51
|
Li J, Helal ZH, Karch CP, Mishra N, Girshick T, Garmendia A, Burkhard P, Khan MI. A self-adjuvanted nanoparticle based vaccine against infectious bronchitis virus. PLoS One 2018; 13:e0203771. [PMID: 30216376 PMCID: PMC6138407 DOI: 10.1371/journal.pone.0203771] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 08/27/2018] [Indexed: 12/21/2022] Open
Abstract
Infectious bronchitis virus (IBV) affects poultry respiratory, renal and reproductive systems. Currently the efficacy of available live attenuated or killed vaccines against IBV has been challenged. We designed a novel IBV vaccine alternative using a highly innovative platform called Self-Assembling Protein Nanoparticle (SAPN). In this vaccine, B cell epitopes derived from the second heptad repeat (HR2) region of IBV spike proteins were repetitively presented in its native trimeric conformation. In addition, flagellin was co-displayed in the SAPN to achieve a self-adjuvanted effect. Three groups of chickens were immunized at four weeks of age with the vaccine prototype, IBV-Flagellin-SAPN, a negative-control construct Flagellin-SAPN or a buffer control. The immunized chickens were challenged with 5x104.7 EID50 IBV M41 strain. High antibody responses were detected in chickens immunized with IBV-Flagellin-SAPN. In ex vivo proliferation tests, peripheral mononuclear cells (PBMCs) derived from IBV-Flagellin-SAPN immunized chickens had a significantly higher stimulation index than that of PBMCs from chickens receiving Flagellin-SAPN. Chickens immunized with IBV-Flagellin-SAPN had a significant reduction of tracheal virus shedding and lesser tracheal lesion scores than did negative control chickens. The data demonstrated that the IBV-Flagellin-SAPN holds promise as a vaccine for IBV.
Collapse
Affiliation(s)
- Jianping Li
- Department of Pathobiology and Veterinary Science University of Connecticut, Storrs, CT, United States of America
| | - Zeinab H. Helal
- Department of Pathobiology and Veterinary Science University of Connecticut, Storrs, CT, United States of America
- Department of Microbiology and Immunology, Faculty of Pharmacy, Al-Azhar University, Cairo, Egypt
| | - Christopher P. Karch
- The Institute of Material Sciences, University of Connecticut, Storrs, CT, United States of America
| | - Neha Mishra
- Department of Pathobiology and Veterinary Science University of Connecticut, Storrs, CT, United States of America
| | - Theodore Girshick
- Charles River Laboratories, Avian vaccine services, North Franklin, CT, United States of America
| | - Antonio Garmendia
- Department of Pathobiology and Veterinary Science University of Connecticut, Storrs, CT, United States of America
| | - Peter Burkhard
- The Institute of Material Sciences, University of Connecticut, Storrs, CT, United States of America
- Department of Molecular Cell Biology, University of Connecticut, Storrs, CT, United States of America
- Alpha-O-Peptides AG, Riehen, Switzerland
| | - Mazhar I. Khan
- Department of Pathobiology and Veterinary Science University of Connecticut, Storrs, CT, United States of America
- * E-mail:
| |
Collapse
|
52
|
Flores-Langarica A, Müller Luda K, Persson EK, Cook CN, Bobat S, Marshall JL, Dahlgren MW, Hägerbrand K, Toellner KM, Goodall MD, Withers DR, Henderson IR, Johansson Lindbom B, Cunningham AF, Agace WW. CD103 +CD11b + mucosal classical dendritic cells initiate long-term switched antibody responses to flagellin. Mucosal Immunol 2018; 11:681-692. [PMID: 29346347 PMCID: PMC5912514 DOI: 10.1038/mi.2017.105] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 10/23/2017] [Indexed: 02/04/2023]
Abstract
Antibody responses induced at mucosal and nonmucosal sites demonstrate a significant level of autonomy. Here, we demonstrate a key role for mucosal interferon regulatory factor-4 (IRF4)-dependent CD103+CD11b+ (DP), classical dendritic cells (cDCs) in the induction of T-dependent immunoglobulin G (IgG) and immunoglobulin A (IgA) responses in the mesenteric lymph node (MLN) following systemic immunization with soluble flagellin (sFliC). In contrast, IRF8-dependent CD103+CD11b- (SP) are not required for these responses. The lack of this response correlated with a complete absence of sFliC-specific plasma cells in the MLN, small intestinal lamina propria, and surprisingly also the bone marrow (BM). Many sFliC-specific plasma cells accumulating in the BM of immunized wild-type mice expressed α4β7+, suggesting a mucosal origin. Collectively, these results suggest that mucosal DP cDC contribute to the generation of the sFliC-specific plasma cell pool in the BM and thus serve as a bridge linking the mucosal and systemic immune system.
Collapse
Affiliation(s)
- A Flores-Langarica
- Institute of Immunology & Immunotherapy, College of Medical & Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - K Müller Luda
- Immunology Section, Lund University, BMC D14 Sölvegatan 19, S-221 84. Lund 22184, Sweden
| | - E K Persson
- Immunology Section, Lund University, BMC D14 Sölvegatan 19, S-221 84. Lund 22184, Sweden
| | - C N Cook
- Institute of Immunology & Immunotherapy, College of Medical & Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - S Bobat
- Institute of Immunology & Immunotherapy, College of Medical & Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - J L Marshall
- Institute of Immunology & Immunotherapy, College of Medical & Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - M W Dahlgren
- Immunology Section, Lund University, BMC D14 Sölvegatan 19, S-221 84. Lund 22184, Sweden
| | - K Hägerbrand
- Immunology Section, Lund University, BMC D14 Sölvegatan 19, S-221 84. Lund 22184, Sweden
| | - K M Toellner
- Institute of Immunology & Immunotherapy, College of Medical & Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - M D Goodall
- Institute of Immunology & Immunotherapy, College of Medical & Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - D R Withers
- Institute of Immunology & Immunotherapy, College of Medical & Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - I R Henderson
- Institute of Microbiology and Infection, College of Medical & Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - B Johansson Lindbom
- Immunology Section, Lund University, BMC D14 Sölvegatan 19, S-221 84. Lund 22184, Sweden
- Division of Immunology and Vaccinology, National Veterinary Institute, Technical University of Denmark (DTU). Kongens Lyngby, Denmark
| | - A F Cunningham
- Institute of Immunology & Immunotherapy, College of Medical & Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK
- Institute of Microbiology and Infection, College of Medical & Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - W W Agace
- Immunology Section, Lund University, BMC D14 Sölvegatan 19, S-221 84. Lund 22184, Sweden
- Division of Immunology and Vaccinology, National Veterinary Institute, Technical University of Denmark (DTU). Kongens Lyngby, Denmark
| |
Collapse
|
53
|
Il Kim M, Lee C, Park J, Jeon BY, Hong M. Crystal structure of Bacillus cereus flagellin and structure-guided fusion-protein designs. Sci Rep 2018; 8:5814. [PMID: 29643437 PMCID: PMC5895748 DOI: 10.1038/s41598-018-24254-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 03/23/2018] [Indexed: 12/16/2022] Open
Abstract
Flagellin is a major component of the flagellar filament. Flagellin also functions as a specific ligand that stimulates innate immunity through direct interaction with Toll-like receptor 5 (TLR5) in the host. Because flagellin activates the immune response, it has been of interest to develop as a vaccine adjuvant in subunit vaccines or antigen fusion vaccines. Despite the widespread application of flagellin fusion in preventing infectious diseases, flagellin-antigen fusion designs have never been biophysically and structurally characterized. Moreover, flagellin from Salmonella species has been used extensively despite containing hypervariable regions not required for TLR5 that can cause an unexpected immune response. In this study, flagellin from Bacillus cereus (BcFlg) was identified as the smallest flagellin molecule containing only the conserved TLR5-activating D0 and D1 domains. The crystal structure of BcFlg was determined to provide a scheme for fusion designs. Through homology-based modeling and comparative structural analyses, diverse fusion strategies were proposed. Moreover, cellular and biophysical analysis of an array of fusion constructs indicated that insertion fusion at BcFlg residues 178–180 does not interfere with the protein stability or TLR5-stimulating capacity of flagellin, suggesting its usefulness in the development and optimization of flagellin fusion vaccines.
