1
|
Han Z, Mai Q, Zhao Y, Liu X, Cui M, Li M, Chen Y, Shu Y, Gan J, Pan W, Sun C. Mosaic neuraminidase-based vaccine induces antigen-specific T cell responses against homologous and heterologous influenza viruses. Antiviral Res 2024; 230:105978. [PMID: 39117282 DOI: 10.1016/j.antiviral.2024.105978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 07/20/2024] [Accepted: 08/04/2024] [Indexed: 08/10/2024]
Abstract
Seasonal influenza is an annually severe crisis for global public health, and an ideal influenza vaccine is expected to provide broad protection against constantly drifted strains. Compared to highly flexible hemagglutinin (HA), increasing data have demonstrated that neuraminidase (NA) might be a potential target against influenza variants. In the present study, a series of genetic algorithm-based mosaic NA were designed, and then cloned into recombinant DNA and replication-defective Vesicular Stomatitis Virus (VSV) vector as a novel influenza vaccine candidate. Our Results showed that DNA prime/VSV boost strategy elicited a robust NA-specific Th1-dominated immune response, but the traditional inactivated influenza vaccine elicited a Th2-dominated immune response. More importantly, the superior NA-specific immunity induced by our strategy could confer both a full protection against lethal homologous influenza challenge and a partial protection against heterologous influenza infection. These findings will provide insights on designing NA-based universal vaccine strategy against influenza variants.
Collapse
Affiliation(s)
- Zirong Han
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, China; Shenzhen Key Laboratory of Pathogenic Microbes and Biosafety, Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
| | - Qianyi Mai
- State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yangguo Zhao
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, China; Shenzhen Key Laboratory of Pathogenic Microbes and Biosafety, Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
| | - Xinglai Liu
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, China; Shenzhen Key Laboratory of Pathogenic Microbes and Biosafety, Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
| | - Mingting Cui
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, China; Shenzhen Key Laboratory of Pathogenic Microbes and Biosafety, Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
| | - Minchao Li
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, China; Shenzhen Key Laboratory of Pathogenic Microbes and Biosafety, Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
| | - Yaoqing Chen
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, China; Shenzhen Key Laboratory of Pathogenic Microbes and Biosafety, Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
| | - Yuelong Shu
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, China; Shenzhen Key Laboratory of Pathogenic Microbes and Biosafety, Shenzhen Campus of Sun Yat-sen University, Shenzhen, 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 & Peking Union Medical College, Beijing, China
| | - Jianhui Gan
- Shenzhen Kangtai Biological Products Co., Ltd, Shenzhen, 518057, China.
| | - Weiqi Pan
- State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| | - Caijun Sun
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, China; Shenzhen Key Laboratory of Pathogenic Microbes and Biosafety, Shenzhen Campus of Sun Yat-sen University, Shenzhen, China; Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, China; State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
2
|
Lanfermeijer J, van de Ven K, Hendriks M, van Dijken H, Lenz S, Vos M, Borghans JAM, van Baarle D, de Jonge J. The Memory-CD8+-T-Cell Response to Conserved Influenza Virus Epitopes in Mice Is Not Influenced by Time Since Previous Infection. Vaccines (Basel) 2024; 12:419. [PMID: 38675801 PMCID: PMC11054904 DOI: 10.3390/vaccines12040419] [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: 03/14/2024] [Revised: 03/24/2024] [Accepted: 04/03/2024] [Indexed: 04/28/2024] Open
Abstract
To protect older adults against influenza A virus (IAV) infection, innovative strategies are imperative to overcome the decrease in protective immune response with age. One approach involves the boosting of CD8+ T cells at middle age that were previously induced by natural infection. At this stage, the immune system is still fit. Given the high conservation of T-cell epitopes within internal viral proteins, such a response may confer lasting protection against evolving influenza strains at older age, also reducing the high number of influenza immunizations currently required. However, at the time of vaccination, some individuals may have been more recently exposed to IAV than others, which could affect the T-cell response. We therefore investigated the fundamental principle of how the interval between the last infection and booster immunization during middle age influences the CD8+ T-cell response. To model this, female mice were infected at either 6 or 9 months of age and subsequently received a heterosubtypic infection booster at middle age (12 months). Before the booster infection, 6-month-primed mice displayed lower IAV-specific CD8+ T-cell responses in the spleen and lung than 9-month-primed mice. Both groups were better protected against the subsequent heterosubtypic booster infection compared to naïve mice. Notably, despite the different CD8+ T-cell levels between the 6-month- and 9-month-primed mice, we observed comparable responses after booster infection, based on IFNγ responses, and IAV-specific T-cell frequencies and repertoire diversity. Lung-derived CD8+ T cells of 6- and 9-month-primed mice expressed similar levels of tissue-resident memory-T-cell markers 30 days post booster infection. These data suggest that the IAV-specific CD8+ T-cell response after boosting is not influenced by the time post priming.
Collapse
Affiliation(s)
- Josien Lanfermeijer
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, 3720 BA Bilthoven, The Netherlands
- Center for Translational Immunology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
- AstraZeneca, 2594 AV Den Haag, The Netherlands
| | - Koen van de Ven
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, 3720 BA Bilthoven, The Netherlands
- DICA (Dutch Institute for Clinical Auditing), 2333 AA Leiden, The Netherlands
| | - Marion Hendriks
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, 3720 BA Bilthoven, The Netherlands
- Deventer Ziekenhuis, 7416 SE Deventer, The Netherlands
| | - Harry van Dijken
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, 3720 BA Bilthoven, The Netherlands
| | - Stefanie Lenz
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, 3720 BA Bilthoven, The Netherlands
- MSD Animal Health, 5830 AA Boxmeer, The Netherlands
| | - Martijn Vos
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, 3720 BA Bilthoven, The Netherlands
| | - José A. M. Borghans
- Center for Translational Immunology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Debbie van Baarle
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, 3720 BA Bilthoven, The Netherlands
- Center for Translational Immunology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
- Virology & Immunology Research, Department Medical Microbiology and Infection Prevention, University Medical Center Groningen, 9700 RB Groningen, The Netherlands
| | - Jørgen de Jonge
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, 3720 BA Bilthoven, The Netherlands
| |
Collapse
|
3
|
Petro-Turnquist E, Corder Kampfe B, Gadeken A, Pekarek MJ, Weaver EA. Multivalent Epigraph Hemagglutinin Vaccine Protects against Influenza B Virus in Mice. Pathogens 2024; 13:97. [PMID: 38392835 PMCID: PMC10892733 DOI: 10.3390/pathogens13020097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 01/18/2024] [Accepted: 01/22/2024] [Indexed: 02/25/2024] Open
Abstract
Influenza B virus is a respiratory pathogen that contributes to seasonal epidemics, accounts for approximately 25% of global influenza infections, and can induce severe disease in young children. While vaccination is the most commonly used method of preventing influenza infections, current vaccines only induce strain-specific responses and have suboptimal efficacy when mismatched from circulating strains. Further, two influenza B virus lineages have been described, B/Yamagata-like and B/Victoria-like, and the limited cross-reactivity between the two lineages provides an additional barrier in developing a universal influenza B virus vaccine. Here, we report a novel multivalent vaccine using computationally designed Epigraph hemagglutinin proteins targeting both the B/Yamagata-like and B/Victoria-like lineages. When compared to the quadrivalent commercial vaccine, the Epigraph vaccine demonstrated increased breadth of neutralizing antibody and T cell responses. After lethal heterologous influenza B virus challenge, mice immunized with the Epigraph vaccine were completely protected against both weight loss and mortality. The superior cross-reactive immunity conferred by the Epigraph vaccine immunogens supports their continued investigation as a universal influenza B virus vaccine.
Collapse
Affiliation(s)
- Erika Petro-Turnquist
- Nebraska Center for Virology, School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
| | - Brigette Corder Kampfe
- Nebraska Center for Virology, School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
- Science Department, North Arkansas College, Harrison, AR 72601, USA
| | - Amber Gadeken
- College of Agricultural Sciences and Natural Resources, University of Nebraska-Lincoln, Lincoln, NE 68583, USA;
| | - Matthew J. Pekarek
- Nebraska Center for Virology, School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
| | - Eric A. Weaver
- Nebraska Center for Virology, School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
| |
Collapse
|
4
|
Trubach D, Muleta KG, Lahl K, Saini SK. Identification and Characterization of Antigen-Specific T-Cells in Viral Infections. Methods Mol Biol 2024; 2813:295-308. [PMID: 38888785 DOI: 10.1007/978-1-0716-3890-3_19] [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] [Indexed: 06/20/2024]
Abstract
Identification and characterization of CD8+ T-cells is important to determine their role in protecting and clearing viral infections. Here we provide details of the peptide-MHC (pMHC) tetramers-based approach to identify antigen-specific T-cells in human and murine samples. This method provides ex vivo quantification and functional characterization of T-cells reactive to specific viral antigens derived from CMV and rotavirus in human blood and in murine intestinal lamina propria samples, respectively.
Collapse
Affiliation(s)
- Darya Trubach
- Section for Experimental and Translational Immunology, Institute for Health Technology, Technical University of Denmark (DTU), Kongens Lyngby, Denmark
| | | | - Katharina Lahl
- Section for Experimental and Translational Immunology, Institute for Health Technology, Technical University of Denmark (DTU), Kongens Lyngby, Denmark
- Immunology Section, Lund University, Lund, Sweden
| | - Sunil Kumar Saini
- Section for Experimental and Translational Immunology, Institute for Health Technology, Technical University of Denmark (DTU), Kongens Lyngby, Denmark.
| |
Collapse
|
5
|
Heng WT, Lim HX, Tan KO, Poh CL. Validation of Multi-epitope Peptides Encapsulated in PLGA Nanoparticles Against Influenza A Virus. Pharm Res 2023; 40:1999-2025. [PMID: 37344603 DOI: 10.1007/s11095-023-03540-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Accepted: 05/19/2023] [Indexed: 06/23/2023]
Abstract
BACKGROUND Influenza is a highly contagious respiratory disease which poses a serious threat to public health globally, causing severe diseases in 3-5 million humans and resulting in 650,000 deaths annually. The current licensed seasonal influenza vaccines lacked cross-reactivity against novel emerging influenza strains as they conferred limited neutralising capabilities. To address the issue, we designed a multi-epitope peptide-based vaccine delivered by the self-adjuvanting PLGA nanoparticles against influenza infections. METHODS A total of six conserved peptides representing B- and T-cell epitopes of Influenza A were identified and they were formulated in either incomplete Freund's adjuvant containing CpG ODN 1826 or being encapsulated in PLGA nanoparticles for the evaluation of immunogenicity in BALB/c mice. RESULTS The self-adjuvanting PLGA nanoparticles encapsulating the six conserved peptides were capable of eliciting the highest levels of IgG and IFN- γ producing cells. In addition, the immunogenicity of the six peptides encapsulated in PLGA nanoparticles showed greater humoral and cellular mediated immune responses elicited by the mixture of six naked peptides formulated in incomplete Freund's adjuvant containing CpG ODN 1826 in the immunized mice. Peptide 3 from the mixture of six peptides was found to exert necrotic effect on CD3+ T-cells and this finding indicated that peptide 3 should be removed from the nanovaccine formulation. CONCLUSION The study demonstrated the self-adjuvanting properties of the PLGA nanoparticles as a delivery system without the need for incorporation of toxic and costly conventional adjuvants in multi-epitope peptide-based vaccines.
Collapse
Affiliation(s)
- Wen Tzuen Heng
- Centre for Virus and Vaccine Research (CVVR), School of Medical and Life Sciences, Sunway University, No.5 Jalan Universiti, 47500, Petaling Jaya, Selangor Darul Ehsan, Malaysia
| | - Hui Xuan Lim
- Centre for Virus and Vaccine Research (CVVR), School of Medical and Life Sciences, Sunway University, No.5 Jalan Universiti, 47500, Petaling Jaya, Selangor Darul Ehsan, Malaysia
| | - Kuan Onn Tan
- Department of Biological Sciences, School of Medical and Life Sciences, Sunway University, No.5 Jalan Universiti, 47500, Petaling Jaya, Selangor Darul Ehsan, Malaysia
| | - Chit Laa Poh
- Centre for Virus and Vaccine Research (CVVR), School of Medical and Life Sciences, Sunway University, No.5 Jalan Universiti, 47500, Petaling Jaya, Selangor Darul Ehsan, Malaysia.
| |
Collapse
|
6
|
Atmar RL, Bernstein DI, Winokur P, Frey SE, Angelo LS, Bryant C, Ben-Yedidia T, Roberts PC, El Sahly HM, Keitel WA. Safety and immunogenicity of Multimeric-001 (M-001) followed by seasonal quadrivalent inactivated influenza vaccine in young adults - A randomized clinical trial. Vaccine 2023; 41:2716-2722. [PMID: 36941155 PMCID: PMC10396309 DOI: 10.1016/j.vaccine.2023.03.023] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 03/01/2023] [Accepted: 03/10/2023] [Indexed: 03/23/2023]
Abstract
BACKGROUND The continuing evolution of influenza viruses poses a challenge to vaccine prevention, highlighting the need for a universal influenza vaccine. We evaluated the safety and immunogenicity of one such candidate, Multimeric-001 (M-001), when used as a priming vaccine prior to administration of quadrivalent inactivated influenza vaccine (IIV4). METHODS Healthy adults 18 to 49 years of age were enrolled in a phase 2 randomized, double-blind placebo-controlled trial. Participants received two doses of either 1.0-mg M-001 or saline placebo (60 per study arm) on Days 1 and 22 followed by a single dose of IIV4 on about Day 172. Safety, reactogenicity, cellular immune responses and influenza hemagglutination inhibition (HAI) and microneutralization (MN) were assessed. RESULTS The M-001 vaccine was safe and had an acceptable reactogenicity profile. Injection site tenderness (39% post-dose 1, 29% post-dose 2) was the most common reaction after M-001 administration. Polyfunctional CD4+ T cell responses (perforin-negative, CD107α-negative, TNF-α+, IFN-γ+, with or without IL-2) to the pool of M-001 peptides increased significantly from baseline to two weeks after the second dose of M-001, and this increase persisted through Day 172. However, there was no enhancement of HAI or MN antibody responses among M-001 recipients following IIV4 administration. CONCLUSIONS M-001 administration induced a subset of polyfunctional CD4+ T cells that persisted through 6 months of follow-up, but it did not improve HAI or MN antibody responses to IIV4. (clinicaltrials.gov NCT03058692).
