for Epidemiology and Microbiology, the Russian Ministry of Health
| | - V Y Kost
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences
| | - N F Lomakina
- N.F. Gamaleya National Research Centre for Epidemiology and Microbiology, the Russian Ministry of Health
| | | | - N I Bunkova
- N.F. Gamaleya National Research Centre for Epidemiology and Microbiology, the Russian Ministry of Health
| | - T A Timofeeva
- N.F. Gamaleya National Research Centre for Epidemiology and Microbiology, the Russian Ministry of Health
| | - M A Balanova
- N.F. Gamaleya National Research Centre for Epidemiology and Microbiology, the Russian Ministry of Health
| | - S A Ionov
- N.F. Gamaleya National Research Centre for Epidemiology and Microbiology, the Russian Ministry of Health
- Mendeleev University of Chemical Technology
| | - D V Gorikov
- N.F. Gamaleya National Research Centre for Epidemiology and Microbiology, the Russian Ministry of Health
- Mendeleev University of Chemical Technology
| | | |
Collapse
|
8
|
Rak A, Isakova-Sivak I, Rudenko L. Nucleoprotein as a Promising Antigen for Broadly Protective Influenza Vaccines. Vaccines (Basel) 2023; 11:1747. [PMID: 38140152 PMCID: PMC10747533 DOI: 10.3390/vaccines11121747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 11/17/2023] [Accepted: 11/22/2023] [Indexed: 12/24/2023] Open
Abstract
Annual vaccination is considered as the main preventive strategy against seasonal influenza. Due to the highly variable nature of major viral antigens, such as hemagglutinin (HA) and neuraminidase (NA), influenza vaccine strains should be regularly updated to antigenically match the circulating viruses. The influenza virus nucleoprotein (NP) is much more conserved than HA and NA, and thus seems to be a promising target for the design of improved influenza vaccines with broad cross-reactivity against antigenically diverse influenza viruses. Traditional subunit or recombinant protein influenza vaccines do not contain the NP antigen, whereas live-attenuated influenza vaccines (LAIVs) express the viral NP within infected cells, thus inducing strong NP-specific antibodies and T-cell responses. Many strategies have been explored to design broadly protective NP-based vaccines, mostly targeted at the T-cell mode of immunity. Although the NP is highly conserved, it still undergoes slow evolutionary changes due to selective immune pressure, meaning that the particular NP antigen selected for vaccine design may have a significant impact on the overall immunogenicity and efficacy of the vaccine candidate. In this review, we summarize existing data on the conservation of the influenza A viral nucleoprotein and review the results of preclinical and clinical trials of NP-targeting influenza vaccine prototypes, focusing on the ability of NP-specific immune responses to protect against diverse influenza viruses.
Collapse
Affiliation(s)
| | | | - Larisa Rudenko
- Department of Virology, Institute of Experimental Medicine, St. Petersburg 197022, Russia; (A.R.); (I.I.-S.)
| |
Collapse
|
9
|
Misplon JA, Lo CY, Crabbs TA, Price GE, Epstein SL. Adenoviral-vectored universal influenza vaccines administered intranasally reduce lung inflammatory responses upon viral challenge 15 months post-vaccination. J Virol 2023; 97:e0067423. [PMID: 37830821 PMCID: PMC10617573 DOI: 10.1128/jvi.00674-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 09/04/2023] [Indexed: 10/14/2023] Open
Abstract
IMPORTANCE Vaccines targeting highly conserved proteins can protect broadly against diverse viral strains. When a vaccine is administered to the respiratory tract, protection against disease is especially powerful. However, it is important to establish that this approach is safe. When vaccinated animals later encounter viruses, does reactivation of powerful local immunity, including T cell responses, damage the lungs? This study investigates the safety of mucosal vaccination of the respiratory tract. Non-replicating adenoviral vaccine vectors expressing conserved influenza virus proteins were given intranasally. This vaccine-induced protection persists for at least 15 months. Vaccination did not exacerbate inflammatory responses or tissue damage upon influenza virus infection. Instead, vaccination with nucleoprotein reduced cytokine responses and histopathology, while neutrophil and T cell responses resolved earlier. The results are promising for safe vaccination at the site of infection and thus have implications for the control of influenza and other respiratory viruses.
Collapse
Affiliation(s)
- Julia A. Misplon
- Division of Cellular and Gene Therapies, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Chia-Yun Lo
- Division of Cellular and Gene Therapies, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Torrie A. Crabbs
- Experimental Pathology Laboratories, Inc., Durham, North Carolina, USA
| | - Graeme E. Price
- Division of Cellular and Gene Therapies, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Suzanne L. Epstein
- Division of Cellular and Gene Therapies, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, USA
| |
Collapse
|
10
|
Dar L, Krishnan A, Kumar R, Dhakad S, Choudekar A, Bagga S, Sharma A, Kumar A, Jethani J, Saha S, Amarchand R, Kumar R, Choudhary A, Narayan VV, Gopal G, Lafond KE, Lindstrom S. Nasal shedding of vaccine viruses after immunization with a Russian-backbone live attenuated influenza vaccine in India. Influenza Other Respir Viruses 2023; 17:e13149. [PMID: 37380175 PMCID: PMC10293783 DOI: 10.1111/irv.13149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 03/24/2023] [Accepted: 05/07/2023] [Indexed: 06/30/2023] Open
Abstract
BACKGROUND We present post-vaccination nasal shedding findings from the phase IV, community-based, triple-blinded RCT conducted to assess efficacy of trivalent LAIV and inactivated influenza vaccines in rural north India. METHODS Children aged 2-10 years received LAIV or intranasal placebo across 2015 and 2016, as per initial allocation. On days 2 and 4 post-vaccination, trained study nurses collected nasal swabs from randomly selected subset of trial participants based on operational feasibility, accounting for 10.0% and 11.4% of enrolled participants in 2015 and 2016, respectively. Swabs were collected in viral transport medium and transported under cold chain to laboratory for testing by reverse transcriptase real-time polymerase chain reaction. RESULTS In year 1, on day 2 post-vaccination, 71.2% (74/104) of LAIV recipients shed at least one of vaccine virus strains compared to 42.3% (44/104) on day 4. During year 1, on day 2 post-vaccination, LAIV-A(H1N1)pdm09 was detected in nasal swabs of 12% LAIV recipients, LAIV-A(H3N2) in 41%, and LAIV-B in 59%. In year 2, virus shedding was substantially lower; 29.6% (32/108) of LAIV recipients shed one of the vaccine virus strains on day 2 compared to 21.3% on day 4 (23/108). CONCLUSION At day 2 post-vaccination in year 1, two-thirds of LAIV recipients were shedding vaccine viruses. Shedding of vaccine viruses varied between strains and was lower in year 2. More research is needed to determine the reason for lower virus shedding and vaccine efficacy for LAIV-A(H1N1)pdm09.
Collapse
Affiliation(s)
- Lalit Dar
- Department of MicrobiologyAll India Institute of Medical SciencesNew DelhiIndia
| | - Anand Krishnan
- Centre for Community MedicineAll India Institute of Medical SciencesNew DelhiIndia
| | - Ramesh Kumar
- Department of MicrobiologyAll India Institute of Medical SciencesNew DelhiIndia
| | - Shivram Dhakad
- Department of MicrobiologyAll India Institute of Medical SciencesNew DelhiIndia
| | - Avinash Choudekar
- Department of MicrobiologyAll India Institute of Medical SciencesNew DelhiIndia
| | - Sumedha Bagga
- Department of MicrobiologyAll India Institute of Medical SciencesNew DelhiIndia
| | - Amrit Sharma
- Department of MicrobiologyAll India Institute of Medical SciencesNew DelhiIndia
| | - Amit Kumar
- Department of MicrobiologyAll India Institute of Medical SciencesNew DelhiIndia
| | - Jyoti Jethani
- Department of MicrobiologyAll India Institute of Medical SciencesNew DelhiIndia
| | - Siddhartha Saha
- Influenza DivisionUS Centers for Disease Control and PreventionNew DelhiIndia
| | - Ritvik Amarchand
- Centre for Community MedicineAll India Institute of Medical SciencesNew DelhiIndia
| | - Rakesh Kumar
- Centre for Community MedicineAll India Institute of Medical SciencesNew DelhiIndia
| | - Aashish Choudhary
- Department of MicrobiologyAll India Institute of Medical SciencesNew DelhiIndia
| | | | - Giridara Gopal
- Centre for Community MedicineAll India Institute of Medical SciencesNew DelhiIndia
| | - Kathryn E. Lafond
- Influenza DivisionUS Centers for Disease Control and PreventionAtlantaGeorgiaUSA
| | - Stephen Lindstrom
- Influenza DivisionUS Centers for Disease Control and PreventionAtlantaGeorgiaUSA
| |
Collapse
|
11
|
Bagga S, Krishnan A, Dar L. Revisiting live attenuated influenza vaccine efficacy among children in developing countries. Vaccine 2023; 41:1009-1017. [PMID: 36604216 DOI: 10.1016/j.vaccine.2022.12.058] [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: 05/23/2022] [Revised: 12/21/2022] [Accepted: 12/22/2022] [Indexed: 01/05/2023]
Abstract
Seasonal influenza epidemics cause significant pediatric mortality and morbidity worldwide. Live attenuated influenza vaccines (LAIVs) can be administered intranasally, induce a broad and robust immune response, demonstrate higher yields during manufacturing as compared to inactivated influenza vaccines (IIVs), and thereby represent an attractive possibility for young children in developing countries. We summarize recent pediatric studies evaluating LAIV efficacy in developing countries where a large proportion of the influenza-virus-associated respiratory disease burden occurs. Recently, two randomized controlled trials (RCTs) assessing Russian-backbone trivalent LAIV in children reported contradictory results; vaccine efficacy varied between Bangladesh (41 %) and Senegal (0.0 %) against all influenza viral strains. Prior to 2013, Ann Arbor-based LAIV demonstrated superior efficacy as compared to IIV. However, due to low effectiveness of the Ann Arbor-based LAIV against influenza A(H1N1)pdm09-like viruses, the CDC Advisory Committee on Immunization Practices (ACIP) recommended against the use of LAIV during the 2016-17 and 2017-18 influenza seasons. Reduced replicative fitness of the A(H1N1)pdm09 LAIV strains is thought to have led to the low effectiveness of the Ann-Arbor-based LAIV. Once the A(H1N1)pdm09 component was updated, the ACIP reintroduced the Ann-Arbor-based LAIV as a vaccine choice for the 2018-19 influenza season. In 2021, results from a 2-year RCT evaluating the Russian-backbone trivalent LAIV in rural north India reported that LAIV demonstrated significantly lower efficacy compared to IIV, but in Year 2, the vaccine efficacy for LAIV and IIV was comparable. A profounder understanding of the mechanisms underlying varied efficacy of LAIV in developing countries is warranted. Assessing replicative fitness, in addition to antigenicity, when selecting annual A(H1N1)pdm09 components in the Russian-backbone trivalent LAIVs is essential and may ultimately, enable widespread utility in resource-poor settings.
