1
|
Hashimoto M, Tsujii K, Nakajima-Yoshida H, Akiyama N, Yoshihara K, Dohi K, Yin Z, Ejima A, Yamamoto-Mizuno S, Nojiri Y, Saiki S, Baba K, Omoto S. A-910823, a squalene-based emulsion adjuvant, enhances robust and broad immune responses of quadrivalent influenza vaccine in ferrets. Vaccine 2025; 49:126780. [PMID: 39889536 DOI: 10.1016/j.vaccine.2025.126780] [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: 11/19/2024] [Revised: 01/10/2025] [Accepted: 01/20/2025] [Indexed: 02/03/2025]
Abstract
Robust and broad protective immunity is typically elicited by co-administration of an adjuvant with vaccine antigens. A-910823, a squalene-based emulsion adjuvant, has been approved in Japan. It effectively enhances the humoral and cellular immunity of S-268019-b, a recombinant COVID-19 vaccine; however, the adjuvant effects of A-910823 on other vaccines, including influenza vaccine, have not been evaluated. Here, a ferret model was employed to investigate the adjuvant effects of A-910823 when combined with split-inactivated quadrivalent influenza vaccine (QIV). We demonstrate that combination of A-910823 with QIV enhances the neutralizing antibody titer and its breadth against non-vaccine strains. Also, the protective efficacy of A-910823-adjuvanted QIV against virus challenge has been confirmed. Of note, QIV combined with A-910823 caused only minor detectable side effects, whereas a significant increase in fever was observed after two doses of mRNA-LNP in ferrets. This study provides information on the effectiveness and safety of A-910823-adjuvanted QIV and suggests the usefulness of the ferret model for evaluating vaccine-induced reactogenicity.
Collapse
MESH Headings
- Animals
- Ferrets/immunology
- Influenza Vaccines/immunology
- Influenza Vaccines/administration & dosage
- Squalene/administration & dosage
- Squalene/immunology
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- Adjuvants, Immunologic/administration & dosage
- Antibodies, Neutralizing/blood
- Antibodies, Neutralizing/immunology
- Emulsions
- Adjuvants, Vaccine/administration & dosage
- Female
- Orthomyxoviridae Infections/prevention & control
- Orthomyxoviridae Infections/immunology
- Male
- Disease Models, Animal
- Immunity, Humoral/drug effects
- COVID-19/prevention & control
- COVID-19/immunology
- Vaccines, Inactivated/immunology
- Vaccines, Inactivated/administration & dosage
Collapse
Affiliation(s)
- Masayuki Hashimoto
- Shionogi & Co., Ltd., Vaccine Business Division, 1-1, Futaba-cho 3-chome, Toyonaka, Osaka 561-0825, Japan.
| | - Kenichiro Tsujii
- Shionogi & Co., Ltd., Vaccine Business Division, 1-1, Futaba-cho 3-chome, Toyonaka, Osaka 561-0825, Japan.
| | - Hitomi Nakajima-Yoshida
- Shionogi & Co., Ltd., Vaccine Business Division, 1-1, Futaba-cho 3-chome, Toyonaka, Osaka 561-0825, Japan.
| | - Nobuteru Akiyama
- Shionogi & Co., Ltd., Vaccine Business Division, 1-1, Futaba-cho 3-chome, Toyonaka, Osaka 561-0825, Japan.
| | - Ken Yoshihara
- Shionogi & Co., Ltd., Vaccine Business Division, 1-1, Futaba-cho 3-chome, Toyonaka, Osaka 561-0825, Japan.
| | - Keiji Dohi
- Shionogi & Co., Ltd., Vaccine Business Division, 1-1, Futaba-cho 3-chome, Toyonaka, Osaka 561-0825, Japan.
| | - Ziwei Yin
- Shionogi & Co., Ltd., Vaccine Business Division, 1-1, Futaba-cho 3-chome, Toyonaka, Osaka 561-0825, Japan.
| | - Aki Ejima
- Shionogi & Co., Ltd., Vaccine Business Division, 1-1, Futaba-cho 3-chome, Toyonaka, Osaka 561-0825, Japan.
| | - Saki Yamamoto-Mizuno
- Shionogi & Co., Ltd., Vaccine Business Division, 1-1, Futaba-cho 3-chome, Toyonaka, Osaka 561-0825, Japan.
| | - Yasushi Nojiri
- Shionogi TechnoAdvance Research Co., Ltd., 1-1, Futaba-cho 3-chome, Toyonaka, Osaka 561-0825, Japan.
| | - Shou Saiki
- Shionogi TechnoAdvance Research Co., Ltd., 1-1, Futaba-cho 3-chome, Toyonaka, Osaka 561-0825, Japan.
| | - Kaoru Baba
- Shionogi TechnoAdvance Research Co., Ltd., 1-1, Futaba-cho 3-chome, Toyonaka, Osaka 561-0825, Japan.
| | - Shinya Omoto
- Shionogi & Co., Ltd., Vaccine Business Division, 1-1, Futaba-cho 3-chome, Toyonaka, Osaka 561-0825, Japan.
| |
Collapse
|
2
|
Berber E, Ross TM. Factors Predicting COVID-19 Vaccine Effectiveness and Longevity of Humoral Immune Responses. Vaccines (Basel) 2024; 12:1284. [PMID: 39591186 PMCID: PMC11598945 DOI: 10.3390/vaccines12111284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 11/09/2024] [Accepted: 11/11/2024] [Indexed: 11/28/2024] Open
Abstract
The COVID-19 pandemic, caused by SARS-CoV-2, prompted global efforts to develop vaccines to control the disease. Various vaccines, including mRNA (BNT162b2, mRNA-1273), adenoviral vector (ChAdOx1, Ad26.COV2.S), and inactivated virus platforms (BBIBP-CorV, CoronaVac), elicit high-titer, protective antibodies against the virus, but long-term antibody durability and effectiveness vary. The objective of this study is to elucidate the factors that influence vaccine effectiveness (VE) and the longevity of humoral immune responses to COVID-19 vaccines through a review of the relevant literature, including clinical and real-world studies. Here, we discuss the humoral immune response to different COVID-19 vaccines and identify factors influencing VE and antibody longevity. Despite initial robust immune responses, vaccine-induced immunity wanes over time, particularly with the emergence of variants, such as Delta and Omicron, that exhibit immune escape mechanisms. Additionally, the durability of the humoral immune responses elicited by different vaccine platforms, along with the identification of essential determinants of long-term protection-like pre-existing immunity, booster doses, hybrid immunity, and demographic factors-are critical for protecting against severe COVID-19. Booster vaccinations substantially restore neutralizing antibody levels, especially against immune-evasive variants, while individuals with hybrid immunity have a more durable and potent immune response. Importantly, comorbidities such as diabetes, cardiovascular disease, chronic kidney disease, and cancer significantly reduce the magnitude and longevity of vaccine-induced protection. Immunocompromised individuals, particularly those undergoing chemotherapy and those with hematologic malignancies, have diminished humoral responses and benefit disproportionately from booster vaccinations. Age and sex also influence immune responses, with older adults experiencing accelerated antibody decline and females generally exhibiting stronger humoral responses compared to males. Understanding the variables affecting immune protection is crucial to improving vaccine strategies and predicting VE and protection against COVID-19.
