1
|
Clark TW, Tregoning JS, Lister H, Poletti T, Amin F, Nguyen-Van-Tam JS. Recent advances in the influenza virus vaccine landscape: a comprehensive overview of technologies and trials. Clin Microbiol Rev 2024; 37:e0002524. [PMID: 39360831 PMCID: PMC11629632 DOI: 10.1128/cmr.00025-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/05/2024] Open
Abstract
SUMMARYIn the United Kingdom (UK) in 2022/23, influenza virus infections returned to the levels recorded before the COVID-19 pandemic, exerting a substantial burden on an already stretched National Health Service (NHS) through increased primary and emergency care visits and subsequent hospitalizations. Population groups ≤4 years and ≥65 years of age, and those with underlying health conditions, are at the greatest risk of influenza-related hospitalization. Recent advances in influenza virus vaccine technologies may help to mitigate this burden. This review aims to summarize advances in the influenza virus vaccine landscape by describing the different technologies that are currently in use in the UK and more widely. The review also describes vaccine technologies that are under development, including mRNA, and universal influenza virus vaccines which aim to provide broader or increased protection. This is an exciting and important era for influenza virus vaccinations, and advances are critical to protect against a disease that still exerts a substantial burden across all populations and disproportionately impacts the most vulnerable, despite it being over 80 years since the first influenza virus vaccines were deployed.
Collapse
Affiliation(s)
- Tristan W. Clark
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - John S. Tregoning
- Department of Infectious Disease, Imperial College London, London, United Kingdom
| | | | | | | | | |
Collapse
|
2
|
Spinardi JR, Thakkar KB, Welch VL, Jagun O, Kyaw MH. The need for novel influenza vaccines in low- and middle-income countries: A narrative review. Braz J Infect Dis 2024; 29:104465. [PMID: 39642677 DOI: 10.1016/j.bjid.2024.104465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 09/27/2024] [Accepted: 10/30/2024] [Indexed: 12/09/2024] Open
Abstract
Influenza viruses cause 3-5 million severe cases and 300,000-600,000 deaths worldwide. Most of the disease burden is in Low- and Middle-Income Countries (LMICs) owing to factors such as high population density, infrastructure challenges, poor quality healthcare, lack of consistent recommendations, less prioritization of all high-risk groups, and prevalent use of trivalent influenza vaccines. Although influenza vaccines are effective in reducing the annual influenza disease burden, existing vaccines have several limitations. In this narrative review, we address the unmet needs of existing influenza vaccines in LMICs in Africa, Asia Pacific, Latin America and the Middle East and discuss the characteristics of novel vaccines in clinical development. We also describe features of a successful vaccination program that LMICs could emulate to improve their current vaccination coverage and reduce the public health burden of influenza.
Collapse
Affiliation(s)
- Julia R Spinardi
- Vaccine Medical and Scientific Affairs, Emerging Markets, Pfizer Inc., São Paulo, SP, Brazil.
| | - Karan B Thakkar
- Vaccine Medical and Scientific Affairs, Emerging Markets, Pfizer Inc., Singapore, Singapore
| | - Verna L Welch
- Vaccine Medical and Scientific Affairs, Pfizer Inc., Pennsylvania, USA
| | - Oladayo Jagun
- Real World Strategy and Innovation, IQVIA Inc., New Jersey, USA
| | - Moe H Kyaw
- Vaccine Medical and Scientific Affairs, Emerging Markets, Pfizer Inc., Virginia, USA
| |
Collapse
|
3
|
Tian Y, Ma Y, Ran J, Yuan L, Zeng X, Tan L, Chen L, Xu Y, Li S, Huang T, Lu H. Protective Impact of Influenza Vaccination on Healthcare Workers. Vaccines (Basel) 2024; 12:1237. [PMID: 39591140 PMCID: PMC11599008 DOI: 10.3390/vaccines12111237] [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: 09/20/2024] [Revised: 10/25/2024] [Accepted: 10/28/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Influenza vaccine uptake among healthcare workers is crucial for preventing influenza infections, yet its effectiveness needs further investigation. OBJECTIVES This prospective observational study aimed to assess the protective effect of influenza vaccination among healthcare workers in Shenzhen. METHODS We enrolled 100 participants, with 50 receiving the 2023-2024 quadrivalent influenza vaccine (QIV) and 50 serving as unvaccinated controls. Epidemiological data were collected when the participants presented influenza-like illness. Serum samples were collected at three time points (pre-vaccination and 28 and 180 days after vaccination). Hemagglutination inhibition (HI) assay was performed against the strains included in the 2023-2024 QIV (H1N1, H3N2, BV and BY strains) to assess antibody protection levels. Demographics comparisons revealed no significant differences between the vaccinated and control groups (p > 0.05), ensuring group comparability. RESULTS The incidence of influenza-like illness was significantly lower in the vaccinated (18%) compared to the control group (36%; p = 0.046; OR = 0.39; 95% CI: 0.15 to 0.98). The vaccinated group also exhibited a higher rate of consecutive two-year vaccinations (48% vs. 24% in the control group, p < 0.05). Additionally, the vaccinated healthcare workers were more inclined to recommend vaccination to their families (80% vs. 48%, p < 0.05). HI titers against H1N1 (p < 0.01), H3N2 (p < 0.01), BV (p < 0.001) and BY (p < 0.01) significantly increased in the vaccinated group at 28 days post-vaccination. Moreover, a marked and sustained increase in HI titers against the H3N2 strain (p < 0.001) was observed at 180 days post-vaccination, highlighting the vaccine's enduring impact on the immune response. The fold change in the HI titers, indicative of the magnitude of the immune response, was significantly higher for H1N1 (p < 0.01), H3N2 (p < 0.001), BV (p < 0.01) and BY (p < 0.05) among the vaccinated individuals compared to the control group, underscoring the vaccine's efficacy in eliciting a robust and sustained antibody response. CONCLUSION Influenza vaccination significantly reduces the incidence of influenza-like illness among healthcare workers and promotes a sustained immune response. The study supports the importance of annual vaccination for this group to enhance personal and public health.
Collapse
Affiliation(s)
- Yimei Tian
- Department of Preventive Medicine and Healthcare-Associated Infection Management, National Clinical Research Center for Infectious Diseases, Third People’s Hospital of Shenzhen and the Second Affiliated Hospital of Southern University of Science and Technology, No 29 Bulan Road, Longgang District, Shenzhen 518112, China; (Y.T.); (Y.M.); (J.R.); (X.Z.); (L.T.); (L.C.); (Y.X.); (S.L.)
| | - Yue Ma
- Department of Preventive Medicine and Healthcare-Associated Infection Management, National Clinical Research Center for Infectious Diseases, Third People’s Hospital of Shenzhen and the Second Affiliated Hospital of Southern University of Science and Technology, No 29 Bulan Road, Longgang District, Shenzhen 518112, China; (Y.T.); (Y.M.); (J.R.); (X.Z.); (L.T.); (L.C.); (Y.X.); (S.L.)
| | - Jianchao Ran
- Department of Preventive Medicine and Healthcare-Associated Infection Management, National Clinical Research Center for Infectious Diseases, Third People’s Hospital of Shenzhen and the Second Affiliated Hospital of Southern University of Science and Technology, No 29 Bulan Road, Longgang District, Shenzhen 518112, China; (Y.T.); (Y.M.); (J.R.); (X.Z.); (L.T.); (L.C.); (Y.X.); (S.L.)
| | - Lifang Yuan
- School of Nursing, Guangdong Pharmaceutical University, 283 Jianghai Avenue, Guangzhou 510310, China;
| | - Xianhu Zeng
- Department of Preventive Medicine and Healthcare-Associated Infection Management, National Clinical Research Center for Infectious Diseases, Third People’s Hospital of Shenzhen and the Second Affiliated Hospital of Southern University of Science and Technology, No 29 Bulan Road, Longgang District, Shenzhen 518112, China; (Y.T.); (Y.M.); (J.R.); (X.Z.); (L.T.); (L.C.); (Y.X.); (S.L.)
| | - Lu Tan
- Department of Preventive Medicine and Healthcare-Associated Infection Management, National Clinical Research Center for Infectious Diseases, Third People’s Hospital of Shenzhen and the Second Affiliated Hospital of Southern University of Science and Technology, No 29 Bulan Road, Longgang District, Shenzhen 518112, China; (Y.T.); (Y.M.); (J.R.); (X.Z.); (L.T.); (L.C.); (Y.X.); (S.L.)
| | - Li Chen
- Department of Preventive Medicine and Healthcare-Associated Infection Management, National Clinical Research Center for Infectious Diseases, Third People’s Hospital of Shenzhen and the Second Affiliated Hospital of Southern University of Science and Technology, No 29 Bulan Road, Longgang District, Shenzhen 518112, China; (Y.T.); (Y.M.); (J.R.); (X.Z.); (L.T.); (L.C.); (Y.X.); (S.L.)
| | - Yifan Xu
- Department of Preventive Medicine and Healthcare-Associated Infection Management, National Clinical Research Center for Infectious Diseases, Third People’s Hospital of Shenzhen and the Second Affiliated Hospital of Southern University of Science and Technology, No 29 Bulan Road, Longgang District, Shenzhen 518112, China; (Y.T.); (Y.M.); (J.R.); (X.Z.); (L.T.); (L.C.); (Y.X.); (S.L.)
| | - Shaxi Li
- Department of Preventive Medicine and Healthcare-Associated Infection Management, National Clinical Research Center for Infectious Diseases, Third People’s Hospital of Shenzhen and the Second Affiliated Hospital of Southern University of Science and Technology, No 29 Bulan Road, Longgang District, Shenzhen 518112, China; (Y.T.); (Y.M.); (J.R.); (X.Z.); (L.T.); (L.C.); (Y.X.); (S.L.)
| | - Ting Huang
- Department of Preventive Medicine and Healthcare-Associated Infection Management, National Clinical Research Center for Infectious Diseases, Third People’s Hospital of Shenzhen and the Second Affiliated Hospital of Southern University of Science and Technology, No 29 Bulan Road, Longgang District, Shenzhen 518112, China; (Y.T.); (Y.M.); (J.R.); (X.Z.); (L.T.); (L.C.); (Y.X.); (S.L.)
| | - Hongzhou Lu
- Department of Preventive Medicine and Healthcare-Associated Infection Management, National Clinical Research Center for Infectious Diseases, Third People’s Hospital of Shenzhen and the Second Affiliated Hospital of Southern University of Science and Technology, No 29 Bulan Road, Longgang District, Shenzhen 518112, China; (Y.T.); (Y.M.); (J.R.); (X.Z.); (L.T.); (L.C.); (Y.X.); (S.L.)
| |
Collapse
|
4
|
Cowling BJ, Wong SS, Santos JJS, Touyon L, Ort JT, Ye N, Kwok NKM, Ho F, Cheng SMS, Ip DKM, Peiris M, Webby RJ, Wilson PC, Valkenburg SA, Tsang JS, Leung NHL, Hensley SE, Cobey S. Preliminary Findings From the Dynamics of the Immune Responses to Repeat Influenza Vaccination Exposures (DRIVE I) Study: A Randomized Controlled Trial. Clin Infect Dis 2024; 79:901-909. [PMID: 39041887 PMCID: PMC11478574 DOI: 10.1093/cid/ciae380] [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: 05/17/2024] [Revised: 06/28/2024] [Accepted: 07/18/2024] [Indexed: 07/24/2024] Open
Abstract
BACKGROUND Studies have reported that repeated annual vaccination may influence influenza vaccination effectiveness in the current season. METHODS We established a 5-year randomized placebo-controlled trial of repeated influenza vaccination (Flublok; Sanofi Pasteur) in adults 18-45 years of age. In the first 2 years, participants were randomized to receive vaccine or saline placebo as follows: placebo-placebo (P-P), placebo-vaccine (P-V), or vaccine-vaccine (V-V). Serum samples were collected each year just before vaccination and after 30 and 182 days. A subset of serum samples collected at 5 time points from 95 participants were tested for antibodies against vaccine strains. RESULTS From 23 October 2020 through 11 March 2021 we enrolled and randomized 447 adults. Among vaccinated individuals, antibody titers increased between days 0 and 30 against each of the vaccine strains, with smaller increases for repeat vaccinees who on average had higher prevaccination titers in year 2. There were statistically significant differences in the proportions of participants achieving ≥4-fold rises in antibody titer for the repeat vaccinees for influenza A(H1N1), B/Victoria, and B/Yamagata, but not for A(H3N2). Among participants who received vaccination in year 2, there were no significant differences between the P-V and V-V groups in geometric mean titers at day 30 or the proportions of participants with antibody titers ≥40 at day 30 for any of the vaccine strains. CONCLUSIONS In the first 2 years, during which influenza did not circulate, repeat and first-time vaccinees had similar postvaccination geometric mean titers to all 4 vaccine strains, indicative of similar levels of clinical protection. Clinical Trials Registration. NCT04576377.
Collapse
Affiliation(s)
- Benjamin J Cowling
- WHO 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, China
- Laboratory of Data Discovery for Health Limited, Hong Kong Science and Technology Park, New Territories, Hong Kong Special Administrative Region, China
| | - Sook-San Wong
- WHO 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, China
- HKU-Pasteur Research Pole, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Jefferson J S Santos
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Lisa Touyon
- WHO 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, China
- HKU-Pasteur Research Pole, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Jordan T Ort
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Naiqing Ye
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Natalie K M Kwok
- WHO 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, China
| | - Faith Ho
- WHO 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, China
| | - Samuel M S Cheng
- WHO 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, China
| | - Dennis K M Ip
- WHO 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, China
| | - Malik Peiris
- WHO 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, China
| | - Richard J Webby
- Department of Infectious Diseases, St Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Patrick C Wilson
- Gale and Ira Drukier Institute for Children's Health, Weill Cornell Medicine, New York, New York, USA
| | - Sophie A Valkenburg
- HKU-Pasteur Research Pole, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
| | - John S Tsang
- Yale Center for Systems and Engineering Immunology and Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Nancy H L Leung
- WHO 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, China
- Laboratory of Data Discovery for Health Limited, Hong Kong Science and Technology Park, New Territories, Hong Kong Special Administrative Region, China
| | - Scott E Hensley
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Sarah Cobey
- Department of Ecology and Evolution, University of Chicago, Chicago, Illinois, USA
| |
Collapse
|
5
|
Uno N, Ebensen T, Guzman CA, Ross TM. Intranasal administration of octavalent next-generation influenza vaccine elicits protective immune responses against seasonal and pre-pandemic viruses. J Virol 2024; 98:e0035424. [PMID: 39171925 PMCID: PMC11406897 DOI: 10.1128/jvi.00354-24] [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: 02/23/2024] [Accepted: 06/27/2024] [Indexed: 08/23/2024] Open
Abstract
Development of next-generation influenza virus vaccines is crucial to improve protection against circulating and emerging viruses. Current vaccine formulations have to be updated annually due to mutations in seasonal strains and do not offer protection against strains with pandemic potential. Computationally optimized broadly reactive antigen (COBRA) methodology has been utilized by our group to generate broadly reactive immunogens for individual influenza subtypes, which elicit protective immune responses against a broad range of strains over numerous seasons. Octavalent mixtures of COBRA hemagglutinin (HA) (H1, H2, H3, H5, H7, and influenza B virus) plus neuraminidase (NA) (N1 and N2) recombinant proteins mixed with c-di-AMP adjuvant were administered intranasally to naive or pre-immune ferrets in prime-boost fashion. Four weeks after final vaccination, collected sera were analyzed for breadth of antibody response, and the animals were challenged with seasonal or pre-pandemic strains. The octavalent COBRA vaccine elicited antibodies that recognized a broad panel of strains representing different subtypes, and these vaccinated animals were protected against influenza virus challenges. Overall, this study demonstrated that the mixture of eight COBRA HA/NA proteins mixed with an intranasal adjuvant is a promising candidate for a universal influenza vaccine. IMPORTANCE Influenza is a respiratory virus which infects around a billion people globally every year, with millions experiencing severe illness. Commercial vaccine efficacy varies year to year and can be low due to mismatch of circulating virus strains. Thus, the formulation of current vaccines has to be adapted accordingly every year. The development of a broadly reactive influenza vaccine would lessen the global economic and public health burden caused by the different types of influenza viruses. The significance of our research is producing a promising universal vaccine candidate which provides protection against a wider range of virus strains over a wider range of time.