Collapse
Affiliation(s)
- Robert L Atmar
- Department of Medicine, Baylor College of Medicine, Houston, TX, United States; Department of Molecular Virology & Microbiology, Baylor College of Medicine, Houston, TX, United States.
| | - David I Bernstein
- Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Patricia Winokur
- University of Iowa College of Medicine, Iowa City, IA, United States
| | - Sharon E Frey
- Saint Louis University School of Medicine, St. Louis, MO, United States
| | - Laura S Angelo
- Department of Molecular Virology & Microbiology, Baylor College of Medicine, Houston, TX, United States
| | | | | | - Paul C Roberts
- Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Hana M El Sahly
- Department of Medicine, Baylor College of Medicine, Houston, TX, United States; Department of Molecular Virology & Microbiology, Baylor College of Medicine, Houston, TX, United States
| | - Wendy A Keitel
- Department of Medicine, Baylor College of Medicine, Houston, TX, United States; Department of Molecular Virology & Microbiology, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
7
|
Ogura H, Gohda J, Lu X, Yamamoto M, Takesue Y, Son A, Doi S, Matsushita K, Isobe F, Fukuda Y, Huang TP, Ueno T, Mambo N, Murakami H, Kawaguchi Y, Inoue JI, Shirai K, Yamasaki S, Hirata JI, Ishido S. Dysfunctional Sars-CoV-2-M protein-specific cytotoxic T lymphocytes in patients recovering from severe COVID-19. Nat Commun 2022; 13:7063. [PMID: 36526616 PMCID: PMC9758236 DOI: 10.1038/s41467-022-34655-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 11/02/2022] [Indexed: 12/23/2022] Open
Abstract
Although the importance of virus-specific cytotoxic T lymphocytes (CTL) in virus clearance is evident in COVID-19, the characteristics of virus-specific CTLs related to disease severity have not been fully explored. Here we show that the phenotype of virus-specific CTLs against immunoprevalent epitopes in COVID-19 convalescents might differ according to the course of the disease. We establish a cellular screening method that uses artificial antigen presenting cells, expressing HLA-A*24:02, the costimulatory molecule 4-1BBL, SARS-CoV-2 structural proteins S, M, and N and non-structural proteins ORF3a and nsp6/ORF1a. The screen implicates SARS-CoV-2 M protein as a frequent target of IFNγ secreting CD8+ T cells, and identifies M198-206 as an immunoprevalent epitope in our cohort of HLA-A*24:02 positive convalescent COVID-19 patients recovering from mild, moderate and severe disease. Further exploration of M198-206-specific CD8+ T cells with single cell RNA sequencing reveals public TCRs in virus-specific CD8+ T cells, and shows an exhausted phenotype with less differentiated status in cells from the severe group compared to cells from the moderate group. In summary, this study describes a method to identify T cell epitopes, indicate that dysfunction of virus-specific CTLs might be an important determinant of clinical outcomes.
Collapse
Affiliation(s)
- Hideki Ogura
- grid.272264.70000 0000 9142 153XDepartment of Microbiology, Hyogo Medical University, Hyogo, Japan
| | - Jin Gohda
- grid.26999.3d0000 0001 2151 536XResearch Center for Asian Infectious Diseases, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Xiuyuan Lu
- grid.136593.b0000 0004 0373 3971Laboratory of Molecular Immunology, Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Mizuki Yamamoto
- grid.26999.3d0000 0001 2151 536XResearch Center for Asian Infectious Diseases, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Yoshio Takesue
- grid.272264.70000 0000 9142 153XDepartment of Infection Control and Prevention, Hyogo Medical University, Hyogo, Japan ,Tokoname City Hospital, Aichi, Japan
| | - Aoi Son
- grid.272264.70000 0000 9142 153XDepartment of Microbiology, Hyogo Medical University, Hyogo, Japan
| | - Sadayuki Doi
- grid.513274.60000 0004 0569 8532Kawanishi City Hospital, Hyogo, Japan
| | | | - Fumitaka Isobe
- Kyowa Marina Hospital/Wellhouse Nishinomiya, Hyogo, Japan
| | | | | | - Takamasa Ueno
- grid.274841.c0000 0001 0660 6749Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan
| | - Naomi Mambo
- grid.272264.70000 0000 9142 153XDepartment of Emergency and Critical Care Medicine, Hyogo Medical University, Hyogo, Japan
| | - Hiromoto Murakami
- grid.272264.70000 0000 9142 153XDepartment of Emergency and Critical Care Medicine, Hyogo Medical University, Hyogo, Japan
| | - Yasushi Kawaguchi
- grid.26999.3d0000 0001 2151 536XResearch Center for Asian Infectious Diseases, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan ,grid.26999.3d0000 0001 2151 536XDivision of Molecular Virology, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Jun-ichiro Inoue
- grid.26999.3d0000 0001 2151 536XResearch Platform Office, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Kunihiro Shirai
- grid.272264.70000 0000 9142 153XDepartment of Emergency and Critical Care Medicine, Hyogo Medical University, Hyogo, Japan
| | - Sho Yamasaki
- grid.136593.b0000 0004 0373 3971Laboratory of Molecular Immunology, Immunology Frontier Research Center, Osaka University, Suita, Japan ,grid.136593.b0000 0004 0373 3971Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Suita, Japan ,grid.177174.30000 0001 2242 4849Division of Molecular Design, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan ,grid.136304.30000 0004 0370 1101Division of Molecular Immunology, Medical Mycology Research Center, Chiba University, Chiba, Japan
| | - Jun-Ichi Hirata
- grid.272264.70000 0000 9142 153XDepartment of Emergency and Critical Care Medicine, Hyogo Medical University, Hyogo, Japan
| | - Satoshi Ishido
- grid.272264.70000 0000 9142 153XDepartment of Microbiology, Hyogo Medical University, Hyogo, Japan
| |
Collapse
|
8
|
Hartwell M, Greiner BH, Nicks S. Examination of Homologies between COVID-19 Vaccines and Common Allergens: The Potential for T Cell-mediated Responses for Allergic Rhinitis and Asthma. INFECTIOUS DISEASES & IMMUNITY 2022. [PMID: 37520897 PMCID: PMC9612419 DOI: 10.1097/id9.0000000000000056] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
As the SARS-CoV-2 virus shares relatively large protein sequences homologous to grass pollens, dust mites, and molds, our objective was to assess the potential overlap between the COVID-19 mRNA vaccines from Pfizer-BioNtech and Moderna and known allergens. We found 7 common allergens with potential for cross-reactivity with the Pfizer vaccine and 19 with the Moderna vaccine, including common grasses, molds, and dust mites. T-cell mediated antigen cross-reactivity between viruses and allergens is a relatively new area of study in clinical immunology; a discipline that may be particularly useful regarding the SARS-CoV-2 virus and the allergic response in humans. These results suggest that vaccination with the Pfizer-BioNtech and Moderna COVID-19 vaccines may contribute to T-cell cross-reactivity with allergens that impact allergic asthma and allergic rhinitis. Further research should assess the clinical implications of COVID-19 vaccination on the severity and symptomatology of the allergic disease, in addition to natural viral infection.
Collapse
|
9
|
Leroux-Roels I, Waerlop G, Tourneur J, De Boever F, Maes C, Bruhwyler J, Guyon-Gellin D, Moris P, Del Campo J, Willems P, Leroux-Roels G, Le Vert A, Nicolas F. Randomized, Double-Blind, Reference-Controlled, Phase 2a Study Evaluating the Immunogenicity and Safety of OVX836, A Nucleoprotein-Based Influenza Vaccine. Front Immunol 2022; 13:852904. [PMID: 35464450 PMCID: PMC9022189 DOI: 10.3389/fimmu.2022.852904] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 03/09/2022] [Indexed: 11/13/2022] Open
Abstract
OVX836 is a recombinant protein-based vaccine targeting the highly conserved influenza nucleoprotein (NP), which aims to confer a broad-spectrum protection against influenza. In a Phase 1 study, OVX836, administered intramuscularly, has been found safe and immunogenic. The 90µg and 180µg dose levels were selected to be further evaluated in this randomized, monocenter, reference-controlled (Influvac Tetra™: quadrivalent seasonal influenza subunit vaccine), parallel group, double-blind, Phase 2a study in 300 healthy volunteers, aged 18-65 years, during the 2019/2020 flu season. Safety, influenza-like illness episodes (ILI; based on the Flu-PRO® questionnaire) and immunogenicity were assessed up to 180 days post-vaccination. OVX836 was safe and presented a reactogenicity profile similar to Influvac Tetra. It induced a significant increase in terms of NP-specific interferon-gamma (IFNγ) spot forming cells (SFCs), NP-specific CD4+ T-cells (essentially polyfunctional cells) and anti-NP IgG responses. OVX836 was superior to Influvac Tetra for all immunological parameters related to NP, and the 180µg dose was significantly superior to the 90µg dose for SFCs and CD4+ T-cells expressing IFNγ. Both the CD4+ T-cell and the anti-NP IgG responses persisted up to Day 180. An efficacy signal was observed with OVX836 at 180µg through reduction of ILI episodes occurring during the flu season as of 14 days post-vaccination. In conclusion, these results encourage further clinical evaluation of OVX836 in order to confirm the signal of efficacy on ILIs and/or laboratory-confirmed influenza cases. NCT04192500 (https://clinicaltrials.gov/ct2/show/study/NCT04192500).
Collapse
Affiliation(s)
- Isabel Leroux-Roels
- Center for Vaccinology (CEVAC), Ghent University and University Hospital, Ghent, Belgium
| | - Gwenn Waerlop
- Center for Vaccinology (CEVAC), Ghent University and University Hospital, Ghent, Belgium
| | | | - Fien De Boever
- Center for Vaccinology (CEVAC), Ghent University and University Hospital, Ghent, Belgium
| | - Catherine Maes
- Center for Vaccinology (CEVAC), Ghent University and University Hospital, Ghent, Belgium
| | | | | | | | | | | | - Geert Leroux-Roels
- Center for Vaccinology (CEVAC), Ghent University and University Hospital, Ghent, Belgium
| | | | | |
Collapse
|
10
|
Blunck BN, Angelo LS, Henke D, Avadhanula V, Cusick M, Ferlic-Stark L, Zechiedrich L, Gilbert BE, Piedra PA. Adult Memory T Cell Responses to the Respiratory Syncytial Virus Fusion Protein During a Single RSV Season (2018-2019). Front Immunol 2022; 13:823652. [PMID: 35422803 PMCID: PMC9002099 DOI: 10.3389/fimmu.2022.823652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Accepted: 03/02/2022] [Indexed: 11/13/2022] Open
Abstract
Respiratory Syncytial Virus (RSV) is ubiquitous and re-infection with both subtypes (RSV/A and RSV/B) is common. The fusion (F) protein of RSV is antigenically conserved, induces neutralizing antibodies, and is a primary target of vaccine development. Insight into the breadth and durability of RSV-specific adaptive immune response, particularly to the F protein, may shed light on susceptibility to re-infection. We prospectively enrolled healthy adult subjects (n = 19) and collected serum and peripheral blood mononuclear cells (PBMCs) during the 2018–2019 RSV season. Previously, we described their RSV-specific antibody responses and identified three distinct antibody kinetic profiles associated with infection status: uninfected (n = 12), acutely infected (n = 4), and recently infected (n = 3). In this study, we measured the longevity of RSV-specific memory T cell responses to the F protein following natural RSV infection. We stimulated PBMCs with overlapping 15-mer peptide libraries spanning the F protein derived from either RSV/A or RSV/B and found that memory T cell responses mimic the antibody responses for all three groups. The uninfected group had stable, robust memory T cell responses and polyfunctionality. The acutely infected group had reduced polyfunctionality of memory T cell response at enrollment compared to the uninfected group, but these returned to comparable levels by end-of-season. The recently infected group, who were unable to maintain high levels of RSV-specific antibody following infection, similarly had decreased memory T cell responses and polyfunctionality during the RSV season. We observed subtype-specific differences in memory T cell responses and polyfunctionality, with RSV/A stimulating stronger memory T cell responses with higher polyfunctionality even though RSV/B was the dominant subtype in circulation. A subset of individuals demonstrated an overall deficiency in the generation of a durable RSV-specific adaptive immune response. Because memory T cell polyfunctionality may be associated with protection against re-infection, this latter group would likely be at greater risk of re-infection. Overall, these results expand our understanding of the longevity of the adaptive immune response to the RSV fusion protein and should be considered in future vaccine development efforts.