Collapse
Affiliation(s)
- Meong Il Kim
- Division of Biological Science and Technology, Yonsei University, Wonju, 26493, Republic of Korea
| | - Choongdeok Lee
- Division of Biological Science and Technology, Yonsei University, Wonju, 26493, Republic of Korea
| | - Jaewan Park
- Division of Biological Science and Technology, Yonsei University, Wonju, 26493, Republic of Korea
| | - Bo-Young Jeon
- Department of Biomedical Laboratory Science, Yonsei University, Wonju, 26493, Republic of Korea
| | - Minsun Hong
- Division of Biological Science and Technology, Yonsei University, Wonju, 26493, Republic of Korea.
| |
Collapse
|
54
|
Stepanova LA, Mardanova ES, Shuklina MA, Blokhina EA, Kotlyarov RY, Potapchuk MV, Kovaleva AA, Vidyaeva IG, Korotkov AV, Eletskaya EI, Ravin NV, Tsybalova LM. Flagellin-fused protein targeting M2e and HA2 induces potent humoral and T-cell responses and protects mice against various influenza viruses a subtypes. J Biomed Sci 2018; 25:33. [PMID: 29631629 PMCID: PMC5891888 DOI: 10.1186/s12929-018-0433-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 03/27/2018] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Current influenza vaccines are mainly strain-specific and have limited efficacy in preventing new, potentially pandemic, influenza strains. Efficient control of influenza A infection can potentially be achieved through the development of broad-spectrum vaccines based on conserved antigens. A current trend in the design of universal flu vaccines is the construction of recombinant proteins based on combinations of various conserved epitopes of viral proteins (M1, M2, HA2, NP). In this study, we compared the immunogenicity and protective action of two recombinant proteins which feature different designs and which target different antigens. RESULTS Balb/c mice were immunized subcutaneously with Flg-HA2-2-4M2ehs or FlgSh-HA2-2-4M2ehs; these constructs differ in the location of hemagglutinin's HA2-2(76-130) insertion into flagellin (FliC). The humoral and T-cell immune responses to these constructs were evaluated. The simultaneous expression of different M2e and HA2-2(76-130) in recombinant protein form induces a strong M2e-specific IgG response and CD4+/ CD8+ T-cell response. The insertion of HA2-2(76-130) into the hypervariable domain of flagellin greatly increases antigen-specific T-cell response, as evidenced by the formation of multi-cytokine-secreting CD4+, CD8+ T-cells, Tem, and Tcm. Both proteins provide full protection from lethal challenge with A/H3N2 and A/H7N9. CONCLUSION Our results show that highly conserved M2e and HA2-2(76-130) can be used as important targets for the development of universal flu vaccines. The location of the HA2-2(76-130) peptide's insertion into the hypervariable domain of flagellin had a significant effect on the T-cell response to influenza antigens, as seen by forming of multi-cytokine-secreting CD4+ and CD8+ T-cells.
Collapse
Affiliation(s)
- Liudmila A Stepanova
- Research Institute of Influenza, Russian Ministry of Health, Prof. Popova str.15/17, 197376, St. Petersburg, Russia.
| | - Eugenia S Mardanova
- Institute of Bioengineering, Research Center of Biotechnology, Russian Academy of Sciences, Leninsky Ave. 33, building 2, 119071, Moscow, Russia
| | - Marina A Shuklina
- Research Institute of Influenza, Russian Ministry of Health, Prof. Popova str.15/17, 197376, St. Petersburg, Russia
| | - Elena A Blokhina
- Institute of Bioengineering, Research Center of Biotechnology, Russian Academy of Sciences, Leninsky Ave. 33, building 2, 119071, Moscow, Russia
| | - Roman Y Kotlyarov
- Institute of Bioengineering, Research Center of Biotechnology, Russian Academy of Sciences, Leninsky Ave. 33, building 2, 119071, Moscow, Russia
| | - Marina V Potapchuk
- Research Institute of Influenza, Russian Ministry of Health, Prof. Popova str.15/17, 197376, St. Petersburg, Russia
| | - Anna A Kovaleva
- Research Institute of Influenza, Russian Ministry of Health, Prof. Popova str.15/17, 197376, St. Petersburg, Russia
| | - Inna G Vidyaeva
- Research Institute of Influenza, Russian Ministry of Health, Prof. Popova str.15/17, 197376, St. Petersburg, Russia
| | - Alexandr V Korotkov
- Research Institute of Influenza, Russian Ministry of Health, Prof. Popova str.15/17, 197376, St. Petersburg, Russia
| | - Elizaveta I Eletskaya
- Research Institute of Influenza, Russian Ministry of Health, Prof. Popova str.15/17, 197376, St. Petersburg, Russia
| | - Nikolai V Ravin
- Institute of Bioengineering, Research Center of Biotechnology, Russian Academy of Sciences, Leninsky Ave. 33, building 2, 119071, Moscow, Russia
| | - Liudmila M Tsybalova
- Research Institute of Influenza, Russian Ministry of Health, Prof. Popova str.15/17, 197376, St. Petersburg, Russia
| |
Collapse
|
55
|
Abstract
INTRODUCTION Bacterial flagellin, as a pathogen-associated molecular pattern (PAMP), can activate both innate and adaptive immunity. Its unique structural characteristics endow an effective and flexible adjuvant activity, which allow the design of different types of vaccine strategies to prevent various diseases. This review will discuss recent progress in the mechanism of action of flagellin and its prospects for use as a vaccine adjuvant. AREAS COVERED Herein we summarize various types of information related to flagellin adjuvants from PubMed, including structures, signaling pathways, natural immunity, and extensive applications in vaccines, and it discusses the immunogenicity, safety, and efficacy of flagellin-adjuvanted vaccines in clinical trials. EXPERT COMMENTARY It is widely accepted that as an adjuvant, flagellin can induce an enhanced antigen-specific immune response. Flagellin adjuvants will allow more effective flagellin-based vaccines to enter clinical trials. Furthermore, vaccine formulations containing PAMPs are crucial to exert the maximum potential of vaccine antigens. Therefore, combinations of flagellin-adjuvanted vaccines with other adjuvants that act in a synergistic manner, particularly TLR ligands, represent a promising method for tailoring targeted vaccines to meet specific requirements.
Collapse
Affiliation(s)
- Baofeng Cui
- a State Key Laboratory of Veterinary Etiological Biology, OIE/National Foot and Mouth Disease Reference Laboratory, Key Laboratory of Animal Virology of Ministry of Agriculture , Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences , Lanzhou , China.,b Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses , Yangzhou , China
| | - Xinsheng Liu
- a State Key Laboratory of Veterinary Etiological Biology, OIE/National Foot and Mouth Disease Reference Laboratory, Key Laboratory of Animal Virology of Ministry of Agriculture , Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences , Lanzhou , China.,b Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses , Yangzhou , China
| | - Yuzhen Fang
- a State Key Laboratory of Veterinary Etiological Biology, OIE/National Foot and Mouth Disease Reference Laboratory, Key Laboratory of Animal Virology of Ministry of Agriculture , Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences , Lanzhou , China.,b Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses , Yangzhou , China
| | - Peng Zhou
- a State Key Laboratory of Veterinary Etiological Biology, OIE/National Foot and Mouth Disease Reference Laboratory, Key Laboratory of Animal Virology of Ministry of Agriculture , Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences , Lanzhou , China.,b Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses , Yangzhou , China
| | - Yongguang Zhang
- a State Key Laboratory of Veterinary Etiological Biology, OIE/National Foot and Mouth Disease Reference Laboratory, Key Laboratory of Animal Virology of Ministry of Agriculture , Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences , Lanzhou , China.,b Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses , Yangzhou , China
| | - Yonglu Wang
- a State Key Laboratory of Veterinary Etiological Biology, OIE/National Foot and Mouth Disease Reference Laboratory, Key Laboratory of Animal Virology of Ministry of Agriculture , Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences , Lanzhou , China.,b Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses , Yangzhou , China
| |
Collapse
|
56
|
Ignacio BJ, Albin TJ, Esser-Kahn AP, Verdoes M. Toll-like Receptor Agonist Conjugation: A Chemical Perspective. Bioconjug Chem 2018; 29:587-603. [PMID: 29378134 PMCID: PMC10642707 DOI: 10.1021/acs.bioconjchem.7b00808] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Toll-like receptors (TLRs) are vital elements of the mammalian immune system that function by recognizing pathogen-associated molecular patterns (PAMPs), bridging innate and adaptive immunity. They have become a prominent therapeutic target for the treatment of infectious diseases, cancer, and allergies, with many TLR agonists currently in clinical trials or approved as immunostimulants. Numerous studies have shown that conjugation of TLR agonists to other molecules can beneficially influence their potency, toxicity, pharmacokinetics, or function. The functional properties of TLR agonist conjugates, however, are highly dependent on the ligation strategy employed. Here, we review the chemical structural requirements for effective functional TLR agonist conjugation. In addition, we provide similar analysis for those that have yet to be conjugated. Moreover, we discuss applications of covalent TLR agonist conjugation and their implications for clinical use.