Collapse
Affiliation(s)
- Naoko Uno
- Center for Vaccines and Immunology, University of Georgia, Athens, Georgia, USA
- Department of Infection Biology, Lehner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Thomas Ebensen
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Carlos A Guzman
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Ted M Ross
- Center for Vaccines and Immunology, University of Georgia, Athens, Georgia, USA
- Department of Infection Biology, Lehner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Florida Research and Innovation Center, Cleveland Clinic, Port Saint Lucie, Florida, USA
- Department of Infectious Diseases, University of Georgia, Athens, Georgia, USA
| |
Collapse
|
6
|
Veroniki AA, Thirugnanasampanthar SS, Konstantinidis M, Dourka J, Ghassemi M, Neupane D, Khan P, Nincic V, Corry M, Robson R, Parker A, Soobiah C, Sinilaite A, Doyon-Plourde P, Gil A, Siu W, Moqueet N, Stevens A, English K, Florez ID, Yepes-Nuñez JJ, Hutton B, Muller M, Moja L, Straus S, Tricco AC. Trivalent and quadrivalent seasonal influenza vaccine in adults aged 60 and older: a systematic review and network meta-analysis. BMJ Evid Based Med 2024; 29:239-254. [PMID: 38604619 PMCID: PMC11287607 DOI: 10.1136/bmjebm-2023-112767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/02/2024] [Indexed: 04/13/2024]
Abstract
OBJECTIVES To compare the efficacy of influenza vaccines of any valency for adults 60 years and older. DESIGN AND SETTING Systematic review with network meta-analysis (NMA) of randomised controlled trials (RCTs). MEDLINE, EMBASE, JBI Evidence-Based Practice (EBP) Database, PsycINFO, and Cochrane Evidence -Based Medicine database were searched from inception to 20 June 20, 2022. Two reviewers screened, abstracted, and appraised articles (Cochrane Risk of Bias (ROB) 2.0 tool) independently. We assessed certainty of findings using Confidence in Network Meta-Analysis and Grading of Recommendations, Assessment, Development and Evaluations approaches. We performed random-effects meta-analysis and network meta-analysis (NMA), and estimated odds ratios (ORs) for dichotomous outcomes and incidence rate ratios (IRRs) for count outcomes along with their corresponding 95% confidence intervals (CIs) and prediction intervals. PARTICIPANTS Older adults (≥60 years old) receiving an influenza vaccine licensed in Canada or the USA (vs placebo, no vaccine, or any other licensed vaccine), at any dose. MAIN OUTCOME MEASURES Laboratory-confirmed influenza (LCI) and influenza-like illness (ILI). Secondary outcomes were the number of vascular adverse events, hospitalisation for acute respiratory infection (ARI) and ILI, inpatient hospitalisation, emergency room (ER) visit for ILI, outpatient visit, and mortality, among others. RESULTS We included 41 RCTs and 15 companion reports comprising 8 vaccine types and 206 032 participants. Vaccines may prevent LCI compared with placebo, with high-dose trivalent inactivated influenza vaccine (IIV3-HD) (NMA: 9 RCTs, 52 202 participants, OR 0.23, 95% confidence interval (CI) (0.11 to 0.51), low certainty of evidence) and recombinant influenza vaccine (RIV) (OR 0.25, 95%CI (0.08 to 0.73), low certainty of evidence) among the most efficacious vaccines. Standard dose trivalent IIV3 (IIV3-SD) may prevent ILI compared with placebo, but the result was imprecise (meta-analysis: 2 RCTs, 854 participants, OR 0.39, 95%CI (0.15 to 1.02), low certainty of evidence). Any HD was associated with prevention of ILI compared with placebo (NMA: 9 RCTs, 65 658 participants, OR 0.38, 95%CI (0.15 to 0.93)). Adjuvanted quadrivalent IIV (IIV4-Adj) may be associated with the least vascular adverse events, but the results were very uncertain (NMA: eight 8 RCTs, 57 677 participants, IRR 0.18, 95%CI (0.07 to 0.43), very low certainty of evidence). RIV on all-cause mortality may be comparable to placebo (NMA: 20 RCTs, 140 577 participants, OR 1.01, 95%CI (0.23 to 4.49), low certainty of evidence). CONCLUSIONS This systematic review demonstrated efficacy associated with IIV3-HD and RIV vaccines in protecting older persons against LCI. RIV vaccine may reduce all-cause mortality when compared with other vaccines, but the evidence is uncertain. Differences in efficacy between influenza vaccines remain uncertain with very low to moderate certainty of evidence. PROSPERO REGISTRATION NUMBER CRD42020177357.
Collapse
Affiliation(s)
- Areti Angeliki Veroniki
- Institute of Health Policy Management and Evaluation, University of Toronto, Toronto, Ontario, Canada
- Knowledge Translation Program, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada
| | - Sai Surabi Thirugnanasampanthar
- Knowledge Translation Program, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada
| | - Menelaos Konstantinidis
- Institute of Health Policy Management and Evaluation, University of Toronto, Toronto, Ontario, Canada
- Knowledge Translation Program, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada
| | - Jasmeen Dourka
- Knowledge Translation Program, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada
| | - Marco Ghassemi
- Knowledge Translation Program, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada
| | - Dipika Neupane
- Knowledge Translation Program, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada
| | - Paul Khan
- Knowledge Translation Program, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada
| | - Vera Nincic
- Knowledge Translation Program, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada
| | - Margarita Corry
- Trinity College Dublin School of Nursing and Midwifery, Dublin, Ireland
| | - Reid Robson
- Knowledge Translation Program, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada
| | - Amanda Parker
- Knowledge Translation Program, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada
| | - Charlene Soobiah
- Institute of Health Policy Management and Evaluation, University of Toronto, Toronto, Ontario, Canada
- Knowledge Translation Program, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada
| | | | | | - Anabel Gil
- Public Health Agency, Ottawa, Ontario, Canada
| | - Winnie Siu
- Public Health Agency, Ottawa, Ontario, Canada
| | | | | | - Kelly English
- Patient Partner, Strategy for Patient Oriented-Research Evidence Alliance (SPOR EA), St Michael's Hospital, Toronto, Ontario, Canada
| | - Ivan D Florez
- Department of Pediatrics, University of Antioquia Faculty of Medicine, Medellin, Colombia
- School of Rehabilitation Science, McMaster University, Hamilton, Ontario, Canada
- Pediatric Intensive Care Unit, Clinica Las Américas-AUNA, Medellin, Colombia
| | - Juan J Yepes-Nuñez
- University of los Andes Faculty of Medicine, Bogota, Cundinamarca, Colombia
- Pulmonology Service, Internal Medicine Section, University Hospital of the Fundacion Santa Fe de Bogota, Bogota, Colombia
| | - Brian Hutton
- Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| | - Matthew Muller
- Institute of Health Policy Management and Evaluation, University of Toronto, Toronto, Ontario, Canada
- Department of Medicine, St Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Lorenzo Moja
- Department of Biomedical Sciences and Technologies, University of Milan, Milano, Lombardia, Italy
| | - Sharon Straus
- Institute of Health Policy Management and Evaluation, University of Toronto, Toronto, Ontario, Canada
- Knowledge Translation Program, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada
- Department of Geriatric Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Andrea C Tricco
- Knowledge Translation Program, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada
- Epidemiology Division & Institute of Health Policy, Management, and Evaluation, University of Toronto Dalla Lana School of Public Health, Toronto, Ontario, Canada
| |
Collapse
|
7
|
Cowling BJ, Wong SS, Santos JJS, Touyon L, Ort J, Ye N, Kwok NKM, Ho F, Cheng SMS, Ip DKM, Peiris M, Webby RJ, Wilson PC, Valkenburg SA, Tsang JS, Leung NHL, Hensley SE, Cobey S. Preliminary findings from the Dynamics of the Immune Responses to Repeat Influenza Vaccination Exposures (DRIVE I) Study: a Randomized Controlled Trial. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.05.16.24307455. [PMID: 38798684 PMCID: PMC11118649 DOI: 10.1101/2024.05.16.24307455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Background Studies have reported that repeated annual vaccination may influence the effectiveness of the influenza vaccination in the current season. The mechanisms underlying these differences are unclear but might include "focusing" of the adaptive immune response to older strains. Methods We established a 5-year randomized placebo-controlled trial of repeated influenza vaccination (Flublok, Sanofi Pasteur) in adults 18-45 years of age. Participants were randomized equally between five groups, with planned annual receipt of vaccination (V) or saline placebo (P) as follows: P-P-P-P-V, P-P-P-V-V, P-P-V-V-V, P-V-V-V-V, or V-V-V-VV. Serum samples were collected each year just before vaccination and after 30 and 182 days. A subset of sera were tested by hemagglutination inhibition assays, focus reduction neutralization tests and enzyme-linked immunosorbent assays against vaccine strains. Results From 23 October 2020 through 11 March 2021 we enrolled and randomized 447 adults. We selected sera from 95 participants at five timepoints from the first two study years for testing. Among vaccinated individuals, antibody titers increased between days 0 and 30 against each of the vaccine strains, with substantial increases for first-time vaccinees and smaller increases for repeat vaccinees, who had higher pre-vaccination titers in year 2. There were statistically significant reductions in the proportion of participants achieving a four-fold greater rise in antibody titer for the repeat vaccinees for A(H1N1), B/Victoria and B/Yamagata, but not for influenza A(H3N2). There were no statistically significant differences between groups in geometric mean titers at day 30 or the proportions of participants with antibody titers ≥40 at day 30 for any of the vaccine strains. Conclusions In the first two years, repeat vaccinees and first-time vaccinees had similar post-vaccination geometric mean titers to all four vaccine strains, indicative of similar levels of clinical protection. The vaccine strains of A(H1N1) and A(H3N2) were updated in year 2, providing an opportunity to explore antigenic distances between those strains in humans in subsequent years.
Collapse
Affiliation(s)
- Benjamin J. Cowling
- WHO 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, China
- Laboratory of Data Discovery for Health Limited, Hong Kong Science and Technology Park, New Territories, Hong Kong Special Administrative Region, China
| | - Sook-San Wong
- WHO 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, China
- HKU-Pasteur Research Pole, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Jefferson J. S. Santos
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Lisa Touyon
- WHO 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, China
- HKU-Pasteur Research Pole, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Jordan Ort
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Naiqing Ye
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Natalie K. M. Kwok
- WHO 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, China
| | - Faith Ho
- WHO 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, China
| | - Samuel M. S. Cheng
- WHO 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, China
| | - Dennis K. M. Ip
- WHO 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, China
| | - Malik Peiris
- WHO 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, China
| | - Richard J. Webby
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, TN 38105
| | - Patrick C. Wilson
- Gale and Ira Drukier Institute for Children’s Health, Weill Cornell Medicine, New York, NY 10021
| | - Sophie A. Valkenburg
- HKU-Pasteur Research Pole, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | - John S. Tsang
- Yale Center for Systems and Engineering Immunology and Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520
| | - Nancy H. L. Leung
- WHO 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, China
- Laboratory of Data Discovery for Health Limited, Hong Kong Science and Technology Park, New Territories, Hong Kong Special Administrative Region, China
| | - Scott E. Hensley
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Sarah Cobey
- Department of Ecology and Evolution, University of Chicago, Chicago, IL 60637
| |
Collapse
|
8
|
Jangra S, Lamoot A, Singh G, Laghlali G, Chen Y, Ye T, García-Sastre A, De Geest BG, Schotsaert M. Lipid nanoparticle composition for adjuvant formulation modulates disease after influenza virus infection in quadrivalent influenza vaccine vaccinated mice. Front Immunol 2024; 15:1370564. [PMID: 38711520 PMCID: PMC11070541 DOI: 10.3389/fimmu.2024.1370564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Accepted: 03/19/2024] [Indexed: 05/08/2024] Open
Abstract
There are considerable avenues through which currently licensed influenza vaccines could be optimized. We tested influenza vaccination in a mouse model with two adjuvants: Sendai virus-derived defective interfering (SDI) RNA, a RIG-I agonist; and an amphiphilic imidazoquinoline (IMDQ-PEG-Chol), a TLR7/8 agonist. The negatively charged SDI RNA was formulated into lipid nanoparticles (LNPs) facilitating direct delivery of SDI RNA to the cytosol, where RIG-I sensing induces inflammatory and type I interferon responses. We previously tested SDI RNA and IMDQ-PEG-Chol as standalone and combination adjuvants for influenza and SARS-CoV-2 vaccines. Here, we tested two different ionizable lipids, K-Ac7-Dsa and S-Ac7-Dog, for LNP formulations. The LNPs were incorporated with SDI RNA to determine its potential as a combination adjuvant with IMDQ-PEG-Chol by evaluating the host immune response to vaccination and infection in immunized BALB/c mice. Adjuvanticity of IMDQ-PEG-Chol with and without empty or SDI-loaded LNPs was validated with quadrivalent inactivated influenza vaccine (QIV), showing robust induction of antibody titers and T-cell responses. Depending on the adjuvant combination and LNP formulation, humoral and cellular vaccine responses could be tailored towards type 1 or type 2 host responses with specific cytokine profiles that correlated with the protective responses to viral infection. The extent of protection conferred by different vaccine/LNP/adjuvant combinations was tested by challenging mice with a vaccine-matched strain of influenza A virus A/Singapore/gp1908/2015 IVR-180 (H1N1). Groups that received either LNP formulated with SDI or IMDQ-PEG-Chol, or both, showed very low levels of viral replication in their lungs at 5 days post-infection (DPI). These studies provide evidence that the combination of vaccines with LNPs and/or adjuvants promote antigen-specific cellular responses that can contribute to protection upon infection. Interestingly, we observed differences in humoral and cellular responses to vaccination between different groups receiving K-Ac7-Dsa or S-Ac7-Dog lipids in LNP formulations. The differences were also reflected in inflammatory responses in lungs of vaccinated animals to infection, depending on LNP formulations. Therefore, this study suggests that the composition of the LNPs, particularly the ionizable lipid, plays an important role in inducing inflammatory responses in vivo, which is important for vaccine safety and to prevent adverse effects upon viral exposure.
Collapse
Affiliation(s)
- Sonia Jangra
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | | | - Gagandeep Singh
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Gabriel Laghlali
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Pharmaceutics, Ghent University, Ghent, Belgium
| | - Yong Chen
- Department of Pharmaceutics, Ghent University, Ghent, Belgium
| | - Tingting Ye
- Department of Pharmaceutics, Ghent University, Ghent, Belgium
| | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | | | - Michael Schotsaert
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
9
|
Youhanna J, Tran V, Hyer R, Domnich A. Immunogenicity of Enhanced Influenza Vaccines Against Mismatched Influenza Strains in Older Adults: A Review of Randomized Controlled Trials. Influenza Other Respir Viruses 2024; 18:e13286. [PMID: 38594827 PMCID: PMC11004266 DOI: 10.1111/irv.13286] [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: 10/23/2023] [Revised: 03/15/2024] [Accepted: 03/18/2024] [Indexed: 04/11/2024] Open
Abstract
Antigenic drift is a major driver of viral evolution and a primary reason why influenza vaccines must be reformulated annually. Mismatch between vaccine and circulating viral strains negatively affects vaccine effectiveness and often contributes to higher rates of influenza-related hospitalizations and deaths, particularly in years dominated by A(H3N2). Several countries recommend enhanced influenza vaccines for older adults, who are at the highest risk of severe influenza complications and mortality. The immunogenicity of enhanced vaccines against heterologous A(H3N2) strains has been examined in nine studies to date. In six studies, an enhanced, licensed MF59-adjuvanted trivalent inactivated influenza vaccine (aIIV3) consistently increased heterologous antibody titers relative to standard influenza vaccine, with evidence of a broad heterologous immune response across multiple genetic clades. In one study, licensed high-dose trivalent inactivated influenza vaccine (HD-IIV3) also induced higher heterologous antibody titers than standard influenza vaccine. In a study comparing a higher dose licensed quadrivalent recombinant influenza vaccine (RIV4) with HD-IIV3 and aIIV3, no significant differences in antibody titers against a heterologous strain were observed, although seroconversion rates were higher with RIV4 versus comparators. With the unmet medical need for improved influenza vaccines, the paucity of studies especially with enhanced vaccines covering mismatched strains highlights a need for further investigation of cross-protection in older adults.