Collapse
Affiliation(s)
- Brittani N Blunck
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States
| | - Laura S Angelo
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States
| | - David Henke
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States
| | - Vasanthi Avadhanula
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States
| | - Matthew Cusick
- Department of Pathology, University of Michigan, Ann Arbor, MI, United States
| | - Laura Ferlic-Stark
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States
| | - Lynn Zechiedrich
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States.,Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX, United States.,Department of Pharmacology and Chemical Biology, Baylor College of Medicine, Houston, TX, United States
| | - Brian E Gilbert
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States
| | - Pedro A Piedra
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States.,Department of Pediatrics, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
11
|
Tate MC, Roy A, Pinchinat M, Lund E, Fox JB, Cottrill S, Vaccaro A, Stein LAR. Impact of Being a Peer Recovery Specialist on Work and Personal Life: Implications for Training and Supervision. Community Ment Health J 2022; 58:193-204. [PMID: 33677802 DOI: 10.1007/s10597-021-00811-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Accepted: 02/24/2021] [Indexed: 11/30/2022]
Abstract
Peer recovery specialists are an important resource in community mental health settings. This study, which was part of a larger statewide assessment, evaluates how the role impacts work and personal lives of peers, with implications for improving the training and supervision of this service. The importance of peer work has been investigated through client outcomes, however less work has investigated outcomes on peers themselves, which impacts the work force and service delivery. Nine focus groups were conducted with peer recovery specialists. A two-stage qualitative analysis led to two overarching themes, work and personal, and six subthemes. Findings suggest being a peer presents unique benefits and challenges in work and personal life. Peers benefit from more training and supervision, consistency within the role, and maintaining boundaries. Additionally, work environment roles may be improved by attention to needs of supervisors in terms of skills for effective supervision and clarification of supervisory roles.
Collapse
Affiliation(s)
- Marie C Tate
- University of Rhode Island, 130 Flagg Rd, Kingston, RI, 02881, USA.
| | - Amanda Roy
- University of Rhode Island, 130 Flagg Rd, Kingston, RI, 02881, USA
| | - Meinca Pinchinat
- University of Rhode Island, 130 Flagg Rd, Kingston, RI, 02881, USA
| | - Emma Lund
- University of Rhode Island, 130 Flagg Rd, Kingston, RI, 02881, USA
| | - Judith B Fox
- Department of Behavioral Health, Developmental Disabilities, and Hospitals, 41 West Rd, Cranston, RI, 02920, USA
| | - Sara Cottrill
- University of Rhode Island, 130 Flagg Rd, Kingston, RI, 02881, USA
| | - Annemarie Vaccaro
- University of Rhode Island, 6 Rhody Ram Way, Kingston, RI, 02881, USA
| | - L A R Stein
- University of Rhode Island, 130 Flagg Rd, Kingston, RI, 02881, USA.,Department of Behavioral Health, Developmental Disabilities, and Hospitals, 41 West Rd, Cranston, RI, 02920, USA.,Center for Alcohol & Addiction Studies, and Dept. of Behavioral & Social Sciences, School of Public Health, Brown University, 121 South Main St, Providence, RI, 02903, USA.,Rhode Island Training School, 57 Power Rd, Cranston, RI, 02920, USA
| |
Collapse
|
12
|
Withanage K, De Coster I, Cools N, Viviani S, Tourneur J, Chevandier M, Lambiel M, Willems P, Le Vert A, Nicolas F, Van Damme P. Phase 1 randomized, placebo-controlled, dose-escalating study to evaluate OVX836, a nucleoprotein-based influenza vaccine: intramuscular results. J Infect Dis 2021; 226:119-127. [PMID: 34653245 PMCID: PMC9373130 DOI: 10.1093/infdis/jiab532] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 10/13/2021] [Indexed: 11/12/2022] Open
Abstract
Background OVX836 is a recombinant protein vaccine targeting the highly conserved influenza nucleoprotein (NP), which could confer broad-spectrum protection against this disease. Methods A randomized, placebo-controlled, double-blind, dose-escalating, single- center, first-in-human study was conducted in 36 healthy adults aged 18–49 years. Twelve subjects per cohort (9 vaccine and 3 placebo) received 2 OVX836 intramuscular administrations on days 1 and 28 at the dose level of 30 µg, 90 µg, or 180 µg. Safety and immunogenicity were assessed after each vaccination and for 150 days in total. Results OVX836 was safe and well tolerated at all dose levels, with no difference in solicited local and systemic symptoms, and unsolicited adverse events between the first and second administration, or between dose levels. All subjects presented pre-existing NP-specific immunity at baseline. OVX836 induced a significant increase in NP-specific interferon-gamma T cells and anti-NP immunoglobulin G at all dose levels after the first vaccination. The second vaccination did not further increase the response. There was a trend for a dose effect in the immune response. Conclusions The safety and reactogenicity profile, as well as the humoral and cellular immune responses, encourage further evaluation of OVX836 in a larger Phase 2a study.
Collapse
Affiliation(s)
- Kanchanamala Withanage
- Vaccine and Infectious Disease Institute (VaxInfectio), Faculty of Medicine and Health Sciences, University of Antwerp, Campus Drie Eiken, Universiteitsplein, Antwerpen, Belgium
| | - Ilse De Coster
- Vaccine and Infectious Disease Institute (VaxInfectio), Faculty of Medicine and Health Sciences, University of Antwerp, Campus Drie Eiken, Universiteitsplein, Antwerpen, Belgium
| | - Nathalie Cools
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VaxInfectio), Faculty of Medicine and Health Sciences, University of Antwerp, Universiteitsplein, Antwerpen, Belgium
| | - Simonetta Viviani
- Independent Scientific Advisor on Vaccines, Epidemiology and Public Health, Via Gramsci, I-Monteriggioni, Siena, Italy. Acting as Chief Medical Officer for Osivax at the time of the clinical study
| | | | | | | | | | | | | | - Pierre Van Damme
- Vaccine and Infectious Disease Institute (VaxInfectio), Faculty of Medicine and Health Sciences, University of Antwerp, Campus Drie Eiken, Universiteitsplein, Antwerpen, Belgium
| |
Collapse
|
13
|
Paramsothy A, Lartey Jalloh S, Davies RA, Guttormsen AB, Cox RJ, Mohn KGI. Humoral and cellular immune responses in critically ill influenza A/H1N1-infected patients. Scand J Immunol 2021; 94:e13045. [PMID: 33891354 DOI: 10.1111/sji.13045] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 03/10/2021] [Accepted: 04/11/2021] [Indexed: 12/31/2022]
Abstract
There is limited knowledge of influenza-specific immune responses and their kinetics in critically ill patients. We investigated humoral and cellular immune responses after critical influenza A/H1N1 infection and hypothesized that dysfunctionality or absence of immune responses could contribute to more severe illness. We followed 12 patients hospitalized with severe influenza infection; the majority admitted to intensive care unit (ICU). Blood samples were collected at days 10 and 19 and at 5 months. Antibody responses to surface glycoproteins haemagglutinin (HA) and neuraminidase (NA) of A/H1N1pdm09 were quantified by haemagglutination inhibition (HAI), microneutralization (MN), Enzyme-linked immunosorbent assay (ELISA) and Enzyme-linked lectin assay (ELLA). Influenza-specific antibody levels and avidity were measured separately for head and stalk domains of H1. Cytokine secreting CD4+ and CD8+ T cell responses to conserved influenza epitopes (M1, NP and PB1) were analysed by FluoroSpot. Overall, the patients retained a high level of functional HA- and NA-specific antibodies over the study period. During the acute phase (up to 3 weeks from symptom onset), antibodies specific to H1 stalk increased earlier and were present in higher amount compared with H1 head-specific antibodies. The NA-specific antibodies and the non-neutralizing HA-specific antibody response for H1 head and H1 full-length showed a significant decline from acute to convalescent phase. Despite high total IgG concentrations, avidity to H1 head and H1 full-length protein remained low at all time points. Similarly, CD8+ T cell responses were continuously measured at low levels. In conclusion, our study found that critically ill patients were characterized by low HA-specific antibody avidity and CD8+ T cell response.
Collapse
Affiliation(s)
- Abira Paramsothy
- Department of Clinical Sciences, Influenza Centre, University of Bergen, Norway
| | - Sarah Lartey Jalloh
- Department of Clinical Sciences, Influenza Centre, University of Bergen, Norway
| | - Richard A Davies
- Department of Clinical Sciences, Influenza Centre, University of Bergen, Norway
| | - Anne-Berit Guttormsen
- Department of Anesthesia and Intensive Care, Haukeland University Hospital, Bergen, Norway.,Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Rebecca J Cox
- Department of Clinical Sciences, Influenza Centre, University of Bergen, Norway.,Department of Microbiology, Haukeland University Hospital, Bergen, Norway
| | - Kristin G-I Mohn
- Department of Clinical Sciences, Influenza Centre, University of Bergen, Norway.,Emergency Care Clinic, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
14
|
Zuo J, Dowell AC, Pearce H, Verma K, Long HM, Begum J, Aiano F, Amin-Chowdhury Z, Hoschler K, Brooks T, Taylor S, Hewson J, Hallis B, Stapley L, Borrow R, Linley E, Ahmad S, Parker B, Horsley A, Amirthalingam G, Brown K, Ramsay ME, Ladhani S, Moss P. Robust SARS-CoV-2-specific T cell immunity is maintained at 6 months following primary infection. Nat Immunol 2021; 22:620-626. [PMID: 33674800 PMCID: PMC7610739 DOI: 10.1038/s41590-021-00902-8] [Citation(s) in RCA: 255] [Impact Index Per Article: 63.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Accepted: 02/19/2021] [Indexed: 12/21/2022]
Abstract
The immune response to SARS-CoV-2 is critical in controlling disease, but there is concern that waning immunity may predispose to reinfection. We analyzed the magnitude and phenotype of the SARS-CoV-2-specific T cell response in 100 donors at 6 months following infection. T cell responses were present by ELISPOT and/or intracellular cytokine staining analysis in all donors and characterized by predominant CD4+ T cell responses with strong interleukin (IL)-2 cytokine expression. Median T cell responses were 50% higher in donors who had experienced a symptomatic infection, indicating that the severity of primary infection establishes a 'set point' for cellular immunity. T cell responses to spike and nucleoprotein/membrane proteins were correlated with peak antibody levels. Furthermore, higher levels of nucleoprotein-specific T cells were associated with preservation of nucleoprotein-specific antibody level although no such correlation was observed in relation to spike-specific responses. In conclusion, our data are reassuring that functional SARS-CoV-2-specific T cell responses are retained at 6 months following infection.
Collapse
Affiliation(s)
- Jianmin Zuo
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Alexander C Dowell
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Hayden Pearce
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Kriti Verma
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Heather M Long
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Jusnara Begum
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Felicity Aiano
- Immunisation and Countermeasures Division, National Infection Service, London, UK
| | - Zahin Amin-Chowdhury
- Immunisation and Countermeasures Division, National Infection Service, London, UK
| | - Katja Hoschler
- Immunisation and Countermeasures Division, National Infection Service, London, UK
| | - Tim Brooks
- Immunoassay Laboratory, National Infection Service, Porton Down, UK
| | - Stephen Taylor
- Immunoassay Laboratory, National Infection Service, Porton Down, UK
| | - Jacqueline Hewson
- Immunisation and Countermeasures Division, National Infection Service, London, UK
| | - Bassam Hallis
- Immunoassay Laboratory, National Infection Service, Porton Down, UK
| | - Lorrain Stapley
- Immunoassay Laboratory, National Infection Service, Porton Down, UK
| | - Ray Borrow
- Sero-epidemiology Unit, Public Health England, Public Health Laboratory Manchester, Manchester Medical Microbiology Partnership, Manchester Royal Infirmary, Manchester, UK
| | - Ezra Linley
- Sero-epidemiology Unit, Public Health England, Public Health Laboratory Manchester, Manchester Medical Microbiology Partnership, Manchester Royal Infirmary, Manchester, UK
| | - Shazaad Ahmad
- Department of Virology, Manchester Medical Microbiology Partnership, Manchester Foundation Trust, Manchester Academic Health Sciences Centre, Manchester, UK
| | - Ben Parker
- The NIHR Manchester Clinical Research Facility, Manchester University NHS Foundation Trust, Manchester, UK
- Kellgren Centre for Rheumatology, NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester, UK
| | - Alex Horsley
- The NIHR Manchester Clinical Research Facility, Manchester University NHS Foundation Trust, Manchester, UK
- Division of Infection, Immunity and Respiratory Medicine, University of Manchester, Manchester, UK
| | | | - Kevin Brown
- Immunisation and Countermeasures Division, National Infection Service, London, UK
| | - Mary E Ramsay
- Immunisation and Countermeasures Division, National Infection Service, London, UK
| | - Shamez Ladhani
- Immunisation and Countermeasures Division, National Infection Service, London, UK.