Collapse
Affiliation(s)
- Bob J. Ignacio
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Tyler J. Albin
- Department of Chemistry, University of California, Irvine, Irvine, California 92697, United States
| | - Aaron P. Esser-Kahn
- Department of Chemistry, University of California, Irvine, Irvine, California 92697, United States
- Institute for Molecular Engineering, University of Chicago, Chicago, Illinois 60637, United States
| | - Martijn Verdoes
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| |
Collapse
|
57
|
Chua BY, Sekiya T, Jackson DC. Opinion: Making Inactivated and Subunit-Based Vaccines Work. Viral Immunol 2018; 31:150-158. [PMID: 29369750 DOI: 10.1089/vim.2017.0146] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Empirically derived vaccines have in the past relied on the isolation and growth of disease-causing microorganisms that are then inactivated or attenuated before being administered. This is often done without prior knowledge of the mechanisms involved in conferring protective immunity. Recent advances in scientific technologies and in our knowledge of how protective immune responses are induced enable us to rationally design novel and safer vaccination strategies. Such advances have accelerated the development of inactivated whole-organism- and subunit-based vaccines. In this review, we discuss ideal attributes and criteria that need to be considered for the development of vaccines and some existing vaccine platforms. We focus on inactivated vaccines against influenza virus and ways by which vaccine efficacy can be improved with the use of adjuvants and Toll-like receptor-2 signaling.
Collapse
Affiliation(s)
- Brendon Y Chua
- 1 Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne , Melbourne, Australia .,2 Research Center for Zoonosis Control, Hokkaido University , Sapporo, Japan .,3 Global Institution for Collaborative Research and Education, Hokkaido University , Sapporo, Japan
| | - Toshiki Sekiya
- 2 Research Center for Zoonosis Control, Hokkaido University , Sapporo, Japan .,3 Global Institution for Collaborative Research and Education, Hokkaido University , Sapporo, Japan
| | - David C Jackson
- 1 Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne , Melbourne, Australia .,2 Research Center for Zoonosis Control, Hokkaido University , Sapporo, Japan .,3 Global Institution for Collaborative Research and Education, Hokkaido University , Sapporo, Japan
| |
Collapse
|
58
|
Xu Z, Moyle PM. Bioconjugation Approaches to Producing Subunit Vaccines Composed of Protein or Peptide Antigens and Covalently Attached Toll-Like Receptor Ligands. Bioconjug Chem 2017; 29:572-586. [PMID: 28891637 DOI: 10.1021/acs.bioconjchem.7b00478] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Traditional vaccines derived from attenuated or inactivated pathogens are effective at inducing antibody-based protective immune responses but tend to be highly reactogenic, causing notable adverse effects. Vaccines with superior safety profiles can be produced by subunit approaches, utilizing molecularly defined antigens (e.g., proteins and polysaccharides). These antigens, however, often elicit poor immunological responses, necessitating the use of adjuvants. Immunostimulatory adjuvants have the capacity to activate antigen presenting cells directly through specific receptors (e.g., Toll-like receptors (TLRs)), resulting in enhanced presentation of antigens as well as the secretion of proinflammatory chemokines and cytokines. Consequently, innate immune responses are amplified and adaptive immunity is generated. Recently, site-specific conjugation of such immunostimulatory adjuvants (e.g., TLR ligands) onto defined antigens has shown superior efficacy over unconjugated mixtures, suggesting that the development of chemically characterized immunostimulatory adjuvants and optimized approaches for their conjugation with antigens may provide a better opportunity for the development of potent, novel vaccines. This review briefly summarizes various TLR agonists utilized as immunostimulatory adjuvants and focuses on the development of techniques (e.g., recombinant, synthetic, and semisynthetic) for generating adjuvant-antigen fusion vaccines incorporating peptide or protein antigens.
Collapse
Affiliation(s)
- Zhenghui Xu
- School of Pharmacy , The University of Queensland , Woolloongabba 4102 , Queensland , Australia
| | - Peter Michael Moyle
- School of Pharmacy , The University of Queensland , Woolloongabba 4102 , Queensland , Australia
| |
Collapse
|
59
|
Schülke S, Kuttich K, Wolfheimer S, Duschek N, Wangorsch A, Reuter A, Briza P, Pablos I, Gadermaier G, Ferreira F, Vieths S, Toda M, Scheurer S. Conjugation of wildtype and hypoallergenic mugwort allergen Art v 1 to flagellin induces IL-10-DC and suppresses allergen-specific TH2-responses in vivo. Sci Rep 2017; 7:11782. [PMID: 28924222 PMCID: PMC5603567 DOI: 10.1038/s41598-017-11972-w] [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: 01/24/2017] [Accepted: 08/16/2017] [Indexed: 11/09/2022] Open
Abstract
Allergies to weed pollen including members of the Compositae family, such as mugwort, ragweed, and feverfew are spreading worldwide. To efficiently treat these newly arising allergies, allergen specific immunotherapy needs to be improved. Therefore, we generated novel vaccine candidates consisting of the TLR5-ligand Flagellin A from Listeria and the major mugwort allergen Art v 1 including either the wild type Art v 1 sequence (rFlaA:Artv1) or a hypoallergenic variant (rFlaA:Artv1hyp) with reduced IgE-binding capacity. Immune modulating capacity of these constructs and respective controls was evaluated in vitro and in vivo. Incorporation of hypoallergenic Art v 1 derivative did not interfere with the resulting fusion proteins’ immune stimulatory capacity. Both rFlaA:Artv1 and rFlaA:Artv1hyp induced a prominent, mTOR-dependent, IL-10 secretion from murine dendritic cells, and suppressed allergen-specific TH2-cytokine secretion in vitro and in vivo. Both conjugates retained the capacity to induce rFlaA-specific antibody responses while efficiently inducing production of Art v 1-specific IgG1 and IgG2a antibodies in mice. Interestingly, only the suppression of TH2-cytokine secretion by rFlaA:Artv1 (but not rFlaA:Artv1hyp) was paralleled by a strong secretion of IFN-γ. In summary, we provided evidence that incorporating hypoallergens into flagellin:allergen fusion proteins is a suitable strategy to further improve these promising vaccine candidates.
Collapse
Affiliation(s)
- Stefan Schülke
- Section Molecular Allergology, Paul-Ehrlich-Institut, Langen, Hessen, Germany.