Collapse
Affiliation(s)
- Engin Berber
- Infection Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA;
| | - Ted M. Ross
- Infection Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA;
- Florida Research and Innovation Center, Cleveland Clinic, Florida, FL 34986, 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
|
Sevim Bayrak C, Forst CV, Jones DR, Gresham DJ, Pushalkar S, Wu S, Vogel C, Mahal LK, Ghedin E, Ross T, García-Sastre A, Zhang B. Patient subtyping analysis of baseline multi-omic data reveals distinct pre-immune states associated with antibody response to seasonal influenza vaccination. Clin Immunol 2024; 266:110333. [PMID: 39089348 PMCID: PMC11340208 DOI: 10.1016/j.clim.2024.110333] [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: 06/18/2024] [Revised: 07/26/2024] [Accepted: 07/27/2024] [Indexed: 08/03/2024]
Abstract
Understanding the molecular mechanisms underpinning diverse vaccination responses is critical for developing efficient vaccines. Molecular subtyping can offer insights into heterogeneous nature of responses and aid in vaccine design. We analyzed multi-omic data from 62 haemagglutinin seasonal influenza vaccine recipients (2019-2020), including transcriptomics, proteomics, glycomics, and metabolomics data collected pre-vaccination. We performed a subtyping analysis on the integrated data revealing five subtypes with distinct molecular signatures. These subtypes differed in the expression of pre-existing adaptive or innate immunity signatures, which were linked to significant variation in baseline immunoglobulin A (IgA) and hemagglutination inhibition (HAI) titer levels. It is worth noting that these differences persisted through day 28 post-vaccination, indicating the effect of initial immune state on vaccination response. These findings highlight the significance of interpersonal variation in baseline immune status as a crucial factor in determining the effectiveness of seasonal vaccines. Ultimately, incorporating molecular profiling could enable personalized vaccine optimization.
Collapse
Affiliation(s)
- Cigdem Sevim Bayrak
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mt Sinai, New York, NY, USA; Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Christian V Forst
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mt Sinai, New York, NY, USA; Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Microbiology, Icahn School of Medicine at Mt Sinai, New York, NY, USA
| | - Drew R Jones
- Department of Biochemistry and Molecular Pharmacology, New York University Langone Health, NY, New York, USA
| | - David J Gresham
- Center for Genomics and Systems Biology, Department of Biology, New York University, New York, NY, USA
| | - Smruti Pushalkar
- Center for Genomics and Systems Biology, Department of Biology, New York University, New York, NY, USA
| | - Shaohuan Wu
- Center for Genomics and Systems Biology, Department of Biology, New York University, New York, NY, USA
| | - Christine Vogel
- Center for Genomics and Systems Biology, Department of Biology, New York University, New York, NY, USA
| | - Lara K Mahal
- Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada
| | - Elodie Ghedin
- Systems Genomics Section, Laboratory of Parasitic Diseases, NIAID, NIH, Bethesda, MD, USA
| | - Ted Ross
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, USA; Department of Infectious Diseases, University of Georgia, Athens, GA, USA
| | - 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 Pathology, Molecular and Cell-Based Medicine, 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
| | - Bin Zhang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mt Sinai, New York, NY, USA; Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
5
|
Wiggins KB, Winston SM, Reeves IL, Gaevert J, Spence Y, Brimble MA, Livingston B, Morton CL, Thomas PG, Sant AJ, Ross TM, Davidoff AM, Schultz-Cherry S. rAAV expressing a COBRA-designed influenza hemagglutinin generates a protective and durable adaptive immune response with a single dose. J Virol 2024; 98:e0078124. [PMID: 39078191 PMCID: PMC11338075 DOI: 10.1128/jvi.00781-24] [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: 04/30/2024] [Accepted: 05/13/2024] [Indexed: 07/31/2024] Open
Abstract
Influenza remains a worldwide public health threat. Although seasonal influenza vaccines are currently the best means of preventing severe disease, the standard-of-care vaccines require frequent updating due to antigenic drift and can have low efficacy, particularly in vulnerable populations. Here, we demonstrate that a single administration of a recombinant adenovirus-associated virus (rAAV) vector expressing a computationally optimized broadly reactive antigen (COBRA)-derived influenza H1 hemagglutinin (HA) induces strongly neutralizing and broadly protective antibodies in naïve mice and ferrets with pre-existing influenza immunity. Following a lethal viral challenge, the rAAV-COBRA vaccine allowed for significantly reduced viral loads in the upper and lower respiratory tracts and complete protection from morbidity and mortality that lasted for at least 5 months post-vaccination. We observed no signs of antibody waning during this study. CpG motif enrichment of the antigen can act as an internal adjuvant to further enhance the immune responses to allow for lower vaccine dosages with the induction of unique interferon-producing CD4+ and CD8+ T cells specific to HA head and stem peptide sequences. Our studies highlight the utility of rAAV as an effective platform to improve seasonal influenza vaccines. IMPORTANCE Developing an improved seasonal influenza vaccine remains an ambitious goal of researchers and clinicians alike. With influenza routinely causing severe epidemics with the potential to rise to pandemic levels, it is critical to create an effective, broadly protective, and durable vaccine to improve public health worldwide. As a potential solution, we created a rAAV viral vector expressing a COBRA-optimized influenza hemagglutinin antigen with modestly enriched CpG motifs to evoke a robust and long-lasting immune response after a single intramuscular dose without needing boosts or adjuvants. Importantly, the rAAV vaccine boosted antibody breadth to future strains in ferrets with pre-existing influenza immunity. Together, our data support further investigation into the utility of viral vectors as a potential avenue to improve our seasonal influenza vaccines.
Collapse
Affiliation(s)
- Kristin B. Wiggins
- St. Jude Graduate
School of Biomedical Sciences,
Memphis, Tennessee, USA
- Department of
Host-Microbe Interactions, St. Jude Children’s Research
Hospital, Memphis,
Tennessee, USA
| | - Stephen M. Winston
- St. Jude Graduate
School of Biomedical Sciences,
Memphis, Tennessee, USA
- Department of Surgery,
St. Jude Children’s Research
Hospital, Memphis,
Tennessee, USA
| | - Isaiah L. Reeves
- St. Jude Graduate
School of Biomedical Sciences,
Memphis, Tennessee, USA
- Department of Surgery,
St. Jude Children’s Research
Hospital, Memphis,
Tennessee, USA
| | - Jessica Gaevert
- St. Jude Graduate
School of Biomedical Sciences,
Memphis, Tennessee, USA
- Department of
Host-Microbe Interactions, St. Jude Children’s Research
Hospital, Memphis,
Tennessee, USA
| | - Yunyu Spence
- Department of Surgery,
St. Jude Children’s Research
Hospital, Memphis,
Tennessee, USA
| | - Mark A. Brimble
- Department of
Host-Microbe Interactions, St. Jude Children’s Research
Hospital, Memphis,
Tennessee, USA
| | - Brandi Livingston
- Department of
Host-Microbe Interactions, St. Jude Children’s Research
Hospital, Memphis,
Tennessee, USA
| | - Christopher L. Morton
- Department of Surgery,
St. Jude Children’s Research
Hospital, Memphis,
Tennessee, USA
| | - Paul G. Thomas
- Department of
Host-Microbe Interactions, St. Jude Children’s Research
Hospital, Memphis,
Tennessee, USA
| | - Andrea J. Sant
- David H. Smith Center
for Vaccine Biology and Immunology, Department of Microbiology and
Immunology, University of Rochester Medical
Center, Rochester, New
York, USA
| | - Ted M. Ross
- Department of
Infectious Biology, Cleveland Clinic,
Cleveland, Ohio, USA
- Cleveland Clinic,
Florida Research and Innovation Center,
Port St. Lucie, Florida,
USA
| | - Andrew M. Davidoff
- St. Jude Graduate
School of Biomedical Sciences,
Memphis, Tennessee, USA
- Department of Surgery,
St. Jude Children’s Research
Hospital, Memphis,
Tennessee, USA
| | - Stacey Schultz-Cherry
- St. Jude Graduate
School of Biomedical Sciences,
Memphis, Tennessee, USA
- Department of
Host-Microbe Interactions, St. Jude Children’s Research
Hospital, Memphis,
Tennessee, USA
| |
Collapse
|
6
|
Yuan L, Zhang S, Bi R, Liu X, Han Z, Li M, Liao X, Xie T, Bai S, Xie Q, Luo C, Jiang Y, Yuan J, Luo H, Yan H, Sun C, Shu Y. A broad-spectrum multiepitope vaccine against seasonal influenza A and B viruses in mice. EBioMedicine 2024; 106:105269. [PMID: 39111250 DOI: 10.1016/j.ebiom.2024.105269] [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: 03/03/2024] [Revised: 07/23/2024] [Accepted: 07/23/2024] [Indexed: 08/18/2024] Open
Abstract
BACKGROUND Influenza viruses pose a persistent threat to global public health, necessitating the development of innovative and broadly effective vaccines. METHODS This study focuses on a multiepitope vaccine (MEV) designed to provide broad-spectrum protection against different influenza viruses. The MEV, containing 19 B-cell linear epitopes, 7 CD4+ T cells, and 11 CD8+ T cells epitopes identified through enzyme-linked immunospot assay (ELISPOT) in influenza viruses infected mice, was administered through a regimen of two doses of DNA vaccine followed by one dose of a protein vaccine in C57BL/6 female mice. FINDINGS Upon lethal challenge with both seasonal circulating strains (H1N1, H3N2, BV, and BY) and historical strains (H1N1-PR8 and H3N2-X31), MEV demonstrated substantial protection against different influenza seasonal strains, with partial efficacy against historical strains. Notably, the increased germinal centre B cells and antibody-secreting cells, along with robust T cell immune responses, highlighted the comprehensive immune defence elicited by MEV. Elevated hemagglutinin inhibition antibody was also observed against seasonal circulating and historical strains. Additionally, mice vaccinated with MEV exhibited significantly lower counts of inflammatory cells in the lungs compared to negative control groups. INTERPRETATION Our results demonstrated the efficacy of a broad-spectrum MEV against influenza viruses in mice. Conducting long-term studies to evaluate the durability of MEV-induced immune responses and explore its potential application in diverse populations will offer valuable insights for the continued advancement of this promising vaccine. FUNDING Funding bodies are described in the Acknowledgments section.