- Paediatric Infectious Diseases Research Group (PIDRG), St. Georges University of London (SGUL), London, UK.
| | - Paul Moss
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK.
| |
Collapse
|
15
|
Topham DJ, DeDiego ML, Nogales A, Sangster MY, Sant A. Immunity to Influenza Infection in Humans. Cold Spring Harb Perspect Med 2021; 11:a038729. [PMID: 31871226 PMCID: PMC7919402 DOI: 10.1101/cshperspect.a038729] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
This review discusses the human immune responses to influenza infection with some insights from studies using animal models, such as experimental infection of mice. Recent technological advances in the study of human immune responses have greatly added to our knowledge of the infection and immune responses, and therefore much of the focus is on recent studies that have moved the field forward. We consider the complexity of the adaptive response generated by many sequential encounters through infection and vaccination.
Collapse
Affiliation(s)
- David J Topham
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York 14642, USA
| | - Marta L DeDiego
- Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Cientificas, 28049 Madrid, Spain
| | - Aitor Nogales
- Instituto Nacional de Investigación y Tecnologia Agraria y Ailmentaria, 28040 Madrid, Spain
| | - Mark Y Sangster
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York 14642, USA
| | - Andrea Sant
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York 14642, USA
| |
Collapse
|
16
|
Mohn KGI, Brokstad KA, Islam S, Oftung F, Tøndel C, Aarstad HJ, Cox RJ. Early Induction of Cross-Reactive CD8+ T-Cell Responses in Tonsils After Live-Attenuated Influenza Vaccination in Children. J Infect Dis 2021; 221:1528-1537. [PMID: 32255493 PMCID: PMC7137893 DOI: 10.1093/infdis/jiz583] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 12/10/2019] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Live-attenuated influenza vaccine (LAIV) was licensed for prophylaxis of children 2-17 years old in Europe in 2012 and is administered as a nasal spray. Live-attenuated influenza vaccine induces both mucosal and systemic antibodies and systemic T-cell responses. Tonsils are the lymph nodes serving the upper respiratory tract, acting as both induction and effector site for mucosal immunity. METHODS Here, we have studied the early tonsillar T-cell responses induced in children after LAIV. Thirty-nine children were immunized with trivalent LAIV (containing A/H1N1, A/H3N2, and B viruses) at days 3, 7, and 14 before tonsillectomy. Nonvaccinated controls were included for comparison. Tonsils and peripheral blood (pre- and postvaccination) were collected to study T-cell responses. RESULTS Tonsillar and systemic T-cell responses differed between influenza strains, and both were found against H3N2 and B viruses, whereas only systemic responses were observed against A/H1N1. A significant increase in cross-reactive tonsillar CD8+ T cells recognizing conserved epitopes from a broad range of seasonal and pandemic viruses occurred at day 14. Tonsillar T cells showed significant cytokine responses (Th1, Th2, and granulocyte-macrophage colony-stimulating factor). CONCLUSIONS Our findings support the use of LAIV in children to elicit broadly cross-reactive T cells, which are not induced by traditional inactivated influenza vaccines and may provide protection to novel virus strains.
Collapse
Affiliation(s)
- K G-I Mohn
- Influenza Centre, University of Bergen, Bergen, Norway.,K. G. Jebsen Center for Influenza Vaccines, University of Bergen, Bergen, Norway.,Emergency Care Clinic, Haukeland University Hospital, Bergen, Norway
| | - K A Brokstad
- Broegelmann Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - S Islam
- Influenza Centre, University of Bergen, Bergen, Norway.,K. G. Jebsen Center for Influenza Vaccines, University of Bergen, Bergen, Norway
| | - F Oftung
- Department of Infectious Disease Immunology, Norwegian Institute of Public Health, Oslo, Norway
| | - C Tøndel
- Department of Pediatrics, Haukeland University Hospital, Bergen, Norway
| | - H J Aarstad
- Department of Otorhinolaryngology/Head and Neck Surgery, Haukeland University Hospital, Bergen, Norway
| | - R J Cox
- Influenza Centre, University of Bergen, Bergen, Norway.,K. G. Jebsen Center for Influenza Vaccines, University of Bergen, Bergen, Norway.,Department of Research and Development, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
17
|
Sant AJ. The Way Forward: Potentiating Protective Immunity to Novel and Pandemic Influenza Through Engagement of Memory CD4 T Cells. J Infect Dis 2020; 219:S30-S37. [PMID: 30715376 PMCID: PMC6452298 DOI: 10.1093/infdis/jiy666] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Potentially pandemic strains of influenza pose an undeniable threat to human populations. Therefore, it is essential to develop better strategies to enhance vaccine design and predict parameters that identify susceptible humans. CD4 T cells are a central component of protective immunity to influenza, delivering direct effector function and potentiating responses of other lymphoid cells. Humans have highly diverse influenza-specific CD4 T-cell populations that vary in stimulation history, specificity, and functionality. These complexities constitute a formidable obstacle to predicting immune responses to pandemic strains of influenza and derivation of optimal vaccine strategies. We suggest that more precise efforts to identify and enumerate both the positive and negative contributors of immunity in the CD4 T-cell compartment will aid in both predicting susceptible hosts and in development of vaccination strategies that will poise most human subjects to respond to pandemic influenza strains with protective immune responses.
Collapse
Affiliation(s)
- Andrea J Sant
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, New York
| |
Collapse
|
18
|
Heterologous viral protein interactions within licensed seasonal influenza virus vaccines. NPJ Vaccines 2020; 5:3. [PMID: 31934357 PMCID: PMC6954117 DOI: 10.1038/s41541-019-0153-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 10/16/2019] [Indexed: 01/10/2023] Open
Abstract
Currently, licensed influenza virus vaccines are designed and tested only for their ability to elicit hemagglutinin (HA)-reactive, neutralizing antibodies. Despite this, the purification process in vaccine manufacturing often does not completely remove other virion components. In the studies reported here, we have examined the viral protein composition of a panel of licensed vaccines from different manufacturers and licensed in different years. Using western blotting, we found that, beyond HA proteins, there are detectable quantities of neuraminidase (NA), nucleoprotein (NP), and matrix proteins (M1) from both influenza A and influenza B viruses in the vaccines but that the composition differed by source and method of vaccine preparation. We also found that disparities in viral protein composition were associated with distinct patterns of elicited antibody specificities. Strikingly, our studies also revealed that many viral proteins contained in the vaccine form heterologous complexes. When H1 proteins were isolated by immunoprecipitation, NA (N1), M1 (M1-A), H3, and HA-B proteins were co-isolated with the H1. Further biochemical studies suggest that these interactions persist for at least 4 h at 37 °C and that the membrane/intracytoplasmic domains in the intact HA proteins are important for the intermolecular interactions detected. These studies indicate that, if such interactions persist after vaccines reach the draining lymph node, both dendritic cells and HA-specific B cells may take up multiple viral proteins simultaneously. Whether these interactions are beneficial or harmful to the developing immune response will depend on the functional potential of the elicited virus-specific CD4 T cells. Licensed influenza virus vaccines are evaluated for their ability to elicit neutralizing antibodies specific for hemagglutinin (HA), but the manufacturing process does not completely exclude other virion components from the formulations. Andrea Sant and colleagues now report the presence of several viral proteins, such as M1, NA, H3, and HA-B, in licensed formulations from different manufacturers and spanning stocks from several years. These viral proteins form heterologous complexes, and immunization of mice with some of the formulations analyzed elicited antibody responses specific to these viral proteins. These findings reveal heterogeneity across licensed influenza virus vaccine formulations, potentially due to variations in production processes, and raise the possibility that the presence of these additional viral protein complexes could influence the elicited immune responses following immunization, particularly in the context of multivalent strategies involving mixing of different formulations.
Collapse
|
19
|
Corder BN, Bullard BL, DeBeauchamp JL, Ilyushina NA, Webby RJ, Weaver EA. Influenza H1 Mosaic Hemagglutinin Vaccine Induces Broad Immunity and Protection in Mice. Vaccines (Basel) 2019; 7:vaccines7040195. [PMID: 31771231 PMCID: PMC6963302 DOI: 10.3390/vaccines7040195] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 11/19/2019] [Accepted: 11/20/2019] [Indexed: 12/15/2022] Open
Abstract
Annually, influenza A virus (IAV) infects ~5-10% of adults and 20-30% of children worldwide. The primary resource to protect against infection is by vaccination. However, vaccination only induces strain-specific and transient immunity. Vaccine strategies that induce cross-protective immunity against the broad diversity of IAV are needed. Here we developed and tested a novel mosaic H1 HA immunogen. The mosaic immunogen was optimized in silico to include the most potential B and T cell epitopes (PBTE) across a diverse population of human H1 IAV. Phylogenetic analysis showed that the mosaic HA localizes towards the non-pandemic 2009 strains which encompasses the broadest diversity in the H1 IAV population. We compared the mosaic H1 immunogen to wild-type HA immunogens and the commercial inactivated influenza vaccine, Fluzone. When analyzed by ELISA, the mosaic immunogen induced stronger antibody responses against all four diverse H1 HA proteins. When analyzing T cell responses, again the mosaic immunogen induced stronger cellular immunity against all 4 diverse HA strains. Not only was the magnitude of T cell responses strongest in mosaic immunized mice, the number of epitopes recognized was also greater. The mosaic vaccinated mice showed strong cross-protection against challenges with three divergent IAV strains. These data show that the mosaic immunogen induces strong cross-protective immunity and should be investigated further as a universal influenza vaccine.
Collapse
Affiliation(s)
- Brigette N. Corder
- School of Biological Sciences, Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, NE 68503, USA; (B.N.C.); (B.L.B.)
| | - Brianna L. Bullard
- School of Biological Sciences, Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, NE 68503, USA; (B.N.C.); (B.L.B.)
| | - Jennifer L. DeBeauchamp
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (J.L.D.); (R.J.W.)
| | - Natalia A. Ilyushina
- Division of Biotechnology Review and Research II, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD 20993, USA;
| | - Richard J. Webby
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (J.L.D.); (R.J.W.)
| | - Eric A. Weaver
- School of Biological Sciences, Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, NE 68503, USA; (B.N.C.); (B.L.B.)
- Correspondence:
| |
Collapse
|
20
|
Knowlden ZAG, Richards KA, Moritzky SA, Sant AJ. Peptide Epitope Hot Spots of CD4 T Cell Recognition Within Influenza Hemagglutinin During the Primary Response to Infection. Pathogens 2019; 8:pathogens8040220. [PMID: 31694141 PMCID: PMC6963931 DOI: 10.3390/pathogens8040220] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 10/22/2019] [Accepted: 10/29/2019] [Indexed: 01/15/2023] Open
Abstract
Antibodies specific for the hemagglutinin (HA) protein of influenza virus are critical for protective immunity to infection. Our studies show that CD4 T cells specific for epitopes derived from HA are the most effective in providing help for the HA-specific B cell responses to infection and vaccination. In this study, we asked whether HA epitopes recognized by CD4 T cells in the primary response to infection are equally distributed across the HA protein or if certain segments are enriched in CD4 T cell epitopes. Mice that collectively expressed eight alternative MHC (Major Histocompatibility Complex) class II molecules, that would each have different peptide binding specificities, were infected with an H1N1 influenza virus. CD4 T cell peptide epitope specificities were identified by cytokine EliSpots. These studies revealed that the HA-specific CD4 T cell epitopes cluster in two distinct regions of HA and that some segments of HA are completely devoid of CD4 T cell epitopes. When located on the HA structure, it appears that the regions that most poorly recruit CD4 T cells are sequestered within the interior of the HA trimer, perhaps inaccessible to the proteolytic machinery inside the endosomal compartments of antigen presenting cells.