Collapse
Affiliation(s)
- Sumedha Bagga
- Department of Microbiology, All India Institute of Medical Sciences, New Delhi, India
| | - Anand Krishnan
- Centre for Community Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Lalit Dar
- Department of Microbiology, All India Institute of Medical Sciences, New Delhi, India.
| |
Collapse
|
12
|
Gandhi L, Maisnam D, Rathore D, Chauhan P, Bonagiri A, Venkataramana M. Respiratory illness virus infections with special emphasis on COVID-19. Eur J Med Res 2022; 27:236. [PMID: 36348452 PMCID: PMC9641310 DOI: 10.1186/s40001-022-00874-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 10/26/2022] [Indexed: 11/10/2022] Open
Abstract
Viruses that emerge pose challenges for treatment options as their uniqueness would not know completely. Hence, many viruses are causing high morbidity and mortality for a long time. Despite large diversity, viruses share common characteristics for infection. At least 12 different respiratory-borne viruses are reported belonging to various virus taxonomic families. Many of these viruses multiply and cause damage to the upper and lower respiratory tracts. The description of these viruses in comparison with each other concerning their epidemiology, molecular characteristics, disease manifestations, diagnosis and treatment is lacking. Such information helps diagnose, differentiate, and formulate the control measures faster. The leading cause of acute illness worldwide is acute respiratory infections (ARIs) and are responsible for nearly 4 million deaths every year, mostly in young children and infants. Lower respiratory tract infections are the fourth most common cause of death globally, after non-infectious chronic conditions. This review aims to present the characteristics of different viruses causing respiratory infections, highlighting the uniqueness of SARS-CoV-2. We expect this review to help understand the similarities and differences among the closely related viruses causing respiratory infections and formulate specific preventive or control measures.
Collapse
Affiliation(s)
- Lekha Gandhi
- Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Hyderabad, 500046, Telangana, India
| | - Deepti Maisnam
- Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Hyderabad, 500046, Telangana, India
| | - Deepika Rathore
- Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Hyderabad, 500046, Telangana, India
| | - Preeti Chauhan
- Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Hyderabad, 500046, Telangana, India
| | - Anvesh Bonagiri
- Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Hyderabad, 500046, Telangana, India
| | - Musturi Venkataramana
- Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Hyderabad, 500046, Telangana, India.
| |
Collapse
|
13
|
Choy RKM, Bourgeois AL, Ockenhouse CF, Walker RI, Sheets RL, Flores J. Controlled Human Infection Models To Accelerate Vaccine Development. Clin Microbiol Rev 2022; 35:e0000821. [PMID: 35862754 PMCID: PMC9491212 DOI: 10.1128/cmr.00008-21] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The timelines for developing vaccines against infectious diseases are lengthy, and often vaccines that reach the stage of large phase 3 field trials fail to provide the desired level of protective efficacy. The application of controlled human challenge models of infection and disease at the appropriate stages of development could accelerate development of candidate vaccines and, in fact, has done so successfully in some limited cases. Human challenge models could potentially be used to gather critical information on pathogenesis, inform strain selection for vaccines, explore cross-protective immunity, identify immune correlates of protection and mechanisms of protection induced by infection or evoked by candidate vaccines, guide decisions on appropriate trial endpoints, and evaluate vaccine efficacy. We prepared this report to motivate fellow scientists to exploit the potential capacity of controlled human challenge experiments to advance vaccine development. In this review, we considered available challenge models for 17 infectious diseases in the context of the public health importance of each disease, the diversity and pathogenesis of the causative organisms, the vaccine candidates under development, and each model's capacity to evaluate them and identify correlates of protective immunity. Our broad assessment indicated that human challenge models have not yet reached their full potential to support the development of vaccines against infectious diseases. On the basis of our review, however, we believe that describing an ideal challenge model is possible, as is further developing existing and future challenge models.
Collapse
Affiliation(s)
- Robert K. M. Choy
- PATH, Center for Vaccine Innovation and Access, Seattle, Washington, USA
| | - A. Louis Bourgeois
- PATH, Center for Vaccine Innovation and Access, Seattle, Washington, USA
| | | | - Richard I. Walker
- PATH, Center for Vaccine Innovation and Access, Seattle, Washington, USA
| | | | - Jorge Flores
- PATH, Center for Vaccine Innovation and Access, Seattle, Washington, USA
| |
Collapse
|
14
|
Deviatkin AA, Simonov RA, Trutneva KA, Maznina AA, Khavina EM, Volchkov PY. Universal Flu mRNA Vaccine: Promises, Prospects, and Problems. Vaccines (Basel) 2022; 10:vaccines10050709. [PMID: 35632465 PMCID: PMC9145388 DOI: 10.3390/vaccines10050709] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 04/26/2022] [Accepted: 04/28/2022] [Indexed: 02/05/2023] Open
Abstract
The seasonal flu vaccine is, essentially, the only known way to prevent influenza epidemics. However, this approach has limited efficacy due to the high diversity of influenza viruses. Several techniques could potentially overcome this obstacle. A recent first-in-human study of a chimeric hemagglutinin-based universal influenza virus vaccine demonstrated promising results. The coronavirus pandemic triggered the development of fundamentally new vaccine platforms that have demonstrated their effectiveness in humans. Currently, there are around a dozen messenger RNA and self-amplifying RNA flu vaccines in clinical or preclinical trials. However, the applicability of novel approaches for a universal influenza vaccine creation remains unclear. The current review aims to cover the current state of this problem and to suggest future directions for RNA-based flu vaccine development.
Collapse
Affiliation(s)
- Andrei A. Deviatkin
- The National Medical Research Center for Endocrinology, 117036 Moscow, Russia; (A.A.D.); (K.A.T.)
- Genome Engineering Lab, Life Sciences Research Center, Moscow Institute of Physics and Technology (National Research University), 141700 Dolgoprudniy, Russia; (R.A.S.); (A.A.M.); (E.M.K.)
| | - Ruslan A. Simonov
- Genome Engineering Lab, Life Sciences Research Center, Moscow Institute of Physics and Technology (National Research University), 141700 Dolgoprudniy, Russia; (R.A.S.); (A.A.M.); (E.M.K.)
| | - Kseniya A. Trutneva
- The National Medical Research Center for Endocrinology, 117036 Moscow, Russia; (A.A.D.); (K.A.T.)
- Genome Engineering Lab, Life Sciences Research Center, Moscow Institute of Physics and Technology (National Research University), 141700 Dolgoprudniy, Russia; (R.A.S.); (A.A.M.); (E.M.K.)
| | - Anna A. Maznina
- Genome Engineering Lab, Life Sciences Research Center, Moscow Institute of Physics and Technology (National Research University), 141700 Dolgoprudniy, Russia; (R.A.S.); (A.A.M.); (E.M.K.)
| | - Elena M. Khavina
- Genome Engineering Lab, Life Sciences Research Center, Moscow Institute of Physics and Technology (National Research University), 141700 Dolgoprudniy, Russia; (R.A.S.); (A.A.M.); (E.M.K.)
| | - Pavel Y. Volchkov
- The National Medical Research Center for Endocrinology, 117036 Moscow, Russia; (A.A.D.); (K.A.T.)
- Genome Engineering Lab, Life Sciences Research Center, Moscow Institute of Physics and Technology (National Research University), 141700 Dolgoprudniy, Russia; (R.A.S.); (A.A.M.); (E.M.K.)