Collapse
Affiliation(s)
- Lifang Yuan
- School of Public Health (Shenzhen), Shenzhen Key Laboratory of Pathogenic Microbes and Biosafetuy, Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, PR China.
| | - Shengze Zhang
- School of Public Health (Shenzhen), Shenzhen Key Laboratory of Pathogenic Microbes and Biosafetuy, Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, PR China.
| | - Rongjun Bi
- School of Public Health (Shenzhen), Shenzhen Key Laboratory of Pathogenic Microbes and Biosafetuy, Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, PR China.
| | - Xuejie Liu
- School of Public Health (Shenzhen), Shenzhen Key Laboratory of Pathogenic Microbes and Biosafetuy, Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, PR China.
| | - Zirong Han
- School of Public Health (Shenzhen), Shenzhen Key Laboratory of Pathogenic Microbes and Biosafetuy, Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, PR China.
| | - Minchao Li
- School of Public Health (Shenzhen), Shenzhen Key Laboratory of Pathogenic Microbes and Biosafetuy, Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, PR China.
| | - Xinzhong Liao
- School of Public Health (Shenzhen), Shenzhen Key Laboratory of Pathogenic Microbes and Biosafetuy, Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, PR China.
| | - Ting Xie
- School of Public Health (Shenzhen), Shenzhen Key Laboratory of Pathogenic Microbes and Biosafetuy, Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, PR China.
| | - Shaohui Bai
- School of Public Health (Shenzhen), Shenzhen Key Laboratory of Pathogenic Microbes and Biosafetuy, Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, PR China.
| | - Qian Xie
- School of Public Health (Shenzhen), Shenzhen Key Laboratory of Pathogenic Microbes and Biosafetuy, Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, PR China.
| | - Chuming Luo
- School of Public Health (Shenzhen), Shenzhen Key Laboratory of Pathogenic Microbes and Biosafetuy, Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, PR China.
| | - Ying Jiang
- Shenzhen Nanshan Centre for Disease Control and Prevention, Shenzhen, 518054, PR China.
| | - Jianhui Yuan
- Shenzhen Nanshan Centre for Disease Control and Prevention, Shenzhen, 518054, PR China.
| | - Huanle Luo
- School of Public Health (Shenzhen), Shenzhen Key Laboratory of Pathogenic Microbes and Biosafetuy, Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, PR China; Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, PR China.
| | - Huacheng Yan
- Centre for Disease Control and Prevention of Southern Military Theatre, 510610, Guangzhou, PR China.
| | - Caijun Sun
- School of Public Health (Shenzhen), Shenzhen Key Laboratory of Pathogenic Microbes and Biosafetuy, Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, PR China; Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, PR China.
| | - Yuelong Shu
- School of Public Health (Shenzhen), Shenzhen Key Laboratory of Pathogenic Microbes and Biosafetuy, Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, PR China; Key Laboratory of Pathogen Infection Prevention and Control (MOE), State Key Laboratory of Respiratory Health and Multimorbidity, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 102629, PR China.
| |
Collapse
|
7
|
Papadatou I, Geropeppa M, Piperi C, Spoulou V, Adamopoulos C, Papavassiliou AG. Deciphering Immune Responses to Immunization via Transcriptional Analysis: A Narrative Review of the Current Evidence towards Personalized Vaccination Strategies. Int J Mol Sci 2024; 25:7095. [PMID: 39000206 PMCID: PMC11240890 DOI: 10.3390/ijms25137095] [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: 05/20/2024] [Revised: 06/25/2024] [Accepted: 06/27/2024] [Indexed: 07/16/2024] Open
Abstract
The development of vaccines has drastically reduced the mortality and morbidity of several diseases. Despite the great success of vaccines, the immunological processes involved in protective immunity are not fully understood and several issues remain to be elucidated. Recently, the advent of high-throughput technologies has enabled a more in-depth investigation of the immune system as a whole and the characterization of the interactions of numerous components of immunity. In the field of vaccinology, these tools allow for the exploration of the molecular mechanisms by which vaccines can induce protective immune responses. In this review, we aim to describe current data on transcriptional responses to vaccination, focusing on similarities and differences of vaccine-induced transcriptional responses among vaccines mostly in healthy adults, but also in high-risk populations, such as the elderly and children. Moreover, the identification of potential predictive biomarkers of vaccine immunogenicity, the effect of age on transcriptional response and future perspectives for the utilization of transcriptomics in the field of vaccinology will be discussed.
Collapse
Affiliation(s)
- Ioanna Papadatou
- Immunobiology and Vaccinology Research Laboratory, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (I.P.); (M.G.); (V.S.)
- First Department of Pediatrics, “Aghia Sophia” Children’s Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Maria Geropeppa
- Immunobiology and Vaccinology Research Laboratory, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (I.P.); (M.G.); (V.S.)
| | - Christina Piperi
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (C.P.); (A.G.P.)
| | - Vana Spoulou
- Immunobiology and Vaccinology Research Laboratory, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (I.P.); (M.G.); (V.S.)
- First Department of Pediatrics, “Aghia Sophia” Children’s Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Christos Adamopoulos
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (C.P.); (A.G.P.)
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Athanasios G. Papavassiliou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (C.P.); (A.G.P.)
| |
Collapse
|
8
|
Bayrak CS, Forst C, Jones DR, Gresham D, Pushalkar S, Wu S, Vogel C, Mahal L, Ghedin E, Ross T, García-Sastre A, Zhang B. Patient Subtyping Analysis of Baseline Multi-omic Data Reveals Distinct Pre-immune States Predictive of Vaccination Responses. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.18.576213. [PMID: 38328256 PMCID: PMC10849502 DOI: 10.1101/2024.01.18.576213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
Understanding the molecular mechanisms that underpin diverse vaccination responses is a critical step toward developing efficient vaccines. Molecular subtyping approaches can offer valuable insights into the heterogeneous nature of responses and aid in the design of more effective vaccines. In order to explore the molecular signatures associated with the vaccine response, we analyzed baseline transcriptomics data from paired samples of whole blood, proteomics and glycomics data from serum, and metabolomics data from urine, obtained from influenza vaccine recipients (2019-2020 season) prior to vaccination. After integrating the data using a network-based model, we performed a subtyping analysis. The integration of multiple data modalities from 62 samples resulted in five baseline molecular subtypes with distinct molecular signatures. These baseline subtypes differed in the expression of pre-existing adaptive or innate immunity signatures, which were linked to significant variation across subtypes in baseline immunoglobulin A (IgA) and hemagglutination inhibition (HAI) titer levels. It is worth noting that these significant differences persisted through day 28 post-vaccination, indicating the effect of initial immune state on vaccination response. These findings highlight the significance of interpersonal variation in baseline immune status as a crucial factor in determining vaccine response and efficacy. Ultimately, incorporating molecular profiling could enable personalized vaccine optimization.