Collapse
|
21
|
Protein Vaccination Directs the CD4 + T Cell Response toward Shared Protective Epitopes That Can Be Recalled after Influenza Virus Infection. J Virol 2019; 93:JVI.00947-19. [PMID: 31341045 DOI: 10.1128/jvi.00947-19] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 07/16/2019] [Indexed: 12/20/2022] Open
Abstract
Vaccination is widely used to generate protective immunity against influenza virus. CD4+ T cells contribute in diverse ways to protective immunity, most notably, in the provision of help for the production of neutralizing antibodies. Several recent reports have suggested that influenza virus infection elicits CD4+ T cells whose specificity only partially overlaps that of T cells elicited by vaccination. This finding has raised serious concerns regarding the utility of currently licensed inactivated influenza virus vaccines and novel protein-based vaccines. Here, using controlled animal models that allowed a broad sampling of the CD4+ T cell repertoire, we evaluated protein vaccine- versus infection-generated CD4+ T cell epitopes. Our studies revealed that all the infection-elicited CD4+ T cell epitope specificities are also elicited by protein vaccination, although the immunodominance hierarchies can differ. Finally, using a reverse-engineered influenza virus and a heterologous protein vaccination and infection challenge strategy, we show that protein vaccine-elicited CD4+ memory T cells are recalled and boosted after infection and provide early help to accelerate hemagglutinin (HA)-specific antibody responses. The early CD4+ T cell response and HA-specific antibody production are associated with lowered viral titers during the infection challenge. Our data lend confidence to the ability of current protein-based vaccines to elicit influenza virus-specific CD4+ T cells that can potentiate protective immunity upon influenza virus infection.IMPORTANCE Most current and new influenza vaccine candidates consist of a single influenza virus protein or combinations of influenza virus proteins. For these vaccines to elicit CD4+ T cells that can be recalled after infection, the peptide epitopes should be shared between the two modes of confrontation. Recently, questions regarding the relatedness of epitope selection by influenza virus infection and protein vaccination have been raised. However, the studies reported here show that the specificity of CD4+ T cells elicited by protein-based vaccines overlaps that of T cells elicited by infection and that CD4+ T cells primed by protein vaccines are recalled and contribute to protection of the host from a future infection.
Collapse
|
22
|
Jansen JM, Gerlach T, Elbahesh H, Rimmelzwaan GF, Saletti G. Influenza virus-specific CD4+ and CD8+ T cell-mediated immunity induced by infection and vaccination. J Clin Virol 2019; 119:44-52. [DOI: 10.1016/j.jcv.2019.08.009] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 08/20/2019] [Accepted: 08/21/2019] [Indexed: 01/13/2023]
|
23
|
Islam S, Zhou F, Lartey S, Mohn KGI, Krammer F, Cox RJ, Brokstad KA. Functional immune response to influenza H1N1 in children and adults after live attenuated influenza virus vaccination. Scand J Immunol 2019; 90:e12801. [PMID: 31269273 DOI: 10.1111/sji.12801] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 06/20/2019] [Accepted: 06/28/2019] [Indexed: 02/02/2023]
Abstract
Influenza virus is a major respiratory pathogen, and vaccination is the main method of prophylaxis. In 2012, the trivalent live attenuated influenza vaccine (LAIV) was licensed in Europe for use in children. Vaccine-induced antibodies directed against the main viral surface glycoproteins, haemagglutinin (HA) and neuraminidase (NA) play important roles in limiting virus infection. The objective of this study was to dissect the influenza-specific antibody responses in children and adults, and T cell responses in children induced after LAIV immunization to the A/H1N1 virus. Blood samples were collected pre- and at 28 and 56 days post-vaccination from 20 children and 20 adults. No increase in micro-neutralization (MN) antibodies against A/H1N1 was observed after vaccination. A/H1N1 stalk-specific neutralizing and NA-inhibiting (NI) antibodies were boosted in children after LAIV. Interferon γ-producing T cells increased significantly in children, and antibody-dependent cellular-mediated cytotoxic (ADCC) cell activity increased slightly in children after vaccination, although this change was not significant. The results indicate that the NI assay is more sensitive to qualitative changes in serum antibodies after LAIV. There was a considerable difference in the immune response in children and adults after vaccination, which may be related to priming and previous influenza history. Our findings warrant further studies for evaluating LAIV vaccination immunogenicity.
Collapse
Affiliation(s)
- Shahinul Islam
- Department of Clinical Science, Influenza Centre, University of Bergen, Bergen, Norway.,Department of Clinical Science, K.G. Jebsen Centre for Influenza Vaccine Research, University of Bergen, Bergen, Norway
| | - Fan Zhou
- Department of Clinical Science, Influenza Centre, University of Bergen, Bergen, Norway.,Department of Clinical Science, K.G. Jebsen Centre for Influenza Vaccine Research, University of Bergen, Bergen, Norway
| | - Sarah Lartey
- Department of Clinical Science, Influenza Centre, University of Bergen, Bergen, Norway.,Department of Clinical Science, K.G. Jebsen Centre for Influenza Vaccine Research, University of Bergen, Bergen, Norway
| | - Kristin G I Mohn
- Department of Clinical Science, Influenza Centre, University of Bergen, Bergen, Norway.,Emergency Care Clinic, Haukeland University Hospital, Bergen, Norway
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Rebecca Jane Cox
- Department of Clinical Science, Influenza Centre, University of Bergen, Bergen, Norway.,Department of Clinical Science, K.G. Jebsen Centre for Influenza Vaccine Research, University of Bergen, Bergen, Norway.,Department of Research & Development, Haukeland University Hospital, Bergen, Norway
| | - Karl Albert Brokstad
- Department of Clinical Science, Broegelmann Research Laboratory, University of Bergen, Bergen, Norway
| |
Collapse
|
24
|
Eickhoff CS, Terry FE, Peng L, Meza KA, Sakala IG, Van Aartsen D, Moise L, Martin WD, Schriewer J, Buller RM, De Groot AS, Hoft DF. Highly conserved influenza T cell epitopes induce broadly protective immunity. Vaccine 2019; 37:5371-5381. [PMID: 31331771 DOI: 10.1016/j.vaccine.2019.07.033] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 06/26/2019] [Accepted: 07/08/2019] [Indexed: 12/30/2022]
Abstract
Influenza world-wide causes significant morbidity and mortality annually, and more severe pandemics when novel strains evolve to which humans are immunologically naïve. Because of the high viral mutation rate, new vaccines must be generated based on the prevalence of circulating strains every year. New approaches to induce more broadly protective immunity are urgently needed. Previous research has demonstrated that influenza-specific T cells can provide broadly heterotypic protective immunity in both mice and humans, supporting the rationale for developing a T cell-targeted universal influenza vaccine. We used state-of-the art immunoinformatic tools to identify putative pan-HLA-DR and HLA-A2 supertype-restricted T cell epitopes highly conserved among > 50 widely diverse influenza A strains (representing hemagglutinin types 1, 2, 3, 5, 7 and 9). We found influenza peptides that are highly conserved across influenza subtypes that were also predicted to be class I epitopes restricted by HLA-A2. These peptides were found to be immunoreactive in HLA-A2 positive but not HLA-A2 negative individuals. Class II-restricted T cell epitopes that were highly conserved across influenza subtypes were identified. Human CD4+ T cells were reactive with these conserved CD4 epitopes, and epitope expanded T cells were responsive to both H1N1 and H3N2 viruses. Dendritic cell vaccines pulsed with conserved epitopes and DNA vaccines encoding these epitopes were developed and tested in HLA transgenic mice. These vaccines were highly immunogenic, and more importantly, vaccine-induced immunity was protective against both H1N1 and H3N2 influenza challenges. These results demonstrate proof-of-principle that conserved T cell epitopes expressed by widely diverse influenza strains can induce broadly protective, heterotypic influenza immunity, providing strong support for further development of universally relevant multi-epitope T cell-targeting influenza vaccines.
Collapse
Affiliation(s)
- Christopher S Eickhoff
- Saint Louis University, Division of Infectious Diseases, Allergy, and Immunology, Department of Internal Medicine, 1100 S. Grand Blvd., Edward A. Doisy Research Center - 8th Floor, Saint Louis, MO 63104, United States
| | - Frances E Terry
- EpiVax, Inc., 188 Valley Street, Suite 424, Providence, RI 02909, United States
| | - Linda Peng
- Saint Louis University, Division of Infectious Diseases, Allergy, and Immunology, Department of Internal Medicine, 1100 S. Grand Blvd., Edward A. Doisy Research Center - 8th Floor, Saint Louis, MO 63104, United States
| | - Krystal A Meza
- Saint Louis University, Division of Infectious Diseases, Allergy, and Immunology, Department of Internal Medicine, 1100 S. Grand Blvd., Edward A. Doisy Research Center - 8th Floor, Saint Louis, MO 63104, United States
| | - Isaac G Sakala
- Saint Louis University, Division of Infectious Diseases, Allergy, and Immunology, Department of Internal Medicine, 1100 S. Grand Blvd., Edward A. Doisy Research Center - 8th Floor, Saint Louis, MO 63104, United States
| | - Daniel Van Aartsen
- Saint Louis University, Division of Infectious Diseases, Allergy, and Immunology, Department of Internal Medicine, 1100 S. Grand Blvd., Edward A. Doisy Research Center - 8th Floor, Saint Louis, MO 63104, United States
| | - Leonard Moise
- EpiVax, Inc., 188 Valley Street, Suite 424, Providence, RI 02909, United States; University of Rhode Island, Institute for Immunology and Informatics, Department of Cell and Molecular Biology, 80 Washington Street, Providence, RI 02903, United States
| | - William D Martin
- EpiVax, Inc., 188 Valley Street, Suite 424, Providence, RI 02909, United States
| | - Jill Schriewer
- Saint Louis University, Department of Molecular Microbiology & Immunology, 1100 S. Grand Blvd., Edward A. Doisy Research Center - 8th Floor, Saint Louis, MO 63104, United States
| | - R Mark Buller
- Saint Louis University, Department of Molecular Microbiology & Immunology, 1100 S. Grand Blvd., Edward A. Doisy Research Center - 8th Floor, Saint Louis, MO 63104, United States
| | - Anne S De Groot
- EpiVax, Inc., 188 Valley Street, Suite 424, Providence, RI 02909, United States; University of Rhode Island, Institute for Immunology and Informatics, Department of Cell and Molecular Biology, 80 Washington Street, Providence, RI 02903, United States
| | - Daniel F Hoft
- Saint Louis University, Division of Infectious Diseases, Allergy, and Immunology, Department of Internal Medicine, 1100 S. Grand Blvd., Edward A. Doisy Research Center - 8th Floor, Saint Louis, MO 63104, United States; Saint Louis University, Department of Molecular Microbiology & Immunology, 1100 S. Grand Blvd., Edward A. Doisy Research Center - 8th Floor, Saint Louis, MO 63104, United States.
| |
Collapse
|
25
|
Nelson SA, Sant AJ. Imprinting and Editing of the Human CD4 T Cell Response to Influenza Virus. Front Immunol 2019; 10:932. [PMID: 31134060 PMCID: PMC6514101 DOI: 10.3389/fimmu.2019.00932] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 04/11/2019] [Indexed: 12/22/2022] Open
Abstract
Immunity to influenza is unique among pathogens, in that immune memory is established both via intermittent lung localized infections with highly variable influenza virus strains and by intramuscular vaccinations with inactivated protein-based vaccines. Studies in the past decades have suggested that the B cell responses to influenza infection and vaccination are highly biased by an individual's early history of influenza infection. This reactivity likely reflects both the competitive advantage that memory B cells have in an immune response and the relatively limited diversity of epitopes in influenza hemagglutinin that are recognized by B cells. In contrast, CD4 T cells recognize a wide array of epitopes, with specificities that are heavily influenced by the diversity of influenza antigens available, and a multiplicity of functions that are determined by both priming events and subsequent confrontations with antigens. Here, we consider the events that prime and remodel the influenza-specific CD4 T cell response in humans that have highly diverse immune histories and how the CD4 repertoire may be edited in terms of functional potential and viral epitope specificity. We discuss the consequences that imprinting and remodeling may have on the potential of different human hosts to rapidly respond with protective cellular immunity to infection. Finally, these issues are discussed in the context of future avenues of investigation and vaccine strategies.
Collapse
Affiliation(s)
| | - Andrea J. Sant
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester Medical Center, Rochester, NY, United States
| |
Collapse
|
26
|
Sant AJ, DiPiazza AT, Nayak JL, Rattan A, Richards KA. CD4 T cells in protection from influenza virus: Viral antigen specificity and functional potential. Immunol Rev 2019; 284:91-105. [PMID: 29944766 DOI: 10.1111/imr.12662] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
CD4 T cells convey a number of discrete functions to protective immunity to influenza, a complexity that distinguishes this arm of adaptive immunity from B cells and CD8 T cells. Although the most well recognized function of CD4 T cells is provision of help for antibody production, CD4 T cells are important in many aspects of protective immunity. Our studies have revealed that viral antigen specificity is a key determinant of CD4 T cell function, as illustrated both by mouse models of infection and human vaccine responses, a factor whose importance is due at least in part to events in viral antigen handling. We discuss research that has provided insight into the diverse viral epitope specificity of CD4 T cells elicited after infection, how this primary response is modified as CD4 T cells home to the lung, establish memory, and after challenge with a secondary and distinct influenza virus strain. Our studies in human subjects point out the challenges facing vaccine efforts to facilitate responses to novel and avian strains of influenza, as well as strategies that enhance the ability of CD4 T cells to promote protective antibody responses to both seasonal and potentially pandemic strains of influenza.