- Correspondence:
| |
Collapse
|
15
|
Krishnan A, Dar L, Saha S, Narayan VV, Kumar R, Kumar R, Amarchand R, Dhakad S, Chokker R, Choudekar A, Gopal G, Choudhary A, Potdar V, Chadha M, Lafond KE, Lindstrom S, Widdowson MA, Jain S. Efficacy of live attenuated and inactivated influenza vaccines among children in rural India: A 2-year, randomized, triple-blind, placebo-controlled trial. PLoS Med 2021; 18:e1003609. [PMID: 33914729 PMCID: PMC8118535 DOI: 10.1371/journal.pmed.1003609] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 05/13/2021] [Accepted: 04/04/2021] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Influenza is a cause of febrile acute respiratory infection (FARI) in India; however, few influenza vaccine trials have been conducted in India. We assessed absolute and relative efficacy of live attenuated influenza vaccine (LAIV) and inactivated influenza vaccine (IIV) among children aged 2 to 10 years in rural India through a randomized, triple-blind, placebo-controlled trial conducted over 2 years. METHODS AND FINDINGS In June 2015, children were randomly allocated to LAIV, IIV, intranasal placebo, or inactivated polio vaccine (IPV) in a 2:2:1:1 ratio. In June 2016, vaccination was repeated per original allocation. Overall, 3,041 children received LAIV (n = 1,015), IIV (n = 1,010), nasal placebo (n = 507), or IPV (n = 509). Mean age of children was 6.5 years with 20% aged 9 to 10 years. Through weekly home visits, nasal and throat swabs were collected from children with FARI and tested for influenza virus by polymerase chain reaction. The primary outcome was laboratory-confirmed influenza-associated FARI; vaccine efficacy (VE) was calculated using modified intention-to-treat (mITT) analysis by Cox proportional hazards model (PH) for each year. In Year 1, VE was 40.0% (95% confidence interval (CI) 25.2 to 51.9) for LAIV and 59.0% (95% CI 47.8 to 67.9) for IIV compared with controls; relative efficacy of LAIV compared with IIV was -46.2% (95% CI -88.9 to -13.1). In Year 2, VE was 51.9% (95% CI 42.0 to 60.1) for LAIV and 49.9% (95% CI 39.2 to 58.7) for IIV; relative efficacy of LAIV compared with IIV was 4.2% (95% CI -19.9 to 23.5). No serious adverse vaccine-attributable events were reported. Study limitations include differing dosage requirements for children between nasal and injectable vaccines (single dose of LAIV versus 2 doses of IIV) in Year 1 and the fact that immunogenicity studies were not conducted. CONCLUSIONS In this study, we found that LAIV and IIV vaccines were safe and moderately efficacious against influenza virus infection among Indian children. TRIAL REGISTRATION Clinical Trials Registry of India CTRI/2015/06/005902.
Collapse
Affiliation(s)
- Anand Krishnan
- Centre for Community Medicine, All India Institute of Medical Sciences, Delhi, India
- * E-mail:
| | - Lalit Dar
- Microbiology Department, All India Institute of Medical Sciences, Delhi, India
| | - Siddhartha Saha
- Influenza Program, Centers for Disease Control and Prevention, New Delhi, India
| | | | - Rakesh Kumar
- Centre for Community Medicine, All India Institute of Medical Sciences, Delhi, India
| | - Ramesh Kumar
- Microbiology Department, All India Institute of Medical Sciences, Delhi, India
| | - Ritvik Amarchand
- Centre for Community Medicine, All India Institute of Medical Sciences, Delhi, India
| | - Shivram Dhakad
- Microbiology Department, All India Institute of Medical Sciences, Delhi, India
| | - Reshmi Chokker
- Centre for Community Medicine, All India Institute of Medical Sciences, Delhi, India
| | - Avinash Choudekar
- Microbiology Department, All India Institute of Medical Sciences, Delhi, India
| | - Giridara Gopal
- Centre for Community Medicine, All India Institute of Medical Sciences, Delhi, India
| | - Aashish Choudhary
- Microbiology Department, All India Institute of Medical Sciences, Delhi, India
| | | | | | - Kathryn E. Lafond
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Stephen Lindstrom
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Marc-Alain Widdowson
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Seema Jain
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| |
Collapse
|
16
|
Stepanova E, Krutikova E, Wong PF, Matyushenko V, Bazhenova E, Isakova-Sivak I, Rudenko L. Safety, Immunogenicity, and Protective Efficacy of a Chimeric A/B Live Attenuated Influenza Vaccine in a Mouse Model. Microorganisms 2021; 9:microorganisms9020259. [PMID: 33513862 PMCID: PMC7910998 DOI: 10.3390/microorganisms9020259] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 01/06/2021] [Accepted: 01/25/2021] [Indexed: 01/20/2023] Open
Abstract
Influenza A and B viruses cause significant morbidity and mortality worldwide. Current influenza vaccines are composed of three or four strains: A/H1N1, A/H3N2, and B (Victoria and Yamagata lineages). It is of great interest if immunization against both type A and B influenza viruses can be combined in a single vaccine strain, thus reducing the cost of vaccine production and the possibility of strain interference within the multicomponent vaccine. In the current study, we developed an experimental live cold-adapted influenza intertype reassortant (influenza A and B) vaccine on the live attenuated influenza vaccine (LAIV) A/Leningrad/134/17/57 backbone. Hemagglutinin (HA) and neuraminidase (NA) functional domains were inherited from the influenza B/Brisbane/60/2008 strain, whereas their packaging signals were substituted with appropriate fragments of influenza A virus genes. The recombinant A/B virus efficiently replicated in eggs and Madin–Darby Canine Kidney (MDCK) cells under optimal conditions, temperature-sensitive phenotype was maintained, and its antigenic properties matched the influenza B parental virus. The chimeric vaccine was attenuated in mice: after intranasal immunization, viral replication was seen only in nasal turbinates but not in the lungs. Immunological studies demonstrated the induction of IgG antibody responses against the influenza A and B virus, whereas hemagglutination inhibition (HAI) and neutralizing antibodies were detected only against the influenza B virus, resulting in significant protection of immunized animals against influenza B virus challenge. IFNγ-secreting CD8 effector memory T cells (CD44+CD62L−) were detected in mouse splenocytes after stimulation with the specific influenza A peptide (NP366); however, the T-cell response was not sufficient to protect animals against infection with a high-dose mouse-adapted A/California/07/2009 (H1N1pdm09) virus, most probably due to the mismatch of key T-cell epitopes of the H1N1 virus and the LAIV backbone. Overall, generation of the chimeric A/B LAIV virus on a licensed LAIV backbone demonstrated prospects for the development of safe and efficacious vaccine candidates that afford combined protection against both type A and type B influenza viruses; however, further optimization of the T-cell epitope content within the LAIV backbone may be required.
Collapse
|
17
|
Mara K, Dai M, Brice AM, Alexander MR, Tribolet L, Layton DS, Bean AGD. Investigating the Interaction between Negative Strand RNA Viruses and Their Hosts for Enhanced Vaccine Development and Production. Vaccines (Basel) 2021; 9:vaccines9010059. [PMID: 33477334 PMCID: PMC7830660 DOI: 10.3390/vaccines9010059] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Accepted: 01/13/2021] [Indexed: 11/30/2022] Open
Abstract
The current pandemic has highlighted the ever-increasing risk of human to human spread of zoonotic pathogens. A number of medically-relevant zoonotic pathogens are negative-strand RNA viruses (NSVs). NSVs are derived from different virus families. Examples like Ebola are known for causing severe symptoms and high mortality rates. Some, like influenza, are known for their ease of person-to-person transmission and lack of pre-existing immunity, enabling rapid spread across many countries around the globe. Containment of outbreaks of NSVs can be difficult owing to their unpredictability and the absence of effective control measures, such as vaccines and antiviral therapeutics. In addition, there remains a lack of essential knowledge of the host–pathogen response that are induced by NSVs, particularly of the immune responses that provide protection. Vaccines are the most effective method for preventing infectious diseases. In fact, in the event of a pandemic, appropriate vaccine design and speed of vaccine supply is the most critical factor in protecting the population, as vaccination is the only sustainable defense. Vaccines need to be safe, efficient, and cost-effective, which is influenced by our understanding of the host–pathogen interface. Additionally, some of the major challenges of vaccines are the establishment of a long-lasting immunity offering cross protection to emerging strains. Although many NSVs are controlled through immunisations, for some, vaccine design has failed or efficacy has proven unreliable. The key behind designing a successful vaccine is understanding the host–pathogen interaction and the host immune response towards NSVs. In this paper, we review the recent research in vaccine design against NSVs and explore the immune responses induced by these viruses. The generation of a robust and integrated approach to development capability and vaccine manufacture can collaboratively support the management of outbreaking NSV disease health risks.
Collapse
|
18
|
Ganguly M, Yeolekar L, Tyagi P, Sagar U, Narale S, Anaspure Y, Tupe S, Wadkar K, Ingle N, Dhere R, Scorza FB, Mahmood K. Evaluation of manufacturing feasibility and safety of an MDCK cell-based live attenuated influenza vaccine (LAIV) platform. Vaccine 2020; 38:8379-8386. [PMID: 33229107 DOI: 10.1016/j.vaccine.2020.10.092] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 10/24/2020] [Accepted: 10/31/2020] [Indexed: 12/23/2022]
Abstract
Cell culture based live attenuated influenza vaccines (LAIV) as an alternative to egg-based LAIV have been explored because of lack of easy access to SPF eggs for large scale production. In this study, feasibility of MDCK platform was assessed by including multiple LAIV strains covering both type A (H1 and H3) and type B seasonal strains as well as the candidate pandemic potential strains like A/H5 and A/H7 for the growth in MDCK cells. A risk assessment study was conducted on the cell banks to evaluate safety concerns related to tumorigenicity with a regulatory perspective. Tumorigenic potential of the MDCK cells was evaluated in nude mice (107cells/mouse) model system. The 50% tumor producing dose (TPD50) of MDCK cells was studied in SCID mice to determine the amount of cells required for induction of tumors. Further, we conducted an oncogenicity study in three sensitive rodent species as per the requirements specified in the WHO guidelines. We determined TPD50 value of 1.9 X 104 cells/mice through subcutaneous route. Our results suggest that, the intranasal route of administration of the cell culture based LAIV pose minimal to no risk of tumorigenicity associated with the host cells. Also, non-oncogenic nature of MDCK cells was demonstrated. Host cell DNA in the vaccine formulations was < 10 ng/dose which ensures vaccine safety. Production efficiency and consistency were characterized and the observed titer values of the viral harvest and the processed bulk were comparable to the expansion in embryonated eggs. The present study clearly establishes the suitability of MDCK cells as a substrate for the manufacture of a safe and viable LAIV.