Collapse
|
9
|
Sintusek P, Buranapraditkun S, Khunsri S, Polsawat W, Vichaiwattana P, Poovorawan Y. Antibody persistence of standard versus double three-dose hepatitis B vaccine in liver transplant children: a randomized controlled trial. Sci Rep 2024; 14:499. [PMID: 38177354 PMCID: PMC10767042 DOI: 10.1038/s41598-024-51149-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 01/01/2024] [Indexed: 01/06/2024] Open
Abstract
Rapid hepatitis B (HB) surface antibody (anti-HBs) loss is prevalent after liver transplantation (LT). Herein, we evaluated anti-HBs persistence after HB vaccination using two regimens in LT children. We recruited 66 previously immunized LT children with anti-HBs level of < 100 mIU/mL. Participants were randomly reimmunized with standard-three-dose (SD) and double-three-dose (DD) intramuscular HB vaccination at 0, 1, and 6 months. Anti-HBs were assessed at every outpatient visit. Antibody loss defined as anti-HBs levels < 100 mIU/mL after three-dose vaccination. After three-dose vaccination, 81.8% and 78.7% of participants in the SD and DD groups, had anti-HBs levels > 100 mIU/mL, with a geometric mean titer (GMT) of 601.68 and 668.01 mIU/mL (P = 0.983). After a mean follow-up of 2.31 years, the anti-HBs GMT was 209.81 and 212.61 mIU/mL in the SD and DD groups (P = 0.969). The number of immunosuppressants used and an anti-HBs level < 1 mIU/mL at baseline were independently associated with anti-HB loss. The DD regimen strongly increased the risk of anti-HBs loss (adjusted hazard ratio, 2.97 [1.21-7.31]; P = 0.018). The SD HB reimmunization regimen effectively maintained protective anti-HBs levels in children undergoing LT, making it the preferred regimen for such children with anti-HB loss.Trial registration: TCTR20180723002.
Collapse
Affiliation(s)
- Palittiya Sintusek
- Center of Excellence in Thai Pediatric Gastroenterology, Hepatology and Immunology (TPGHAI), Department of Pediatrics, Faculty of Medicine, King Chulalongkorn Memorial Hospital and the Thai Red Cross Society, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Supranee Buranapraditkun
- Center of Excellence in Thai Pediatric Gastroenterology, Hepatology and Immunology (TPGHAI), Department of Pediatrics, Faculty of Medicine, King Chulalongkorn Memorial Hospital and the Thai Red Cross Society, Chulalongkorn University, Bangkok, 10330, Thailand
- Division of Allergy and Clinical Immunology, Department of Medicine, King Chulalongkorn Memorial Hospital, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Siriporn Khunsri
- Center of Excellence in Thai Pediatric Gastroenterology, Hepatology and Immunology (TPGHAI), Department of Pediatrics, Faculty of Medicine, King Chulalongkorn Memorial Hospital and the Thai Red Cross Society, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Warunee Polsawat
- Excellence Center for Organ Transplantation, King Chulalongkorn Memorial Hospital and the Thai Red Cross Society, Bangkok, 10330, Thailand
| | - Preeyaporn Vichaiwattana
- Excellence Center of Clinical Virology, Department of Pediatrics, Faculty of Medicine, King Chulalongkorn Memorial Hospital, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Yong Poovorawan
- Excellence Center of Clinical Virology, Department of Pediatrics, Faculty of Medicine, King Chulalongkorn Memorial Hospital, Chulalongkorn University, Bangkok, 10330, Thailand.
| |
Collapse
|
10
|
Yuan L, Li X, Li M, Bi R, Li Y, Song J, Li W, Yan M, Luo H, Sun C, Shu Y. In silico design of a broad-spectrum multiepitope vaccine against influenza virus. Int J Biol Macromol 2024; 254:128071. [PMID: 37967595 DOI: 10.1016/j.ijbiomac.2023.128071] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 11/10/2023] [Accepted: 11/10/2023] [Indexed: 11/17/2023]
Abstract
Influenza remains a global health concern due to its potential to cause pandemics as a result of rapidly mutating influenza virus strains. Existing vaccines often struggle to keep up with these rapidly mutating flu viruses. Therefore, the development of a broad-spectrum peptide vaccine that can stimulate an optimal antibody response has emerged as an innovative approach to addressing the influenza threat. In this study, an immunoinformatic approach was employed to rapidly predict immunodominant epitopes from different antigens, aiming to develop an effective multiepitope influenza vaccine (MEV). The immunodominant B-cell linear epitopes of seasonal influenza strains hemagglutinin (HA) and neuraminidase (NA) were predicted using an antibody-peptide microarray, involving a human cohort including vaccinees and infected patients. On the other hand, bioinformatics tools were used to predict immunodominant cytotoxic T-cell (CTL) and helper T-cell (HTL) epitopes. Subsequently, these epitopes were evaluated by various immunoinformatic tools. Epitopes with high antigenicity, high immunogenicity, non-allergenicity, non-toxicity, as well as exemplary conservation were then connected in series with appropriate linkers and adjuvants to construct a broad-spectrum MEV. Moreover, the structural analysis revealed that the MEV candidates exhibited good stability, and the docking results demonstrated their strong affinity to Toll-like receptors 4 (TLR4). In addition, molecular dynamics simulation confirmed the stable interaction between TLR4 and MEVs. Three injections with MEVs showed a high level of B-cell and T-cell immune responses according to the immunological simulations in silico. Furthermore, in-silico cloning was performed, and the results indicated that the MEVs could be produced in considerable quantities in Escherichia coli (E. coli). Based on these findings, it is reasonable to create a broad-spectrum MEV against different subtypes of influenza A and B viruses in silico.
Collapse
Affiliation(s)
- Lifang Yuan
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China; School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou 510275, PR China.
| | - Xu Li
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China; School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou 510275, PR China; Department of Pathogenic Biology and Immunology, School of Basic Medicine, Xiangnan University, Chenzhou, Hunan, PR China.
| | - Minchao Li
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China; School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou 510275, PR China.
| | - Rongjun Bi
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China; School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou 510275, PR China
| | - Yingrui Li
- Shenzhen Digital Life Institute, Shenzhen, Guangdong 518000, PR China.
| | - Jiaping Song
- Shenzhen Digital Life Institute, Shenzhen, Guangdong 518000, PR China.
| | - Wei Li
- Shenzhen Digital Life Institute, Shenzhen, Guangdong 518000, PR China.
| | - Mingchen Yan
- Shenzhen Digital Life Institute, Shenzhen, Guangdong 518000, PR China
| | - Huanle Luo
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China; School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou 510275, PR China; Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou 510080, PR China.
| | - Caijun Sun
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China; School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou 510275, PR China; Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou 510080, PR China.
| | - Yuelong Shu
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China; School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou 510275, PR China; Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou 510080, PR China; Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100176, PR China.
| |
Collapse
|
11
|
Heida R, Frijlink HW, Hinrichs WLJ. Inhalation of vaccines and antiviral drugs to fight respiratory virus infections: reasons to prioritize the pulmonary route of administration. mBio 2023; 14:e0129523. [PMID: 37768057 PMCID: PMC10653782 DOI: 10.1128/mbio.01295-23] [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: 09/29/2023] Open
Abstract
Many of the current pandemic threats are caused by viruses that infect the respiratory tract. Remarkably though, the majority of vaccines and antiviral drugs are administered via alternative routes. In this perspective, we argue that the pulmonary route of administration deserves more attention in the search for novel therapeutic strategies against respiratory virus infections. Firstly, vaccines administered at the viral portal of entry can induce a broader immune response, employing the mucosal arm of the immune system; secondly, direct administration of antiviral drugs at the target site leads to superior bioavailability, enabling lower dosing and reducing the chance of side effects. We further elaborate on why the pulmonary route may induce a superior effect compared to the intranasal route of administration and provide reasons why dry powder formulations for inhalation have significant advantages over standard liquid formulations.