Collapse
Affiliation(s)
| | - Vy Tran
- CSL Seqirus LtdSummitNew JerseyUSA
| | - Randall Hyer
- Baruch S. Blumberg InstituteDoylestownPennsylvaniaUSA
| | | |
Collapse
|
10
|
Pavia G, Scarpa F, Ciccozzi A, Romano C, Branda F, Quirino A, Marascio N, Matera G, Sanna D, Ciccozzi M. Changing and Evolution of Influenza Virus: Is It a Trivial Flu? Chemotherapy 2024; 69:185-193. [PMID: 38508151 DOI: 10.1159/000538382] [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: 02/24/2024] [Accepted: 03/09/2024] [Indexed: 03/22/2024]
Abstract
BACKGROUND Influenza viruses are etiological agents which cause contagious respiratory, seasonal epidemics and, for influenza A subtypes, pandemics. The clinical picture of influenza has undergone continuous change over the years, due to intrinsic viral evolution as well as "reassortment" of its genomic segments. The history of influenza highlights its ability to adapt and to rapidly evolve, without specific circumstances. This reflects the complexity of this pathology and poses the fundamental question about its assumption as a "common illness" and its impact on public health. SUMMARY The global influenza epidemics and pandemics claimed millions of deaths, leaving an indelible mark on public health and showing the need for a better comprehension of the influenza virus. The clear understanding of genetic variations during the influenza seasonal epidemics is a crucial point for developing effective strategies for prevention, treatment, and vaccine design. The recent advance in next-generation sequencing approaches, model systems to virus culture, and bioinformatics pipeline played a key role in the rapid characterization of circulating influenza strains. In particular, the increase in computational power allowed the performance of complex tasks in healthcare settings through machine learning algorithms, which analyze different variables, such as medical and laboratory outputs, to optimize medical research and improve public health systems. The early detection of emerging and reemerging pathogens is a matter of importance to prevent future pandemics. KEY MESSAGES The perception of influenza as a "trivial flu" or a more serious public health concern is a subject of ongoing debate, reflecting the multifaceted nature of this infectious disease. The variability in the severity of influenza sheds light on the unpredictability of the viral characteristics, coupled with the challenges in accurately predicting circulating strains. This adds complexity to the public health burden of influenza and highlights the need for targeted interventions.
Collapse
Affiliation(s)
- Grazia Pavia
- Unit of Clinical Microbiology, Department of Health Sciences, "Magna Græcia" University Hospital, Catanzaro, Italy
| | - Fabio Scarpa
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | | | - Chiara Romano
- Unit of Medical Statistics and Molecular Epidemiology, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Francesco Branda
- Unit of Medical Statistics and Molecular Epidemiology, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Angela Quirino
- Unit of Clinical Microbiology, Department of Health Sciences, "Magna Græcia" University Hospital, Catanzaro, Italy
| | - Nadia Marascio
- Unit of Clinical Microbiology, Department of Health Sciences, "Magna Græcia" University Hospital, Catanzaro, Italy
| | - Giovanni Matera
- Unit of Clinical Microbiology, Department of Health Sciences, "Magna Græcia" University Hospital, Catanzaro, Italy
| | - Daria Sanna
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Massimo Ciccozzi
- Unit of Medical Statistics and Molecular Epidemiology, Università Campus Bio-Medico di Roma, Rome, Italy
| |
Collapse
|
11
|
Croucher NJ. Immune interface interference vaccines: An evolution-informed approach to anti-bacterial vaccine design. Microb Biotechnol 2024; 17:e14446. [PMID: 38536702 PMCID: PMC10970203 DOI: 10.1111/1751-7915.14446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 03/01/2024] [Indexed: 10/17/2024] Open
Abstract
Developing protein-based vaccines against bacteria has proved much more challenging than producing similar immunisations against viruses. Currently, anti-bacterial vaccines are designed using methods based on reverse vaccinology. These identify broadly conserved, immunogenic proteins using a combination of genomic and high-throughput laboratory data. While this approach has successfully generated multiple rationally designed formulations that show promising immunogenicity in animal models, few have been licensed. The difficulty of inducing protective immunity in humans with such vaccines mirrors the ability of many bacteria to recolonise individuals despite recognition by natural polyvalent antibody repertoires. As bacteria express too many antigens to evade all adaptive immune responses through mutation, they must instead inhibit the efficacy of such host defences through expressing surface structures that interface with the immune system. Therefore, 'immune interface interference' (I3) vaccines that target these features should synergistically directly target bacteria and prevent them from inhibiting responses to other surface antigens. This approach may help us understand the efficacy of the two recently introduced immunisations against serotype B meningococci, which both target the Factor H-binding protein (fHbp) that inhibits complement deposition on the bacterial surface. Therefore, I3 vaccine designs may help overcome the current challenges of developing protein-based vaccines to prevent bacterial infections.
Collapse
Affiliation(s)
- Nicholas J. Croucher
- MRC Centre for Global Infectious Disease Analysis, Department of Infectious Disease Epidemiology, School of Public HealthImperial College LondonLondonUK
| |
Collapse
|
12
|
Chatterjee A, Ambrose K, Canaday DH, Delair S, Ezike N, Huber VC, Jhaveri R, Nyquist AC, Sporer A, Varman M, Vivekanandan R, Wojcik R, Jandhyala R. The association between influenza vaccine effectiveness and egg-based manufacturing technology: literature review and US expert consensus. Curr Med Res Opin 2024; 40:335-343. [PMID: 38054898 DOI: 10.1080/03007995.2023.2284386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 11/13/2023] [Indexed: 12/07/2023]
Abstract
BACKGROUND Influenza is associated with significant disease burden in the US and is currently best controlled by vaccination programs. Influenza vaccine effectiveness (VE) is low and may be reduced by several factors, including egg adaptations. Although non-egg-based influenza vaccines reportedly have greater VE in egg-adapted seasons, evidence for egg adaptations' reduction of VE is indirect and dissociated, apart from two previous European consensuses. METHODS This study replicated the methodology used in a 2020 literature review and European consensus, providing an updated review and consensus opinion of 10 US experts on the evidence for a mechanistic basis for reduction of VE due to egg-based manufacturing methods. A mechanistic basis was assumed if sufficient evidence was found for underlying principles proposed to give rise to such an effect. Evidence for each principle was brought forward from the 2020 review and identified here by structured literature review and expert panel. Experts rated the strength of support for each principle and a mechanistic basis for reduction of VE due to egg-based influenza vaccine manufacture in a consensus method (consensus for strong/very strong evidence = ≥ 3.5 on 5-point Likert scale). RESULTS Experts assessed 251 references (from previous study: 185; this study: 66). The majority of references for all underlying principles were rated as strong or very strong supporting evidence (52-86%). Global surveillance, WHO candidate vaccine virus selection, and manufacturing stages involving eggs were identified as most likely to impact influenza VE. CONCLUSION After review of extensive evidence for reduction of VE due to egg-based influenza vaccine manufacture, influenza experts in the US joined those in Europe in unanimous agreement for a mechanistic basis for the effect. Vaccine providers and administrators should consider use of non-egg-based influenza vaccine manufacture to reduce the risk of egg adaptations and likely impact on VE.
Collapse
Affiliation(s)
- Archana Chatterjee
- Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | | | | | | | | | | | - Ravi Jhaveri
- Feinberg School of Medicine, Northwestern, IL, USA
| | | | | | | | | | | | - Ravi Jandhyala
- Medialis Ltd, Milton Keynes, UK
- King's College London, London, UK
| |
Collapse
|
13
|
Jangra S, Lamoot A, Singh G, Laghlali G, Chen Y, Yz T, García-Sastre A, De Geest BG, Schotsaert M. Lipid nanoparticle composition for adjuvant formulation modulates disease after influenza virus infection in QIV vaccinated mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.14.575599. [PMID: 38293047 PMCID: PMC10827098 DOI: 10.1101/2024.01.14.575599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
Adjuvants can enhance vaccine effectiveness of currently licensed influenza vaccines. We tested influenza vaccination in a mouse model with two adjuvants: Sendai virus derived defective interfering (SDI) RNA, a RIG-I agonist, and an amphiphilic imidazoquinoline (IMDQ-PEG-Chol), TLR7/8 adjuvant. The negatively charged SDI RNA was formulated into lipid nanoparticles (LNPs) facilitating the direct delivery of a RIG-I agonist to the cytosol. We have previously tested SDI and IMDQ-PEG-Chol as standalone and combination adjuvants for influenza and SARS-CoV-2 vaccines. Here we tested two different ionizable lipids, K-Ac7-Dsa and S-Ac7-Dog, for LNP formulations. The adjuvanticity of IMDQ-PEG-Chol with and without empty or SDI-loaded LNPs was validated in a licensed vaccine setting (quadrivalent influenza vaccine or QIV) against H1N1 influenza virus, showing robust induction of antibody titres and T cell responses. Depending on the adjuvant combination and LNP lipid composition (K-Ac7-Dsa or S-Ac7-Dog lipids), humoral and cellular vaccine responses could be tailored towards type 1 or type 2 host responses with specific cytokine profiles that correlated with protection during viral infection. The extent of protection conferred by different vaccine/LNP/adjuvant combinations was examined against challenge with the vaccine-matching strain of H1N1 influenza A virus. Groups that received either LNP formulated with SDI, IMDQ-PEG-Chol or both showed very low levels of viral replication in their lungs at five days post virus infection. LNP ionizable lipid composition as well as loading (empty versus SDI) also skewed host responses to infection, as reflected in the cytokine and chemokine levels in lungs of vaccinated animals upon infection. These studies show the potential of LNPs as adjuvant delivery vehicles for licensed vaccines and illustrate the importance of LNP composition for subsequent host responses to infection, an important point of consideration for vaccine safety.
Collapse
Affiliation(s)
- Sonia Jangra
- Department of Microbiology, 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
| | | | - Gagandeep Singh
- Department of Microbiology, 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
| | - Gabriel Laghlali
- Department of Microbiology, 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
- Department of Pharmaceutics, Ghent University, Ghent Belgium
| | - Yong Chen
- Department of Pharmaceutics, Ghent University, Ghent Belgium
| | - Tingting Yz
- Department of Pharmaceutics, Ghent University, Ghent Belgium
| | - Adolfo García-Sastre
- Department of Microbiology, 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
- Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai New York, NY, USA
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai New York, NY, USA
| | | | - Michael Schotsaert
- Department of Microbiology, 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
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
14
|
de Waure C, Gärtner BC, Lopalco PL, Puig-Barbera J, Nguyen-Van-Tam JS. Real world evidence for public health decision-making on vaccination policies: perspectives from an expert roundtable. Expert Rev Vaccines 2024; 23:27-38. [PMID: 38084895 DOI: 10.1080/14760584.2023.2290194] [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: 11/04/2023] [Accepted: 11/28/2023] [Indexed: 12/18/2023]
Abstract
INTRODUCTION Influenza causes significant morbidity and mortality, but influenza vaccine uptake remains below most countries' targets. Vaccine policy recommendations vary, as do procedures for reviewing and appraising the evidence. AREAS COVERED During a series of roundtable discussions, we reviewed procedures and methodologies used by health ministries in four European countries to inform vaccine recommendations. We review the type of evidence currently recommended by each health ministry and the range of approaches toward considering randomized controlled trials (RCTs) and real-world evidence (RWE) studies when setting influenza vaccine recommendations. EXPERT OPINION Influenza vaccine recommendations should be based on data from both RCTs and RWE studies of efficacy, effectiveness, and safety. Such data should be considered alongside health-economic, cost-effectiveness, and budgetary factors. Although RCT data are more robust and less prone to bias, well-designed RWE studies permit timely evaluation of vaccine benefits, effectiveness comparisons over multiple seasons in large populations, and detection of rare adverse events, under real-world conditions. Given the variability of vaccine effectiveness due to influenza virus mutations and increasing diversification of influenza vaccines, we argue that consideration of both RWE and RCT evidence is the best approach to more nuanced and timely updates of influenza vaccine recommendations.
Collapse
Affiliation(s)
- Chiara de Waure
- Public Health, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Barbara C Gärtner
- Department and Institute of Microbiology, Saarland University Hospital, Homburg, Germany
| | | | - Joan Puig-Barbera
- Foundation for the Promotion of Health and Biomedical Research of the Valencian Region, Valencia, Spain
| | | |
Collapse
|
15
|
Park J, Champion JA. Development of Self-Assembled Protein Nanocage Spatially Functionalized with HA Stalk as a Broadly Cross-Reactive Influenza Vaccine Platform. ACS NANO 2023; 17:25045-25060. [PMID: 38084728 PMCID: PMC10753887 DOI: 10.1021/acsnano.3c07669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 11/29/2023] [Accepted: 12/01/2023] [Indexed: 12/27/2023]
Abstract
There remains a need for the development of a universal influenza vaccine, as current seasonal influenza vaccines exhibit limited protection against mismatched, mutated, or pandemic influenza viruses. A desirable approach to developing an effective universal influenza vaccine is the incorporation of highly conserved antigens in a multivalent scaffold that enhances their immunogenicity. Here, we develop a broadly cross-reactive influenza vaccine by functionalizing self-assembled protein nanocages (SAPNs) with multiple copies of the hemagglutinin stalk on the outer surface and matrix protein 2 ectodomain on the inner surface. SAPNs were generated by engineering short coiled coils, and the design was simulated by MD GROMACS. Due to the short sequences, off-target immune responses against empty SAPN scaffolds were not seen in immunized mice. Vaccination with the multivalent SAPNs induces high levels of broadly cross-reactive antibodies of only external antigens, demonstrating tight spatial control over the designed antigen placement. This work demonstrates the use of SAPNs as a potential influenza vaccine.
Collapse
Affiliation(s)
- Jaeyoung Park
- School of Chemical and Biomolecular
Engineering, Georgia Institute of Technology, 950 Atlantic Dr. NW, Atlanta, Georgia 30332-2000, United States
| | - Julie A. Champion
- School of Chemical and Biomolecular
Engineering, Georgia Institute of Technology, 950 Atlantic Dr. NW, Atlanta, Georgia 30332-2000, United States
| |
Collapse
|
16
|
Miwa H, Antao OQ, Kelly‐Scumpia KM, Baghdasarian S, Mayer DP, Shang L, Sanchez GM, Archang MM, Scumpia PO, Weinstein JS, Di Carlo D. Improved Humoral Immunity and Protection against Influenza Virus Infection with a 3d Porous Biomaterial Vaccine. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2302248. [PMID: 37750461 PMCID: PMC10625058 DOI: 10.1002/advs.202302248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 08/03/2023] [Indexed: 09/27/2023]
Abstract
New vaccine platforms that activate humoral immunity and generate neutralizing antibodies are required to combat emerging pathogens, including influenza virus. A slurry of antigen-loaded hydrogel microparticles that anneal to form a porous scaffold with high surface area for antigen uptake by infiltrating immune cells as the biomaterial degrades is demonstrated to enhance humoral immunity. Antigen-loaded-microgels elicited a robust cellular humoral immune response, with increased CD4+ T follicular helper (Tfh) cells and prolonged germinal center (GC) B cells comparable to the commonly used adjuvant, aluminum hydroxide (Alum). Increasing the weight fraction of polymer material led to increased material stiffness and antigen-specific antibody titers superior to Alum. Vaccinating mice with inactivated influenza virus loaded into this more highly cross-linked formulation elicited a strong antibody response and provided protection against a high dose viral challenge. By tuning physical and chemical properties, adjuvanticity can be enhanced leading to humoral immunity and protection against a pathogen, leveraging two different types of antigenic material: individual protein antigen and inactivated virus. The flexibility of the platform may enable design of new vaccines to enhance innate and adaptive immune cell programming to generate and tune high affinity antibodies, a promising approach to generate long-lasting immunity.