Collapse
Affiliation(s)
- Milan Ganguly
- Serum Institute of India Private Limited, 212/2, Hadapsar, Pune, India.
| | - Leena Yeolekar
- Serum Institute of India Private Limited, 212/2, Hadapsar, Pune, India
| | - Parikshit Tyagi
- Serum Institute of India Private Limited, 212/2, Hadapsar, Pune, India
| | - Umesh Sagar
- Serum Institute of India Private Limited, 212/2, Hadapsar, Pune, India
| | - Swapnil Narale
- Serum Institute of India Private Limited, 212/2, Hadapsar, Pune, India
| | | | - Sham Tupe
- Serum Institute of India Private Limited, 212/2, Hadapsar, Pune, India
| | - Kuntinath Wadkar
- Serum Institute of India Private Limited, 212/2, Hadapsar, Pune, India
| | - Nilesh Ingle
- Serum Institute of India Private Limited, 212/2, Hadapsar, Pune, India
| | - Rajeev Dhere
- Serum Institute of India Private Limited, 212/2, Hadapsar, Pune, India
| | | | | |
Collapse
|
19
|
Lewis KDC, Ortiz JR, Rahman MZ, Levine MZ, Rudenko L, Wright PF, Katz JM, Dally L, Rahman M, Isakova-Sivak I, Ilyushina NA, Matyushenko V, Fry AM, Lindstrom SE, Bresee JS, Brooks WA, Neuzil KM. Immunogenicity and Viral Shedding of Russian-Backbone, Seasonal, Trivalent, Live, Attenuated Influenza Vaccine in a Phase II, Randomized, Placebo-Controlled Trial Among Preschool-Aged Children in Urban Bangladesh. Clin Infect Dis 2020; 69:777-785. [PMID: 30481272 PMCID: PMC6695509 DOI: 10.1093/cid/ciy1003] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 11/23/2018] [Indexed: 12/03/2022] Open
Abstract
Background We evaluated a Russian-backbone, live, attenuated influenza vaccine (LAIV) for immunogenicity and viral shedding in a randomized, placebo-controlled trial among Bangladeshi children. Methods Healthy children received a single, intranasal dose of LAIV containing the 2011–2012 recommended formulation or placebo. Nasopharyngeal wash (NPW) specimens were collected on days 0, 2, 4, and 7. Reverse transcription polymerase chain reactions and sequencing identified the influenza virus (vaccine or wild-type). On days 0 and 21, blood specimens were collected to assess immunogenicity using hemagglutination inhibition, microneutralization, and immunoglobulin A (IgA) and G enzyme-linked immunosorbent assays (ELISAs); NPW specimens were also collected to assess mucosal immunogenicity using kinetic IgA ELISA. Results We enrolled 300 children aged 24 through 59 months in the immunogenicity and viral shedding analyses. Among children receiving LAIV, 45% and 67% shed A/H3N2 and B vaccine strains, respectively. No child shed A/H1N1 vaccine strain. There were significantly higher day 21 geometric mean titers (GMTs) for the LAIV, as compared to the placebo groups, in all immunoassays for A/H3N2 and B (log10 titer P < .0001; GMT Ratio >2.0). Among immunoassays for A/H1N1, only the mucosal IgA GMT was significantly higher than placebo at day 21 (log10 titer P = .0465). Conclusions Children vaccinated with LAIV had serum and mucosal antibody responses to A/H3N2 and B, but only a mucosal IgA response to A/H1N1. Many children shed A/H3N2 and B vaccine strains, but none shed A/H1N1. More research is needed to determine the reason for decreased LAIV A/H1N1 immunogenicity and virus shedding. Clinical Trials Registration NCT01625689.
Collapse
Affiliation(s)
| | - Justin R Ortiz
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore
| | - Mohammed Z Rahman
- Infectious Diseases Division, International Centre for Diarrheal Disease Research, Bangladesh, Dhaka
| | - Min Z Levine
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Larisa Rudenko
- Department of Virology, Institute of Experimental Medicine, St Petersburg, Russia
| | | | - Jacqueline M Katz
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia
| | | | - Mustafizur Rahman
- Infectious Diseases Division, International Centre for Diarrheal Disease Research, Bangladesh, Dhaka
| | - Irina Isakova-Sivak
- Department of Virology, Institute of Experimental Medicine, St Petersburg, Russia
| | - Natalia A Ilyushina
- Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring
| | - Victoria Matyushenko
- Department of Virology, Institute of Experimental Medicine, St Petersburg, Russia
| | - Alicia M Fry
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Stephen E Lindstrom
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Joseph S Bresee
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia
| | | | - Kathleen M Neuzil
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore
| |
Collapse
|
20
|
Emerging Role of Mucosal Vaccine in Preventing Infection with Avian Influenza A Viruses. Viruses 2020; 12:v12080862. [PMID: 32784697 PMCID: PMC7472103 DOI: 10.3390/v12080862] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 08/05/2020] [Accepted: 08/05/2020] [Indexed: 12/13/2022] Open
Abstract
Avian influenza A viruses (AIVs), as a zoonotic agent, dramatically impacts public health and the poultry industry. Although low pathogenic avian influenza virus (LPAIV) incidence and mortality are relatively low, the infected hosts can act as a virus carrier and provide a resource pool for reassortant influenza viruses. At present, vaccination is the most effective way to eradicate AIVs from commercial poultry. The inactivated vaccines can only stimulate humoral immunity, rather than cellular and mucosal immune responses, while failing to effectively inhibit the replication and spread of AIVs in the flock. In recent years, significant progresses have been made in the understanding of the mechanisms underlying the vaccine antigen activities at the mucosal surfaces and the development of safe and efficacious mucosal vaccines that mimic the natural infection route and cut off the AIVs infection route. Here, we discussed the current status and advancement on mucosal immunity, the means of establishing mucosal immunity, and finally a perspective for design of AIVs mucosal vaccines. Hopefully, this review will help to not only understand and predict AIVs infection characteristics in birds but also extrapolate them for distinction or applicability in mammals, including humans.
Collapse
|
21
|
Immunogenicity and efficacy comparison of MDCK cell-based and egg-based live attenuated influenza vaccines of H5 and H7 subtypes in ferrets. Vaccine 2020; 38:6280-6290. [PMID: 32741671 DOI: 10.1016/j.vaccine.2020.07.043] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 06/30/2020] [Accepted: 07/20/2020] [Indexed: 11/22/2022]
Abstract
During a pandemic, the availability of specific pathogen free chicken eggs is a major bottleneck for up-scaling response to the demand for influenza vaccine. This has led us to explore the use of Madin-Darby Canine Kidney (MDCK) cells for the manufacture of live attenuated influenza vaccine (LAIV) that provides production flexibility and speed. The present study reports the comparison of the immunogenicity and efficacy of two MDCK-based LAIVs against two egg-based LAIVs prepared from the same pandemic potential strains of H5 and H7 subtypes after a single dose of the vaccine followed by a challenge with a homologous wild type strain. The vaccine strains have been generated by classical method of reassortment using the A/Leningrad/134/17/57 master donor strain. Additionally, a prime-boost regimen of the MDCK-based vaccine followed by a challenge with a homologous wild type strain for H5 and H7 immunized ferrets and also a heterologous wild type strain for the H5 immunized animals was studied. No difference in the hemagglutination inhibition and virus neutralization antibody titers against the homologous virus was observed following a single dose of either egg-based or MDCK-based H5 and H7 LAIV vaccine. A second dose of MDCK-based vaccine significantly boosted antibody titers in the vaccinated animals. Both a single dose or two doses of LAIV provided complete protection from lower respiratory tract infection and resulted in a significant reduction in the virus titers recovered from the throat, nasal turbinates and lungs after challenge with the homologous wild type strain. Protection from a challenge with a heterologous strain of H5 was also observed after two doses of the MDCK-based LAIVs. This data strongly supports the use of MDCK as a substrate for the manufacture of LAIV which ensures reliable quality, safety, production flexibility, speed and breadth of protection, features that are highly critical during a pandemic.
Collapse
|
22
|
Krutikova EV, Stepanova EA, Wong PF, Kiseleva IV, Grigor'eva EP, Rudenko LG. Genetic Basis of Attenuation of Cold-Adapted Influenza Strain B/Leningrad/14/17/55 - Backup Master Donor Virus for Influenza Type B Live Attenuated Vaccines. Bull Exp Biol Med 2020; 168:669-672. [PMID: 32248454 DOI: 10.1007/s10517-020-04777-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Indexed: 10/24/2022]
Abstract
The reassortant vaccine strain of live attenuated influenza vaccine inherits temperature sensitivity and areactogenicity from cold-adapted attenuated master donor virus. In Russia, B/ USSR/60/69 master donor virus (B60) is currently in use for the preparation of live attenuated type B influenza vaccine candidates. Trivalent live attenuated influenza vaccine based on A/ Leningrad/134/17/57 and B60 are licensed for the use in Russia for single dose vaccination of adults and children over 3 years. B/Leningrad/14/17/55 (B14) cold-adapted virus is a backup master donor virus for live attenuated type B influenza vaccine. According to our preliminary estimates, it is more attenuated than B60, which can allow expanding applicability of this vaccine for children under 3 years of age. In this paper, the role of B14 genes in its attenuation was assessed. Representative collection of reassortants of B14 with epidemic influenza B viruses was obtained, a phenotypic analysis of reassortants was performed, and their pathogenicity for animals was assessed. The leading role of PB2 and PA genes in attenuation of B14 master donor virus was proven.