Collapse
Affiliation(s)
- Andrea J Sant
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, USA
| | - Anthony T DiPiazza
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, USA
| | - Jennifer L Nayak
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, USA.,Division of Infectious Diseases, Department of Pediatrics, University of Rochester Medical Center, Rochester, NY, USA
| | - Ajitanuj Rattan
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, USA
| | - Katherine A Richards
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
27
|
Bodin J, Mihret A, Holm-Hansen C, Dembinski JL, Trieu MC, Tessema B, Tarekegne A, Yimer SA, Cox R, Aseffa A, Haneberg B, Mjaaland S. Vitamin D Deficiency is Associated with Increased Use of Antimicrobials among Preschool Girls in Ethiopia. Nutrients 2019; 11:nu11030575. [PMID: 30866564 PMCID: PMC6471093 DOI: 10.3390/nu11030575] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 02/25/2019] [Accepted: 03/01/2019] [Indexed: 12/12/2022] Open
Abstract
Preschool children in Addis Ababa, Ethiopia, are highly exposed to influenza viruses. Factors related to infections, nutrition, and environmental conditions that might explain the burden of influenza among these children were investigated. Ninety-five preschool children, 48 girls and 47 boys, were followed clinically for 12 months. Illness and immune responses to influenza; three other respiratory viruses; five airway pathogenic bacteria; and levels of vitamins D, A, and B12 were assessed. Most of the children had antibodies to numerous respiratory viral and bacterial agents at study start, and many were infected during follow-up. Twenty-five girls and 25 boys fell ill during the study, and were treated with one or more courses of systemic antimicrobials. Ninety percent of both girls and boys had 25-hydroxyvitamin D [25(OH)D] levels below the recommended levels. While there was no overall difference in the levels of vitamins D, A, and B12 between girls and boys, treated girls had significantly lower 25(OH)D levels than non-treated girls and treated boys. There was a considerable number of short for age children, but only the short treated girls had significantly lower 25(OH)D levels than the non-treated children. Preschool girls with low 25(OH)D levels were more vulnerable to pathogenic microbes than boys.
Collapse
Affiliation(s)
- Johanna Bodin
- Department of Infectious Disease Immunology, Norwegian Institute of Public Health, 0456 Oslo, Norway.
- K.G. Jebsen Centre for Influenza Vaccine Research, University of Oslo, 0316 Oslo, Norway.
| | - Adane Mihret
- Armauer Hansen Research Institute, 1005 Addis Ababa, Ethiopia.
| | - Carol Holm-Hansen
- Department of Infectious Disease Immunology, Norwegian Institute of Public Health, 0456 Oslo, Norway.
| | - Jennifer L Dembinski
- Department of Infectious Disease Immunology, Norwegian Institute of Public Health, 0456 Oslo, Norway.
- Department of Virology, Norwegian Institute of Public Health, 0456 Oslo, Norway.
| | - Mai-Chi Trieu
- K.G. Jebsen Centre for Influenza Vaccine Research, University of Oslo, 0316 Oslo, Norway.
- The Influenza Centre and Department of Clinical Science, University of Bergen, 5020 Bergen, Norway.
| | - Bamlak Tessema
- Armauer Hansen Research Institute, 1005 Addis Ababa, Ethiopia.
| | - Azeb Tarekegne
- Armauer Hansen Research Institute, 1005 Addis Ababa, Ethiopia.
| | - Solomon A Yimer
- Coalition for Epidemic Preparedness Innovations (CEPI), 0306 Oslo, Norway.
- Department of Microbiology, University of Oslo, 0316 Oslo, Norway.
| | - Rebecca Cox
- K.G. Jebsen Centre for Influenza Vaccine Research, University of Oslo, 0316 Oslo, Norway.
- The Influenza Centre and Department of Clinical Science, University of Bergen, 5020 Bergen, Norway.
| | - Abraham Aseffa
- Armauer Hansen Research Institute, 1005 Addis Ababa, Ethiopia.
| | - Bjørn Haneberg
- Department of Infectious Disease Immunology, Norwegian Institute of Public Health, 0456 Oslo, Norway.
| | - Siri Mjaaland
- Department of Infectious Disease Immunology, Norwegian Institute of Public Health, 0456 Oslo, Norway.
- K.G. Jebsen Centre for Influenza Vaccine Research, University of Oslo, 0316 Oslo, Norway.
| |
Collapse
|
28
|
Trieu MC, Zhou F, Lartey SL, Sridhar S, Mjaaland S, Cox RJ. Augmented CD4 + T-cell and humoral responses after repeated annual influenza vaccination with the same vaccine component A/H1N1pdm09 over 5 years. NPJ Vaccines 2018; 3:37. [PMID: 30131880 PMCID: PMC6092382 DOI: 10.1038/s41541-018-0069-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Revised: 05/10/2018] [Accepted: 05/23/2018] [Indexed: 12/11/2022] Open
Abstract
Annual seasonal influenza vaccination is recommended for high-risk populations and often occupational groups such as healthcare workers (HCWs). Repeated annual vaccination has been reported to either have no impact or reduce antibody responses or protection. However, whether repeated vaccination influences T-cell responses has not been sufficiently studied, despite the increasing evidence of the protective roles of T-cell immunity. Here, we explored the impact of repeated annual vaccination with the same vaccine strain (H1N1pdm09) over multiple seasons in the post-2009 pandemic era and showed that repeated vaccination increased both T-cell and humoral responses. Using the T-cell FluroSpot and intracellular cytokine-staining, the hemagglutination inhibition (HI), and the memory B-cell (MBC) ELISpot assays, we investigated pre- and postvaccination T cells, antibodies, and MBCs in a cohort of HCWs repeatedly vaccinated with H1N1pdm09 for 5 years (pandemic vaccination in 2009 and subsequently annual seasonal vaccination containing H1N1pdm09 during 2010-2013). We found that the prevaccination H1N1pdm09-specific T cells, antibodies, and MBCs were significantly increased after 3-4 repeated vaccinations and maintained at high levels throughout seasons 2012 and 2013. The cross-reactive IFN-γ-secreting CD4+ cells recognizing conserved viral external or internal epitopes were also maintained throughout 2012 and 2013. Repeated vaccination improved the multifunctional memory CD4+ responses. Particularly, the IFN-γ+TNF-α+CD4+ T cells were boosted following each vaccination. HI antibodies were significantly induced after each vaccination over 5 years. Our findings indicate a broad impact of repeated annual vaccination, even with the same vaccine component, on the influenza-specific T-cell and humoral immunity and support the continuing recommendation of annual influenza vaccination. Despite the widespread implementation of annual influenza vaccination, the effect of repeated vaccinations on the immune response in subsequent seasons is poorly understood. A team led by Rebecca Jane Cox at the University of Bergen examined the humoral and T-cell response elicited by 2009's H1N1pdm09 seasonal vaccine. Since the H1N1pdm09 strain continued to circulate in subsequent years it was included in later vaccine formulations and the authors could therefore examine the effects of repeated annual vaccination over multiple seasons. They observed that H1N1pdm09-specific polyfunctional T-cell responses and antibodies were enhanced by multiple annual vaccinations. In particular, T cells showed progressive skewing to IFN-γ+TNF+ double producers, but the magnitude of the T-cell response tended to plateau after 3-4 repeated vaccinations. The findings suggest that including the same component in subsequent annual vaccines can significantly impact the influenza immune response.
Collapse
Affiliation(s)
- Mai-Chi Trieu
- 1The Influenza Centre, Department of Clinical Science, University of Bergen, Bergen, Norway.,2K.G. Jebsen Centre for Influenza Vaccine Research, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Fan Zhou
- 1The Influenza Centre, Department of Clinical Science, University of Bergen, Bergen, Norway.,2K.G. Jebsen Centre for Influenza Vaccine Research, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Sarah Larteley Lartey
- 1The Influenza Centre, Department of Clinical Science, University of Bergen, Bergen, Norway.,2K.G. Jebsen Centre for Influenza Vaccine Research, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Saranya Sridhar
- 3Jenner Institute, University of Oxford, Oxford, UK.,6Present Address: Sanofi Pasteur, 1541 Avenue Marcel Mérieux, 69280 Marcy l'Etoile, France
| | - Siri Mjaaland
- 2K.G. Jebsen Centre for Influenza Vaccine Research, Department of Clinical Science, University of Bergen, Bergen, Norway.,4Department of Infectious Disease Immunology, Norwegian Institute of Public Health, Oslo, Norway
| | - Rebecca Jane Cox
- 1The Influenza Centre, Department of Clinical Science, University of Bergen, Bergen, Norway.,2K.G. Jebsen Centre for Influenza Vaccine Research, Department of Clinical Science, University of Bergen, Bergen, Norway.,5Department of Research and Development, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
29
|
Sant AJ, Richards KA, Nayak J. Distinct and complementary roles of CD4 T cells in protective immunity to influenza virus. Curr Opin Immunol 2018; 53:13-21. [PMID: 29621639 PMCID: PMC6141328 DOI: 10.1016/j.coi.2018.03.019] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 03/17/2018] [Accepted: 03/19/2018] [Indexed: 02/01/2023]
Abstract
CD4 T cells play a multiplicity of roles in protective immunity to influenza. Included in these functions are help for high affinity antibody production, enhancement of CD8 T cell expansion, function and memory, acceleration of the early innate response to infection and direct cytotoxicity. The influenza-specific CD4 T cell repertoire in humans established through exposures to infection and vaccination has been found to be highly variable in abundance, specificity and functionality. Deficits in particular subsets of CD4 T cells recruited into the response result in diminished antibody responses and protection from infection. Therefore, improved strategies for vaccination should include better methods to identify deficiencies in the circulating CD4 T cell repertoire, and vaccine constructs that increase the representation of CD4 T cells of the correct specificity and functionality.
Collapse
Affiliation(s)
- Andrea J Sant
- David H. Smith Center for Vaccine Biology and Immunology, University of Rochester Medical Center, USA; Department of Microbiology and Immunology, University of Rochester Medical Center, USA.
| | - Katherine A Richards
- David H. Smith Center for Vaccine Biology and Immunology, University of Rochester Medical Center, USA
| | - Jennifer Nayak
- David H. Smith Center for Vaccine Biology and Immunology, University of Rochester Medical Center, USA; Department of Microbiology and Immunology, University of Rochester Medical Center, USA; Department of Pediatrics, Division of Infectious Diseases, University of Rochester Medical Center, USA
| |
Collapse
|
30
|
Schmidt ME, Varga SM. The CD8 T Cell Response to Respiratory Virus Infections. Front Immunol 2018; 9:678. [PMID: 29686673 PMCID: PMC5900024 DOI: 10.3389/fimmu.2018.00678] [Citation(s) in RCA: 255] [Impact Index Per Article: 36.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 03/20/2018] [Indexed: 12/19/2022] Open
Abstract
Humans are highly susceptible to infection with respiratory viruses including respiratory syncytial virus (RSV), influenza virus, human metapneumovirus, rhinovirus, coronavirus, and parainfluenza virus. While some viruses simply cause symptoms of the common cold, many respiratory viruses induce severe bronchiolitis, pneumonia, and even death following infection. Despite the immense clinical burden, the majority of the most common pulmonary viruses lack long-lasting efficacious vaccines. Nearly all current vaccination strategies are designed to elicit broadly neutralizing antibodies, which prevent severe disease following a subsequent infection. However, the mucosal antibody response to many respiratory viruses is not long-lasting and declines with age. CD8 T cells are critical for mediating clearance following many acute viral infections in the lung. In addition, memory CD8 T cells are capable of providing protection against secondary infections. Therefore, the combined induction of virus-specific CD8 T cells and antibodies may provide optimal protective immunity. Herein, we review the current literature on CD8 T cell responses induced by respiratory virus infections. Additionally, we explore how this knowledge could be utilized in the development of future vaccines against respiratory viruses, with a special emphasis on RSV vaccination.