Collapse
Affiliation(s)
- Hiromi Miwa
- Department of BioengineeringUniversity of California Los AngelesLos AngelesCA90095USA
| | - Olivia Q. Antao
- Center for Immunity and InflammationRutgers New Jersey Medical SchoolNewarkNJ07103USA
| | - Kindra M. Kelly‐Scumpia
- Division of CardiologyDepartment of MedicineDavid Geffen School of MedicineUniversity of California Los AngelesLos AngelesCA90095USA
| | - Sevana Baghdasarian
- Department of BioengineeringUniversity of California Los AngelesLos AngelesCA90095USA
| | - Daniel P. Mayer
- Center for Immunity and InflammationRutgers New Jersey Medical SchoolNewarkNJ07103USA
| | - Lily Shang
- Department of BioengineeringUniversity of California Los AngelesLos AngelesCA90095USA
| | - Gina M. Sanchez
- Center for Immunity and InflammationRutgers New Jersey Medical SchoolNewarkNJ07103USA
| | - Maani M. Archang
- Department of BioengineeringUniversity of California Los AngelesLos AngelesCA90095USA
- MSTP ProgramDavid Geffen School of MedicineUniversity of California Los AngelesLos AngelesCA90095USA
| | - Philip O. Scumpia
- Division of DermatologyDepartment of MedicineDavid Geffen School of MedicineUniversity of California Los AngelesLos AngelesCA90095USA
- Department of DermatologyVA Greater Los Angeles Healthcare SystemLos AngelesCA90073USA
- Jonsson Comprehensive Cancer CenterUniversity of California Los AngelesLos AngelesCA90095USA
| | - Jason S Weinstein
- Center for Immunity and InflammationRutgers New Jersey Medical SchoolNewarkNJ07103USA
| | - Dino Di Carlo
- Department of BioengineeringUniversity of California Los AngelesLos AngelesCA90095USA
- Jonsson Comprehensive Cancer CenterUniversity of California Los AngelesLos AngelesCA90095USA
- Department of Mechanical and Aerospace EngineeringUniversity of California Los AngelesLos AngelesCA90095USA
- California Nano Systems Institute (CNSI)University of California Los AngelesLos AngelesCA90095USA
| |
Collapse
|
17
|
Sun W, Zhang Z, Chen M, Liu X, Wang Y, Yao S, Li L. Discovery of a potent inhibitor targeting the cap-binding domain of the PB2 subunit of influenza RNA-dependent RNA polymerase. Biochem Biophys Res Commun 2023; 678:97-101. [PMID: 37625270 DOI: 10.1016/j.bbrc.2023.08.027] [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: 07/26/2023] [Accepted: 08/12/2023] [Indexed: 08/27/2023]
Abstract
Influenza pandemics have emerged as a significant global public health and security concern. PB2, a crucial subunit of the influenza RNA-dependent RNA polymerase (RdRP), has been identified as a promising target for influenza treatment. We herein report the discovery of a potent novel PB2 inhibitor, 7-51A, with a KD value of 1.64 nM as determined by ITC. The high activity of 7-51A was elucidated by the co-crystal structure of the PB2-7-51A complex, and comparative analysis revealed unique interactions that had never been observed before. The preliminary pharmacological evaluation indicated that 7-51A exhibited commendable cellular safety, hepatic microsomal metabolic safety and stability. Collectively, 7-51A was found to be an effective PB2 inhibitor and could be used as a lead compound for further studies.
Collapse
Affiliation(s)
- Weining Sun
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ziling Zhang
- Key Laboratory of Drug Targeting and Drug Delivery System of Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Mingxin Chen
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xinlei Liu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yifei Wang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Shaohua Yao
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Linli Li
- Key Laboratory of Drug Targeting and Drug Delivery System of Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
18
|
Kackos CM, DeBeauchamp J, Davitt CJH, Lonzaric J, Sealy RE, Hurwitz JL, Samsa MM, Webby RJ. Seasonal quadrivalent mRNA vaccine prevents and mitigates influenza infection. NPJ Vaccines 2023; 8:157. [PMID: 37828126 PMCID: PMC10570305 DOI: 10.1038/s41541-023-00752-5] [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: 01/05/2023] [Accepted: 09/25/2023] [Indexed: 10/14/2023] Open
Abstract
Annually, seasonal influenza is responsible for millions of infections and hundreds of thousands of deaths. The current method for managing influenza is vaccination using a standardized amount of the influenza virus' primary surface antigen, hemagglutinin (HA), as the intended target of the immune response. This vaccination strategy results in vaccines with variable efficacy year to year due to antigenic drift of HA, which can be further exacerbated by manufacturing processes optimizing growth of vaccine virus in eggs. Due to these limitations, alternative vaccine platforms are actively being explored to improve influenza vaccine efficacy, including cell-based, recombinant protein, and mRNA vaccines. mRNA's rapid, in vitro production makes it an appealing platform for influenza vaccination, and the success of SARS-CoV-2 mRNA vaccines in the clinic has encouraged the development of mRNA vaccines for other pathogens. Here, the immunogenicity and protective efficacy of a quadrivalent mRNA vaccine encoding HA from four seasonal influenza viruses, A/California/07/2009 (H1N1), A/Hong Kong/4801/2014 (H3N2), B/Brisbane/60/2008 (B-Victoria lineage), and B/Phuket/3073/2013 (B-Yamagata lineage), was evaluated. In mice, a 120 μg total dose of this quadrivalent mRNA vaccine induced robust antibody titers against each subtype that were commensurate with titers when each antigen was administered alone. Following A/California/04/2009 challenge, mice were fully protected from morbidity and mortality, even at doses as low as 1 μg of each antigen. Additionally, a single administration of 10 μg of quadrivalent mRNA was sufficient to prevent weight loss caused by A/California/04/2009. These results support the promise of this mRNA vaccine for prevention and mitigation of influenza vaccine.
Collapse
Affiliation(s)
- Christina M Kackos
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN, USA
- St. Jude Children's Research Hospital Graduate School of Biomedical Sciences, Memphis, TN, USA
| | - Jennifer DeBeauchamp
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN, USA
| | | | | | - Robert E Sealy
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Julia L Hurwitz
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN, USA
| | | | - Richard J Webby
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN, USA.
| |
Collapse
|
19
|
Rikhi N, Sei CJ, Rao M, Schuman RF, Kroscher KA, Matyas GR, Muema K, Lange C, Assiaw-Dufu A, Hussin E, Jobe O, Alving CR, Fischer GW. Unconjugated Multi-Epitope Peptides Adjuvanted with ALFQ Induce Durable and Broadly Reactive Antibodies to Human and Avian Influenza Viruses. Vaccines (Basel) 2023; 11:1468. [PMID: 37766144 PMCID: PMC10537791 DOI: 10.3390/vaccines11091468] [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: 07/20/2023] [Revised: 08/30/2023] [Accepted: 09/01/2023] [Indexed: 09/29/2023] Open
Abstract
An unconjugated composite peptide vaccine targeting multiple conserved influenza epitopes from hemagglutinin, neuraminidase, and matrix protein and formulated with a safe and highly potent adjuvant, Army Liposome formulation (ALFQ), generated broad and durable immune responses in outbred mice. The antibodies recognized specific epitopes in influenza peptides and several human, avian, and swine influenza viruses. Comparable antibody responses to influenza viruses were observed with intramuscular and intradermal routes of vaccine administration. The peptide vaccine induced cross-reactive antibodies that recognized influenza virus subtypes A/H1N1, A/H3N2, A/H5N1, B/Victoria, and B/Yamagata. In addition, immune sera neutralized seasonal and pandemic influenza strains (Group 1 and Group 2). This composite multi-epitope peptide vaccine, formulated with ALFQ and administered via intramuscular and intradermal routes, provides a high-performance supra-seasonal vaccine that would be cost-effective and easily scalable, thus moving us closer to a viable strategy for a universal influenza vaccine and pandemic preparedness.
Collapse
Affiliation(s)
- Nimisha Rikhi
- Longhorn Vaccines and Diagnostics, Gaithersburg, MD 20878, USA; (N.R.); (K.A.K.); (K.M.); (A.A.-D.); (G.W.F.)
| | - Clara J. Sei
- Longhorn Vaccines and Diagnostics, Gaithersburg, MD 20878, USA; (N.R.); (K.A.K.); (K.M.); (A.A.-D.); (G.W.F.)
| | - Mangala Rao
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA; (M.R.); (G.R.M.); (C.L.); (E.H.); (O.J.); (C.R.A.)
| | | | - Kellie A. Kroscher
- Longhorn Vaccines and Diagnostics, Gaithersburg, MD 20878, USA; (N.R.); (K.A.K.); (K.M.); (A.A.-D.); (G.W.F.)
| | - Gary R. Matyas
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA; (M.R.); (G.R.M.); (C.L.); (E.H.); (O.J.); (C.R.A.)
| | - Kevin Muema
- Longhorn Vaccines and Diagnostics, Gaithersburg, MD 20878, USA; (N.R.); (K.A.K.); (K.M.); (A.A.-D.); (G.W.F.)
| | - Camille Lange
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA; (M.R.); (G.R.M.); (C.L.); (E.H.); (O.J.); (C.R.A.)
- Henry M Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD 20817, USA
| | - Aba Assiaw-Dufu
- Longhorn Vaccines and Diagnostics, Gaithersburg, MD 20878, USA; (N.R.); (K.A.K.); (K.M.); (A.A.-D.); (G.W.F.)
| | - Elizabeth Hussin
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA; (M.R.); (G.R.M.); (C.L.); (E.H.); (O.J.); (C.R.A.)
- Henry M Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD 20817, USA
| | - Ousman Jobe
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA; (M.R.); (G.R.M.); (C.L.); (E.H.); (O.J.); (C.R.A.)
- Henry M Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD 20817, USA
| | - Carl R. Alving
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA; (M.R.); (G.R.M.); (C.L.); (E.H.); (O.J.); (C.R.A.)
| | - Gerald W. Fischer
- Longhorn Vaccines and Diagnostics, Gaithersburg, MD 20878, USA; (N.R.); (K.A.K.); (K.M.); (A.A.-D.); (G.W.F.)
| |
Collapse
|
20
|
Hendy DA, Lifshits LM, Batty CJ, Carlock MA, Ross TM, Mousa JJ, Bachelder EM, Ainslie KM. Zinc Carnosine Metal-Organic Coordination Polymer as a Potent Broadly Active Influenza Vaccine Platform with In Vitro Shelf-Stability. Mol Pharm 2023; 20:4687-4697. [PMID: 37603310 PMCID: PMC11654055 DOI: 10.1021/acs.molpharmaceut.3c00424] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/22/2023]
Abstract
Current seasonal influenza vaccines are limited in that they need to be reformulated every year in order to account for the constant mutation of the virus. Hemagglutinin (HA) immunogens have been developed using a computationally optimized broadly reactive antigen (COBRA) methodology, which are able to elicit an antibody response that neutralizes antigenically distinct influenza strains; however, subunit proteins are not immunogenic enough on their own to generate a substantial immune response. Due to this, different delivery strategies and adjuvants can be used to improve immunogenicity. Recently, we reported a new coordination polymer composed of the dipeptide carnosine and zinc (ZnCar) that is able to deliver protein antigens along with CpG to generate a potent immune response. In the present work, ZnCar was used to deliver the COBRA HA immunogen Y2 and the adjuvant CpG. We incorporated Y2 into ZnCar using two different methods to assess which would be the most immunogenic. Mice vaccinated with Y2 and CpG complexed with ZnCar showed an improved humoral and cellular response when compared to mice vaccinated with soluble Y2 and CpG. Further, we demonstrate in vitro that when Y2 and CpG are coordinated with ZnCar, they are protected from degradation at 40 °C for 3 months or 24 °C for 6 months. Overall, ZnCar shows promise as a delivery vehicle for subunit vaccines, given its superior immunogenicity and in vitro storage stability.
Collapse
Affiliation(s)
- Dylan A. Hendy
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, USA
| | - Liubov M. Lifshits
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, USA
| | - Cole J. Batty
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, USA
| | - Michael A. Carlock
- Florida Research and Innovation Center, Port Saint, Cleveland Clinic Florida, Port St. Lucie, FL, USA
| | - Ted M. Ross
- Florida Research and Innovation Center, Port Saint, Cleveland Clinic Florida, Port St. Lucie, FL, USA
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, USA
- Department of Infectious Diseases, University of Georgia, Athens, GA, USA
| | - Jarrod J. Mousa
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, USA
- Department of Infectious Diseases, University of Georgia, Athens, GA, USA
| | - Eric M. Bachelder
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, USA
| | - Kristy M. Ainslie
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, USA
- Department of Microbiology and Immunology, UNC School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
21
|
Luo J, Zhang Z, Zhao S, Gao R. A Comparison of Etiology, Pathogenesis, Vaccinal and Antiviral Drug Development between Influenza and COVID-19. Int J Mol Sci 2023; 24:ijms24076369. [PMID: 37047339 PMCID: PMC10094131 DOI: 10.3390/ijms24076369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 03/15/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
Influenza virus and coronavirus, two kinds of pathogens that exist widely in nature, are common emerging pathogens that cause respiratory tract infections in humans. In December 2019, a novel coronavirus SARS-CoV-2 emerged, causing a severe respiratory infection named COVID-19 in humans, and raising a global pandemic which has persisted in the world for almost three years. Influenza virus, a seasonally circulating respiratory pathogen, has caused four global pandemics in humans since 1918 by the emergence of novel variants. Studies have shown that there are certain similarities in transmission mode and pathogenesis between influenza and COVID-19, and vaccination and antiviral drugs are considered to have positive roles as well as several limitations in the prevention and control of both diseases. Comparative understandings would be helpful to the prevention and control of these diseases. Here, we review the study progress in the etiology, pathogenesis, vaccine and antiviral drug development for the two diseases.
Collapse
|
22
|
Li X, Xu Y, Li W, Che J, Zhao X, Cao R, Li X, Li S. Design, Synthesis, Biological Evaluation, and Molecular Dynamics Simulation of Influenza Polymerase PB2 Inhibitors. Molecules 2023; 28:molecules28041849. [PMID: 36838837 PMCID: PMC9960307 DOI: 10.3390/molecules28041849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 01/15/2023] [Accepted: 01/29/2023] [Indexed: 02/18/2023] Open
Abstract
The PB2 subunit of the influenza RNA-dependent RNA polymerase (RdRp) has been identified as a promising target for the treatment of influenza. To expand the chemical space of the known influenza polymerase PB2 inhibitor-pimodivir (formerly VX-787) and improve its pharmacokinetic profile, two pimodivir analogs containing 2,3-dihydro-imidazopyridine fragment (comp. I and comp. II) were designed, synthesized, and evaluated for anti-influenza virus activity. In the cytopathic effect (CPE) inhibition assay, comp. I and comp. II showed IC50 values of 0.07 and 0.09 μM for A/Puerto Rico/8/34 (H1N1) and 0.04 and 0.07 μM for A/Hong Kong/8/68 (H3N2), respectively. Protein-binding affinity assay results showed a concentration-dependent association and dissociation pattern, with KD values of 1.398 and 1.670 μM, respectively. In vitro metabolic stability assays showed that comp. I and comp. II exhibited good stability to liver microsomes and considerably less sensitivity to aldehyde oxidase compared to pimodivir. The binding modes of comp. I and comp. II were similar to those of VX-787; however, comp. I and comp. II had lower structural adaptability to PB2 than VX-787. Our results provide helpful information regarding the structure-activity relationship for the design of novel PB2 inhibitors and a reference for the development of drugs containing 2,3-dihydro-imidazopyridine fragments.