Collapse
Affiliation(s)
- E V Krutikova
- A. A. Smorodintsev Department of Virology, Institute of Experimental Medicine, St. Petersburg, Russia.
| | - E A Stepanova
- A. A. Smorodintsev Department of Virology, Institute of Experimental Medicine, St. Petersburg, Russia
| | - P F Wong
- A. A. Smorodintsev Department of Virology, Institute of Experimental Medicine, St. Petersburg, Russia
| | - I V Kiseleva
- A. A. Smorodintsev Department of Virology, Institute of Experimental Medicine, St. Petersburg, Russia
| | - E P Grigor'eva
- A. A. Smorodintsev Department of Virology, Institute of Experimental Medicine, St. Petersburg, Russia
| | - L G Rudenko
- A. A. Smorodintsev Department of Virology, Institute of Experimental Medicine, St. Petersburg, Russia
| |
Collapse
|
23
|
Affiliation(s)
- John Treanor
- Department of Medicine, University of Rochester School of Medicine and Dentistry
| |
Collapse
|
24
|
Isakova-Sivak I, Grigorieva E, Rudenko L. Insights into current clinical research on the immunogenicity of live attenuated influenza vaccines. Expert Rev Vaccines 2020; 19:43-55. [PMID: 31903816 DOI: 10.1080/14760584.2020.1711056] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Introduction: Live attenuated influenza vaccines (LAIVs) have been in use for more than three decades and are now licensed in many countries. There is evidence that LAIVs can have greater efficacy than inactivated influenza vaccines, especially against mismatched influenza, however, in recent years, a number of trials have found a lack of LAIV efficacy, mainly in relation to the H1N1 virus.Areas covered: In this review, we summarize the results of clinical research published in the past 5 years on the immunogenicity of LAIVs, with special attention to the mechanisms of establishing protective immunity and some factors that may influence immunogenicity and efficacy.Expert opinion: A number of recent clinical studies confirmed that the immune responses to LAIVs are multifaceted, involving different immune mechanisms. These trials suggest that the intrinsic replicative properties of each LAIV component should be taken into account, and the precise effects of adding a fourth vaccine strain to trivalent LAIV formulations are still to be identified. In addition, new data are emerging regarding the impact of pre-vaccination conditions, such as preexisting immunity or concurrent asymptomatic viral and bacterial respiratory infections, on LAIV immunogenicity, suggesting the importance of monitoring them during clinical trials or vaccination campaigns.
Collapse
Affiliation(s)
- Irina Isakova-Sivak
- Department of Virology, Institute of Experimental Medicine, St. Petersburg, Russia
| | - Elena Grigorieva
- Department of Virology, Institute of Experimental Medicine, St. Petersburg, Russia
| | - Larisa Rudenko
- Department of Virology, Institute of Experimental Medicine, St. Petersburg, Russia
| |
Collapse
|
25
|
Biswas A, Chakrabarti AK, Dutta S. Current challenges: from the path of “original antigenic sin” towards the development of universal flu vaccines. Int Rev Immunol 2019; 39:21-36. [DOI: 10.1080/08830185.2019.1685990] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Asim Biswas
- Virology, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, India
| | - Alok K. Chakrabarti
- Virology, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, India
| | - Shanta Dutta
- Bacteriology, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, India
| |
Collapse
|
26
|
Comparative Study of the Temperature Sensitive, Cold Adapted and Attenuated Mutations Present in the Master Donor Viruses of the Two Commercial Human Live Attenuated Influenza Vaccines. Viruses 2019; 11:v11100928. [PMID: 31658679 PMCID: PMC6832241 DOI: 10.3390/v11100928] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 10/03/2019] [Accepted: 10/08/2019] [Indexed: 12/28/2022] Open
Abstract
Influenza viruses cause annual, seasonal infection across the globe. Vaccination represents the most effective strategy to prevent such infections and/or to reduce viral disease. Two major types of influenza vaccines are approved for human use: inactivated influenza vaccines (IIVs) and live attenuated influenza vaccines (LAIVs). Two Master Donor Virus (MDV) backbones have been used to create LAIVs against influenza A virus (IAV): the United States (US) A/Ann Arbor/6/60 (AA) and the Russian A/Leningrad/134/17/57 (Len) H2N2 viruses. The mutations responsible for the temperature sensitive (ts), cold-adapted (ca) and attenuated (att) phenotypes of the two MDVs have been previously identified and genetically mapped. However, a direct comparison of the contribution of these residues to viral attenuation, immunogenicity and protection efficacy has not been conducted. Here, we compared the In vitro and in vivo phenotype of recombinant influenza A/Puerto Rico/8/34 H1N1 (PR8) viruses containing the ts, ca and att mutations of the US (PR8/AA) and the Russian (PR8/Len) MDVs. Our results show that PR8/Len is more attenuated in vivo than PR8/AA, although both viruses induced similar levels of humoral and cellular responses, and protection against homologous and heterologous viral challenges. Our findings support the feasibility of using a different virus backbone as MDV for the development of improved LAIVs for the prevention of IAV infections.
Collapse
|
27
|
Gianchecchi E, Manenti A, Kistner O, Trombetta C, Manini I, Montomoli E. How to assess the effectiveness of nasal influenza vaccines? Role and measurement of sIgA in mucosal secretions. Influenza Other Respir Viruses 2019; 13:429-437. [PMID: 31225704 PMCID: PMC6692539 DOI: 10.1111/irv.12664] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 05/27/2019] [Accepted: 05/29/2019] [Indexed: 01/07/2023] Open
Abstract
Secretory IgAs (sIgA) constitute the principal isotype of antibodies present in nasal and mucosal secretions. They are secreted by plasma cells adjacent to the mucosal epithelial cells, the site where infection occurs, and are the main humoral mediator of mucosal immunity. Mucosally delivered vaccines, such as live attenuated influenza vaccine (LAIV), are able to mimic natural infection without causing disease or virus transmission and mainly elicit a local immune response. The measurement of sIgA concentrations in nasal swab/wash and saliva samples is therefore a valuable tool for evaluating their role in the effectiveness of such vaccines. Here, we describe two standardized assays (enzyme‐linked immunosorbent assay and microneutralization) available for the quantification of sIgA and discuss the advantages and limitations of their use.
Collapse
Affiliation(s)
| | | | | | - Claudia Trombetta
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Ilaria Manini
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Emanuele Montomoli
- VisMederi Srl, Siena, Italy.,VisMederi Research Srl, Siena, Italy.,Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| |
Collapse
|
28
|
Shcherbik S, Pearce N, Carney P, Bazhenova E, Larionova N, Kiseleva I, Rudenko L, Kumar A, Goldsmith CS, Dugan V, Stevens J, Wentworth DE, Bousse T. Evaluation of A(H1N1)pdm09 LAIV vaccine candidates stability and replication efficiency in primary human nasal epithelial cells. Vaccine X 2019; 2:100031. [PMID: 31384746 PMCID: PMC6668239 DOI: 10.1016/j.jvacx.2019.100031] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 06/11/2019] [Accepted: 06/17/2019] [Indexed: 01/06/2023] Open
Abstract
The recent reduction of live attenuated influenza vaccine (LAIV) effectiveness in multivalent formulations was particularly associated with the A(H1N1)pdm09 component. In the 2017 the WHO vaccine composition committee changed its recommendations for the A(H1N1)pdm09 component to include an A/Michigan/45/2015-like virus. We evaluated effectiveness and quality of newly developed and previous A(H1N1)pdm09 LAIV reassortants through assessment of their thermal and pH stability, receptor binding specificity and replication fitness in primary human airway epithelial cells of nasal origin (hAECN). Our analysis showed that LAIV expressed hemagglutinin (HA) and neuraminidase (NA) from an A/Michigan/45/2015-like strain A/New York/61/2015 (A/New York/61/2015-CDC-LV16A, NY-LV16A), exhibit higher thermal and pH stability compared to the previous vaccine candidates expressing HA and NA from A/California/07/2009 and A/Bolivia/559/2013 (A17/Cal09 and A17/Bol13). Reassortants A/South Africa/3626/2013-CDC-LV14A (SA-LV14A) and NY-LV16A showed preferential binding to α2,6 sialic acid (SA) receptors and replicated at higher titers and more extensively in hAECN compared to A17/Cal09 and A17/Bol13, which had an α2,3 SA receptor binding preference. Our data analysis supports selection of A/New York/61/2015-CDC-LV16A for LAIV formulation and the introduction of new assays for LAIV characterization.