Collapse
Affiliation(s)
- Rick Heida
- Department of Pharmaceutical Technology and Biopharmacy, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, The Netherlands
| | - Henderik W. Frijlink
- Department of Pharmaceutical Technology and Biopharmacy, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, The Netherlands
| | - Wouter L. J. Hinrichs
- Department of Pharmaceutical Technology and Biopharmacy, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
12
|
Xie Y, Tian X, Zhang X, Yao H, Wu N. Immune interference in effectiveness of influenza and COVID-19 vaccination. Front Immunol 2023; 14:1167214. [PMID: 37153582 PMCID: PMC10154574 DOI: 10.3389/fimmu.2023.1167214] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 04/06/2023] [Indexed: 05/09/2023] Open
Abstract
Vaccines are known to function as the most effective interventional therapeutics for controlling infectious diseases, including polio, smallpox, rabies, tuberculosis, influenza and SARS-CoV-2. Smallpox has been eliminated completely and polio is almost extinct because of vaccines. Rabies vaccines and Bacille Calmette-Guérin (BCG) vaccines could effectively protect humans against respective infections. However, both influenza vaccines and COVID-19 vaccines are unable to eliminate these two infectious diseases of their highly variable antigenic sites in viral proteins. Vaccine effectiveness (VE) could be negatively influenced (i.e., interfered with) by immune imprinting of previous infections or vaccinations, and repeated vaccinations could interfere with VE against infections due to mismatch between vaccine strains and endemic viral strains. Moreover, VE could also be interfered with when more than one kind of vaccine is administrated concomitantly (i.e., co-administrated), suggesting that the VE could be modulated by the vaccine-induced immunity. In this review, we revisit the evidence that support the interfered VE result from immune imprinting or repeated vaccinations in influenza and COVID-19 vaccine, and the interference in co-administration of these two types of vaccines is also discussed. Regarding the development of next-generation COVID-19 vaccines, the researchers should focus on the induction of cross-reactive T-cell responses and naive B-cell responses to overcome negative effects from the immune system itself. The strategy of co-administrating influenza and COVID-19 vaccine needs to be considered more carefully and more clinical data is needed to verify this strategy to be safe and immunogenic.
Collapse
Affiliation(s)
- Yiwen Xie
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, Shandong, China
| | - Xuebin Tian
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, Shandong, China
| | - Xiaodi Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, Shandong, China
| | - Hangping Yao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, Shandong, China
| | - Nanping Wu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, Shandong, China
| |
Collapse
|
13
|
Forst CV, Chung M, Hockman M, Lashua L, Adney E, Hickey A, Carlock M, Ross T, Ghedin E, Gresham D. Vaccination History, Body Mass Index, Age, and Baseline Gene Expression Predict Influenza Vaccination Outcomes. Viruses 2022; 14:2446. [PMID: 36366544 PMCID: PMC9697051 DOI: 10.3390/v14112446] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/24/2022] [Accepted: 10/28/2022] [Indexed: 11/06/2022] Open
Abstract
Seasonal influenza is a primary public health burden in the USA and globally. Annual vaccination programs are designed on the basis of circulating influenza viral strains. However, the effectiveness of the seasonal influenza vaccine is highly variable between seasons and among individuals. A number of factors are known to influence vaccination effectiveness including age, sex, and comorbidities. Here, we sought to determine whether whole blood gene expression profiling prior to vaccination is informative about pre-existing immunological status and the immunological response to vaccine. We performed whole transcriptome analysis using RNA sequencing (RNAseq) of whole blood samples obtained prior to vaccination from 275 participants enrolled in an annual influenza vaccine trial. Serological status prior to vaccination and 28 days following vaccination was assessed using the hemagglutination inhibition assay (HAI) to define baseline immune status and the response to vaccination. We find evidence that genes with immunological functions are increased in expression in individuals with higher pre-existing immunity and in those individuals who mount a greater response to vaccination. Using a random forest model, we find that this set of genes can be used to predict vaccine response with a performance similar to a model that incorporates physiological and prior vaccination status alone. A model using both gene expression and physiological factors has the greatest predictive power demonstrating the potential utility of molecular profiling for enhancing prediction of vaccine response. Moreover, expression of genes that are associated with enhanced vaccination response may point to additional biological pathways that contribute to mounting a robust immunological response to the seasonal influenza vaccine.
Collapse
Affiliation(s)
- Christian V. Forst
- Department of Genetics and Genomic Sciences, Department of Microbiology, Icahn School of Medicine at Mt Sinai, One Gustave L. Levy Place, Box 1498, New York, NY 10029-6574, USA
| | - Matthew Chung
- Systems Genomics Section, Laboratory of Parasitic Diseases, NIAID, NIH, Bethesda, MD 20894, USA
| | - Megan Hockman
- Center for Genomics and Systems Biology, Department of Biology, New York University, New York, NY 10003, USA
| | - Lauren Lashua
- Center for Genomics and Systems Biology, Department of Biology, New York University, New York, NY 10003, USA
| | - Emily Adney
- Center for Genomics and Systems Biology, Department of Biology, New York University, New York, NY 10003, USA
| | - Angela Hickey
- Center for Genomics and Systems Biology, Department of Biology, New York University, New York, NY 10003, USA
| | - Michael Carlock
- Center for Vaccines and Immunology, University of Georgia, Athens, GA 30602, USA
- Department of Infectious Diseases, University of Georgia, Athens, GA 30602, USA
| | - Ted Ross
- Center for Vaccines and Immunology, University of Georgia, Athens, GA 30602, USA
- Department of Infectious Diseases, University of Georgia, Athens, GA 30602, USA
| | - Elodie Ghedin
- Systems Genomics Section, Laboratory of Parasitic Diseases, NIAID, NIH, Bethesda, MD 20894, USA
| | - David Gresham
- Center for Genomics and Systems Biology, Department of Biology, New York University, New York, NY 10003, USA
| |
Collapse
|
14
|
Rcheulishvili N, Papukashvili D, Liu C, Ji Y, He Y, Wang PG. Promising strategy for developing mRNA-based universal influenza virus vaccine for human population, poultry, and pigs- focus on the bigger picture. Front Immunol 2022; 13:1025884. [PMID: 36325349 PMCID: PMC9618703 DOI: 10.3389/fimmu.2022.1025884] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 10/03/2022] [Indexed: 08/08/2023] Open
Abstract
Since the first outbreak in the 19th century influenza virus has remained emergent owing to the huge pandemic potential. Only the pandemic of 1918 caused more deaths than any war in world history. Although two types of influenza- A (IAV) and B (IBV) cause epidemics annually, influenza A deserves more attention as its nature is much wilier. IAVs have a large animal reservoir and cause the infection manifestation not only in the human population but in poultry and domestic pigs as well. This many-sided characteristic of IAV along with the segmented genome gives rise to the antigenic drift and shift that allows evolving the new strains and new subtypes, respectively. As a result, the immune system of the body is unable to recognize them. Importantly, several highly pathogenic avian IAVs have already caused sporadic human infections with a high fatality rate (~60%). The current review discusses the promising strategy of using a potentially universal IAV mRNA vaccine based on conserved elements for humans, poultry, and pigs. This will better aid in averting the outbreaks in different susceptible species, thus, reduce the adverse impact on agriculture, and economics, and ultimately, prevent deadly pandemics in the human population.