Collapse
Affiliation(s)
- Xinhong Li
- National Engineering Research Center for the Emergency Strategic Drug, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Yijie Xu
- National Engineering Research Center for the Emergency Strategic Drug, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Wei Li
- National Engineering Research Center for the Emergency Strategic Drug, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Jinjing Che
- National Engineering Research Center for the Emergency Strategic Drug, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Xu Zhao
- Department of Hepatology, Fifth Medical Center of Chinese PLA General Hospital, Beijing 100039, China
- China Military Institute of Chinese Materia, Fifth Medical Center of Chinese PLA General Hospital, Beijing 100053, China
| | - Ruyuan Cao
- National Engineering Research Center for the Emergency Strategic Drug, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
- Correspondence: (R.C.); (X.L.); (S.L.); Tel.: +86-10-66930673 (ext. 717) (R.C.); +86-10-66930634 (X.L.); +86-10-66930250 (S.L.)
| | - Xingzhou Li
- National Engineering Research Center for the Emergency Strategic Drug, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
- Correspondence: (R.C.); (X.L.); (S.L.); Tel.: +86-10-66930673 (ext. 717) (R.C.); +86-10-66930634 (X.L.); +86-10-66930250 (S.L.)
| | - Song Li
- National Engineering Research Center for the Emergency Strategic Drug, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
- Correspondence: (R.C.); (X.L.); (S.L.); Tel.: +86-10-66930673 (ext. 717) (R.C.); +86-10-66930634 (X.L.); +86-10-66930250 (S.L.)
| |
Collapse
|
23
|
Rumfelt KE, Fitzsimmons WJ, Truscon R, Monto AS, Martin ET, Lauring AS. A rapid and flexible microneutralization assay for serological assessment of influenza viruses. Influenza Other Respir Viruses 2023; 17:e13141. [PMID: 37127782 PMCID: PMC10174083 DOI: 10.1111/irv.13141] [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: 02/03/2023] [Revised: 04/12/2023] [Accepted: 04/13/2023] [Indexed: 05/03/2023] Open
Abstract
BACKGROUND Serological responses from influenza vaccination or infection are typically measured by hemagglutinin inhibition (HAI) or microneutralization (MN). Both methods are limited in feasibility, standardization, and generalizability to recent strains. We developed a luciferase MN (LMN) assay that combines the advantages of the conventional MN assay with the ease of the HAI assay. METHODS Sera were obtained from the HIVE study, a Michigan household cohort. Reverse genetics was used to generate recombinant influenza viruses expressing the hemagglutinin and neuraminidase of test strains, all other viral proteins from an A/WSN/1933 backbone, and a NanoLuc reporter. Serum neutralization of luciferase-expressing targets was quantified as a reduction in light emission from infected cells. Neutralization titers were measured for cell- and egg-adapted versions of A/Hong Kong/4801/2014 and A/Singapore/INFIMH-16-0019/2016 and compared to HAI titers against egg-grown antigens. RESULTS Three hundred thirty-three sera were collected from 259 participants between May 2016 and July 2018. Sampled participants were 7-68 years of age, and >80% were vaccinated against influenza. HAI and LMN titers were correlated for A/Hong Kong/4801/2014 (ρ = 0.52, p ≤ 0.01) and A/Singapore/INFIMH-16-0019/2016 (ρ = 0.79, p ≤ 0.01). LMN titers were lower for cell strains compared to egg strains (A/Hong Kong/4801/2014 mean log2 fold change = -2.66, p ≤ 0.01 and A/Singapore/INFIMH-16-0019/2016 mean log2 fold change = -3.15, p ≤ 0.01). CONCLUSIONS The LMN assay was feasible using limited sample volumes and able to differentiate small antigenic differences between egg-adapted and cell-derived strains. The correspondence of these results with the commonly used HAI confirms the utility of this assay for high-throughput studies of correlates of protection and vaccine response.
Collapse
Affiliation(s)
- Kalee E. Rumfelt
- Department of EpidemiologyUniversity of MichiganAnn ArborMichiganUSA
| | - William J. Fitzsimmons
- Division of Infectious Diseases, Department of Internal MedicineUniversity of MichiganAnn ArborMichiganUSA
| | - Rachel Truscon
- Department of EpidemiologyUniversity of MichiganAnn ArborMichiganUSA
| | - Arnold S. Monto
- Department of EpidemiologyUniversity of MichiganAnn ArborMichiganUSA
| | - Emily T. Martin
- Department of EpidemiologyUniversity of MichiganAnn ArborMichiganUSA
| | - Adam S. Lauring
- Division of Infectious Diseases, Department of Internal MedicineUniversity of MichiganAnn ArborMichiganUSA
- Department of Microbiology and ImmunologyUniversity of MichiganAnn ArborMichiganUSA
| |
Collapse
|
24
|
Rockman S, Laurie K, Ong C, Rajaram S, McGovern I, Tran V, Youhanna J. Cell-Based Manufacturing Technology Increases Antigenic Match of Influenza Vaccine and Results in Improved Effectiveness. Vaccines (Basel) 2022; 11:52. [PMID: 36679895 PMCID: PMC9861528 DOI: 10.3390/vaccines11010052] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 12/19/2022] [Accepted: 12/22/2022] [Indexed: 12/28/2022] Open
Abstract
To ensure that vaccination offers the best protection against an infectious disease, sequence identity between the vaccine and the circulating strain is paramount. During replication of nucleic acid, random mutations occur due to the level of polymerase fidelity. In traditional influenza vaccine manufacture, vaccine viruses are propagated in fertilized chicken eggs, which can result in egg-adaptive mutations in the antigen-encoding genes. Whilst this improves infection and replication in eggs, mutations may reduce the effectiveness of egg-based influenza vaccines against circulating human viruses. In contrast, egg-adaptive mutations are avoided when vaccine viruses are propagated in Madin-Darby canine kidney (MDCK) cell lines during manufacture of cell-based inactivated influenza vaccines. The first mammalian cell-only strain was included in Flucelvax® Quadrivalent in 2017. A sequence analysis of the viruses selected for inclusion in this vaccine (n = 15 vaccine strains, containing both hemagglutinin and neuraminidase) demonstrated that no mutations occur in the antigenic sites of either hemagglutinin or neuraminidase, indicating that cell adaptation does not occur during production of this cell-based vaccine. The development of this now entirely mammalian-based vaccine system, which incorporates both hemagglutinin and neuraminidase, ensures that the significant protective antigens are equivalent to the strains recommended by the World Health Organization (WHO) in both amino acid sequence and glycosylation pattern. The inclusion of both proteins in a vaccine may provide an advantage over recombinant vaccines containing hemagglutinin alone. Findings from real world effectiveness studies support the use of cell-based influenza vaccines.
Collapse
Affiliation(s)
- Steven Rockman
- CSL Seqirus Ltd., Parkville, VIC 3050, Australia
- Department of Immunology and Microbiology, The University of Melbourne, Parkville, VIC 3050, Australia
| | - Karen Laurie
- CSL Seqirus Ltd., Parkville, VIC 3050, Australia
| | - Chi Ong
- CSL Seqirus Ltd., Parkville, VIC 3050, Australia
| | | | | | - Vy Tran
- CSL Seqirus Ltd., Kirkland, QC H9H 4M7, Canada
| | | |
Collapse
|
25
|
Rahman S, Hasan M, Alam MS, Uddin KMM, Moni S, Rahman M. The evolutionary footprint of influenza A subtype H3N2 strains in Bangladesh: implication of vaccine strain selection. Sci Rep 2022; 12:16186. [PMID: 36171388 PMCID: PMC9519982 DOI: 10.1038/s41598-022-20179-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 09/09/2022] [Indexed: 11/09/2022] Open
Abstract
In February each year, World Health Organization (WHO) recommends candidate vaccine viruses for the forthcoming northern hemisphere (NH) season; however, the influenza season in the temperate zone of NH begins in October. During egg- or cell culture-propagation, the vaccine viruses become too old to confer the highest match with the latest strains, impacting vaccine effectiveness. Therefore, an alternative strategy like mRNA-based vaccine using the most recent strains should be considered. We analyzed influenza A subtype H3N2 strains circulating in NH during the last 10 years (2009-2020). Phylogenetic analysis revealed multiple clades of influenza strains circulating every season, which had substantial mismatches with WHO-recommended vaccine strains. The clustering pattern suggests that influenza A subtype H3N2 strains are not fixed to the specific geographical region but circulate globally in the same season. By analyzing 39 seasons from eight NH countries with the highest vaccine coverage, we also provide evidence that the influenza A, subtype H3N2 strains from South and Southeast Asia, including Bangladesh, had the highest genetic proximity to the NH strains. Furthermore, insilico analysis showed minimal effect on the Bangladeshi HA protein structure, indicating the stability of Bangladeshi strains. Therefore, we propose that Bangladeshi influenza strains represent genetic makeup that may better fit and serve as the most suitable candidate vaccine viruses for the forthcoming NH season.
Collapse
Affiliation(s)
- Sezanur Rahman
- Virology Laboratory, Infectious Diseases Division, icddr,b, Mohakhali, 68 Shaheed Tajuddin Ahmed Sarani, Dhaka, 1212, Bangladesh
| | - Mehedi Hasan
- Virology Laboratory, Infectious Diseases Division, icddr,b, Mohakhali, 68 Shaheed Tajuddin Ahmed Sarani, Dhaka, 1212, Bangladesh
| | - Md Shaheen Alam
- Virology Laboratory, Infectious Diseases Division, icddr,b, Mohakhali, 68 Shaheed Tajuddin Ahmed Sarani, Dhaka, 1212, Bangladesh
| | - K M Main Uddin
- Virology Laboratory, Infectious Diseases Division, icddr,b, Mohakhali, 68 Shaheed Tajuddin Ahmed Sarani, Dhaka, 1212, Bangladesh
| | - Sayra Moni
- Virology Laboratory, Infectious Diseases Division, icddr,b, Mohakhali, 68 Shaheed Tajuddin Ahmed Sarani, Dhaka, 1212, Bangladesh
| | - Mustafizur Rahman
- Virology Laboratory, Infectious Diseases Division, icddr,b, Mohakhali, 68 Shaheed Tajuddin Ahmed Sarani, Dhaka, 1212, Bangladesh.
- Genomics Centre, icddr,b, Mohakhali, Dhaka, 1212, Bangladesh.
| |
Collapse
|
26
|
Sinilaite A, Papenburg J. Summary of the National Advisory Committee on Immunization (NACI) Seasonal Influenza Vaccine Statement for 2022-2023. CANADA COMMUNICABLE DISEASE REPORT = RELEVE DES MALADIES TRANSMISSIBLES AU CANADA 2022; 48:373-382. [PMID: 38116046 PMCID: PMC10729780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/21/2023]
Abstract
Background The National Advisory Committee on Immunization (NACI) reviews the evolving evidence on influenza immunization and provides annual recommendations regarding the use of authorized seasonal influenza vaccines to the Public Health Agency of Canada. Objective To summarize the NACI seasonal influenza vaccine recommendations for 2022-2023 and to highlight new recommendations and supporting evidence. Methods In the preparation of the Statement on Seasonal Influenza Vaccine for 2022-2023, NACI's Influenza Working Group followed the NACI evidence-based process for developing recommendations. The recommendations were then considered and approved by NACI in light of the available evidence. Results The following key updates and new recommendations have been made for the 2022-2023 season: 1) updated information/guidance on influenza vaccination in the context of the coronavirus disease 2019 (COVID-19) has been incorporated; 2) Supemtek™ recombinant influenza vaccine may be considered for use among the quadrivalent influenza vaccines offered to adults 18 years of age and older for annual influenza immunization; and 3) Flucelvax® Quad may be considered among the quadrivalent influenza vaccines offered to adults and children two years of age and older. Conclusion NACI continues to recommend that an age-appropriate influenza vaccine should be offered annually for all individuals aged six months of age and older who do not have contraindications to the vaccine, with particular focus on people at high risk of influenza-related complications or hospitalization, people capable of transmitting influenza to those at high risk, and other groups for whom influenza vaccination is particularly recommended.
Collapse
Affiliation(s)
- Angela Sinilaite
- Centre for Immunization Readiness, Public Health Agency of Canada, Ottawa, ON
| | - Jesse Papenburg
- NACI Influenza Working Group Chair
- Division of Pediatric Infectious Diseases, Department of Pediatrics, Montréal Children's Hospital of the McGill University Health Centre, Montréal, QC
- Division of Microbiology, Department of Clinical Laboratory Medicine, Optilab Montréal - McGill University Health Centre, Montréal, QC
- Department of Epidemiology, Biostatistics, and Occupational Health, School of Population and Global Health, McGill University, Montréal, QC
| |
Collapse
|
27
|
Ju H, Hou L, Zhao F, Zhang Y, Jia R, Guizzo L, Bonomini A, Zhang J, Gao Z, Liang R, Bertagnin C, Kong X, Ma X, Kang D, Loregian A, Huang B, Liu X, Zhan P. Iterative Optimization and Structure-Activity Relationship Studies of Oseltamivir Amino Derivatives as Potent and Selective Neuraminidase Inhibitors via Targeting 150-Cavity. J Med Chem 2022; 65:11550-11573. [PMID: 35939763 DOI: 10.1021/acs.jmedchem.1c01970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
With our continuous endeavors in seeking neuraminidase (NA) inhibitors, we reported herein three series of novel oseltamivir amino derivatives with the goal of exploring the druggable chemical space inside the 150-cavity of influenza virus NAs. Among them, around half of the compounds in series C were demonstrated to be better inhibitors against both wild-type and oseltamivir-resistant group-1 NAs than oseltamivir carboxylate (OSC). Notably, compounds 12d, 12e, 15e, and 15i showed more potent or equipotent antiviral activity against H1N1, H5N1, and H5N8 viruses compared to OSC in cellular assays. Furthermore, compounds 12e and 15e exhibited high metabolic stability in human liver microsomes (HLMs) and low inhibitory effect on main cytochrome P450 (CYP) enzymes, as well as low acute/subacute toxicity and certain antiviral efficacy in vivo. Also, pharmacokinetic (PK) and molecular docking studies were performed. Overall, 12e and 15e possess great potential to serve as anti-influenza candidates and are worthy of further investigation.
Collapse
Affiliation(s)
- Han Ju
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, 250012 Jinan, Shandong, P. R. China
| | - Lingxin Hou
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, 250012 Jinan, Shandong, P. R. China
| | - Fabao Zhao
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, 250012 Jinan, Shandong, P. R. China
| | - Ying Zhang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, 250012 Jinan, Shandong, P. R. China
| | - Ruifang Jia
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, 250012 Jinan, Shandong, P. R. China
| | - Laura Guizzo
- Department of Molecular Medicine, University of Padova, Via Gabelli 63, 35121 Padova, Italy
| | - Anna Bonomini
- Department of Molecular Medicine, University of Padova, Via Gabelli 63, 35121 Padova, Italy
| | - Jiwei Zhang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, 250012 Jinan, Shandong, P. R. China
| | - Zhen Gao
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, 250012 Jinan, Shandong, P. R. China
| | - Ruipeng Liang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, 250012 Jinan, Shandong, P. R. China
| | - Chiara Bertagnin
- Department of Molecular Medicine, University of Padova, Via Gabelli 63, 35121 Padova, Italy
| | - Xiujie Kong
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, 250012 Jinan, Shandong, P. R. China
| | - Xiuli Ma
- Institute of Poultry Science, Shandong Academy of Agricultural Sciences, 202 North Gongye Road, 250100 Jinan, Shandong, P. R. China
| | - Dongwei Kang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, 250012 Jinan, Shandong, P. R. China
| | - Arianna Loregian
- Department of Molecular Medicine, University of Padova, Via Gabelli 63, 35121 Padova, Italy
| | - Bing Huang
- Institute of Poultry Science, Shandong Academy of Agricultural Sciences, 202 North Gongye Road, 250100 Jinan, Shandong, P. R. China
| | - Xinyong Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, 250012 Jinan, Shandong, P. R. China
| | - Peng Zhan
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, 250012 Jinan, Shandong, P. R. China
| |
Collapse
|
28
|
Lee J, Kim KS, Na K. Intranasal administration of an aronia extract and carrageenan nanocomposite for the prevention of influenza A H1N1 virus infection. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2022. [DOI: 10.1007/s40005-022-00591-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
29
|
Wang WC, Sayedahmed EE, Sambhara S, Mittal SK. Progress towards the Development of a Universal Influenza Vaccine. Viruses 2022; 14:v14081684. [PMID: 36016306 PMCID: PMC9415875 DOI: 10.3390/v14081684] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 07/22/2022] [Accepted: 07/28/2022] [Indexed: 11/21/2022] Open
Abstract
Influenza viruses are responsible for millions of cases globally and significantly threaten public health. Since pandemic and zoonotic influenza viruses have emerged in the last 20 years and some of the viruses have resulted in high mortality in humans, a universal influenza vaccine is needed to provide comprehensive protection against a wide range of influenza viruses. Current seasonal influenza vaccines provide strain-specific protection and are less effective against mismatched strains. The rapid antigenic drift and shift in influenza viruses resulted in time-consuming surveillance and uncertainty in the vaccine protection efficacy. Most recent universal influenza vaccine studies target the conserved antigen domains of the viral surface glycoproteins and internal proteins to provide broader protection. Following the development of advanced vaccine technologies, several innovative strategies and vaccine platforms are being explored to generate robust cross-protective immunity. This review provides the latest progress in the development of universal influenza vaccines.