Collapse
Affiliation(s)
- Svetlana Shcherbik
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road, Atlanta, GA 30333, United States
| | - Nicholas Pearce
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road, Atlanta, GA 30333, United States
- Battelle, Atlanta, GA 30329, United States
| | - Paul Carney
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road, Atlanta, GA 30333, United States
| | - Ekaterina Bazhenova
- Institute of Experimental Medicine, Department of Virology, St. Petersburg, Russia
| | - Natalie Larionova
- Institute of Experimental Medicine, Department of Virology, St. Petersburg, Russia
| | - Irina Kiseleva
- Institute of Experimental Medicine, Department of Virology, St. Petersburg, Russia
| | - Larisa Rudenko
- Institute of Experimental Medicine, Department of Virology, St. Petersburg, Russia
| | - Amrita Kumar
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road, Atlanta, GA 30333, United States
- Battelle, Atlanta, GA 30329, United States
| | - Cynthia S. Goldsmith
- Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road, Atlanta, GA 30333, United States
| | - Vivien Dugan
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road, Atlanta, GA 30333, United States
| | - James Stevens
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road, Atlanta, GA 30333, United States
| | - David E. Wentworth
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road, Atlanta, GA 30333, United States
| | - Tatiana Bousse
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road, Atlanta, GA 30333, United States
- Corresponding author.
| |
Collapse
|
29
|
Pan SC, Hsieh SM, Lin CF, Hsu YS, Chang M, Chang SC. A randomized, double-blind, controlled clinical trial to evaluate the safety and immunogenicity of an intranasally administered trivalent inactivated influenza vaccine with adjuvant LTh(αK): A phase I study. Vaccine 2019; 37:1994-2003. [DOI: 10.1016/j.vaccine.2019.02.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 02/01/2019] [Accepted: 02/08/2019] [Indexed: 11/27/2022]
|
30
|
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: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 11/07/2018] [Indexed: 01/04/2023]
|
31
|
Rudenko L, Kiseleva I, Krutikova E, Stepanova E, Rekstin A, Donina S, Pisareva M, Grigorieva E, Kryshen K, Muzhikyan A, Makarova M, Sparrow EG, Torelli G, Kieny MP. Rationale for vaccination with trivalent or quadrivalent live attenuated influenza vaccines: Protective vaccine efficacy in the ferret model. PLoS One 2018; 13:e0208028. [PMID: 30507951 PMCID: PMC6277076 DOI: 10.1371/journal.pone.0208028] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 11/10/2018] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND AND AIM The majority of seasonal influenza vaccines are trivalent, containing two A virus strains (H1N1 and H3N2) and one B virus strain. The co-circulation of two distinct lineages of B viruses can lead to mismatch between the influenza B virus strain recommended for the trivalent seasonal vaccine and the circulating B virus. This has led some manufacturers to produce quadrivalent influenza vaccines containing one strain from each B lineage in addition to H1N1 and H3N2 strains. However, it is also important to know whether vaccines containing a single influenza B strain can provide cross-protectivity against viruses of the antigenically distinct lineage. The aim of this study was to assess in naïve ferrets the potential cross-protective activity of trivalent live attenuated influenza vaccine (T-LAIV) against challenge with a heterologous wild-type influenza B virus belonging to the genetically different lineage and to compare this activity with effectiveness of quadrivalent LAIV (Q-LAIV) in the ferret model. METHODS AND RESULTS Ferrets were vaccinated with either one dose of trivalent LAIV containing B/Victoria or B/Yamagata lineage virus, or quadrivalent LAIV (containing both B lineages), or placebo. They were then challenged with B/Victoria or B/Yamagata lineage wild-type virus 28 days after vaccination. The ferrets were monitored for clinical signs and morbidity. Nasal swabs and lung tissue samples were analyzed for the presence of challenge virus. Antibody response to vaccination was assessed by routine hemagglutination inhibition assay. All LAIVs tested were found to be safe and effective against wild-type influenza B viruses based on clinical signs, and virological and histological data. The absence of interference between vaccine strains in trivalent and quadrivalent vaccine formulations was confirmed. Trivalent LAIVs were shown to have the potential to be cross-protective against infection with genetically different influenza B/Victoria and B/Yamagata lineages. CONCLUSIONS In this ferret model, quadrivalent vaccine provided higher protection to challenge against both B/Victoria and B/Yamagata lineage viruses. However, T-LAIV provided some cross-protection in the case of a mismatch between circulating and vaccine type B strains. Notably, B/Victoria-based T-LAIV was more protective compared to B/Yamagata-based T-LAIV.
Collapse
MESH Headings
- Administration, Intranasal
- Animals
- Antibodies, Viral/blood
- Cross Protection/genetics
- Cross Protection/immunology
- Disease Models, Animal
- Female
- Ferrets
- Humans
- Immunogenicity, Vaccine
- Influenza A Virus, H1N1 Subtype/genetics
- Influenza A Virus, H1N1 Subtype/immunology
- Influenza A Virus, H1N1 Subtype/pathogenicity
- Influenza A Virus, H3N2 Subtype/genetics
- Influenza A Virus, H3N2 Subtype/immunology
- Influenza A Virus, H3N2 Subtype/pathogenicity
- Influenza B virus/genetics
- Influenza B virus/immunology
- Influenza B virus/pathogenicity
- Influenza Vaccines/administration & dosage
- Influenza Vaccines/immunology
- Influenza, Human/blood
- Influenza, Human/immunology
- Influenza, Human/prevention & control
- Influenza, Human/virology
- Vaccination/methods
- Vaccines, Attenuated/administration & dosage
- Vaccines, Attenuated/immunology
Collapse
Affiliation(s)
- Larisa Rudenko
- Department of Virology, Institute of Experimental Medicine, St Petersburg, Russia
| | - Irina Kiseleva
- Department of Virology, Institute of Experimental Medicine, St Petersburg, Russia
| | - Elena Krutikova
- Department of Virology, Institute of Experimental Medicine, St Petersburg, Russia
| | - Ekaterina Stepanova
- Department of Virology, Institute of Experimental Medicine, St Petersburg, Russia
| | - Andrey Rekstin
- Department of Virology, Institute of Experimental Medicine, St Petersburg, Russia
| | - Svetlana Donina
- Department of Virology, Institute of Experimental Medicine, St Petersburg, Russia
| | - Maria Pisareva
- Department of Virology, Institute of Experimental Medicine, St Petersburg, Russia
| | - Elena Grigorieva
- Department of Virology, Institute of Experimental Medicine, St Petersburg, Russia
| | - Kirill Kryshen
- Department of Toxicology and Microbiology, Institute of Preclinical Research Ltd, St Petersburg, Russia
| | - Arman Muzhikyan
- Department of Toxicology and Microbiology, Institute of Preclinical Research Ltd, St Petersburg, Russia
| | - Marina Makarova
- Department of Toxicology and Microbiology, Institute of Preclinical Research Ltd, St Petersburg, Russia
| | - Erin Grace Sparrow
- Universal Health Coverage and Health Systems, World Health Organization, Geneva, Switzerland
| | - Guido Torelli
- Universal Health Coverage and Health Systems, World Health Organization, Geneva, Switzerland
| | - Marie-Paule Kieny
- International Institutional Cooperation, Institut national de la santé et de la recherche médicale (INSERM), Paris, France
| |
Collapse
|
32
|
Rudenko L, Kiseleva I, Krutikova E, Stepanova E, Isakova-Sivak I, Donina S, Rekstin A, Pisareva M, Bazhenova E, Kotomina T, Katelnikova A, Muzhikyan A, Makarov V, Sparrow EG, Torelli G. Two Live Attenuated Vaccines against Recent Low⁻and Highly Pathogenic H7N9 Influenza Viruses Are Safe and Immunogenic in Ferrets. Vaccines (Basel) 2018; 6:vaccines6040074. [PMID: 30388790 PMCID: PMC6313887 DOI: 10.3390/vaccines6040074] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 09/21/2018] [Accepted: 09/27/2018] [Indexed: 12/11/2022] Open
Abstract
Influenza H7N9 virus is a potentially pandemic subtype to which most people are immunologically naïve. To be better prepared for the potential occurrence of an H7N9 pandemic, in 2017 the World Health Organization recommended developing candidate vaccine viruses from two new H7N9 viruses, A/Guangdong/17SF003/2016 (A/GD) and A/Hong Kong/125/2017 (A/HK). This report describes the development of live attenuated influenza vaccine (LAIV) candidates against A/GD and A/HK viruses and study of their safety and immunogenicity in the ferret model in order to choose the most promising one for a phase I clinical trial. The A/HK-based vaccine candidate (A/17/HK) was developed by classical reassortment in eggs. The A/GD-based vaccine candidate (A/17/GD) was generated by reverse genetics. Ferrets were vaccinated with two doses of LAIV or phosphate-buffered saline. Both H7N9 LAIVs tested were safe for ferrets, as shown by absence of clinical signs, and by virological and histological data; they were immunogenic after a single vaccination. These results provide a compelling argument for further testing of these vaccines in volunteers. Since the A/HK virus represents the cluster that has caused the majority of human cases, and because the A/HK-based LAIV candidate was developed by classical reassortment, this is the preferred candidate for a phase I clinical trial.