Collapse
Affiliation(s)
| | | | | | | | - Yunjiao He
- *Correspondence: Yunjiao He, ; Peng George Wang,
| | | |
Collapse
|
15
|
Association between influenza vaccination and mortality due to COVID-19. VACUNAS (ENGLISH EDITION) 2022. [PMCID: PMC9247262 DOI: 10.1016/j.vacune.2022.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
Background It has recently been suggested that influenza vaccination may be a factor associated with decreased COVID-19 mortality. Methods An age-matched case–control study based on hospital cases. We included subjects aged 18 years and older with a diagnosis of moderate to severe COVID-19. Infection was corroborated by RT-PCR test for SARS-COV-2. Deceased subjects were considered cases, controls were patients discharged due to improvement of acute symptoms. We used bivariate analysis to determine factors associated with death from COVID-19, and calculated odds ratios and 95% confidence intervals. Results A total of 560 patients were included in the study, 214 (38.2%) were considered cases and 346 (61.7%) controls. A significant difference was observed with the presence of type 2 diabetes mellitus [54% vs. 39.3% between cases and controls, respectively (P = 0.04)] and having received influenza vaccination (P = 0.02). Type 2 diabetes mellitus was associated with higher COVID-19 mortality [OR 1.8 (95% CI 1.2–2.5) P = 0.01], whereas having been immunised against influenza in 2019 was associated with lower mortality in this group of patients [OR 0.6 (95% CI 0.4–0.9) P = 0.02]. Conclusions Influenza vaccination in the previous year appears to be associated with lower mortality from COVID-19; whereas type 2 diabetes mellitus is confirmed as a condition associated with higher mortality.
Collapse
|
16
|
Arce-Salinas CA, Esquivel-Torruco YN, Bejarano-Juvera AA, Bustamante-Flores AK, Aguilar-Martínez N, Azcorra-López JG, Cabañas-Espinosa B, Luna-Rivera EM, Hernández-Alarcón A, Reyna Figueroa J. Asociación entre la vacunación contra influenza y la mortalidad por COVID-19. VACUNAS 2022; 23:113-118. [PMID: 34751213 PMCID: PMC8566694 DOI: 10.1016/j.vacun.2021.09.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 09/06/2021] [Indexed: 12/23/2022]
Affiliation(s)
- C A Arce-Salinas
- Servicio de Medicina Interna, Hospital Central Sur Petróleos Mexicanos, Ciudad de México, México
| | - Y N Esquivel-Torruco
- Servicio de Pediatría, Hospital Central Sur Petróleos Mexicanos, Ciudad de México, México
| | - A A Bejarano-Juvera
- Servicio de Pediatría, Hospital Central Sur Petróleos Mexicanos, Ciudad de México, México
| | - A K Bustamante-Flores
- Servicio de Pediatría, Hospital Central Sur Petróleos Mexicanos, Ciudad de México, México
| | - N Aguilar-Martínez
- Servicio de Pediatría, Hospital Central Sur Petróleos Mexicanos, Ciudad de México, México
| | - J G Azcorra-López
- Servicio de Pediatría, Hospital Central Sur Petróleos Mexicanos, Ciudad de México, México
| | - B Cabañas-Espinosa
- Servicio de Pediatría, Hospital Central Sur Petróleos Mexicanos, Ciudad de México, México
| | - E M Luna-Rivera
- Departamento de Enseñanza e Investigación, Hospital Central Sur Petróleos Mexicanos, Ciudad de México, México
| | - A Hernández-Alarcón
- Servicio de Consulta Externa, Hospital Central Sur Petróleos Mexicanos, Ciudad de México, México
| | - J Reyna Figueroa
- Departamento de Enseñanza e Investigación, Hospital Central Sur Petróleos Mexicanos, Ciudad de México, México
| |
Collapse
|
17
|
Krivitskaya VZ, Kuznecova EV, Maiorova VG, Petrova ER, Sominina AA, Danilenko DM. Influenza vaccination influencing level of specific humoral immunity in healthy individuals. RUSSIAN JOURNAL OF INFECTION AND IMMUNITY 2022. [DOI: 10.15789/2220-7619-ivi-1750] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
To assess an effect of vaccination on the level of humoral anti-influenza herd immunity, 2955 sera were collected and analyzed by HIT in the 2019–2020 and 2020–2021 epidemiological seasons. All sera were obtained from healthy adult donors residing in various cities of the Russian Federation. Among them, 1057 volunteers were vaccinated with seasonal influenza trivalent inactivated vaccine. Characteristics of humoral anti-influenza immunity (average geometric antibody titers and the proportion of individuals seropositive for the vaccine viruses) obtained in autumn 2019 and 2020 (1–2 months after vaccination) in vaccinated individuals vs. unvaccinated subjects were found to be markedly higher evidencing about a positive vaccination-related contribution to developing herd immunity against influenza in the preepidemic periods. After the 2019–2020 influenza epidemic, in spring 2020 (6–7 months after vaccination), the levels of antibodies to all vaccine components decreased by 2.6–3.5-fold in vaccinated donors compared to the pre-epidemic period in 2019 autumn. Antibody titers became substantially lower than the protective level (titer by HIT 1/40). At the same time, no significant differences between the groups of vaccinated vs. unvaccinated individuals were observed afterwards. This indicates instability of post-vaccination anti-influenza humoral immunity. As a result, it may decrease an influenza-resistant population cohort of working age on the eve of new epidemic season. The immunogenicity of the inactivated trivalent seasonal influenza vaccine was estimated by HIT while analyzing paired sera obtained from 295 and 112 healthy individuals of various ages vaccinated in autumn 2019 and 2020, respectively. The response to the vaccine was found to be age-related. Children aged 3–14 years vs. older subjects showed a more efficient response. Insufficient immunogenicity of influenza B virus vaccine components was shown. In all age groups, average geometric titers for influenza B virus antibodies were lower (2–8-fold) than for current A(H1N1)pdm09-like strains and influenza A(H3N2) viruses 1–1.5 months post-vaccination. Analyzing vaccine immunogenicity showed a significant inverse relationship between the level of preexisting strain-specific serum antibodies before vaccination and formation of antibodies to the corresponding vaccine virus 1–1.5 months after vaccination. Seroconversion to each vaccine component was remarkably more frequent in individuals with a low preexisting level of antibodies specific to the corresponding virus.
Collapse
|
18
|
Shapiro JR, Li H, Morgan R, Chen Y, Kuo H, Ning X, Shea P, Wu C, Merport K, Saldanha R, Liu S, Abrams E, Chen Y, Kelly DC, Sheridan-Malone E, Wang L, Zeger SL, Klein SL, Leng SX. Sex-specific effects of aging on humoral immune responses to repeated influenza vaccination in older adults. NPJ Vaccines 2021; 6:147. [PMID: 34887436 PMCID: PMC8660902 DOI: 10.1038/s41541-021-00412-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 11/02/2021] [Indexed: 01/16/2023] Open
Abstract
Older adults (≥65 years of age) bear a significant burden of severe disease and mortality associated with influenza, despite relatively high annual vaccination coverage and substantial pre-existing immunity to influenza. To test the hypothesis that host factors, including age and sex, play a role in determining the effect of repeated vaccination and levels of pre-existing humoral immunity to influenza, we evaluated pre- and post-vaccination strain-specific hemagglutination inhibition (HAI) titers in adults over 75 years of age who received a high-dose influenza vaccine in at least four out of six influenza seasons. Pre-vaccination titers, rather than host factors and repeated vaccination were significantly associated with post-vaccination HAI titer outcomes, and displayed an age-by-sex interaction. Pre-vaccination titers to H1N1 remained constant with age. Titers to H3N2 and influenza B viruses decreased substantially with age in males, whereas titers in females remained constant with age. Our findings highlight the importance of pre-existing immunity in this highly vaccinated older adult population and suggest that older males are particularly vulnerable to reduced pre-existing humoral immunity to influenza.