Collapse
Affiliation(s)
- Wen-Chien Wang
- Department of Comparative Pathobiology, Purdue Institute for Immunology, Inflammation and Infectious Disease, and Purdue University Center for Cancer Research, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47907, USA; (W.-C.W.); (E.E.S.)
| | - Ekramy E. Sayedahmed
- Department of Comparative Pathobiology, Purdue Institute for Immunology, Inflammation and Infectious Disease, and Purdue University Center for Cancer Research, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47907, USA; (W.-C.W.); (E.E.S.)
| | - Suryaprakash Sambhara
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA
- Correspondence: (S.S.); (S.K.M.)
| | - Suresh K. Mittal
- Department of Comparative Pathobiology, Purdue Institute for Immunology, Inflammation and Infectious Disease, and Purdue University Center for Cancer Research, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47907, USA; (W.-C.W.); (E.E.S.)
- Correspondence: (S.S.); (S.K.M.)
| |
Collapse
|
30
|
Abstract
Vaccines targeting SARS-CoV-2 have been shown to be highly effective; however, the breadth against emerging variants and the longevity of protection remains unclear. Postimmunization boosting has been shown to be beneficial for disease protection, and as new variants continue to emerge, periodic (and perhaps annual) vaccination will likely be recommended. New seasonal influenza virus vaccines currently need to be developed every year due to continual antigenic drift, an undertaking made possible by a robust global vaccine production and distribution infrastructure. To create a seasonal combination vaccine targeting both influenza viruses and SARS-CoV-2 that is also amenable to frequent reformulation, we have developed an influenza A virus (IAV) genetic platform that allows the incorporation of an immunogenic domain of the SARS-CoV-2 spike (S) protein onto IAV particles. Vaccination with this combination vaccine elicited neutralizing antibodies and provided protection from lethal challenge with both pathogens in mice. This approach may allow the leveraging of established influenza vaccine infrastructure to generate a cost-effective and scalable seasonal vaccine solution for both influenza and coronaviruses. IMPORTANCE The rapid emergence of SARS-CoV-2 variants since the onset of the pandemic has highlighted the need for both periodic vaccination “boosts” and a platform that can be rapidly reformulated to manufacture new vaccines. In this work, we report an approach that can utilize current influenza vaccine manufacturing infrastructure to generate combination vaccines capable of protecting from both influenza virus- and SARS-CoV-2-induced disease. The production of a combined influenza/SARS-CoV-2 vaccine may represent a practical solution to boost immunity to these important respiratory viruses without the increased cost and administration burden of multiple independent vaccines.
Collapse
|
31
|
Trombetta CM, Marchi S, Montomoli E. The baculovirus expression vector system: a modern technology for the future of influenza vaccine manufacturing. Expert Rev Vaccines 2022; 21:1233-1242. [DOI: 10.1080/14760584.2022.2085565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Affiliation(s)
| | - Serena Marchi
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Emanuele Montomoli
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
- VisMederi srl, Siena, Italy
- VisMederi Research srl, Siena, Italy
| |
Collapse
|
32
|
Abstract
mRNA vaccines have brought about a great revolution in the vaccine fields owing to their simplicity and adaptability in antigen design, potential to induce both humoral and cell-mediated immune responses and demonstrated high efficacy, and rapid and low-cost production by using the same manufacturing platform for different mRNA vaccines. Multiple mRNA vaccines have been investigated for both infectious diseases and cancers, showing significant superiority to other types of vaccines. Although great success of mRNA vaccines has been achieved in the control of the coronavirus disease 2019 pandemic, there are still multiple challenges for the future development of mRNA vaccines. In this review, the most recent developments of mRNA vaccines against both infectious diseases and cancers are summarized for an overview of this field. Moreover, the challenges are also discussed on the basis of these developments.
Collapse
Affiliation(s)
- Jinjin Chen
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, USA;
| | - Jianzhu Chen
- Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA;
| | - Qiaobing Xu
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, USA;
| |
Collapse
|
33
|
Goyal G, Prabhala P, Mahajan G, Bausk B, Gilboa T, Xie L, Zhai Y, Lazarovits R, Mansour A, Kim MS, Patil A, Curran D, Long JM, Sharma S, Junaid A, Cohen L, Ferrante TC, Levy O, Prantil‐Baun R, Walt DR, Ingber DE. Ectopic Lymphoid Follicle Formation and Human Seasonal Influenza Vaccination Responses Recapitulated in an Organ-on-a-Chip. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2103241. [PMID: 35289122 PMCID: PMC9109055 DOI: 10.1002/advs.202103241] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 12/08/2021] [Indexed: 05/13/2023]
Abstract
Lymphoid follicles (LFs) are responsible for generation of adaptive immune responses in secondary lymphoid organs and form ectopically during chronic inflammation. A human model of ectopic LF formation will provide a tool to understand LF development and an alternative to non-human primates for preclinical evaluation of vaccines. Here, it is shown that primary human blood B- and T-lymphocytes autonomously assemble into ectopic LFs when cultured in a 3D extracellular matrix gel within one channel of a two-channel organ-on-a-chip microfluidic device. Superfusion via a parallel channel separated by a microporous membrane is required for LF formation and prevents lymphocyte autoactivation. These germinal center-like LFs contain B cells expressing Activation-Induced Cytidine Deaminase and exhibit plasma cell differentiation upon activation. To explore their utility for seasonal vaccine testing, autologous monocyte-derived dendritic cells are integrated into LF Chips. The human LF chips demonstrate improved antibody responses to split virion influenza vaccination compared to 2D cultures, which are enhanced by a squalene-in-water emulsion adjuvant, and this is accompanied by increases in LF size and number. When inoculated with commercial influenza vaccine, plasma cell formation and production of anti-hemagglutinin IgG are observed, as well as secretion of cytokines similar to vaccinated humans over clinically relevant timescales.
Collapse
Affiliation(s)
- Girija Goyal
- Wyss Institute for Biologically Inspired Engineering at Harvard UniversityBostonMA02115USA
| | - Pranav Prabhala
- Wyss Institute for Biologically Inspired Engineering at Harvard UniversityBostonMA02115USA
| | - Gautam Mahajan
- Wyss Institute for Biologically Inspired Engineering at Harvard UniversityBostonMA02115USA
| | - Bruce Bausk
- Wyss Institute for Biologically Inspired Engineering at Harvard UniversityBostonMA02115USA
- Department of PathologyBrigham and Women's Hospital and Harvard Medical SchoolBostonMA02115USA
| | - Tal Gilboa
- Wyss Institute for Biologically Inspired Engineering at Harvard UniversityBostonMA02115USA
- Department of PathologyBrigham and Women's Hospital and Harvard Medical SchoolBostonMA02115USA
| | - Liangxia Xie
- Wyss Institute for Biologically Inspired Engineering at Harvard UniversityBostonMA02115USA
- Department of PathologyBrigham and Women's Hospital and Harvard Medical SchoolBostonMA02115USA
| | - Yunhao Zhai
- Wyss Institute for Biologically Inspired Engineering at Harvard UniversityBostonMA02115USA
| | - Roey Lazarovits
- Wyss Institute for Biologically Inspired Engineering at Harvard UniversityBostonMA02115USA
| | - Adam Mansour
- Wyss Institute for Biologically Inspired Engineering at Harvard UniversityBostonMA02115USA
| | - Min Sun Kim
- Wyss Institute for Biologically Inspired Engineering at Harvard UniversityBostonMA02115USA
| | - Aditya Patil
- Wyss Institute for Biologically Inspired Engineering at Harvard UniversityBostonMA02115USA
| | - Danielle Curran
- Wyss Institute for Biologically Inspired Engineering at Harvard UniversityBostonMA02115USA
| | - Jaclyn M. Long
- Wyss Institute for Biologically Inspired Engineering at Harvard UniversityBostonMA02115USA
| | - Sanjay Sharma
- Wyss Institute for Biologically Inspired Engineering at Harvard UniversityBostonMA02115USA
| | - Abidemi Junaid
- Wyss Institute for Biologically Inspired Engineering at Harvard UniversityBostonMA02115USA
| | - Limor Cohen
- Wyss Institute for Biologically Inspired Engineering at Harvard UniversityBostonMA02115USA
- Department of PathologyBrigham and Women's Hospital and Harvard Medical SchoolBostonMA02115USA
| | - Thomas C. Ferrante
- Wyss Institute for Biologically Inspired Engineering at Harvard UniversityBostonMA02115USA
| | - Oren Levy
- Wyss Institute for Biologically Inspired Engineering at Harvard UniversityBostonMA02115USA
| | - Rachelle Prantil‐Baun
- Wyss Institute for Biologically Inspired Engineering at Harvard UniversityBostonMA02115USA
| | - David R. Walt
- Wyss Institute for Biologically Inspired Engineering at Harvard UniversityBostonMA02115USA
- Department of PathologyBrigham and Women's Hospital and Harvard Medical SchoolBostonMA02115USA
| | - Donald E. Ingber
- Wyss Institute for Biologically Inspired Engineering at Harvard UniversityBostonMA02115USA
- Vascular Biology Program and Department of SurgeryBoston Children's Hospital and Harvard Medical SchoolBostonMA02115USA
- Harvard John A. Paulson School of Engineering and Applied SciencesHarvard UniversityCambridgeMA02139USA
| |
Collapse
|
34
|
Flitter BA, Braun MR, Tucker SN. Drop the Needle; A Temperature Stable Oral Tablet Vaccine Is Protective against Respiratory Viral Pathogens. Vaccines (Basel) 2022; 10:593. [PMID: 35455342 PMCID: PMC9031097 DOI: 10.3390/vaccines10040593] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 04/04/2022] [Accepted: 04/06/2022] [Indexed: 02/07/2023] Open
Abstract
To effectively combat emerging infections and prevent future pandemics, next generation vaccines must be developed quickly, manufactured rapidly, and most critically, administered easily. Next generation vaccines need innovative approaches that prevent infection, severe disease, and reduce community transmission of respiratory pathogens such as influenza and SARS-CoV-2. Here we review an oral vaccine tablet that can be manufactured and released in less than 16 weeks of antigen design and deployed without the need for cold chain. The oral Ad5 modular vaccine platform utilizes a non-replicating adenoviral vector (rAd5) containing a novel molecular TLR3 adjuvant that is delivered by tablet, not by needle. This enterically coated, room temperature-stable vaccine tablet elicits robust antigen-specific IgA in the gastrointestinal and respiratory tracts and upregulates mucosal homing adhesion molecules on circulating B and T cells. Several influenza antigens have been tested using this novel vaccine approach and demonstrated efficacy in both preclinical animal models and in phase I/II clinical trials, including in a human challenge study. This oral rAd5 vaccine platform technology offers a promising new avenue for aiding in rapid pandemic preparedness and equitable worldwide vaccine distribution.
Collapse
Affiliation(s)
- Becca A. Flitter
- Vaxart, Inc., South San Francisco, CA 94080, USA; (M.R.B.); (S.N.T.)
| | | | | |
Collapse
|
35
|
Divino V, Ruthwik Anupindi V, DeKoven M, Mould-Quevedo J, Pelton SI, Postma MJ, Levin MJ. A Real-World Clinical and Economic Analysis of Cell-Derived Quadrivalent Influenza Vaccine Compared to Standard Egg-Derived Quadrivalent Influenza Vaccines During the 2019-2020 Influenza Season in the United States. Open Forum Infect Dis 2022; 9:ofab604. [PMID: 35028334 PMCID: PMC8753033 DOI: 10.1093/ofid/ofab604] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 12/01/2021] [Indexed: 11/13/2022] Open
Abstract
Background Cell-derived influenza vaccines are not subject to egg-adaptive mutations that have potential to decrease vaccine effectiveness. This retrospective analysis estimated the relative vaccine effectiveness (rVE) of cell-derived quadrivalent influenza vaccine (IIV4c) compared to standard egg-derived quadrivalent influenza vaccines (IIV4e) among recipients aged 4-64 years in the United States during the 2019-2020 influenza season. Methods The IQVIA PharMetrics Plus administrative claims database was utilized. Study outcomes were assessed postvaccination through the end of the study period (7 March 2020). Inverse probability of treatment weighting (IPTW) was implemented to adjust for covariate imbalance. Adjusted rVE against influenza-related hospitalizations/emergency room (ER) visits and other clinical outcomes was estimated through IPTW-weighted Poisson regression models for the IIV4c and IIV4e cohorts and for the subgroup with ≥1 high-risk condition. Sensitivity analyses modifying the outcome assessment period as well as a doubly-robust analysis were also conducted. IPTW-weighted generalized linear models were used to estimate predicted annualized all-cause costs. Results The final sample comprised 1 150 134 IIV4c and 3 924 819 IIV4e recipients following IPTW adjustment. IIV4c was more effective in preventing influenza-related hospitalizations/ER visits as well as respiratory-related hospitalizations/ER visits compared to IIV4e. IIV4c was also more effective for the high-risk subgroup and across the sensitivity analyses. IIV4c was also associated with significantly lower annualized all-cause total costs compared to IIV4e (-$467), driven by lower costs for outpatient medical services and inpatient hospitalizations. Conclusions IIV4c was significantly more effective in preventing influenza-related hospitalizations/ER visits compared to IIV4e and was associated with significantly lower all-cause costs.
Collapse
Affiliation(s)
| | | | - Mitch DeKoven
- Real World Solutions, IQVIA, Falls Church, Virginia, USA
| | | | - Stephen I Pelton
- Department of Pediatrics, Boston University School of Medicine, Boston, Massachusetts, USA.,Division of Pediatric Infectious Diseases, Maxwell Finland Laboratory, Boston Medical Center, Boston, Massachusetts, USA
| | - Maarten J Postma
- Department of PharmacoTherapy, Epidemiology and Economics (PTE2), Groningen Research Institute of Pharmacy, University of Groningen, Groningen, The Netherlands.,Department of Health Sciences, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands.,Department of Economics, Econometrics and Finance, Faculty of Economics and Business, University of Groningen, Groningen, The Netherlands
| | - Myron J Levin
- Departments of Pediatrics and Medicine, Anschutz Medical Campus, University of Colorado, Aurora, Colorado, USA
| |
Collapse
|
36
|
Kang M, Zanin M, Wong SS. Subtype H3N2 Influenza A Viruses: An Unmet Challenge in the Western Pacific. Vaccines (Basel) 2022; 10:vaccines10010112. [PMID: 35062773 PMCID: PMC8778411 DOI: 10.3390/vaccines10010112] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 01/07/2022] [Accepted: 01/07/2022] [Indexed: 02/04/2023] Open
Abstract
Subtype H3N2 influenza A viruses (A(H3N2)) have been the dominant strain in some countries in the Western Pacific region since the 2009 influenza A(H1N1) pandemic. Vaccination is the most effective way to prevent influenza; however, low vaccine effectiveness has been reported in some influenza seasons, especially for A(H3N2). Antigenic mismatch introduced by egg-adaptation during vaccine production between the vaccine and circulating viral stains is one of the reasons for low vaccine effectiveness. Here we review the extent of this phenomenon, the underlying molecular mechanisms and discuss recent strategies to ameliorate this, including new vaccine platforms that may provide better protection and should be considered to reduce the impact of A(H3N2) in the Western Pacific region.