| | - Kirsten Kuttich
- Section Molecular Allergology, Paul-Ehrlich-Institut, Langen, Hessen, Germany
| | - Sonja Wolfheimer
- Section Molecular Allergology, Paul-Ehrlich-Institut, Langen, Hessen, Germany
| | - Nadine Duschek
- Section Molecular Allergology, Paul-Ehrlich-Institut, Langen, Hessen, Germany
| | - Andrea Wangorsch
- Section Molecular Allergology, Paul-Ehrlich-Institut, Langen, Hessen, Germany
| | - Andreas Reuter
- Division of Allergology, Paul-Ehrlich-Institut, Langen, Hessen, Germany
| | - Peter Briza
- Department of Molecular Biology, Division of Allergy and Immunology, University of Salzburg, Salzburg, Austria
| | - Isabel Pablos
- Department of Molecular Biology, Division of Allergy and Immunology, University of Salzburg, Salzburg, Austria
| | - Gabriele Gadermaier
- Department of Molecular Biology, Division of Allergy and Immunology, University of Salzburg, Salzburg, Austria
| | - Fatima Ferreira
- Department of Molecular Biology, Division of Allergy and Immunology, University of Salzburg, Salzburg, Austria
| | - Stefan Vieths
- Section Molecular Allergology, Paul-Ehrlich-Institut, Langen, Hessen, Germany
| | - Masako Toda
- Section Molecular Allergology, Paul-Ehrlich-Institut, Langen, Hessen, Germany
| | - Stephan Scheurer
- Section Molecular Allergology, Paul-Ehrlich-Institut, Langen, Hessen, Germany
| |
Collapse
|
60
|
El Bissati K, Zhou Y, Paulillo SM, Raman SK, Karch CP, Roberts CW, Lanar DE, Reed S, Fox C, Carter D, Alexander J, Sette A, Sidney J, Lorenzi H, Begeman IJ, Burkhard P, McLeod R. Protein nanovaccine confers robust immunity against Toxoplasma. NPJ Vaccines 2017; 2:24. [PMID: 29263879 PMCID: PMC5627305 DOI: 10.1038/s41541-017-0024-6] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 06/20/2017] [Accepted: 06/21/2017] [Indexed: 11/08/2022] Open
Abstract
We designed and produced a self-assembling protein nanoparticle. This self-assembling protein nanoparticle contains five CD8+ HLA-A03-11 supertypes-restricted epitopes from antigens expressed during Toxoplasma gondii's lifecycle, the universal CD4+ T cell epitope PADRE, and flagellin as a scaffold and TLR5 agonist. These CD8+ T cell epitopes were separated by N/KAAA spacers and optimized for proteasomal cleavage. Self-assembling protein nanoparticle adjuvanted with TLR4 ligand-emulsion GLA-SE were evaluated for their efficacy in inducing IFN-γ responses and protection of HLA-A*1101 transgenic mice against T. gondii. Immunization, using self-assembling protein nanoparticle-GLA-SE, activated CD8+ T cells to produce IFN-γ. Self-assembling protein nanoparticle-GLA-SE also protected HLA-A*1101 transgenic mice against subsequent challenge with Type II parasites. Hence, combining CD8+ T cell-eliciting peptides and PADRE into a multi-epitope protein that forms a nanoparticle, administered with GLA-SE, leads to efficient presentation by major histocompatibility complex Class I and II molecules. Furthermore, these results suggest that activation of TLR4 and TLR5 could be useful for development of vaccines that elicit T cells to prevent toxoplasmosis in humans.
Collapse
Affiliation(s)
- Kamal El Bissati
- Departments of OVS, The University of Chicago, 5841S Maryland Ave, Chicago, IL 60637 USA
| | - Ying Zhou
- Departments of OVS, The University of Chicago, 5841S Maryland Ave, Chicago, IL 60637 USA
| | | | | | - Christopher P. Karch
- Institute of Materials Science and Department of Molecular and Cell Biology, University of Connecticut, 97 North Eagleville Road, Storrs, CT 06269 USA
| | - Craig W. Roberts
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, G4 0RE UK
| | - David E. Lanar
- Walter Reed Army Institute of Research, 503 Robert Grant Ave, Silver Spring, MD 20910 USA
| | - Steve Reed
- Infectious Diseases Research Institute, 1616 Eastlake Ave E #400, Seattle, WA 98102 USA
| | - Chris Fox
- Infectious Diseases Research Institute, 1616 Eastlake Ave E #400, Seattle, WA 98102 USA
| | - Darrick Carter
- Infectious Diseases Research Institute, 1616 Eastlake Ave E #400, Seattle, WA 98102 USA
| | - Jeff Alexander
- PaxVax, 3985-A Sorrento Valley Blvd, San Diego, CA 92121 USA
| | - Alessandro Sette
- La Jolla Institute of Allergy and Immunology, 9420 Athena Cir, La Jolla, CA 92037 USA
| | - John Sidney
- La Jolla Institute of Allergy and Immunology, 9420 Athena Cir, La Jolla, CA 92037 USA
| | - Hernan Lorenzi
- J. Craig Venter Institute, 9714 Medical Center Drive, Rockville, MD 20850 USA
| | - Ian J. Begeman
- Departments of OVS, The University of Chicago, 5841S Maryland Ave, Chicago, IL 60637 USA
| | - Peter Burkhard
- Alpha-O Peptides AG, Lörracherstrasse 50, 4125 Riehen, Switzerland
- Institute of Materials Science and Department of Molecular and Cell Biology, University of Connecticut, 97 North Eagleville Road, Storrs, CT 06269 USA
| | - Rima McLeod
- Departments of OVS, The University of Chicago, 5841S Maryland Ave, Chicago, IL 60637 USA
- Pediatrics (Infectious Diseases), The University of Chicago, 5841S Maryland Ave, Chicago, IL 60637 USA
| |
Collapse
|
61
|
Bacterial flagellin-a potent immunomodulatory agent. Exp Mol Med 2017; 49:e373. [PMID: 28860663 PMCID: PMC5628280 DOI: 10.1038/emm.2017.172] [Citation(s) in RCA: 208] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 04/23/2017] [Accepted: 05/01/2017] [Indexed: 01/07/2023] Open
Abstract
Flagellin is a subunit protein of the flagellum, a whip-like appendage that enables bacterial motility. Traditionally, flagellin was viewed as a virulence factor that contributes to the adhesion and invasion of host cells, but now it has emerged as a potent immune activator, shaping both the innate and adaptive arms of immunity during microbial infections. In this review, we summarize our understanding of bacterial flagellin and host immune system interactions and the role flagellin as an adjuvant, anti-tumor and radioprotective agent, and we address important areas of future research interests.
Collapse
|
62
|
Molony RD, Malawista A, Montgomery RR. Reduced dynamic range of antiviral innate immune responses in aging. Exp Gerontol 2017; 107:130-135. [PMID: 28822811 PMCID: PMC5815956 DOI: 10.1016/j.exger.2017.08.019] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 08/11/2017] [Accepted: 08/12/2017] [Indexed: 01/04/2023]
Abstract
The worldwide population aged ≥ 65 years is increasing and the average life span is expected to increase another 10 years by 2050. This extended lifespan is associated with a progressive decline in immune function and a paradoxical state of low-grade, chronic inflammation that may contribute to susceptibility to viral infection, and reduced responses to vaccination. Here we review the effects of aging on innate immune responses to viral pathogens including elements of recognition, signaling, and production of inflammatory mediators. We specifically focus on age-related changes in key pattern recognition receptor signaling pathways, converging on altered cytokine responses, including a notable impairment of antiviral interferon responses. We highlight an emergent change in innate immunity that arises during aging – the dampening of the dynamic range of responses to multiple sources of stimulation – which may underlie reduced efficiency of immune responses in aging. We review the effects of aging on innate antiviral immunity, including recognition, signaling, and cytokine responses. Lower Toll-like receptor expression leads to impaired signaling and responses upon activation of these sensors. Effects of aging on cytosolic nucleic acid sensing receptors and inflammasomes remains incompletely characterized. In aging the dynamic range of innate immunity is compressed, with increased basal activation of many signaling pathways. Interferon production is impaired with age, which may lead to the increased viral susceptibility of older persons.
Collapse
Affiliation(s)
- Ryan D Molony
- Departments of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, United States
| | - Anna Malawista
- Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, United States
| | - Ruth R Montgomery
- Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, United States; Human Translational Immunology, Yale University School of Medicine, New Haven, CT 06520, United States.
| |
Collapse
|
63
|
Kitzmüller C, Kalser J, Mutschlechner S, Hauser M, Zlabinger GJ, Ferreira F, Bohle B. Fusion proteins of flagellin and the major birch pollen allergen Bet v 1 show enhanced immunogenicity, reduced allergenicity, and intrinsic adjuvanticity. J Allergy Clin Immunol 2017; 141:293-299.e6. [PMID: 28456624 DOI: 10.1016/j.jaci.2017.02.044] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 02/03/2017] [Accepted: 02/22/2017] [Indexed: 11/28/2022]
Abstract
BACKGROUND Recombinant fusion proteins of flagellin and antigens have been demonstrated to induce strong innate and adaptive immune responses. Such fusion proteins can enhance the efficacy of allergen-specific immunotherapy. OBJECTIVE We sought to characterize different fusion proteins of flagellin and the major birch pollen allergen Bet v 1 for suitability as allergy vaccines. METHODS A truncated version of flagellin (NtCFlg) was genetically fused to the N- or C-terminus of Bet v 1. Toll-like receptor (TLR) 5 binding was assessed with HEK293 cells expressing TLR5. Upregulation of CD40, CD80, CD83, and CD86 on monocyte-derived dendritic cells from allergic patients was analyzed by using flow cytometry. The T cell-stimulatory capacity of the fusion proteins was assessed with naive and Bet v 1-specific T cells. IgE binding was tested in inhibition ELISAs and basophil activation tests. Mice were immunized with the fusion proteins in the absence and presence of aluminum hydroxide. Cellular and antibody responses were monitored. Murine antibodies were tested for blocking capacity in basophil activation tests. RESULTS Both fusion proteins matured monocyte-derived dendritic cells through TLR5. Compared with Bet v 1, the fusion proteins showed stronger T cell-stimulatory and reduced IgE-binding capacity and induced murine Bet v 1-specific antibodies in the absence of aluminum hydroxide. However, only antibodies induced by means of immunization with NtCFlg fused to the C-terminus of Bet v 1 inhibited binding of patients' IgE antibodies to Bet v 1. CONCLUSION Bet v 1-flagellin fusion proteins show enhanced immunogenicity, reduced allergenicity, and intrinsic adjuvanticity and thus represent promising vaccines for birch pollen allergen-specific immunotherapy. However, the sequential order of allergen and adjuvant within a fusion protein determines its immunologic characteristics.