Collapse
Affiliation(s)
- Larisa Rudenko
- Institute of Experimental Medicine, St. Petersburg 197376, Russia.
| | - Irina Kiseleva
- Institute of Experimental Medicine, St. Petersburg 197376, Russia.
| | - Elena Krutikova
- Institute of Experimental Medicine, St. Petersburg 197376, Russia.
| | | | | | - Svetlana Donina
- Institute of Experimental Medicine, St. Petersburg 197376, Russia.
| | - Andrey Rekstin
- Institute of Experimental Medicine, St. Petersburg 197376, Russia.
| | - Maria Pisareva
- Institute of Experimental Medicine, St. Petersburg 197376, Russia.
| | | | - Tatiana Kotomina
- Institute of Experimental Medicine, St. Petersburg 197376, Russia.
| | | | - Arman Muzhikyan
- Institute of Preclinical Research Ltd., St. Petersburg 188663, Russia.
| | - Valery Makarov
- Institute of Preclinical Research Ltd., St. Petersburg 188663, Russia.
| | | | - Guido Torelli
- World Health Organization, 1211 Geneva, Switzerland.
| |
Collapse
|
33
|
Nigwekar PV, Kumar A, Padbidri VV, Choudhury A, Chaudhari AB, Kulkarni PS. Safety of Russian-Backbone Trivalent, Live Attenuated Seasonal Influenza Vaccine in Healthy Subjects: Open-Label, Non-randomized Phase 4 Study. Drug Saf 2018; 41:171-177. [PMID: 29027148 DOI: 10.1007/s40264-017-0605-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
INTRODUCTION AND AIM A trivalent live attenuated influenza vaccine (Nasovac-S®) was developed and licensed in India. A phase 4 study was conducted to assess safety. METHODOLOGY This non-randomized, open-label, single-arm study among individuals ≥ 2 years of age involved administration of 0.5 mL of Nasovac-S intranasally, with a 1-month follow-up after vaccination. Adverse events (AEs) were collected via structured diaries. RESULTS Among 500 vaccinated subjects, 160 were between 2 and 17 years of age, 240 were 18-49 years old and 100 were 50 years and older. A total of 533 solicited reactions were reported. The majority of these reactions were mild, and almost all of them resolved without any sequelae. A total of 20% of subjects reported at least one local solicited reaction, and 23% reported at least one systemic solicited reaction. None of the 45 unsolicited AEs reported by 37 subjects (7.4%) were causally related to the study vaccine. CONCLUSIONS The data from the study adds to the existing safety database of Nasovac-S. REGISTRY Clinical Trials Registry of India (CTRI/2015/08/006074).
Collapse
Affiliation(s)
| | - Anuj Kumar
- Pravara Institute of Medical Sciences, Loni, India
| | | | | | - Amol B Chaudhari
- Serum Institute of India Pvt. Ltd, 212/2, Hadapsar, Pune, India.
| | | |
Collapse
|
34
|
The Future of Influenza Vaccines: A Historical and Clinical Perspective. Vaccines (Basel) 2018; 6:vaccines6030058. [PMID: 30200179 PMCID: PMC6160951 DOI: 10.3390/vaccines6030058] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Revised: 08/21/2018] [Accepted: 08/27/2018] [Indexed: 12/16/2022] Open
Abstract
For centuries, the development of vaccines to prevent infectious disease was an empirical process. From smallpox variolation in Song dynasty China, through the polysaccharide capsule vaccines developed in the 1970s, vaccines were made either from the pathogen itself, treated in some way to render it attenuated or non-infectious, or from a closely related non-pathogenic strain. In recent decades, new scientific knowledge and technologies have enabled rational vaccine design in a way that was unimaginable before. However, vaccines optimal against some infectious diseases, influenza among them, have remained elusive. This review will highlight the challenges that influenza viruses pose for rational vaccine design. In particular, it will consider the clinically beneficial endpoints, beyond complete sterilizing immunity, that have been achieved with vaccines against other infectious diseases, as well as the barriers to achieving similar success against influenza.
Collapse
|
35
|
Shcherbik S, Carney P, Pearce N, Stevens J, Dugan VG, Wentworth DE, Bousse T. Monoclonal antibody against N2 neuraminidase of cold adapted A/Leningrad/134/17/57 (H2N2) enables efficient generation of live attenuated influenza vaccines. Virology 2018; 522:65-72. [PMID: 30014859 DOI: 10.1016/j.virol.2018.07.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 07/05/2018] [Accepted: 07/05/2018] [Indexed: 01/13/2023]
Abstract
Cold adapted influenza virus A/Leningrad/134/17/57 (H2N2) is a reliable master donor virus (Len/17-MDV) for preparing live attenuated influenza vaccines (LAIV). LAIVs are 6:2 reasortants that contain 6 segments of Len/17-MDV and the hemagglutinin (HA) and neuraminidase (NA) of contemporary circulating influenza A viruses. The problem with the classical reassortment procedure used to generate LAIVs is that there is limited selection pressure against NA of the Len/17-MDV resulting in 7:1 reassortants with desired HA only, which are not suitable LAIVs. The monoclonal antibodies (mAb) directed against the N2 of Len/17-MDV were generated. 10C4-8E7 mAb inhibits cell-to-cell spread of viruses containing the Len/17-MDV N2, but not viruses with the related N2 from contemporary H3N2 viruses. 10C4-8E7 antibody specifically inhibited the Len/17-MDV replication in vitro and in ovo but didn't inhibit replication of H3N2 or H1N1pdm09 reassortants. Our data demonstrate that addition of 10C4-8E7 in the classical reassortment improves efficiency of LAIV production.
Collapse
Affiliation(s)
- Svetlana Shcherbik
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road, Atlanta, GA 30333, United States
| | - Paul Carney
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road, Atlanta, GA 30333, United States
| | | | - James Stevens
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road, Atlanta, GA 30333, United States
| | - Vivien G Dugan
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road, Atlanta, GA 30333, United States
| | - David E Wentworth
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road, Atlanta, GA 30333, United States
| | - Tatiana Bousse
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road, Atlanta, GA 30333, United States.
| |
Collapse
|
36
|
Isakova-Sivak I, Korenkov D, Smolonogina T, Kotomina T, Donina S, Matyushenko V, Mezhenskaya D, Krammer F, Rudenko L. Broadly protective anti-hemagglutinin stalk antibodies induced by live attenuated influenza vaccine expressing chimeric hemagglutinin. Virology 2018; 518:313-323. [PMID: 29574336 DOI: 10.1016/j.virol.2018.03.013] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 03/12/2018] [Accepted: 03/15/2018] [Indexed: 12/31/2022]
Abstract
The development of influenza vaccines that can provide broad protection against all drifted seasonal virus variants, zoonotic infections and emerging pandemic strains, has been a priority for two decades. Here we propose a strategy of inducing broadly-reactive anti-stalk antibody by sequential immunizations with live attenuated influenza vaccines (LAIVs) expressing chimeric HAs (cHAs). These vaccines are designed to contain identical hemagglutinin stalk domains from H1N1 virus but antigenically unrelated globular head domains from avian influenza virus subtypes H5, H8 and H9. Mouse experiments demonstrated enhanced cross-protection of cHA-containing LAIVs compared to the relevant vaccine viruses expressing natural HAs, and this enhanced protection was driven by stalk-HA-reactive IgG antibodies. The establishment of fully functional cross-protective immunity after two doses of cHA LAIV vaccination in naïve animals suggests that a similar effect might be expected after a single cHA LAIV dose in primed individuals, or after two to three doses in naïve children.
Collapse
Affiliation(s)
- Irina Isakova-Sivak
- Department of Virology, Institute of Experimental Medicine, 12 Acad. Pavlov Street, St Petersburg, Russia.
| | - Daniil Korenkov
- Department of Virology, Institute of Experimental Medicine, 12 Acad. Pavlov Street, St Petersburg, Russia
| | - Tatiana Smolonogina
- Department of Virology, Institute of Experimental Medicine, 12 Acad. Pavlov Street, St Petersburg, Russia
| | - Tatiana Kotomina
- Department of Virology, Institute of Experimental Medicine, 12 Acad. Pavlov Street, St Petersburg, Russia
| | - Svetlana Donina
- Department of Virology, Institute of Experimental Medicine, 12 Acad. Pavlov Street, St Petersburg, Russia
| | - Victoria Matyushenko
- Department of Virology, Institute of Experimental Medicine, 12 Acad. Pavlov Street, St Petersburg, Russia
| | - Daria Mezhenskaya
- Department of Virology, Institute of Experimental Medicine, 12 Acad. Pavlov Street, St Petersburg, Russia
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Larisa Rudenko
- Department of Virology, Institute of Experimental Medicine, 12 Acad. Pavlov Street, St Petersburg, Russia
| |
Collapse
|
37
|
Rajão DS, Pérez DR. Universal Vaccines and Vaccine Platforms to Protect against Influenza Viruses in Humans and Agriculture. Front Microbiol 2018; 9:123. [PMID: 29467737 PMCID: PMC5808216 DOI: 10.3389/fmicb.2018.00123] [Citation(s) in RCA: 108] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 01/18/2018] [Indexed: 01/22/2023] Open
Abstract
Influenza virus infections pose a significant threat to public health due to annual seasonal epidemics and occasional pandemics. Influenza is also associated with significant economic losses in animal production. The most effective way to prevent influenza infections is through vaccination. Current vaccine programs rely heavily on the vaccine's ability to stimulate neutralizing antibody responses to the hemagglutinin (HA) protein. One of the biggest challenges to an effective vaccination program lies on the fact that influenza viruses are ever-changing, leading to antigenic drift that results in escape from earlier immune responses. Efforts toward overcoming these challenges aim at improving the strength and/or breadth of the immune response. Novel vaccine technologies, the so-called universal vaccines, focus on stimulating better cross-protection against many or all influenza strains. However, vaccine platforms or manufacturing technologies being tested to improve vaccine efficacy are heterogeneous between different species and/or either tailored for epidemic or pandemic influenza. Here, we discuss current vaccines to protect humans and animals against influenza, highlighting challenges faced to effective and uniform novel vaccination strategies and approaches.