Collapse
Affiliation(s)
- Janna R Shapiro
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Huifen Li
- Division of Geriatric Medicine and Gerontology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Rosemary Morgan
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Yiyin Chen
- Guangdong Geriatrics Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Helen Kuo
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Xiaoxuan Ning
- Department of Geriatrics, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Patrick Shea
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Cunjin Wu
- Department of Geriatrics, The Second Hospital of Tianjin Medical University, Tianjin, Hebei, China
| | - Katherine Merport
- Zanvyl Krieger School of Arts and Science, Johns Hopkins University, Baltimore, MD, USA
| | - Rayna Saldanha
- Zanvyl Krieger School of Arts and Science, Johns Hopkins University, Baltimore, MD, USA
| | - Suifeng Liu
- Zhongshan Hospital, Xiamen University, Xiamen, Fujian, China
| | - Engle Abrams
- Division of Geriatric Medicine and Gerontology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yan Chen
- Department of Geriatrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, Sichuan, China
| | - Denise C Kelly
- Division of Geriatric Medicine and Gerontology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Eileen Sheridan-Malone
- Division of Geriatric Medicine and Gerontology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lan Wang
- Department of Geriatrics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Scott L Zeger
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Sabra L Klein
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
| | - Sean X Leng
- Division of Geriatric Medicine and Gerontology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
| |
Collapse
|
19
|
Wild K, Smits M, Killmer S, Strohmeier S, Neumann-Haefelin C, Bengsch B, Krammer F, Schwemmle M, Hofmann M, Thimme R, Zoldan K, Boettler T. Pre-existing immunity and vaccine history determine hemagglutinin-specific CD4 T cell and IgG response following seasonal influenza vaccination. Nat Commun 2021; 12:6720. [PMID: 34795301 PMCID: PMC8602312 DOI: 10.1038/s41467-021-27064-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 11/02/2021] [Indexed: 11/09/2022] Open
Abstract
Effectiveness of seasonal influenza vaccination varies between individuals and might be affected by vaccination history among other factors. Here we show, by monitoring frequencies of CD4 T cells specific to the conserved hemagglutinin epitope HA118-132 and titres of IgG against the corresponding recombinant hemagglutinin protein, that antigen-specific CD4 T cell and antibody responses are closely linked to pre-existing immunity and vaccine history. Upon immunization, a strong early reaction is observed in all vaccine naïve participants and also in vaccine experienced individuals who have not received the respective seasonal vaccine in the previous year. This response is characterized by HA118-132 specific CD4 T cells with a follicular helper T cell phenotype and by ascending titers of hemagglutinin-specific antibodies from baseline to day 28 following vaccination. This trend was observed in only a proportion of those participants who received the seasonal vaccine the year preceding the study. Regardless of history, levels of pre-existing antibodies and CD127 expression on CD4 T cells at baseline were the strongest predictors of robust early response. Thus, both pre-existing immunity and vaccine history contribute to the response to seasonal influenza vaccines.
Collapse
Affiliation(s)
- Katharina Wild
- Department of Medicine II, Medical Center - University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Faculty of Pharmacy, University of Freiburg, Freiburg, Germany
| | - Maike Smits
- Department of Medicine II, Medical Center - University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Saskia Killmer
- Department of Medicine II, Medical Center - University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Shirin Strohmeier
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Christoph Neumann-Haefelin
- Department of Medicine II, Medical Center - University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Bertram Bengsch
- Department of Medicine II, Medical Center - University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Martin Schwemmle
- Institute of Virology, Freiburg University Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Maike Hofmann
- Department of Medicine II, Medical Center - University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Robert Thimme
- Department of Medicine II, Medical Center - University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Katharina Zoldan
- Department of Medicine II, Medical Center - University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Tobias Boettler
- Department of Medicine II, Medical Center - University of Freiburg, Freiburg, Germany.
- Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
20
|
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
|
21
|
Ntouros PA, Vlachogiannis NI, Pappa M, Nezos A, Mavragani CP, Tektonidou MG, Souliotis VL, Sfikakis PP. Effective DNA damage response after acute but not chronic immune challenge: SARS-CoV-2 vaccine versus Systemic Lupus Erythematosus. Clin Immunol 2021; 229:108765. [PMID: 34089859 PMCID: PMC8171000 DOI: 10.1016/j.clim.2021.108765] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 05/29/2021] [Accepted: 05/31/2021] [Indexed: 12/27/2022]
Abstract
Whether and how an acute immune challenge may affect DNA Damage Response (DDR) is unknown. By studying vaccinations against Influenza and SARS-CoV-2 (mRNA-based) we found acute increases of type-I interferon-inducible gene expression, oxidative stress and DNA damage accumulation in blood mononuclear cells of 9 healthy controls, coupled with effective anti-SARS-CoV-2 neutralizing antibody production in all. Increased DNA damage after SARS-CoV-2 vaccine, partly due to increased oxidative stress, was transient, whereas the inherent DNA repair capacity was found intact. In contrast, in 26 patients with Systemic Lupus Erythematosus, who served as controls in the context of chronic immune activation, we validated increased DNA damage accumulation, increased type-I interferon-inducible gene expression and induction of oxidative stress, however aberrant DDR was associated with deficiencies in nucleotide excision repair pathways. These results indicate that acute immune challenge can indeed activate DDR pathways, whereas, contrary to chronic immune challenge, successful repair of DNA lesions occurs.
Collapse
Affiliation(s)
- Panagiotis A Ntouros
- First Department of Propaedeutic Internal Medicine and Joint Rheumatology Program, National and Kapodistrian University of Athens Medical School, Athens, Greece.
| | - Nikolaos I Vlachogiannis
- First Department of Propaedeutic Internal Medicine and Joint Rheumatology Program, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Maria Pappa
- First Department of Propaedeutic Internal Medicine and Joint Rheumatology Program, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Adrianos Nezos
- Department of Physiology, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Clio P Mavragani
- Department of Physiology, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Maria G Tektonidou
- First Department of Propaedeutic Internal Medicine and Joint Rheumatology Program, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Vassilis L Souliotis
- First Department of Propaedeutic Internal Medicine and Joint Rheumatology Program, National and Kapodistrian University of Athens Medical School, Athens, Greece; Institute of Chemical Biology, National Hellenic Research Foundation, Athens, Greece
| | - Petros P Sfikakis
- First Department of Propaedeutic Internal Medicine and Joint Rheumatology Program, National and Kapodistrian University of Athens Medical School, Athens, Greece.
| |
Collapse
|
22
|
The efficacy of vaccination to prevent human papilloma viruses infection at anal and oral: a systematic review and meta-analysis. Public Health 2021; 196:165-171. [PMID: 34229128 DOI: 10.1016/j.puhe.2021.05.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 05/10/2021] [Accepted: 05/12/2021] [Indexed: 01/21/2023]
Abstract
OBJECTIVE The objective of this study was to review evidence on the effectiveness of vaccination in the prevention of human papilloma virus (HPV) infection at the cervix, anal, and oral. STUDY DESIGN Systematic review and meta-analysis. METHODS The key search limitations are as follows: "Human Papilloma Virus", "Papilloma Virus, Human" "Human Papillomavirus Virus", "HPV" and "oral", "anus", "anal", "penis", "cervical," and "vaccine". Randomized controlled studies were searched and analyzed the risk ratio by Review Manager 5.3; funnel plot was adopted for publication bias analysis. RESULTS Five randomized controlled studies enrolling 13,686 participants were retrieved, analyzed, and showed that HPV vaccination can effectively block HPV infection at cervical, anal, and oral. Subgroup analysis, moreover, proved that HPV 16/18 is more effective than HPV 6/11/16/18 in preventing anal and oral infections. CONCLUSION HPV vaccine is efficacious in preventing HPV infection not only at cervical but also at anal and oral, as evidence supported by relevant studies.
Collapse
|
23
|
Wiggins KB, Smith MA, Schultz-Cherry S. The Nature of Immune Responses to Influenza Vaccination in High-Risk Populations. Viruses 2021; 13:v13061109. [PMID: 34207924 PMCID: PMC8228336 DOI: 10.3390/v13061109] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 05/31/2021] [Accepted: 06/02/2021] [Indexed: 12/12/2022] Open
Abstract
The current pandemic has brought a renewed appreciation for the critical importance of vaccines for the promotion of both individual and public health. Influenza vaccines have been our primary tool for infection control to prevent seasonal epidemics and pandemics such as the 2009 H1N1 influenza A virus pandemic. Certain high-risk populations, including the elderly, people with obesity, and individuals with comorbidities such as type 2 diabetes mellitus, are more susceptible to increased disease severity and decreased vaccine efficacy. High-risk populations have unique microenvironments and immune responses that contribute to increased vulnerability for influenza infections. This review focuses on these differences as we investigate the variations in immune responses to influenza vaccination. In order to develop better influenza vaccines, it is critical to understand how to improve responses in our ever-growing high-risk populations.