Collapse
Affiliation(s)
- Megan E Schmidt
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA, United States
| | - Steven M Varga
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA, United States.,Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, United States.,Department of Pathology, University of Iowa, Iowa City, IA, United States
| |
Collapse
|
31
|
Rose NR. Negative selection, epitope mimicry and autoimmunity. Curr Opin Immunol 2017; 49:51-55. [DOI: 10.1016/j.coi.2017.08.014] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 08/26/2017] [Indexed: 12/17/2022]
|
32
|
Savic M, Dembinski JL, Laake I, Hungnes O, Cox R, Oftung F, Trogstad L, Mjaaland S. Distinct T and NK cell populations may serve as immune correlates of protection against symptomatic pandemic influenza A(H1N1) virus infection during pregnancy. PLoS One 2017; 12:e0188055. [PMID: 29145441 PMCID: PMC5690673 DOI: 10.1371/journal.pone.0188055] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 10/31/2017] [Indexed: 11/24/2022] Open
Abstract
Maternal influenza infection during pregnancy is associated with increased risk of morbidity and mortality. However, the link between the anti-influenza immune responses and health-related risks during infection is not well understood. We have analyzed memory T and NK cell mediated immunity (CMI) responses in pandemic influenza A(H1N1)pdm09 (pdm09) virus infected non-vaccinated pregnant women participating in the Norwegian Influenza Pregnancy Cohort (NorFlu). The cohort includes information on immunization, self-reported health and disease status, and biological samples (plasma and PBMC). Infected cases (N = 75) were defined by having a serum hemagglutination inhibition (HI) titer > = 20 to influenza pdm09 virus at the time of delivery, while controls (N = 75) were randomly selected among non-infected pregnant women (HI titer <10). In ELISpot assays cases had higher frequencies of IFNγ+ CD8+ T cells responding to pdm09 virus or conserved CD8 T cell-restricted influenza A virus epitopes, compared to controls. Within this T cell population, frequencies of CD95+ late effector (CD45RA+CCR7-) and naive (CD45RA+CCR7+) CD8+ memory T cells correlated inversely with self-reported influenza illness (ILI) symptoms. ILI symptoms in infected women were also associated with lower numbers of poly-functional (IFNγ+TNFα+, IL2+IFNγ+, IL2+IFNγ+TNFα+) CD4+ T cells and increased frequencies of IFNγ+CD3-CD7+ NK cells compared to asymptomatic cases, or controls, after stimulation with the pdm09 virus. Taken together, virus specific and functionally distinct T and NK cell populations may serve as cellular immune correlates of clinical outcomes of pandemic influenza disease in pregnant women. Our results may provide information important for future universal influenza vaccine design.
Collapse
Affiliation(s)
- Miloje Savic
- Infection Control and Environmental Health, Norwegian Institute of Public Health, Oslo, Norway
- KG Jebsen Centre for Influenza Vaccine Research, Oslo-Bergen, Norway
- * E-mail: ;
| | - Jennifer L. Dembinski
- Infection Control and Environmental Health, Norwegian Institute of Public Health, Oslo, Norway
- KG Jebsen Centre for Influenza Vaccine Research, Oslo-Bergen, Norway
| | - Ida Laake
- Infection Control and Environmental Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Olav Hungnes
- Infection Control and Environmental Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Rebecca Cox
- KG Jebsen Centre for Influenza Vaccine Research, Oslo-Bergen, Norway
- Influenza Centre, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Research and Development, Haukeland University Hospital, Bergen, Norway
| | - Fredrik Oftung
- Infection Control and Environmental Health, Norwegian Institute of Public Health, Oslo, Norway
- KG Jebsen Centre for Influenza Vaccine Research, Oslo-Bergen, Norway
| | - Lill Trogstad
- Infection Control and Environmental Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Siri Mjaaland
- Infection Control and Environmental Health, Norwegian Institute of Public Health, Oslo, Norway
- KG Jebsen Centre for Influenza Vaccine Research, Oslo-Bergen, Norway
- * E-mail: ;
| |
Collapse
|
33
|
Skevaki C, Hudemann C, Matrosovich M, Möbs C, Paul S, Wachtendorf A, Alashkar Alhamwe B, Potaczek DP, Hagner S, Gemsa D, Garn H, Sette A, Renz H. Influenza-derived peptides cross-react with allergens and provide asthma protection. J Allergy Clin Immunol 2017; 142:804-814. [PMID: 29132960 DOI: 10.1016/j.jaci.2017.07.056] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 07/04/2017] [Accepted: 07/19/2017] [Indexed: 10/18/2022]
Abstract
BACKGROUND The hygiene hypothesis is the leading concept to explain the current asthma epidemic, which is built on the observation that a lack of bacterial contact early in life induces allergic TH2 immune responses. OBJECTIVE Because little is known about the contribution of respiratory tract viruses in this context, we evaluated the effect of prior influenza infection on the development of allergic asthma. METHODS Mice were infected with influenza and, once recovered, subjected to an ovalbumin- or house dust mite-induced experimental asthma protocol. Influenza-polarized effector memory T (Tem) cells were transferred adoptively to allergen-sensitized animals before allergen challenge. A comprehensive in silico analysis assessed homologies between virus- and allergen-derived proteins. Influenza-polarized Tem cells were stimulated ex vivo with candidate peptides. Mice were immunized with a pool of virus-derived T-cell epitopes. RESULTS In 2 murine models we found a long-lasting preventive effect against experimental asthma features. Protection could be attributed about equally to CD4+ and CD8+ Tem cells from influenza-infected mice. An in silico bioinformatic analysis identified 4 influenza- and 3 allergen-derived MHC class I and MHC class II candidate T-cell epitopes with potential antigen-specific cross-reactivity between influenza and allergens. Lymphocytes from influenza-infected mice produced IFN-γ and IL-2 but not IL-5 on stimulation with the aforementioned peptides. Immunization with a mixture of the influenza peptides conferred asthma protection, and peptide-immunized mice transferred protection through CD4+ and CD8+ Tem cells. CONCLUSION For the first time, our results illustrate heterologous immunity of virus-infected animals toward allergens. This finding extends the original hygiene hypothesis.
Collapse
Affiliation(s)
- Chrysanthi Skevaki
- Institute of Laboratory Medicine and Pathobiochemistry, Member of the German Center for Lung Research (DZL), Philipps University Marburg, Marburg, Germany
| | - Christoph Hudemann
- Institute of Laboratory Medicine and Pathobiochemistry, Member of the German Center for Lung Research (DZL), Philipps University Marburg, Marburg, Germany
| | | | - Christian Möbs
- Department of Dermatology and Allergology, Philipps University Marburg, Marburg, Germany
| | - Sinu Paul
- La Jolla Institute for Allergy and Immunology, La Jolla, Calif
| | - Andreas Wachtendorf
- Institute of Laboratory Medicine and Pathobiochemistry, Member of the German Center for Lung Research (DZL), Philipps University Marburg, Marburg, Germany
| | - Bilal Alashkar Alhamwe
- Institute of Laboratory Medicine and Pathobiochemistry, Member of the German Center for Lung Research (DZL), Philipps University Marburg, Marburg, Germany
| | - Daniel P Potaczek
- Institute of Laboratory Medicine and Pathobiochemistry, Member of the German Center for Lung Research (DZL), Philipps University Marburg, Marburg, Germany; John Paul II Hospital, Krakow, Poland
| | - Stefanie Hagner
- Institute of Laboratory Medicine and Pathobiochemistry, Member of the German Center for Lung Research (DZL), Philipps University Marburg, Marburg, Germany
| | - Diethard Gemsa
- Institute of Laboratory Medicine and Pathobiochemistry, Member of the German Center for Lung Research (DZL), Philipps University Marburg, Marburg, Germany
| | - Holger Garn
- Institute of Laboratory Medicine and Pathobiochemistry, Member of the German Center for Lung Research (DZL), Philipps University Marburg, Marburg, Germany
| | | | - Harald Renz
- Institute of Laboratory Medicine and Pathobiochemistry, Member of the German Center for Lung Research (DZL), Philipps University Marburg, Marburg, Germany.
| |
Collapse
|
34
|
Muraro E, Merlo A, Martorelli D, Cangemi M, Dalla Santa S, Dolcetti R, Rosato A. Fighting Viral Infections and Virus-Driven Tumors with Cytotoxic CD4 + T Cells. Front Immunol 2017; 8:197. [PMID: 28289418 PMCID: PMC5327441 DOI: 10.3389/fimmu.2017.00197] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 02/09/2017] [Indexed: 12/18/2022] Open
Abstract
CD4+ T cells have been and are still largely regarded as the orchestrators of immune responses, being able to differentiate into distinct T helper cell populations based on differentiation signals, transcription factor expression, cytokine secretion, and specific functions. Nonetheless, a growing body of evidence indicates that CD4+ T cells can also exert a direct effector activity, which depends on intrinsic cytotoxic properties acquired and carried out along with the evolution of several pathogenic infections. The relevant role of CD4+ T cell lytic features in the control of such infectious conditions also leads to their exploitation as a new immunotherapeutic approach. This review aims at summarizing currently available data about functional and therapeutic relevance of cytotoxic CD4+ T cells in the context of viral infections and virus-driven tumors.
Collapse
Affiliation(s)
- Elena Muraro
- Immunopathology and Cancer Biomarkers, Traslational Research Department, IRCCS, C.R.O. National Cancer Institute, Aviano, Pordenone, Italy
| | - Anna Merlo
- Department of Immunology and Blood Transfusions, San Bortolo Hospital, Vicenza, Italy
| | - Debora Martorelli
- Immunopathology and Cancer Biomarkers, Traslational Research Department, IRCCS, C.R.O. National Cancer Institute, Aviano, Pordenone, Italy
| | - Michela Cangemi
- Immunopathology and Cancer Biomarkers, Traslational Research Department, IRCCS, C.R.O. National Cancer Institute, Aviano, Pordenone, Italy
| | | | - Riccardo Dolcetti
- Immunopathology and Cancer Biomarkers, Traslational Research Department, IRCCS, C.R.O. National Cancer Institute, Aviano, Pordenone, Italy
- Translational Research Institute, University of Queensland Diamantina Institute, Brisbane, QLD, Australia
| | - Antonio Rosato
- Istituto Oncologico Veneto IOV-IRCCS, Padova, Italy
- Department of Surgery, Oncology and Gastroenterology, Oncology and Immunology Section, University of Padova, Padova, Italy
| |
Collapse
|
35
|
Dembinski JL, Mihret A, Yimer SA, Tessema B, Trieu MC, Tarekegn A, Getachew N, Cox RJ, Oftung F, Haneberg B, Aseffa A, Mjaaland S. High Prevalence of Humoral and Cellular Immunity to Influenza Viruses in Preschool Children Living in Addis Ababa, Ethiopia. Open Forum Infect Dis 2017; 4:ofx026. [PMID: 28480294 PMCID: PMC5414001 DOI: 10.1093/ofid/ofx026] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 02/06/2017] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND Influenza in children who reside in tropical and subtropical regions has until recently been regarded as insignificant. However, new evidence suggests that it significantly impacts hospitalization and promotes secondary bacterial coinfections. Ethiopia is situated in a subtropical area where influenza viruses are likely to circulate year round. METHODS Clinical data were recorded in a cohort of 103 healthy preschool children recruited in Addis Ababa, Ethiopia. Humoral and cellular immune responses to influenza virus were determined by hemagglutination inhibition (HI) and interferon-γ enzyme-linked immunospot assays. RESULTS Ninety-six percent of the children (2-5 years old) had pre-existing HI antibody responses to 1 or more of the circulating influenza A subtypes, H1N1 (51%), H3N2 (86%), or influenza B (51%) strains. At the age of 4, all children had been infected with at least 1 strain, and 75% had been infected with 2-4 different viral strains. CD4+ and CD8+ T-cell responses against conserved viral antigens increased with repeated exposures, indicating boosting of cross-reactive cellular immunity. Malnutrition did not seem to affect these immune responses to influenza. CONCLUSIONS Influenza is highly prevalent among children in this area of Ethiopia. Due to the risk of secondary bacterial pneumonia, increased influenza awareness might benefit child health.