Collapse
Affiliation(s)
- Min Kang
- School of Public Health, Southern Medical University, Guangzhou 510515, China;
- Guangdong Center for Disease Control and Prevention, Guangzhou 511430, China
| | - Mark Zanin
- State Key Laboratory for Respiratory Diseases and National Clinical Research Centre for Respiratory Disease, Guangzhou Medical University, 195 Dongfengxi Road, Guangzhou 511436, China;
- School of Public Health, The University of Hong Kong, 7 Sassoon Road, Pokfulam, Hong Kong, China
| | - Sook-San Wong
- State Key Laboratory for Respiratory Diseases and National Clinical Research Centre for Respiratory Disease, Guangzhou Medical University, 195 Dongfengxi Road, Guangzhou 511436, China;
- School of Public Health, The University of Hong Kong, 7 Sassoon Road, Pokfulam, Hong Kong, China
- Correspondence: ; Tel.: +86-178-2584-6078
| |
Collapse
|
37
|
Zahmati S, Taghizadeh M, Haghighat S, Jalalirad R, Mahdavi M. Recombinant hemagglutinin of swine H1N1 influenza virus expression in the insect cells: Formulation in Montanide ISA71 adjuvant and the potency studies. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2021; 24:1546-1553. [PMID: 35317113 PMCID: PMC8917850 DOI: 10.22038/ijbms.2021.57053.12716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 09/25/2021] [Indexed: 11/10/2022]
Abstract
Objectives Influenza is a highly contagious disease, which affects the respiratory system and seasonal influenza is common throughout the world. Influenza vaccination is an effective way to reduce the risk of death and hospitalization. This study aims at the expression of swine recombinant hemagglutinin protein in the baculovirus expression system and it offers a comparison of the immunologic parameters with the commercial vaccine. Materials and Methods The HA gene from the swine H1N1 strain of the Influenza virus was cloned into the Bac-To-Bac expression system in pFastBAC HTA vector and was transformed into Escherichia coli TOP10 strain. After the confirmation, the vector was transfected into the SF9 insect cell line. The recombinant HA was evaluated by SDS-PAGE and western blot. After formulation in Montanide ISA71 adjuvant, the immunization test was performed comparatively with Alum adjuvant, commercial vaccine in four groups of BALB/c mice, of which one group was control without any vaccination. Two weeks after the last immunization, the antibody response was assessed with HI assay, and experimental mice were challenged with mouse-adapted Influenza A/PR8/34 (H1N1) virus through nasal inhalation. Results The immunoassay results revealed that the candidate vaccine induced the antibody response as the commercial one did but it did not significantly reduce the mortality rate, body loss, and severe fever. Conclusion To summarize, the results showed that the recombinant protein with the MontanideTM ISA- 71 adjuvant developed a more appropriate level of immunity than Alum adjuvant, so it might be used as a safe and reliable vaccine against H1N1 virus for further research.
Collapse
Affiliation(s)
- Sara Zahmati
- Department of Microbiology, Faculty of Advanced science and Technology, Tehran Medical sciences Islamic Azad University, Tehran, Iran
| | - Morteza Taghizadeh
- Department of Research and Development, Razi Vaccine and Serum Research institute, Agricultural Research Education and Extension Organization
| | - Setareh Haghighat
- Department of Microbiology, Faculty of Advanced science and Technology, Tehran Medical sciences Islamic Azad University, Tehran, Iran
| | | | - Mehdi Mahdavi
- Production and Research Complex, Pasteur Institute of Iran, Karaj, Iran
- Advanced Therapy Medicinal Product Department, Breast Cancer Research Center, Motamed Cancer Institute, Academic Center for Education
- Culture and Research, Tehran, Iran
- Recombinant Vaccine Research Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
38
|
Ortiz de Lejarazu-Leonardo R, Montomoli E, Wojcik R, Christopher S, Mosnier A, Pariani E, Trilla Garcia A, Fickenscher H, Gärtner BC, Jandhyala R, Zambon M, Moore C. Estimation of Reduction in Influenza Vaccine Effectiveness Due to Egg-Adaptation Changes-Systematic Literature Review and Expert Consensus. Vaccines (Basel) 2021; 9:1255. [PMID: 34835186 PMCID: PMC8621612 DOI: 10.3390/vaccines9111255] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 10/14/2021] [Accepted: 10/15/2021] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND Influenza vaccines are the main tool to prevent morbidity and mortality of the disease; however, egg adaptations associated with the choice of the manufacturing process may reduce their effectiveness. This study aimed to estimate the impact of egg adaptations and antigenic drift on the effectiveness of trivalent (TIV) and quadrivalent (QIV) influenza vaccines. METHODS Nine experts in influenza virology were recruited into a Delphi-style exercise. In the first round, the experts were asked to answer questions on the impact of antigenic drift and egg adaptations on vaccine match (VM) and influenza vaccine effectiveness (IVE). In the second round, the experts were presented with the data from a systematic literature review on the same subject and aggregated experts' responses to round one questions. The experts were asked to review and confirm or amend their responses before the final summary statistics were calculated. RESULTS The experts estimated that, across Europe, the egg adaptations reduce, on average, VM to circulating viruses by 7-21% and reduce IVE by 4-16%. According to the experts, antigenic drift results in a similar impact on VM (8-24%) and IVE (5-20%). The highest reduction in IVE was estimated for the influenza virus A(H3N2) subtype for the under 65 age group. When asked about the frequency of the phenomena, the experts indicated that, on average, between the 2014 and 19 seasons, egg adaptation and antigenic drift were significant enough to impact IVE that occurred in two and three out of five seasons, respectively. They also agreed that this pattern is likely to reoccur in future seasons. CONCLUSIONS Expert estimates suggest there is a potential for 9% on average (weighted average of "All strains" over three age groups adjusted by population size) and up to a 16% increase in IVE (against A(H3N2), the <65 age group) if egg adaptations that arise when employing the traditional egg-based manufacturing process are avoided.
Collapse
Affiliation(s)
| | - Emanuele Montomoli
- Department of Molecular Medicine, University of Siena, 53100 Siena, Italy;
| | - Radek Wojcik
- Medialis Ltd., Banbury OX16 0AH, UK; (S.C.); (R.J.)
| | | | | | - Elena Pariani
- Department of Biomedical Science for Health, University of Milan, 20122 Milan, Italy;
| | - Antoni Trilla Garcia
- Preventive Medicine and Epidemiology, Hospital Clínic, University of Barcelona, 08007 Barcelona, Spain;
| | - Helmut Fickenscher
- Institute for Infection Medicine, Kiel University, 24118 Kiel, Germany;
- University Medical Center Schleswig-Holstein, 24105 Kiel, Germany
| | - Barbara C. Gärtner
- Institute for Microbiology and Hygiene, Saarland University, Faculty of Medicine and Medical Center, Building 43, 66421 Homburg/Saar, Germany;
| | | | | | - Catherine Moore
- Wales Specialist Virology Centre, Public Health Wales, Cardiff CF14 4XW, UK;
| |
Collapse
|
39
|
Moore KA, Ostrowsky JT, Kraigsley AM, Mehr AJ, Bresee JS, Friede MH, Gellin BG, Golding JP, Hart PJ, Moen A, Weller CL, Osterholm MT. A Research and Development (R&D) roadmap for influenza vaccines: Looking toward the future. Vaccine 2021; 39:6573-6584. [PMID: 34602302 DOI: 10.1016/j.vaccine.2021.08.010] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 07/30/2021] [Accepted: 08/03/2021] [Indexed: 12/24/2022]
Abstract
Improved influenza vaccines are urgently needed to reduce the burden of seasonal influenza and to ensure a rapid and effective public-health response to future influenza pandemics. The Influenza Vaccines Research and Development (R&D) Roadmap (IVR) was created, through an extensive international stakeholder engagement process, to promote influenza vaccine R&D. The roadmap covers a 10-year timeframe and is organized into six sections: virology; immunology; vaccinology for seasonal influenza vaccines; vaccinology for universal influenza vaccines; animal and human influenza virus infection models; and policy, finance, and regulation. Each section identifies barriers, gaps, strategic goals, milestones, and additional R&D priorities germane to that area. The roadmap includes 113 specific R&D milestones, 37 of which have been designated high priority by the IVR expert taskforce. This report summarizes the major issues and priority areas of research outlined in the IVR. By identifying the key issues and steps to address them, the roadmap not only encourages research aimed at new solutions, but also provides guidance on the use of innovative tools to drive breakthroughs in influenza vaccine R&D.
Collapse
Affiliation(s)
- Kristine A Moore
- Center for Infectious Disease Research and Policy, University of Minnesota, Minneapolis, MN, USA; Center for Infectious Disease Research and Policy, C315 Mayo Memorial Building, MMC 263, 420 Delaware Street, SE, Minneapolis, MN 55455, USA.
| | - Julia T Ostrowsky
- Center for Infectious Disease Research and Policy, University of Minnesota, Minneapolis, MN, USA
| | - Alison M Kraigsley
- Center for Infectious Disease Research and Policy, University of Minnesota, Minneapolis, MN, USA
| | - Angela J Mehr
- Center for Infectious Disease Research and Policy, University of Minnesota, Minneapolis, MN, USA
| | - Joseph S Bresee
- The Global Funders Consortium for Universal Influenza Vaccine Development, The Task Force for Global Health, and the US Centers for Disease Control and Prevention, Atlanta, GA, USA
| | | | | | | | | | - Ann Moen
- World Health Organization, Geneva, Switzerland
| | | | - Michael T Osterholm
- Center for Infectious Disease Research and Policy, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
40
|
Targovnik AM, Simonin JA, Mc Callum GJ, Smith I, Cuccovia Warlet FU, Nugnes MV, Miranda MV, Belaich MN. Solutions against emerging infectious and noninfectious human diseases through the application of baculovirus technologies. Appl Microbiol Biotechnol 2021; 105:8195-8226. [PMID: 34618205 PMCID: PMC8495437 DOI: 10.1007/s00253-021-11615-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 09/20/2021] [Accepted: 09/21/2021] [Indexed: 12/23/2022]
Abstract
Abstract
Baculoviruses are insect pathogens widely used as biotechnological tools in different fields of life sciences and technologies. The particular biology of these entities (biosafety viruses 1; large circular double-stranded DNA genomes, infective per se; generally of narrow host range on insect larvae; many of the latter being pests in agriculture) and the availability of molecular-biology procedures (e.g., genetic engineering to edit their genomes) and cellular resources (availability of cell lines that grow under in vitro culture conditions) have enabled the application of baculoviruses as active ingredients in pest control, as systems for the expression of recombinant proteins (Baculovirus Expression Vector Systems—BEVS) and as viral vectors for gene delivery in mammals or to display antigenic proteins (Baculoviruses applied on mammals—BacMam). Accordingly, BEVS and BacMam technologies have been introduced in academia because of their availability as commercial systems and ease of use and have also reached the human pharmaceutical industry, as incomparable tools in the development of biological products such as diagnostic kits, vaccines, protein therapies, and—though still in the conceptual stage involving animal models—gene therapies. Among all the baculovirus species, the Autographa californica multiple nucleopolyhedrovirus has been the most highly exploited in the above utilities for the human-biotechnology field. This review highlights the main achievements (in their different stages of development) of the use of BEVS and BacMam technologies for the generation of products for infectious and noninfectious human diseases. Key points • Baculoviruses can assist as biotechnological tools in human health problems. • Vaccines and diagnosis reagents produced in the baculovirus platform are described. • The use of recombinant baculovirus for gene therapy–based treatment is reviewed.
Collapse
Affiliation(s)
- Alexandra Marisa Targovnik
- Cátedra de Biotecnología, Departamento de Microbiología, Inmunología, Biotecnología y Genética, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junín 956, Buenos Aires, 1113, Argentina.
- Instituto de Nanobiotecnología (NANOBIOTEC), Facultad de Farmacia y Bioquímica, CONICET -Universidad de Buenos Aires, Junín 956, Sexto Piso, C1113AAD, 1113, Buenos Aires, Argentina.
| | - Jorge Alejandro Simonin
- Laboratorio de Ingeniería Genética y Biología Celular y Molecular, Área Virosis de Insectos, Instituto de Microbiología Básica y Aplicada, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Buenos Aires, Argentina
| | - Gregorio Juan Mc Callum
- Cátedra de Biotecnología, Departamento de Microbiología, Inmunología, Biotecnología y Genética, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junín 956, Buenos Aires, 1113, Argentina
- Instituto de Nanobiotecnología (NANOBIOTEC), Facultad de Farmacia y Bioquímica, CONICET -Universidad de Buenos Aires, Junín 956, Sexto Piso, C1113AAD, 1113, Buenos Aires, Argentina
| | - Ignacio Smith
- Cátedra de Biotecnología, Departamento de Microbiología, Inmunología, Biotecnología y Genética, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junín 956, Buenos Aires, 1113, Argentina
- Instituto de Nanobiotecnología (NANOBIOTEC), Facultad de Farmacia y Bioquímica, CONICET -Universidad de Buenos Aires, Junín 956, Sexto Piso, C1113AAD, 1113, Buenos Aires, Argentina
| | - Franco Uriel Cuccovia Warlet
- Laboratorio de Ingeniería Genética y Biología Celular y Molecular, Área Virosis de Insectos, Instituto de Microbiología Básica y Aplicada, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Buenos Aires, Argentina
| | - María Victoria Nugnes
- Laboratorio de Ingeniería Genética y Biología Celular y Molecular, Área Virosis de Insectos, Instituto de Microbiología Básica y Aplicada, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Buenos Aires, Argentina
| | - María Victoria Miranda
- Cátedra de Biotecnología, Departamento de Microbiología, Inmunología, Biotecnología y Genética, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junín 956, Buenos Aires, 1113, Argentina
- Instituto de Nanobiotecnología (NANOBIOTEC), Facultad de Farmacia y Bioquímica, CONICET -Universidad de Buenos Aires, Junín 956, Sexto Piso, C1113AAD, 1113, Buenos Aires, Argentina
| | - Mariano Nicolás Belaich
- Laboratorio de Ingeniería Genética y Biología Celular y Molecular, Área Virosis de Insectos, Instituto de Microbiología Básica y Aplicada, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Buenos Aires, Argentina
| |
Collapse
|
41
|
Yokomizo S, Katagiri W, Maki Y, Sano T, Inoue K, Fukushi M, Atochin DN, Kushibiki T, Kawana A, Kimizuka Y, Kashiwagi S. Brief exposure of skin to near-infrared laser augments early vaccine responses. NANOPHOTONICS 2021; 10:3187-3197. [PMID: 34868804 PMCID: PMC8635068 DOI: 10.1515/nanoph-2021-0133] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Rapid establishment of herd immunity with vaccination is effective to combat emerging infectious diseases. Although the incorporation of adjuvant and intradermal (ID) injection could augment early responses to the vaccine, the current chemical or biological adjuvants are inappropriate for this purpose with their side effects and high reactogenicity in the skin. Recently, a near-infrared (NIR) laser has been shown to augment the immune response to ID vaccination and could be alternatively used for mass vaccination programs. Here, we determined the effect of NIR laser as well as licensed chemical adjuvants on the immunogenicity 1, 2, and 4 weeks after ID influenza vaccination in mice. The NIR laser adjuvant augmented early antibody responses, while the widely used alum adjuvant induced significantly delayed responses. In addition, the oil-in-water and alum adjuvants, but not the NIR laser, elicited escalated TH2 responses with allergenic immunoglobulin E (IgE) responses. The effect of the NIR laser was significantly suppressed in the basic leucine zipper transcription factor ATF-like 3 (Batf3) knockout mice, suggesting a critical role of the cluster of differentiation 103+ (CD103)+ dendritic cells. The current preliminary study suggests that NIR laser adjuvant is an alternative strategy to chemical and biological agents to timely combat emerging infectious diseases. Moreover, its immunomodulatory property could be used to enhance the efficacy of immunotherapy for allergy and cancer.