Collapse
|
24
|
A Randomized Controlled Study to Evaluate the Safety and Reactogenicity of a Novel rVLP-Based Plant Virus Nanoparticle Adjuvant Combined with Seasonal Trivalent Influenza Vaccine Following Single Immunization in Healthy Adults 18-50 Years of Age. Vaccines (Basel) 2020; 8:vaccines8030393. [PMID: 32698532 PMCID: PMC7564144 DOI: 10.3390/vaccines8030393] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 06/20/2020] [Accepted: 07/06/2020] [Indexed: 02/02/2023] Open
Abstract
Inactivated influenza vaccines efficacy is variable and often poor. We conducted a phase 1 trial (NCT02188810), to assess the safety and immunogenicity of a novel nanoparticle Toll-like receptor 7/8 agonist adjuvant (Papaya Mosaic Virus) at different dose levels combined with trivalent influenza vaccine in healthy persons 18–50 years of age. Hemagglutination-inhibition assays, antibody to Influenza A virus nucleoprotein and peripheral blood mononuclear cells for measurement of interferon-gamma ELISPOT response to influenza antigens, Granzyme B and IFNγ:IL-10 ratio were measured. The most common adverse events were transient mild to severe injection site pain and no safety signals were observed. A dose-related adjuvant effect was observed. Geometric mean hemagglutination-inhibition titers increased at day 28 in most groups and waned over time, but fold-antibody responses were poor in all groups. Cell mediated immunity results were consistent with humoral responses. The Papaya Mosaic Virus adjuvant in doses of 30 to 240 µg combined with reduced influenza antigen content was safe with no signals up to 3 years after vaccination. A dose-related adjuvant effect was observed and immunogenicity results suggest that efficacy study should be conducted in influenza antigen-naïve participants.
Collapse
|
25
|
Alcorn JF, Avula R, Chakka AB, Schwarzmann WE, Nowalk MP, Lin CJ, Ortiz MA, Horne WT, Chandran UR, Nagg JP, Zimmerman RK, Cole KS, Moehling KK, Martin JM. Differential gene expression in peripheral blood mononuclear cells from children immunized with inactivated influenza vaccine. Hum Vaccin Immunother 2020; 16:1782-1790. [PMID: 32298194 DOI: 10.1080/21645515.2020.1711677] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The human immune response to inactivated influenza vaccine is dynamic and impacted by age and preexisting immunity. Our goal was to identify postvaccination transcriptomic changes in peripheral blood mononuclear cells from children. Blood samples were obtained before and at 3 or 7 days postvaccination with 2016-2017 quadrivalent inactivated influenza vaccine and RNA sequencing was performed. There were 1,466 differentially expressed genes (DEGs) for the Day 0-Day 3 group and 513 DEGs for the Day 0-Day 7 group. Thirty-three genes were common between the two groups. The majority of the transcriptomic changes at Day 3 represented innate inflammation and apoptosis pathways. Day 7 DEGs were characterized by activation of cellular processes, including the regulation of cytoskeleton, junctions, and metabolism, and increased expression of immunoglobulin genes. DEGs at Day 3 were compared between older and younger children revealing increased inflammatory gene expression in the older group. Vaccine history in the year prior to the study was characterized by robust DEGs at Day 3 with decreased phagosome and dendritic cell maturation in those who had been vaccinated in the previous year. PBMC responses to inactivated influenza vaccination in children differed significantly by the timing of sampling, patient age, and vaccine history. These data provide insight into the expected molecular pathways to be temporally altered by influenza vaccination in children.
Collapse
Affiliation(s)
- John F Alcorn
- Department of Pediatrics, University of Pittsburgh , Pittsburgh, PA, USA
| | - Raghunandan Avula
- Department of Biomedical Informatics, University of Pittsburgh , Pittsburgh, PA, USA
| | - Anish B Chakka
- Department of Biomedical Informatics, University of Pittsburgh , Pittsburgh, PA, USA
| | - William E Schwarzmann
- Department of Biomedical Informatics, University of Pittsburgh , Pittsburgh, PA, USA
| | | | | | - Marianna A Ortiz
- Department of Pediatrics, University of Pittsburgh , Pittsburgh, PA, USA.,Center for Vaccine Research, University of Pittsburgh , Pittsburgh, PA, USA
| | - William T Horne
- Department of Pediatrics, University of Pittsburgh , Pittsburgh, PA, USA
| | - Uma R Chandran
- Department of Biomedical Informatics, University of Pittsburgh , Pittsburgh, PA, USA
| | - Jennifer P Nagg
- Department of Pediatrics, University of Pittsburgh , Pittsburgh, PA, USA
| | - Richard K Zimmerman
- Department of Family Medicine, University of Pittsburgh , Pittsburgh, PA, USA
| | - Kelly S Cole
- Center for Vaccine Research, University of Pittsburgh , Pittsburgh, PA, USA
| | - Krissy K Moehling
- Department of Family Medicine, University of Pittsburgh , Pittsburgh, PA, USA
| | - Judith M Martin
- Department of Pediatrics, University of Pittsburgh , Pittsburgh, PA, USA
| |
Collapse
|
26
|
Isakova-Sivak I, Grigorieva E, Rudenko L. Insights into current clinical research on the immunogenicity of live attenuated influenza vaccines. Expert Rev Vaccines 2020; 19:43-55. [PMID: 31903816 DOI: 10.1080/14760584.2020.1711056] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Introduction: Live attenuated influenza vaccines (LAIVs) have been in use for more than three decades and are now licensed in many countries. There is evidence that LAIVs can have greater efficacy than inactivated influenza vaccines, especially against mismatched influenza, however, in recent years, a number of trials have found a lack of LAIV efficacy, mainly in relation to the H1N1 virus.Areas covered: In this review, we summarize the results of clinical research published in the past 5 years on the immunogenicity of LAIVs, with special attention to the mechanisms of establishing protective immunity and some factors that may influence immunogenicity and efficacy.Expert opinion: A number of recent clinical studies confirmed that the immune responses to LAIVs are multifaceted, involving different immune mechanisms. These trials suggest that the intrinsic replicative properties of each LAIV component should be taken into account, and the precise effects of adding a fourth vaccine strain to trivalent LAIV formulations are still to be identified. In addition, new data are emerging regarding the impact of pre-vaccination conditions, such as preexisting immunity or concurrent asymptomatic viral and bacterial respiratory infections, on LAIV immunogenicity, suggesting the importance of monitoring them during clinical trials or vaccination campaigns.
Collapse
Affiliation(s)
- Irina Isakova-Sivak
- Department of Virology, Institute of Experimental Medicine, St. Petersburg, Russia
| | - Elena Grigorieva
- Department of Virology, Institute of Experimental Medicine, St. Petersburg, Russia
| | - Larisa Rudenko
- Department of Virology, Institute of Experimental Medicine, St. Petersburg, Russia
| |
Collapse
|
27
|
Kelvin AA, Zambon M. Influenza imprinting in childhood and the influence on vaccine response later in life. Euro Surveill 2019; 24:1900720. [PMID: 31796156 PMCID: PMC6891942 DOI: 10.2807/1560-7917.es.2019.24.48.1900720] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2019] [Accepted: 11/27/2019] [Indexed: 12/13/2022] Open
Affiliation(s)
- Alyson A Kelvin
- Department of Pediatrics, Division of Infectious Disease, Faculty of Medicine, Dalhousie University, Halifax, Canada
- Canadian Centre for Vaccinology, IWK Health Centre, Halifax, Canada
| | - Maria Zambon
- National Infection Service, Public Health England, London, United Kingdom
| |
Collapse
|