Collapse
Affiliation(s)
- Jennifer L Dembinski
- Departments of Immunology and.,Vaccine Preventable Diseases, Domain for Infection Control and Environmental Health, Norwegian Institute of Public Health, Oslo, Norway.,KG Jebsen Centre for Influenza Vaccine Research, Department of Immunology, University of Oslo, Norway
| | - Adane Mihret
- Armauer Hansen Research Institute, Addis Ababa, Ethiopia
| | - Solomon A Yimer
- Departments of Immunology and.,Department of Microbiology, Oslo University Hospital, Norway
| | - Bamlak Tessema
- Armauer Hansen Research Institute, Addis Ababa, Ethiopia
| | - Mai-Chi Trieu
- KG Jebsen Centre for Influenza Vaccine Research, Department of Immunology, University of Oslo, Norway.,The Influenza Centre, Department of Clinical Science, University of Bergen, Norway; and
| | - Azeb Tarekegn
- Armauer Hansen Research Institute, Addis Ababa, Ethiopia
| | - Nahom Getachew
- Armauer Hansen Research Institute, Addis Ababa, Ethiopia
| | - Rebecca J Cox
- KG Jebsen Centre for Influenza Vaccine Research, Department of Immunology, University of Oslo, Norway.,The Influenza Centre, Department of Clinical Science, University of Bergen, Norway; and.,Department of Research and Development, Haukeland University Hospital, Bergen, Norway
| | - Fredrik Oftung
- Departments of Immunology and.,KG Jebsen Centre for Influenza Vaccine Research, Department of Immunology, University of Oslo, Norway
| | | | - Abraham Aseffa
- Armauer Hansen Research Institute, Addis Ababa, Ethiopia
| | - Siri Mjaaland
- Departments of Immunology and.,KG Jebsen Centre for Influenza Vaccine Research, Department of Immunology, University of Oslo, Norway
| |
Collapse
|
36
|
Dhakal S, Hiremath J, Bondra K, Lakshmanappa YS, Shyu DL, Ouyang K, Kang KI, Binjawadagi B, Goodman J, Tabynov K, Krakowka S, Narasimhan B, Lee CW, Renukaradhya GJ. Biodegradable nanoparticle delivery of inactivated swine influenza virus vaccine provides heterologous cell-mediated immune response in pigs. J Control Release 2017; 247:194-205. [PMID: 28057521 DOI: 10.1016/j.jconrel.2016.12.039] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 12/12/2016] [Accepted: 12/29/2016] [Indexed: 10/20/2022]
Abstract
Swine influenza virus (SwIV) is one of the important zoonotic pathogens. Current flu vaccines have failed to provide cross-protection against evolving viruses in the field. Poly(lactic-co-glycolic acid) (PLGA) is a biodegradable FDA approved polymer and widely used in drug and vaccine delivery. In this study, inactivated SwIV H1N2 antigens (KAg) encapsulated in PLGA nanoparticles (PLGA-KAg) were prepared, which were spherical in shape with 200 to 300nm diameter, and induced maturation of antigen presenting cells in vitro. Pigs vaccinated twice with PLGA-KAg via intranasal route showed increased antigen specific lymphocyte proliferation and enhanced the frequency of T-helper/memory and cytotoxic T cells (CTLs) in peripheral blood mononuclear cells (PBMCs). In PLGA-KAg vaccinated and heterologous SwIV H1N1 challenged pigs, clinical flu symptoms were absent, while the control pigs had fever for four days. Grossly and microscopically, reduced lung pathology and viral antigenic mass in the lung sections with clearance of infectious challenge virus in most of the PLGA-KAg vaccinated pig lung airways were observed. Immunologically, PLGA-KAg vaccine irrespective of not significantly boosting the mucosal antibody response, it augmented the frequency of IFN-γ secreting total T cells, T-helper and CTLs against both H1N2 and H1N1 SwIV. In summary, inactivated influenza virus delivered through PLGA-NPs reduced the clinical disease and induced cross-protective cell-mediated immune response in a pig model. Our data confirmed the utility of a pig model for intranasal particulate flu vaccine delivery platform to control flu in humans.
Collapse
Affiliation(s)
- Santosh Dhakal
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, 1680 Madison Avenue, Wooster, OH 44691, USA; Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Jagadish Hiremath
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, 1680 Madison Avenue, Wooster, OH 44691, USA; Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Kathryn Bondra
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, 1680 Madison Avenue, Wooster, OH 44691, USA; Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Yashavanth S Lakshmanappa
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, 1680 Madison Avenue, Wooster, OH 44691, USA; Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Duan-Liang Shyu
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, 1680 Madison Avenue, Wooster, OH 44691, USA; Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Kang Ouyang
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, 1680 Madison Avenue, Wooster, OH 44691, USA; Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Kyung-Il Kang
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, 1680 Madison Avenue, Wooster, OH 44691, USA; Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Basavaraj Binjawadagi
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, 1680 Madison Avenue, Wooster, OH 44691, USA; Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Jonathan Goodman
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA 50011, USA
| | - Kairat Tabynov
- The Research Institute for Biological Safety Problems (RIBSP), Zhambylskaya Oblast, Gvardeiskiy 080409, Kazakhstan
| | - Steven Krakowka
- The Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, 1925 Coffey Road, Columbus, OH, USA
| | - Balaji Narasimhan
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA 50011, USA
| | - Chang Won Lee
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, 1680 Madison Avenue, Wooster, OH 44691, USA; Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Gourapura J Renukaradhya
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, 1680 Madison Avenue, Wooster, OH 44691, USA; Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
37
|
DiPiazza A, Richards K, Batarse F, Lockard L, Zeng H, García-Sastre A, Albrecht RA, Sant AJ. Flow Cytometric and Cytokine ELISpot Approaches To Characterize the Cell-Mediated Immune Response in Ferrets following Influenza Virus Infection. J Virol 2016; 90:7991-8004. [PMID: 27356897 PMCID: PMC4988159 DOI: 10.1128/jvi.01001-16] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 06/18/2016] [Indexed: 12/11/2022] Open
Abstract
UNLABELLED Influenza virus infections represent a significant socioeconomic and public health burden worldwide. Although ferrets are considered by many to be ideal for modeling human responses to influenza infection and vaccination, efforts to understand the cellular immune response have been severely hampered by a paucity of standardized procedures and reagents. In this study, we developed flow cytometric and T cell enzyme-linked immunosorbent spot (ELISpot) approaches to characterize the leukocyte composition and antigen-specific T cell response within key lymphoid tissues following influenza virus infection in ferrets. Through a newly designed and implemented set of serological reagents, we used multiparameter flow cytometry to directly quantify the frequency of CD4(+) and CD8(+) T cells, Ig(+) B cells, CD11b(+) myeloid-derived cells, and major histocompatibility complex (MHC) class II-positive antigen-presenting cells (APCs) both prior to and after intranasal infection with A/California/04/09 (H1N1). We found that the leukocyte composition was altered at 10 days postinfection, with notable gains in the frequency of T cells and myeloid cells within the draining lymph node. Furthermore, these studies revealed that the antigen specificity of influenza virus-reactive CD4 and CD8 T cells was very broad, with recognition of the viral HA, NA, M1, NS1, and NP proteins, and that total reactivity to influenza virus postinfection represented approximately 0.1% of the circulating peripheral blood mononuclear cells (PBMC). Finally, we observed distinct patterns of reactivity between individual animals, suggesting heterogeneity at the MHC locus in ferrets within commercial populations, a finding of considerable interest in efforts to move the ferret model forward for influenza vaccine and challenge studies. IMPORTANCE Ferrets are an ideal animal model to study transmission, diseases, and vaccine efficacies of respiratory viruses because of their close anatomical and physiological resemblances to humans. However, a lack of reagents has limited our understanding of the cell-mediated immune response following infection and vaccination. In this study, we used cross-reactive and ferret-specific antibodies to study the leukocyte composition and antigen-specific CD4 and CD8 T cell responses following influenza A/California/04/09 (H1N1) virus infection. These studies revealed strikingly distinct patterns of reactivity between CD4 and CD8 T cells, which were overlaid with differences in protein-specific responses between individual animals. Our results provide a first, in-depth look at the T cell repertoire in response to influenza infection and suggest that there is considerable heterogeneity at the MHC locus, which is akin to that in humans and an area of intense research interest.
Collapse
Affiliation(s)
- Anthony DiPiazza
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester Medical Center, Rochester, New York, USA
| | - Katherine Richards
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester Medical Center, Rochester, New York, USA
| | - Frances Batarse
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester Medical Center, Rochester, New York, USA
| | - Laura Lockard
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester Medical Center, Rochester, New York, USA
| | - Hui Zeng
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mt. Sinai, New York, New York, USA Global Health and Emerging Pathogens Institute at Icahn School of Medicine, New York, New York, USA Department of Medicine, Icahn School of Medicine at Mt. Sinai, New York, New York, USA
| | - Randy A Albrecht
- Department of Microbiology, Icahn School of Medicine at Mt. Sinai, New York, New York, USA Global Health and Emerging Pathogens Institute at Icahn School of Medicine, New York, New York, USA
| | - Andrea J Sant
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester Medical Center, Rochester, New York, USA
| |
Collapse
|
38
|
Mohn KGI, Cox RJ, Tunheim G, Berdal JE, Hauge AG, Jul-Larsen Å, Peters B, Oftung F, Jonassen CM, Mjaaland S. Immune Responses in Acute and Convalescent Patients with Mild, Moderate and Severe Disease during the 2009 Influenza Pandemic in Norway. PLoS One 2015; 10:e0143281. [PMID: 26606759 PMCID: PMC4659565 DOI: 10.1371/journal.pone.0143281] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2015] [Accepted: 11/03/2015] [Indexed: 01/31/2023] Open
Abstract
Increased understanding of immune responses influencing clinical severity during pandemic influenza infection is important for improved treatment and vaccine development. In this study we recruited 46 adult patients during the 2009 influenza pandemic and characterized humoral and cellular immune responses. Those included were either acute hospitalized or convalescent patients with different disease severities (mild, moderate or severe). In general, protective antibody responses increased with enhanced disease severity. In the acute patients, we found higher levels of TNF-α single-producing CD4+T-cells in the severely ill as compared to patients with moderate disease. Stimulation of peripheral blood mononuclear cells (PBMC) from a subset of acute patients with peptide T-cell epitopes showed significantly lower frequencies of influenza specific CD8+ compared with CD4+ IFN-γ T-cells in acute patients. Both T-cell subsets were predominantly directed against the envelope antigens (HA and NA). However, in the convalescent patients we found high levels of both CD4+ and CD8+ T-cells directed against conserved core antigens (NP, PA, PB, and M). The results indicate that the antigen targets recognized by the T-cell subsets may vary according to the phase of infection. The apparent low levels of cross-reactive CD8+ T-cells recognizing internal antigens in acute hospitalized patients suggest an important role for this T-cell subset in protective immunity against influenza.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Antibodies, Neutralizing/immunology
- Antibodies, Viral/immunology
- Cytokines/metabolism
- Epitopes, T-Lymphocyte/immunology
- Female
- Host-Pathogen Interactions/immunology
- Humans
- Immunity
- Immunity, Cellular
- Immunity, Humoral
- Immunoglobulin G/immunology
- Influenza A Virus, H1N1 Subtype/immunology
- Influenza A virus/immunology
- Influenza, Human/diagnosis
- Influenza, Human/epidemiology
- Influenza, Human/immunology
- Male
- Middle Aged
- Neutralization Tests
- Norway/epidemiology
- Pandemics
- Prospective Studies
- Severity of Illness Index
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
- Young Adult
Collapse
Affiliation(s)
- Kristin G.-I. Mohn
- The Influenza Centre, Department of Clinical Science, University of Bergen, Bergen, Norway
- Infectious Diseases Unit, Department of Internal Medicine, Haukeland University Hospital, Bergen, Norway
- K.G. Jebsen Centre for Influenza Vaccine Research, Department of Clinical Science, University of Bergen, Bergen, and The Norwegian Institute of Public Health, Oslo, Norway
- * E-mail: (KGIM); (SM)
| | - Rebecca Jane Cox
- The Influenza Centre, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Research & Development, Haukeland University Hospital, Bergen, Norway
- K.G. Jebsen Centre for Influenza Vaccine Research, Department of Clinical Science, University of Bergen, Bergen, and The Norwegian Institute of Public Health, Oslo, Norway
| | - Gro Tunheim
- Division of Infectious Disease Control, Department of Bacteriology and Immunology, Norwegian Institute of Public Health, Oslo, Norway
- K.G. Jebsen Centre for Influenza Vaccine Research, Department of Clinical Science, University of Bergen, Bergen, and The Norwegian Institute of Public Health, Oslo, Norway
| | - Jan Erik Berdal
- Department of Infectious Diseases, Akershus University Hospital, Nordbyhagen, Norway
| | - Anna Germundsson Hauge
- Section for Virology, Department of Laboratory Services, Norwegian Veterinary Institute, Oslo, Norway
- Division of Infectious Disease Control, Norwegian Institute of Public Health, Oslo, Norway
| | - Åsne Jul-Larsen
- The Influenza Centre, Department of Clinical Science, University of Bergen, Bergen, Norway
| | | | - Bjoern Peters
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, California, United States of America
| | - Fredrik Oftung
- Division of Infectious Disease Control, Department of Bacteriology and Immunology, Norwegian Institute of Public Health, Oslo, Norway
- K.G. Jebsen Centre for Influenza Vaccine Research, Department of Clinical Science, University of Bergen, Bergen, and The Norwegian Institute of Public Health, Oslo, Norway
| | - Christine Monceyron Jonassen
- Genetic Unit, Department of Multidisciplinary Laboratory Medicine and Medical Biochemistry, Akershus University Hospital, Nordbyhagen, Norway
- Genetic Unit, Centre for Laboratory Medicine, Østfold Hospital Trust, Fredrikstad, Norway
- Department of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, Ås, Norway
| | - Siri Mjaaland
- Division of Infectious Disease Control, Department of Bacteriology and Immunology, Norwegian Institute of Public Health, Oslo, Norway
- K.G. Jebsen Centre for Influenza Vaccine Research, Department of Clinical Science, University of Bergen, Bergen, and The Norwegian Institute of Public Health, Oslo, Norway
- * E-mail: (KGIM); (SM)
| |
Collapse
|