Collapse
Affiliation(s)
- Shinya Yokomizo
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, 149 13th Street, Charlestown 02129, MA, USA
- Department of Radiological Sciences, Tokyo Metropolitan University, 7-2-10 Higashi-Ogu, Arakawa 116-8551, Tokyo, Japan
| | - Wataru Katagiri
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, 149 13th Street, Charlestown 02129, MA, USA
- Graduate School of Science and Technology, Keio University, 3-14-1 Hiyoshi, Yokohama 223-8522, Kanagawa, Japan
| | - Yohei Maki
- Division of Infectious Diseases and Respiratory Medicine, Department of Medicine, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama 359-8513, Japan
| | - Tomoya Sano
- Division of Infectious Diseases and Respiratory Medicine, Department of Medicine, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama 359-8513, Japan
| | - Kazumasa Inoue
- Department of Radiological Sciences, Tokyo Metropolitan University, 7-2-10 Higashi-Ogu, Arakawa 116-8551, Tokyo, Japan
| | - Masahiro Fukushi
- Department of Radiological Sciences, Tokyo Metropolitan University, 7-2-10 Higashi-Ogu, Arakawa 116-8551, Tokyo, Japan
| | - Dmitriy N. Atochin
- Cardiovascular Research Center, Department of Medicine, Massachusetts General Hospital, 149 13th Street, Charlestown 02129, MA, USA
| | - Toshihiro Kushibiki
- Department of Medical Engineering, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama 359-8513, Japan
| | - Akihiko Kawana
- Division of Infectious Diseases and Respiratory Medicine, Department of Medicine, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama 359-8513, Japan
| | | | | |
Collapse
|
42
|
An Antigenic Thrift-Based Approach to Influenza Vaccine Design. Vaccines (Basel) 2021; 9:vaccines9060657. [PMID: 34208489 PMCID: PMC8235769 DOI: 10.3390/vaccines9060657] [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: 04/16/2021] [Revised: 06/04/2021] [Accepted: 06/05/2021] [Indexed: 11/19/2022] Open
Abstract
The antigenic drift theory states that influenza evolves via the gradual accumulation of mutations, decreasing a host’s immune protection against previous strains. Influenza vaccines are designed accordingly, under the premise of antigenic drift. However, a paradox exists at the centre of influenza research. If influenza evolved primarily through mutation in multiple epitopes, multiple influenza strains should co-circulate. Such a multitude of strains would render influenza vaccines quickly inefficacious. Instead, a single or limited number of strains dominate circulation each influenza season. Unless additional constraints are placed on the evolution of influenza, antigenic drift does not adequately explain these observations. Here, we explore the constraints placed on antigenic drift and a competing theory of influenza evolution – antigenic thrift. In contrast to antigenic drift, antigenic thrift states that immune selection targets epitopes of limited variability, which constrain the variability of the virus. We explain the implications of antigenic drift and antigenic thrift and explore their current and potential uses in the context of influenza vaccine design.
Collapse
|
43
|
B Carvalho S, Peixoto C, T Carrondo MJ, S Silva RJ. Downstream processing for influenza vaccines and candidates: An update. Biotechnol Bioeng 2021; 118:2845-2869. [PMID: 33913510 DOI: 10.1002/bit.27803] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 04/10/2021] [Accepted: 04/16/2021] [Indexed: 02/07/2023]
Abstract
Seasonal and pandemic influenza outbreaks present severe health and economic burdens. To overcome limitations on influenza vaccines' availability and effectiveness, researchers chase universal vaccines providing broad, long-lasting protection against multiple influenza subtypes, and including pandemic ones. Novel influenza vaccine designs are under development, in clinical trials, or reaching the market, namely inactivated, or live-attenuated virus, virus-like particles, or recombinant antigens, searching for improved effectiveness; all these bring downstream processing (DSP) new challenges. Having to deal with new influenza strains, including pandemics, requires shorter development time, driving the development of faster bioprocesses. To cope with better upstream processes, new regulatory demands for quality and safety, and cost reduction requirements, new unit operations and integrated processes are increasing DSP efficiency for novel vaccine formats. This review covers recent advances in DSP strategies of different influenza vaccine formats. Focus is given to the improvements on relevant state-of-the-art unit operations, from harvest and clarification to purification steps, ending with sterile filtration and formulation. The development of more efficient unit operations to cope with biophysical properties of the new candidates is discussed: emphasis is given to the design of new stationary phases, 3D printing approaches, and continuous processing tools, such as continuous chromatography. The impact of the production platforms and vaccine designs on the downstream operations for the different influenza vaccine formats approved for this season are highlighted.
Collapse
Affiliation(s)
- Sofia B Carvalho
- Animal Cell Technology Unit, iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal.,Animal Cell Technology Unit, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Cristina Peixoto
- Animal Cell Technology Unit, iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal.,Animal Cell Technology Unit, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Manuel J T Carrondo
- Animal Cell Technology Unit, iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
| | - Ricardo J S Silva
- Animal Cell Technology Unit, iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal.,Animal Cell Technology Unit, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| |
Collapse
|
44
|
Engineered influenza virions reveal the contributions of non-hemagglutinin structural proteins to vaccine mediated protection. J Virol 2021; 95:JVI.02021-20. [PMID: 33658342 PMCID: PMC8139674 DOI: 10.1128/jvi.02021-20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The development of improved and universal anti-influenza vaccines would represent a major advance in the protection of human health. In order to facilitate the development of such vaccines, understanding how viral proteins can contribute to protection from disease is critical. Much of the previous work to address these questions relied on reductionist systems (i.e. vaccinating with individual proteins or VLPs that contain only a few viral proteins); thus we have an incomplete understanding of how immunity to different subsets of viral proteins contribute to protection. Here, we report the development of a platform in which a single viral protein can be deleted from an authentic viral particle that retains the remaining full complement of structural proteins and viral RNA. As a first study with this system, we chose to delete the major IAV antigen, the hemagglutinin protein, to evaluate how the other components of the viral particle contribute en masse to protection from influenza disease. Our results show that while anti-HA immunity plays a major role in protection from challenge with a vaccine-matched strain, the contributions from other structural proteins were the major drivers of protection against highly antigenically drifted, homosubtypic strains. This work highlights the importance of evaluating the inclusion of non-HA viral proteins in the development of broadly efficacious and long-lasting influenza vaccines.ImportanceInfluenza virus vaccines currently afford short-term protection from viruses that are closely related to the vaccine strains. There is currently much effort to develop improved, next-generation influenza vaccines that elicit broader and longer-lasting protection. While the hemagglutinin protein is the major viral antigen, in this work, we developed an approach with which to evaluate the contributions of the non-hemagglutinin proteins to vaccine mediated protection. Our results indicate that other structural proteins together may help to mediate broad antiviral protection and should be considered in the development of more universal influenza vaccines.
Collapse
|
45
|
de Lusignan S, Hoang U, Liyanage H, Tripathy M, Sherlock J, Joy M, Ferreira F, Diez-Domingo J, Clark T. Using Point of Care Testing to estimate influenza vaccine effectiveness in the English primary care sentinel surveillance network. PLoS One 2021; 16:e0248123. [PMID: 33705452 PMCID: PMC7951853 DOI: 10.1371/journal.pone.0248123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 02/19/2021] [Indexed: 11/29/2022] Open
Abstract
Introduction Rapid Point of Care Testing (POCT) for influenza could be used to provide information on influenza vaccine effectiveness (IVE) as well as influencing clinical decision-making in primary care. Methods We undertook a test negative case control study to estimate the overall and age-specific (6 months-17 years, 18–64 years, ≥65 years old) IVE against medically attended POCT-confirmed influenza. The study took place over the winter of 2019–2020 and was nested within twelve general practices that are part of the Oxford-Royal College of General Practitioners (RCGP) Research and Surveillance Centre (RSC), the English sentinel surveillance network. Results 648 POCT were conducted. 193 (29.7%) of those who were swabbed had received the seasonal influenza vaccine. The crude unadjusted overall IVE was 46.1% (95% CI: 13.9–66.3). After adjusting for confounders the overall IVE was 26.0% (95% CI: 0–65.5). In total 211 patients were prescribed an antimicrobial after swab testing. Given a positive influenza POCT result, the odds ratio (OR) of receiving an antiviral was 21.1 (95%CI: 2.4–182.2, p = <0.01) and the OR of being prescribed an antibiotic was 0.6 (95%CI: 0.4–0.9, p = <0.01). Discussion Using influenza POCT in a primary care sentinel surveillance network to estimate IVE is feasible and provides comparable results to published IVE estimates. A further advantage is that near patient testing of influenza is associated with improvements in appropriate antiviral and antibiotic use. Larger, randomised studies are needed in primary care to see if these trends are still present and to explore their impact on outcomes.
Collapse
Affiliation(s)
- Simon de Lusignan
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, United Kingdom
- * E-mail:
| | - Uy Hoang
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, United Kingdom
| | - Harshana Liyanage
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, United Kingdom
| | - Manasa Tripathy
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, United Kingdom
| | - Julian Sherlock
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, United Kingdom
| | - Mark Joy
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, United Kingdom
| | - Filipa Ferreira
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, United Kingdom
| | | | - Tristan Clark
- Academic Unit of Clinical and Experimental Sciences, University of Southampton, Southampton, United Kingdom
| |
Collapse
|
46
|
Lu G, Shan S, Zainab B, Ayaz Z, He J, Xie Z, Rashid U, Zhang D, Mehmood Abbasi A. Novel vaccine design based on genomics data analysis: A review. Scand J Immunol 2021; 93:e12986. [PMID: 33043473 DOI: 10.1111/sji.12986] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 10/04/2020] [Accepted: 10/05/2020] [Indexed: 12/28/2022]
Abstract
Modification of pathogenic strains with the passage of time is responsible for evolution in the timeline of vaccine development for last 30 years. Recent advancements in computational vaccinology on the one hand and genome sequencing approaches on the other have generated new hopes in vaccine development. The aim of this review was to discuss the evolution of vaccines, their characteristics and limitations. In this review, we highlighted the evolution of vaccines, from first generation to the current status, pointing out how different vaccines have emerged and different approaches that are being followed up in the development of more rational vaccines against a wide range of diseases. Data were collected using Google Scholar, Web of Science, Science Direct, Web of Knowledge, Scopus and Science Hub, whereas computational tools such as NCBI, GeneMANIA and STRING were used to analyse the pathways of vaccine action. Innovative tools, such as computational tools, recombinant technologies and intra-dermal devices, are currently being investigated in order to improve the immunological response. New technologies enlightened the interactions of host proteins with pathogenic proteins for vaccine candidate development, but still there is a need of integrating transcriptomic and proteomic approaches. Although immunization with genomics data is a successful approach, its advantages must be assessed case by case and its applicability depends on the nature of the agent to be immunized, the nature of the antigen and the type of immune response required to achieve effective protection.
Collapse
Affiliation(s)
- Guangli Lu
- Institute of Business, School of Business, Henan University, Henan, China
| | - Sharui Shan
- The First Affiliated Hospital of Jinan University (Guangzhou Overseas Chinese Hospital), Guangzhou, China
- Department of Rehabilitation, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Bibi Zainab
- Department of Environmental Sciences, COMSATS University Islamabad, Islamabad, Pakistan
| | - Zainab Ayaz
- Department of Environmental Sciences, COMSATS University Islamabad, Islamabad, Pakistan
| | - Jialiang He
- School of Food and Bioengineering, Henan University of Science and Technology, Luoyang, China
| | - Zhenxing Xie
- Basic School of Medicine, Henan University, Kaifeng, China
| | - Umer Rashid
- Department of Chemistry, COMSATS University Islamabad, Islamabad, Pakistan
| | - Dalin Zhang
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center, University of California Davis, Sacramento, CA, USA
| | - Arshad Mehmood Abbasi
- Department of Environmental Sciences, COMSATS University Islamabad, Islamabad, Pakistan
| |
Collapse
|
47
|
Bartoszko J, Loeb M. The burden of influenza in older adults: meeting the challenge. Aging Clin Exp Res 2021; 33:711-717. [PMID: 31347085 DOI: 10.1007/s40520-019-01279-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 07/17/2019] [Indexed: 12/20/2022]
Abstract
Influenza is an acute respiratory infection for which vaccination is our best prevention strategy. Small seasonal changes in circulating influenza viruses (antigenic drift) result in the need for annual influenza vaccination, in which the vaccine formulation is updated to better match the predominant circulating influenza viruses that have undergone important antigenic changes. Although the burden of influenza infection and its complications is the highest in older adults, vaccine effectiveness is the lowest in this vulnerable population. This is largely due to waning of the immune response with age known as "immune senescence", and presents an important, unmet challenge. Possible strategies to tackle this include adjuvant and high-dose vaccines, and herd immunity induced by greater vaccine uptake.
Collapse
Affiliation(s)
- Jessica Bartoszko
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON, L8N 3Z5, Canada
| | - Mark Loeb
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON, L8N 3Z5, Canada.
| |
Collapse
|
48
|
Clinical and Economic Outcomes Associated with Cell-Based Quadrivalent Influenza Vaccine vs. Standard-Dose Egg-Based Quadrivalent Influenza Vaccines during the 2018-19 Influenza Season in the United States. Vaccines (Basel) 2021; 9:vaccines9020080. [PMID: 33498724 PMCID: PMC7911861 DOI: 10.3390/vaccines9020080] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 01/16/2021] [Accepted: 01/17/2021] [Indexed: 12/16/2022] Open
Abstract
Non-egg-based influenza vaccines eliminate the potential for egg-adapted mutations and potentially increase vaccine effectiveness. This retrospective study compared hospitalizations/emergency room (ER) visits and all-cause annualized healthcare costs among subjects aged 4–64 years who received cell-based quadrivalent (QIVc) or standard-dose egg-based quadrivalent (QIVe-SD) influenza vaccine during the 2018–19 influenza season. Administrative claims data (IQVIA PharMetrics® Plus, IQVIA, USA) were utilized to evaluate clinical and economic outcomes. Adjusted relative vaccine effectiveness (rVE) of QIVc vs. QIVe-SD among overall cohort, as well as for three subgroups (age 4–17 years, age 18–64 years, and high-risk) was evaluated using inverse probability of treatment weighting (IPTW) and Poisson regression models. Generalized estimating equation models among the propensity score matched sample were used to estimate annualized all-cause costs. A total of 669,030 recipients of QIVc and 3,062,797 of QIVe-SD were identified after IPTW adjustments. Among the overall cohort, QIVc had higher adjusted rVEs against hospitalizations/ER visits related to influenza, all-cause hospitalizations, and hospitalizations/ER visits associated with any respiratory event compared to QIVe-SD. The adjusted annualized all-cause total costs were higher for QIVe-SD compared to QIVc ((+$461); p < 0.05).
Collapse
|
49
|
Tsilibary EP, Charonis SA, Georgopoulos AP. Vaccines for Influenza. Vaccines (Basel) 2021; 9:vaccines9010047. [PMID: 33466608 PMCID: PMC7828733 DOI: 10.3390/vaccines9010047] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/11/2021] [Accepted: 01/12/2021] [Indexed: 11/16/2022] Open
Affiliation(s)
- Effie-Photini Tsilibary
- Brain Sciences Center, Department of Veterans Affairs Health Care System, Minneapolis, MN 55417, USA; (E.-P.T.); (S.A.C.)
- Department of Neuroscience, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Spyros A. Charonis
- Brain Sciences Center, Department of Veterans Affairs Health Care System, Minneapolis, MN 55417, USA; (E.-P.T.); (S.A.C.)
- Department of Neuroscience, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Apostolos P. Georgopoulos
- Brain Sciences Center, Department of Veterans Affairs Health Care System, Minneapolis, MN 55417, USA; (E.-P.T.); (S.A.C.)
- Department of Neuroscience, University of Minnesota Medical School, Minneapolis, MN 55455, USA
- Correspondence:
| |
Collapse
|
50
|
Wu Y, Jia H, Lai H, Liu X, Tan WS. Highly efficient production of an influenza H9N2 vaccine using MDCK suspension cells. BIORESOUR BIOPROCESS 2020. [DOI: 10.1186/s40643-020-00352-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
AbstractThe use of H9N2 subtype avian influenza vaccines is an effective approach for the control of the virus spread among the poultry, and for the upgrading of vaccine manufacturing, cell culture-based production platform could overcome the limitations of conventional egg-based platform and alternate it. The development of serum-free suspension cell culture could allow even higher virus productivity, where a suspension cell line with good performance and proper culture strategies are required. In this work, an adherent Mardin–Darby canine kidney (MDCK) cell line was adapted to suspension growth to cell concentration up to 12 × 106 cells/mL in a serum-free medium in batch cultures. Subsequently, the H9N2 influenza virus propagation in this MDCK cell line was evaluated with the optimization of infection conditions in terms of MOI and cell concentration for infection. Furthermore, various feed strategies were tested in the infection phase for improved virus titer and a maximum hemagglutinin titer of 13 log2 (HAU/50 μL) was obtained using the 1:2 medium dilution strategy. The evaluation of MDCK cell growth and H9N2 virus production in bioreactors with optimized operating conditions showed comparable cell performance and virus yield compared to shake flasks, with a high cell-specific virus yield above 13,000 virions/cell. With the purified H9N2 virus harvested from the bioreactors, the MDCK cell-derived vaccine was able to induce high titers of neutralizing antibodies in chickens. Overall, the results demonstrate the promising application of the highly efficient MDCK cell-based production platform for the avian influenza vaccine manufacturing.
Collapse
|