Collapse
Affiliation(s)
- Claudia Kitzmüller
- Christian Doppler Laboratory for Immunomodulation, Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
| | - Julia Kalser
- Christian Doppler Laboratory for Immunomodulation, Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
| | - Sonja Mutschlechner
- Christian Doppler Laboratory for Immunomodulation, Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
| | - Michael Hauser
- Department of Molecular Biology, University of Salzburg, Salzburg, Austria
| | | | - Fatima Ferreira
- Department of Molecular Biology, University of Salzburg, Salzburg, Austria
| | - Barbara Bohle
- Christian Doppler Laboratory for Immunomodulation, Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
64
|
Biotechnology approaches to produce potent, self-adjuvanting antigen-adjuvant fusion protein subunit vaccines. Biotechnol Adv 2017; 35:375-389. [PMID: 28288861 DOI: 10.1016/j.biotechadv.2017.03.005] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2017] [Revised: 03/08/2017] [Accepted: 03/08/2017] [Indexed: 01/07/2023]
Abstract
Traditional vaccination approaches (e.g. live attenuated or killed microorganisms) are among the most effective means to prevent the spread of infectious diseases. These approaches, nevertheless, have failed to yield successful vaccines against many important pathogens. To overcome this problem, methods have been developed to identify microbial components, against which protective immune responses can be elicited. Subunit antigens identified by these approaches enable the production of defined vaccines, with improved safety profiles. However, they are generally poorly immunogenic, necessitating their administration with potent immunostimulatory adjuvants. Since few safe and effective adjuvants are currently used in vaccines approved for human use, with those available displaying poor potency, or an inability to stimulate the types of immune responses required for vaccines against specific diseases (e.g. cytotoxic lymphocytes (CTLs) to treat cancers), the development of new vaccines will be aided by the availability of characterized platforms of new adjuvants, improving our capacity to rationally select adjuvants for different applications. One such approach, involves the addition of microbial components (pathogen-associated molecular patterns; PAMPs), that can stimulate strong immune responses, into subunit vaccine formulations. The conjugation of PAMPs to subunit antigens provides a means to greatly increase vaccine potency, by targeting immunostimulation and antigen to the same antigen presenting cell. Thus, methods that enable the efficient, and inexpensive production of antigen-adjuvant fusions represent an exciting mean to improve immunity towards subunit antigens. Herein we review four protein-based adjuvants (flagellin, bacterial lipoproteins, the extra domain A of fibronectin (EDA), and heat shock proteins (Hsps)), which can be genetically fused to antigens to enable recombinant production of antigen-adjuvant fusion proteins, with a focus on their mechanisms of action, structural or sequence requirements for activity, sequence modifications to enhance their activity or simplify production, adverse effects, and examples of vaccines in preclinical or human clinical trials.
Collapse
|
65
|
Steeper M, Plebanski M, Flanagan KL. The global challenge and future strategies for keeping the world's aging population healthy by vaccination. Trans R Soc Trop Med Hyg 2016; 110:427-31. [PMID: 27618919 DOI: 10.1093/trstmh/trw054] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 08/08/2016] [Indexed: 01/03/2023] Open
Affiliation(s)
- Michelle Steeper
- Vaccine and Infectious Diseases Laboratory, Department of Immunology and Pathology, Monash University, Melbourne, Victoria 3181, Australia
| | - Magdalena Plebanski
- Vaccine and Infectious Diseases Laboratory, Department of Immunology and Pathology, Monash University, Melbourne, Victoria 3181, Australia Monash Institute of Medical Engineering, Monash University, Commercial Road, Prahran, Victoria, Australia
| | - Katie L Flanagan
- Vaccine and Infectious Diseases Laboratory, Department of Immunology and Pathology, Monash University, Melbourne, Victoria 3181, Australia School of Medicine, University of Tasmania, Liverpool Street, Hobart, Tasmania, Australia.
| |
Collapse
|
66
|
Holbrook BC, D'Agostino RB, Parks GD, Alexander-Miller MA. Adjuvanting an inactivated influenza vaccine with flagellin improves the function and quantity of the long-term antibody response in a nonhuman primate neonate model. Vaccine 2016; 34:4712-4717. [PMID: 27516064 DOI: 10.1016/j.vaccine.2016.08.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 07/25/2016] [Accepted: 08/02/2016] [Indexed: 01/07/2023]
Abstract
Young infants are at significantly increased risk of developing severe disease following infection with influenza virus. At present there is no approved vaccine for individuals below the age of six months given previous studies showing a failure of these individuals to efficiently seroconvert. Given the major impact of influenza on infant health, it is critical that we develop vaccines that will be safe and effective in this population. Using a nonhuman primate (NHP) model, we have evaluated the ability of an inactivated influenza virus vaccine adjuvanted with flagellin to result in long term immune responses in neonates. To evaluate this critical attribute, neonate NHP were vaccinated and boosted with inactivated influenza virus in combination with either flagellin or a mutant inactive flagellin control. Our studies show that inclusion of flagellin resulted in a significant increase (5-fold, p=0.04) in influenza virus-specific IgG antibody at 6months post-vaccination. In addition, the antibody present at this late time was of higher affinity (2.4-fold, p=0.02). Finally a greater percentage of infants had detectable neutralizing antibody. These results support the use of flagellin in neonates as an adjuvant that promotes long-lived, high affinity antibody responses.
Collapse
Affiliation(s)
- Beth C Holbrook
- Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Ralph B D'Agostino
- Department of Biostatistical Sciences, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Griffith D Parks
- Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Martha A Alexander-Miller
- Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston-Salem, NC, United States.
| |
Collapse
|
67
|
Holbrook BC, Kim JR, Blevins LK, Jorgensen MJ, Kock ND, D'Agostino RB, Aycock ST, Hadimani MB, King SB, Parks GD, Alexander-Miller MA. A Novel R848-Conjugated Inactivated Influenza Virus Vaccine Is Efficacious and Safe in a Neonate Nonhuman Primate Model. THE JOURNAL OF IMMUNOLOGY 2016; 197:555-64. [PMID: 27279374 DOI: 10.4049/jimmunol.1600497] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 05/15/2016] [Indexed: 11/19/2022]
Abstract
Influenza virus infection of neonates poses a major health concern, often resulting in severe disease and hospitalization. At present, vaccines for this at-risk population are lacking. Thus, development of an effective vaccine is an urgent need. In this study, we have used an innovative nonhuman primate neonate challenge model to test the efficacy of a novel TLR 7/8 agonist R848-conjugated influenza virus vaccine. The use of the intact virus represents a step forward in conjugate vaccine design because it provides multiple antigenic targets allowing for elicitation of a broad immune response. Our results show that this vaccine induces high-level virus-specific Ab- and cell-mediated responses in neonates that result in increased virus clearance and reduced lung pathology postchallenge compared with the nonadjuvanted virus vaccine. Surprisingly, the addition of a second TLR agonist (flagellin) did not enhance vaccine protection, suggesting that combinations of TLR that provide increased efficacy must be determined empirically. These data support further exploration of this new conjugate influenza vaccine approach as a platform for use in the at-risk neonate population.