Collapse
Affiliation(s)
- Daniela S. Rajão
- Department of Population Health, University of Georgia, Athens, GA, United States
| | | |
Collapse
|
38
|
Krammer F, Smith GJD, Fouchier RAM, Peiris M, Kedzierska K, Doherty PC, Palese P, Shaw ML, Treanor J, Webster RG, García-Sastre A. Influenza. Nat Rev Dis Primers 2018; 4:3. [PMID: 29955068 PMCID: PMC7097467 DOI: 10.1038/s41572-018-0002-y] [Citation(s) in RCA: 857] [Impact Index Per Article: 142.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Influenza is an infectious respiratory disease that, in humans, is caused by influenza A and influenza B viruses. Typically characterized by annual seasonal epidemics, sporadic pandemic outbreaks involve influenza A virus strains of zoonotic origin. The WHO estimates that annual epidemics of influenza result in ~1 billion infections, 3–5 million cases of severe illness and 300,000–500,000 deaths. The severity of pandemic influenza depends on multiple factors, including the virulence of the pandemic virus strain and the level of pre-existing immunity. The most severe influenza pandemic, in 1918, resulted in >40 million deaths worldwide. Influenza vaccines are formulated every year to match the circulating strains, as they evolve antigenically owing to antigenic drift. Nevertheless, vaccine efficacy is not optimal and is dramatically low in the case of an antigenic mismatch between the vaccine and the circulating virus strain. Antiviral agents that target the influenza virus enzyme neuraminidase have been developed for prophylaxis and therapy. However, the use of these antivirals is still limited. Emerging approaches to combat influenza include the development of universal influenza virus vaccines that provide protection against antigenically distant influenza viruses, but these vaccines need to be tested in clinical trials to ascertain their effectiveness.
Collapse
Affiliation(s)
- Florian Krammer
- 0000 0001 0670 2351grid.59734.3cDepartment of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Gavin J. D. Smith
- 0000 0001 2180 6431grid.4280.eDuke–NUS Medical School, Singapore, Singapore ,0000 0004 1936 7961grid.26009.3dDuke Global Health Institute, Duke University, Durham, NC USA
| | - Ron A. M. Fouchier
- 000000040459992Xgrid.5645.2Department of Viroscience, Erasmus MC, Rotterdam, Netherlands
| | - Malik Peiris
- 0000000121742757grid.194645.bWHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, Hong Kong, China ,0000000121742757grid.194645.bCenter of Influenza Research, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, Hong Kong, China
| | - Katherine Kedzierska
- 0000 0001 2179 088Xgrid.1008.9Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria Australia
| | - Peter C. Doherty
- 0000 0001 2179 088Xgrid.1008.9Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria Australia ,0000 0001 0224 711Xgrid.240871.8Department of Immunology, St Jude Children’s Research Hospital, Memphis, TN USA
| | - Peter Palese
- 0000 0001 0670 2351grid.59734.3cDepartment of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY USA ,0000 0001 0670 2351grid.59734.3cDivision of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Megan L. Shaw
- 0000 0001 0670 2351grid.59734.3cDepartment of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - John Treanor
- 0000 0004 1936 9166grid.412750.5Division of Infectious Diseases, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY USA
| | - Robert G. Webster
- 0000 0001 0224 711Xgrid.240871.8Department of Infectious Diseases, St Jude Children’s Research Hospital, Memphis, TN USA
| | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA. .,Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA. .,Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
39
|
Singanayagam A, Zambon M, Lalvani A, Barclay W. Urgent challenges in implementing live attenuated influenza vaccine. THE LANCET. INFECTIOUS DISEASES 2017; 18:e25-e32. [PMID: 28780285 DOI: 10.1016/s1473-3099(17)30360-2] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Revised: 05/10/2017] [Accepted: 05/25/2017] [Indexed: 12/26/2022]
Abstract
Conflicting reports have emerged about the effectiveness of the live attenuated influenza vaccine. The live attenuated influenza vaccine appears to protect particularly poorly against currently circulating H1N1 viruses that are derived from the 2009 pandemic H1N1 viruses. During the 2015-16 influenza season, when pandemic H1N1 was the predominant virus, studies from the USA reported a complete lack of effectiveness of the live vaccine in children. This finding led to a crucial decision in the USA to recommend that the live vaccine not be used in 2016-17 and to switch to the inactivated influenza vaccine. Other countries, including the UK, Canada, and Finland, however, have continued to recommend the use of the live vaccine. This policy divergence and uncertainty has far reaching implications for the entire global community, given the importance of the production capabilities of the live attenuated influenza vaccine for pandemic preparedness. In this Personal View, we discuss possible explanations for the observed reduced effectiveness of the live attenuated influenza vaccine and highlight the underpinning scientific questions. Further research to understand the reasons for these observations is essential to enable informed public health policy and commercial decisions about vaccine production and development in coming years.
Collapse
Affiliation(s)
- Anika Singanayagam
- Department of Medicine, Imperial College, London, UK; NIHR Health Protection Research Unit in Respiratory Infections, Imperial College, London, UK
| | - Maria Zambon
- Virus Reference Department, National Infection Service, Public Health England, Colindale, London, UK; NIHR Health Protection Research Unit in Respiratory Infections, Imperial College, London, UK
| | - Ajit Lalvani
- National Heart and Lung Institute, Imperial College, London, UK; NIHR Health Protection Research Unit in Respiratory Infections, Imperial College, London, UK
| | - Wendy Barclay
- Department of Medicine, Imperial College, London, UK; NIHR Health Protection Research Unit in Respiratory Infections, Imperial College, London, UK.
| |
Collapse
|
40
|
Development of an Alternative Modified Live Influenza B Virus Vaccine. J Virol 2017; 91:JVI.00056-17. [PMID: 28381580 PMCID: PMC5446642 DOI: 10.1128/jvi.00056-17] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 03/31/2017] [Indexed: 12/27/2022] Open
Abstract
Influenza B virus (IBV) is considered a major human pathogen, responsible for seasonal epidemics of acute respiratory illness. Two antigenically distinct IBV hemagglutinin (HA) lineages cocirculate worldwide with little cross-reactivity. Live attenuated influenza virus (LAIV) vaccines have been shown to provide better cross-protective immune responses than inactivated vaccines by eliciting local mucosal immunity and systemic B cell- and T cell-mediated memory responses. We have shown previously that incorporation of temperature-sensitive (ts) mutations into the PB1 and PB2 subunits along with a modified HA epitope tag in the C terminus of PB1 resulted in influenza A viruses (IAV) that are safe and effective as modified live attenuated (att) virus vaccines (IAV att). We explored whether analogous mutations in the IBV polymerase subunits would result in a stable virus with an att phenotype. The PB1 subunit of the influenza B/Brisbane/60/2008 strain was used to incorporate ts mutations and a C-terminal HA tag. Such modifications resulted in a B/Bris att strain with ts characteristics in vitro and an att phenotype in vivo Vaccination studies in mice showed that a single dose of the B/Bris att candidate stimulated sterilizing immunity against lethal homologous challenge and complete protection against heterologous challenge. These studies show the potential of an alternative LAIV platform for the development of IBV vaccines.IMPORTANCE A number of issues with regard to the effectiveness of the LAIV vaccine licensed in the United States (FluMist) have arisen over the past three seasons (2013-2014, 2014-2015, and 2015-2016). While the reasons for the limited robustness of the vaccine-elicited immune response remain controversial, this problem highlights the critical importance of continued investment in LAIV development and creates an opportunity to improve current strategies so as to develop more efficacious vaccines. Our laboratory has developed an alternative strategy, the incorporation of 2 amino acid mutations and a modified HA tag at the C terminus of PB1, which is sufficient to attenuate the IBV. As a LAIV, this novel vaccine provides complete protection against IBV strains. The availability of attenuated IAV and IBV backbones based on contemporary strains offers alternative platforms for the development of LAIVs that may overcome current limitations.
Collapse
|
41
|
Isakova-Sivak I, Rudenko L. Tackling a novel lethal virus: a focus on H7N9 vaccine development. Expert Rev Vaccines 2017; 16:1-13. [PMID: 28532182 DOI: 10.1080/14760584.2017.1333907] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
INTRODUCTION Avian-origin H7N9 influenza viruses first detected in humans in China in 2013 continue to cause severe human infections with a mortality rate close to 40%. These viruses are acknowledged as the subtype most likely to cause the next influenza pandemic. Areas covered: Here we review published data on the development of H7N9 influenza vaccine candidates and their evaluation in preclinical and clinical trials identified on PubMed database with the term 'H7N9 influenza vaccine'. In addition, a search with the same term was done on ClinicalTrials.gov to find ongoing clinical trials with H7N9 vaccines. Expert commentary: Influenza vaccines are the most powerful tool for protecting the human population from influenza infections, both seasonal and pandemic. During the past four years, a large number of promising H7N9 influenza vaccine candidates have been generated using traditional and advanced gene engineering techniques. In addition, with the support of WHO's GAP program, influenza vaccine production capacities have been established in a number of vulnerable low- and middle-income countries with a high population density, allowing the countries to be independent of vaccine supply from high-income countries. Overall, it is believed that the world is now well prepared for a possible H7N9 influenza pandemic.
Collapse
Affiliation(s)
- Irina Isakova-Sivak
- a Department of Virology , Institute of Experimental Medicine , Saint Petersburg , Russia
| | - Larisa Rudenko
- a Department of Virology , Institute of Experimental Medicine , Saint Petersburg , Russia
| |
Collapse
|
42
|
Use of live attenuated influenza vaccines in young children in resource-poor settings. LANCET GLOBAL HEALTH 2016; 4:e879-e880. [PMID: 27746227 DOI: 10.1016/s2214-109x(16)30247-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 09/19/2016] [Indexed: 11/23/2022]
|