Collapse
Affiliation(s)
- Beth C Holbrook
- Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston-Salem, NC 27101
| | - Jong R Kim
- Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston-Salem, NC 27101
| | - Lance K Blevins
- Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston-Salem, NC 27101
| | - Matthew J Jorgensen
- Department of Pathology, Section of Comparative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157
| | - Nancy D Kock
- Department of Pathology, Section of Comparative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157
| | - Ralph B D'Agostino
- Department of Biostatistical Sciences, Wake Forest School of Medicine, Winston-Salem, NC 27157
| | - S Tyler Aycock
- Animal Resources Program, Wake Forest School of Medicine, Winston-Salem, NC 27157; and
| | | | - S Bruce King
- Department of Chemistry, Wake Forest University, Winston-Salem, NC 27109
| | - Griffith D Parks
- Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston-Salem, NC 27101
| | | |
Collapse
|
68
|
Dowling JK, Mansell A. Toll-like receptors: the swiss army knife of immunity and vaccine development. Clin Transl Immunology 2016; 5:e85. [PMID: 27350884 PMCID: PMC4910119 DOI: 10.1038/cti.2016.22] [Citation(s) in RCA: 178] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 03/31/2016] [Accepted: 04/05/2016] [Indexed: 12/27/2022] Open
Abstract
Innate immune cells have a critical role in defense against infection and disease. Central to this is the broad specificity with which they can detect pathogen-associated patterns and danger-associated patterns via the pattern recognition receptors (PRRs) they express. Several families of PRRs have been identified including: Toll-like receptors (TLRs), C-type lectin-like receptors, retinoic acid-inducible gene-like receptors and nucleotide-binding oligomerization domain-like receptors. TLRs are one of the most largely studied families of PRRs. The binding of ligands to TLRs on antigen presenting cells (APCs), mainly dendritic cells, leads to APC maturation, induction of inflammatory cytokines and the priming of naive T cells to drive acquired immunity. Therefore, activation of TLRs promotes both innate inflammatory responses and the induction of adaptive immunity. Consequently, in the last two decades mounting evidence has inextricably linked TLR activation with the pathogenesis of immune diseases and cancer. It has become advantageous to harness these aspects of TLR signaling therapeutically to accelerate and enhance the induction of vaccine-specific responses and also target TLRs with the use of biologics and small molecule inhibitors for the treatment of disease. In these respects, TLRs may be considered a 'Swiss Army' knife of the immune system, ready to respond in a multitude of infectious and disease states. Here we describe the latest advances in TLR-targeted therapeutics and the use of TLR ligands as vaccine adjuvants.
Collapse
Affiliation(s)
- Jennifer K Dowling
- Pattern Recognition Receptors and Inflammation Research group, Centre for Innate Immunity and Infectious Disease, Hudson Institute of Medical Research, Melbourne, Victoria, Australia; Monash University, Clayton, Victoria, Australia
| | - Ashley Mansell
- Pattern Recognition Receptors and Inflammation Research group, Centre for Innate Immunity and Infectious Disease, Hudson Institute of Medical Research, Melbourne, Victoria, Australia; Monash University, Clayton, Victoria, Australia
| |
Collapse
|
69
|
Shah M, Anwar MA, Kim JH, Choi S. Advances in Antiviral Therapies Targeting Toll-like Receptors. Expert Opin Investig Drugs 2016; 25:437-53. [DOI: 10.1517/13543784.2016.1154040] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
70
|
Domínguez A, Godoy P, Torner N. The Effectiveness of Influenza Vaccination in Different Groups. Expert Rev Vaccines 2016; 15:751-64. [PMID: 26775669 DOI: 10.1586/14760584.2016.1142878] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Annual administration of the seasonal influenza vaccine, especially to persons known to be at elevated risk for developing serious complications, is the focus of current efforts to reduce the impact of influenza. The main factors influencing estimated inactivated influenza vaccine efficacy and effectiveness, the results obtained in different population groups, current vaccination strategies and the possible advantages of new vaccines are discussed. The available evidence suggests that influenza vaccines are less effective in the elderly than in young adults, but vaccination is encouraged by public health institutions due to higher mortality and complications. There is no consensus on universal vaccination of children yet economic studies suggest that yearly paediatric vaccination is cost saving. The benefits of herd immunity generated by paediatric vaccination require further study. Newer vaccines should be more and more-broadly protective, stable, easy to manufacture and administer and highly immunogenic across all population groups.
Collapse
Affiliation(s)
- Angela Domínguez
- a Department de Salut Pública , Universitat de Barcelona , Barcelona , Spain.,b CIBER Epidemiologia y Salut Pública (CIBERESP) , Madrid , Spain
| | - Pere Godoy
- b CIBER Epidemiologia y Salut Pública (CIBERESP) , Madrid , Spain.,c Agencia de Salut Pública de Catalunya , Generalitat de Catalunya , Barcelona , Spain
| | - Nuria Torner
- a Department de Salut Pública , Universitat de Barcelona , Barcelona , Spain.,b CIBER Epidemiologia y Salut Pública (CIBERESP) , Madrid , Spain.,c Agencia de Salut Pública de Catalunya , Generalitat de Catalunya , Barcelona , Spain
| |
Collapse
|
71
|
Tussey L, Strout C, Davis M, Johnson C, Lucksinger G, Umlauf S, Song L, Liu G, Abraham K, White CJ. Phase 1 Safety and Immunogenicity Study of a Quadrivalent Seasonal Flu Vaccine Comprising Recombinant Hemagglutinin-Flagellin Fusion Proteins. Open Forum Infect Dis 2016; 3:ofw015. [PMID: 26925433 PMCID: PMC4766387 DOI: 10.1093/ofid/ofw015] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Accepted: 01/22/2016] [Indexed: 11/14/2022] Open
Abstract
Background. We evaluated the safety and immunogenicity of VAX2012Q, a quadrivalent influenza vaccine comprising 4 hemagglutinin subunits fused to flagellin. Methods. In this dose-ranging, open-label study, healthy adults (18–40 years) were divided into 7 cohorts for evaluation of 5 dose levels and 3 component ratios. Dose levels were as follows: (1) 1 mcg per component of VAX128C (H1N1), VAX181 (H3N2), VAX173 (B-YAM), and VAX172 (B-VIC), respectively; (2) 2 mcg per component, respectively; (3) 2, 4, 4, and 4 mcg of each component, respectively; (4) 2, 4, 6, and 6 mcg of each component, respectively; and (5) 3 mcg per component, respectively. Tolerability and immunogenicity data were analyzed. Results. Three hundred sixteen subjects received VAX2012Q (309 per protocol). At all dose levels, 54% to 65% of subjects reported mild injection site pain, the most common local reaction. Moderate injection site pain increased at dose levels 2 through 5 (22%–42%, compared with 20% at dose level 1). Systemic symptoms were mostly mild to moderate with moderate symptoms increasing in dose levels 3 and 4. Three dose level 3 subjects (6%) reported severe, transient chills and or fever. Mean fold rises in hemagglutination inhibition titers ranged from 2.5 to 6.9 despite high baseline titers. Mean seroprotection rates were ≥90% and mean seroconversion rates were ≥40% for all strains in all groups postvaccination. Conclusions. VAX2012Q elicited immune responses at all dose levels with no significant safety concerns. Doses of 2 or 3 mcg per component provided a favorable balance of tolerability and immunogenicity.
Collapse
Affiliation(s)
| | - Cynthia Strout
- Coastal Carolina Research , Mt. Pleasant, South Carolina
| | | | | | | | | | | | - Ge Liu
- VaxInnate Corporation , Cranbury, New Jersey
| | | | - C Jo White
- Independent Consultant/Medical Monitor , Lower Gwynedd, Pennsylvania
| |
Collapse
|
72
|
Functional properties of flagellin as a stimulator of innate immunity. Sci Rep 2016; 6:18379. [PMID: 26755208 PMCID: PMC4709591 DOI: 10.1038/srep18379] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 10/05/2015] [Indexed: 11/08/2022] Open
Abstract
We report the development of a well-defined flagellin-based nanoparticle stimulator and also provide a new mechanism of action model explaining how flagellin-triggered innate immunity has evolved to favor localized rather than potentially debilitating systemic immune stimulation. Cell-free protein synthesis (CFPS) was used to facilitate mutational analysis and precisely orientated display of flagellin on Hepatitis B core (HBc) protein virus-like particles (VLPs). The need for product stability and an understanding of mechanism of action motivated investigations indicating that the D0 domain of flagellin is sensitive to amino acid sequence independent hydrolysis - apparently due to the need for structural flexibility during natural flagellin polymerization. When D0-stabilized flagellin was attached to HBc VLPs with the D0 domain facing outward, flagellin's tendency to polymerize caused the VLPs to precipitate. However, attaching the D0 domain to the VLP surface produced a stable nanoparticle adjuvant. Surprisingly, attaching only 2 flagellins per VLP provided the same 1 pM potency as did VLPs with about 33 attached flagellins suggesting that the TLR5 receptor is highly effective in delivering its intracellular signal. These observations suggest that flagellin's protease sensitivity, tendency to aggregate, and very high affinity for TLR5 receptors limit its systemic distribution to favor localized immune stimulation.
Collapse
|
73
|
Lim JS, Nguyen KCT, Nguyen CT, Jang I, Han JM, Fabian C, Lee SE, Rhee JH, Cho KA. Flagellin-dependent TLR5/caveolin-1 as a promising immune activator in immunosenescence. Aging Cell 2015. [PMID: 26223660 PMCID: PMC4568978 DOI: 10.1111/acel.12383] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
The age-associated decline of immune responses causes high susceptibility to infections and reduced vaccine efficacy in the elderly. However, the mechanisms underlying age-related deficits are unclear. Here, we found that the expression and signaling of flagellin (FlaB)-dependent Toll-like receptor 5 (TLR5), unlike the other TLRs, were well maintained in old macrophages, similar to young macrophages. The expression and activation of TLR5/MyD88, but not TLR4, were sensitively regulated by the upregulation of caveolin-1 in old macrophages through direct interaction. This interaction was also confirmed using macrophages from caveolin-1 or MyD88 knockout mice. Because TLR5 and caveolin-1 were well expressed in major old tissues including lung, skin, intestine, and spleen, we analyzed in vivo immune responses via a vaccine platform with FlaB as a mucosal adjuvant for the pneumococcal surface protein A (PspA) against Streptococcus pneumoniae infection in young and aged mice. The FlaB-PspA fusion protein induced a significantly higher level of PspA-specific IgG and IgA responses and demonstrated a high protective efficacy against a lethal challenge with live S. pneumoniae in aged mice. These results suggest that caveolin-1/TLR5 signaling plays a key role in age-associated innate immune responses and that FlaB-PspA stimulation of TLR5 may be a new strategy for a mucosal vaccine adjuvant against pneumococcal infection in the elderly.
Collapse
Affiliation(s)
- Jae Sung Lim
- Department of Biochemistry Chonnam National University Medical School Gwangju 501‐746 South Korea
| | - Kim Cuc Thi Nguyen
- Department of Biochemistry Chonnam National University Medical School Gwangju 501‐746 South Korea
- Center for Creative Biomedical Scientists Chonnam National University Medical School Gwangju 501‐746 South Korea
| | - Chung Truong Nguyen
- Clinical Vaccine R&D Center Chonnam National University Hwasun Hospital 160 Ilsim‐Ri Hwasun‐gun Jeonnam 519‐809 South Korea
| | - Ik‐Soon Jang
- Division of Life Science Korea Basic Science Institute Daejeon 305‐333 South Korea
| | - Jung Min Han
- Department of Integrated OMICS for Biomedical Science Yonsei University Seoul 120‐749 South Korea
- College of Pharmacy Yonsei University Incheon 406‐840 South Korea
| | - Claire Fabian
- Department of Immunology Fraunhofer Institute for Cell Therapy and Immunology (IZI) University of Leipzig 04103 Leipzig Germany
- Translational Center for Regenerative Medicine (TRM) University of Leipzig 04103 Leipzig Germany
| | - Shee Eun Lee
- Clinical Vaccine R&D Center Chonnam National University Hwasun Hospital 160 Ilsim‐Ri Hwasun‐gun Jeonnam 519‐809 South Korea
- Dental Science Research Institute School of Dentistry Chonnam National University Gwangju 500‐757 South Korea
| | - Joon Haeng Rhee
- Clinical Vaccine R&D Center Chonnam National University Hwasun Hospital 160 Ilsim‐Ri Hwasun‐gun Jeonnam 519‐809 South Korea
- Department of Microbiology Chonnam National University Medical School Gwangju 501‐746 South Korea
| | - Kyung A Cho
- Department of Biochemistry Chonnam National University Medical School Gwangju 501‐746 South Korea
- Center for Creative Biomedical Scientists Chonnam National University Medical School Gwangju 501‐746 South Korea
- Clinical Vaccine R&D Center Chonnam National University Hwasun Hospital 160 Ilsim‐Ri Hwasun‐gun Jeonnam 519‐809 South Korea
- Research Institute of Medical Sciences Chonnam National University Medical School Gwangju 501‐746 South Korea
| |
Collapse
|
74
|
Vannice KS, Roehrig JT, Hombach J. Next generation dengue vaccines: A review of the preclinical development pipeline. Vaccine 2015; 33:7091-9. [PMID: 26424602 DOI: 10.1016/j.vaccine.2015.09.053] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Revised: 08/26/2015] [Accepted: 09/02/2015] [Indexed: 11/17/2022]
Abstract
Dengue represents a significant and growing public health problem across the globe, with approximately half of the world's population at risk. The increasing and expanding burden of dengue has highlighted the need for new tools to prevent dengue, including development of dengue vaccines. Recently, the first dengue vaccine candidate was evaluated in Phase 3 clinical trials, and other vaccine candidates are under clinical evaluation. There are also a number of candidates in preclinical development, based on diverse technologies, with promising results in animal models and likely to move into clinical trials and could eventually be next-generation dengue vaccines. This review provides an overview of the various technological approaches to dengue vaccine development with specific focus on candidates in preclinical development and with evaluation in non-human primates.
Collapse
Affiliation(s)
- Kirsten S Vannice
- Initiative for Vaccine Research, Department of Immunizations, Vaccines and Biologicals, World Health Organization, Geneva, Switzerland
| | - John T Roehrig
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Joachim Hombach
- Initiative for Vaccine Research, Department of Immunizations, Vaccines and Biologicals, World Health Organization, Geneva, Switzerland.
| |
Collapse
|
75
|
Bitrus Y, Andrew JN, Owolodun OA, Luka PD, Umaru DA. The reoccurrence of H5N1 outbreaks necessitates the development of safe and effective influenza vaccine technologies for the prevention and control of avian influenza in Sub-Saharan Africa. ACTA ACUST UNITED AC 2015. [DOI: 10.5897/bmbr2015.0246] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
|
76
|
Inclusion of Flagellin during Vaccination against Influenza Enhances Recall Responses in Nonhuman Primate Neonates. J Virol 2015; 89:7291-303. [PMID: 25948746 DOI: 10.1128/jvi.00549-15] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 04/28/2015] [Indexed: 02/01/2023] Open
Abstract
UNLABELLED Influenza virus can cause life-threatening infections in neonates and young infants. Although vaccination is a major countermeasure against influenza, current vaccines are not approved for use in infants less than 6 months of age, in part due to the weak immune response following vaccination. Thus, there is a strong need to develop new vaccines with improved efficacy for this vulnerable population. To address this issue, we established a neonatal African green monkey (AGM) nonhuman primate model that could be used to identify effective influenza vaccine approaches for use in young infants. We assessed the ability of flagellin, a Toll-like receptor 5 (TLR5) agonist, to serve as an effective adjuvant in this at-risk population. Four- to 6-day-old AGMs were primed and boosted with inactivated PR8 influenza virus (IPR8) adjuvanted with either wild-type flagellin or inactive flagellin with a mutation at position 229 (m229), the latter of which is incapable of signaling through TLR5. Increased IgG responses were observed following a boost, as well as at early times after challenge, in infants vaccinated with flagellin-adjuvanted IPR8. Inclusion of flagellin during vaccination also resulted in a significantly increased number of influenza virus-specific T cells following challenge compared to the number in infants vaccinated with the m229 adjuvant. Finally, following challenge infants vaccinated with IPR8 plus flagellin exhibited a reduced pathology in the lungs compared to that in infants that received IPR8 plus m229. This study provides the first evidence of flagellin-mediated enhancement of vaccine responses in nonhuman primate neonates. IMPORTANCE Young infants are particularly susceptible to severe disease as a result of influenza virus infection. Compounding this is the lack of effective vaccines for use in this vulnerable population. Here we describe a vaccine approach that results in improved immune responses and protection in young infants. Incorporation of flagellin during vaccination resulted in increased antibody and T cell responses together with reduced disease following virus infection. These results suggest that flagellin may serve as an effective adjuvant for vaccines targeted to this vulnerable population.
Collapse
|
77
|
Immunogenicity and efficacy of flagellin-envelope fusion dengue vaccines in mice and monkeys. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2015; 22:516-25. [PMID: 25761459 DOI: 10.1128/cvi.00770-14] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Accepted: 02/23/2015] [Indexed: 11/20/2022]
Abstract
The envelope (E) protein of flaviviruses includes three domains, EI, EII, and EIII, and is the major protective antigen. Because EIII is rich in type-specific and subcomplex-specific neutralizing epitopes and is easy to express, it is particularly attractive as a recombinant vaccine antigen. VaxInnate has developed a vaccine platform that genetically links vaccine antigens to bacterial flagellin, a Toll-like receptor 5 ligand. Here we report that tetravalent dengue vaccines (TDVs) consisting of four constructs, each containing two copies of EIII fused to flagellin (R3.2x format), elicited robust and long-lived neutralizing antibodies (geometric mean titers of 200 to 3,000), as measured with a 50% focus reduction neutralization test (FRNT50). In an immunogenicity study, rhesus macaques (n = 2) immunized subcutaneously with 10 μg or 90 μg of TDV three or four times, at 4- to 6-week intervals, developed neutralizing antibodies to four dengue virus (DENV) serotypes (mean post-dose 3 FRNT50 titers of 102 to 601). In an efficacy study, rhesus macaques (n = 4) were immunized intramuscularly with 16 μg or 48 μg of TDV or a placebo control three times, at 1-month intervals. The animals that received 48-μg doses of TDV developed neutralizing antibodies against the four serotypes (geometric mean titers of 49 to 258) and exhibited reduced viremia after DENV-2 challenge, with a group mean viremia duration of 1.25 days and 2 of 4 animals being completely protected, compared to the placebo-treated animals, which all developed viremia, with a mean duration of 4 days. In conclusion, flagellin-EIII fusion vaccines are immunogenic and partially protective in a nonhuman primate model.
Collapse
|
78
|
Guo J, Yang Y, Xiao W, Sun W, Yu H, Du L, Lustigman S, Jiang S, Kou Z, Zhou Y. A truncated fragment of Ov-ASP-1 consisting of the core pathogenesis-related-1 (PR-1) domain maintains adjuvanticity as the full-length protein. Vaccine 2015; 33:1974-80. [PMID: 25736195 PMCID: PMC7115538 DOI: 10.1016/j.vaccine.2015.02.053] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Revised: 12/16/2014] [Accepted: 02/18/2015] [Indexed: 11/18/2022]
Abstract
The Onchocerca volvulus activation-associated secreted protein-1 (Ov-ASP-1) has good adjuvanticity for a variety of antigens and vaccines, probably due to its ability activate antigen-processing cells (APCs). However, the functional domain of Ov-ASP-1 as an adjuvant is not clearly defined. Based on the structural prediction of this protein family, we constructed a 16-kDa recombinant protein of Ov-ASP-1 that contains only the core pathogenesis-related-1 (PR-1) domain (residues 10-153), designated ASPPR. We found that ASPPR exhibits adjuvanticity similar to that of the full-length Ov-ASP-1 (residues 10-220) for various antigens, including ovalbumin (OVA), HBsAg protein antigen, and the HIV peptide 5 (Pep5) antigen, but it is more suitable for vaccine design in ASPPR-antigen fusion proteins, and more stable in PBS than Ov-ASP-1 stored at -70 °C. These results suggest that ASPPR might be the functional region of Ov-ASP-1 as an adjuvant, and therefore could be developed as an adjuvant for human use.
Collapse
Affiliation(s)
- Jingjing Guo
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Yi Yang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Wenjun Xiao
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Weilai Sun
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Hong Yu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Lanying Du
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY, USA
| | - Sara Lustigman
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY, USA
| | - Shibo Jiang
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY, USA; Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhihua Kou
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China.
| | - Yusen Zhou
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China.
| |
Collapse
|
79
|
Lockner JW, Eubanks LM, Choi JL, Lively JM, Schlosburg JE, Collins KC, Globisch D, Rosenfeld-Gunn RJ, Wilson IA, Janda KD. Flagellin as carrier and adjuvant in cocaine vaccine development. Mol Pharm 2015; 12:653-62. [PMID: 25531528 PMCID: PMC4319694 DOI: 10.1021/mp500520r] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Cocaine abuse is problematic, directly and indirectly impacting the lives of millions, and yet existing therapies are inadequate and usually ineffective. A cocaine vaccine would be a promising alternative therapeutic option, but efficacy is hampered by variable production of anticocaine antibodies. Thus, new tactics and strategies for boosting cocaine vaccine immunogenicity must be explored. Flagellin is a bacterial protein that stimulates the innate immune response via binding to extracellular Toll-like receptor 5 (TLR5) and also via interaction with intracellular NOD-like receptor C4 (NLRC4), leading to production of pro-inflammatory cytokines. Reasoning that flagellin could serve as both carrier and adjuvant, we modified recombinant flagellin protein to display a cocaine hapten termed GNE. The resulting conjugates exhibited dose-dependent stimulation of anti-GNE antibody production. Moreover, when adjuvanted with alum, but not with liposomal MPLA, GNE-FliC was found to be better than our benchmark GNE-KLH. This work represents a new avenue for exploration in the use of hapten-flagellin conjugates to elicit antihapten immune responses.
Collapse
Affiliation(s)
- Jonathan W Lockner
- Departments of Chemistry, Integrative Structural and Computational Biology, and Immunology and Microbial Science, The Scripps Research Institute , 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | | | | | | | | | | | | | | | | | | |
Collapse
|
80
|
Girard A, Roques E, Massie B, Archambault D. Flagellin in fusion with human rotavirus structural proteins exerts an adjuvant effect when delivered with replicating but non-disseminating adenovectors through the intrarectal route. Mol Biotechnol 2014; 56:394-407. [PMID: 24271565 DOI: 10.1007/s12033-013-9723-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Human rotavirus (HRV) is the worldwide leading cause of gastroenteritis in young children. Two live attenuated HRV vaccines have been approved since 2006. However, these live vaccines still have potential risks including reversion of virulence. Adenoviruses are suitable vectors for mucosal administration of subunit vaccines. In addition to the adjuvant effect of certain adenovirus components, the use of an adjuvant like flagellin is also another means to increase the immune response to the immunogen. The aim of this study was to determine whether flagellin in fusion with HRV structural proteins stimulates the innate immune response and enhances the HRV-specific immune response when delivered through the intrarectal route with replicating but non-disseminating adenovector (R-AdV). Salmonella typhimurium flagellin B (FljB) in fusion with HRV VP4Δ::VP7 protein induced IL-1β production in J774A.1 macrophages exposed to the R-AdV. Intrarectal administration of R-AdVs expressing either VP4Δ::VP7 or VP4Δ::VP7::FljB in BALB/c mice resulted in HRV-specific mixed Th1/Th2 immune responses. The HRV-specific antibody response elicited with the use of R-AdV expressing VP4Δ::VP7::FljB was higher than that with R-AdV expressing VP4Δ::VP7. The results also show that the replication capability of R-AdVs contributed to enhance the HRV-specific immune response as compared with that obtained with non-replicative AdVs. This work lays the foundation for using the R-AdV system and FljB-adjuvanted formulation to elicit a mucosal immune response specific to HRV.
Collapse
Affiliation(s)
- Aurélie Girard
- Department of Biological Sciences, University of Québec at Montréal, P.O. Box 8888, Succursale Centre-Ville, Montreal, QC, H3C 3P8, Canada
| | | | | | | |
Collapse
|
81
|
|
82
|
Dormitzer P, Tsai T, Del Giudice G. New technologies for influenza vaccines. Hum Vaccin Immunother 2014; 8:45-58. [DOI: 10.4161/hv.8.1.18859] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
|
83
|
Boraschi D, Italiani P. Immunosenescence and vaccine failure in the elderly: Strategies for improving response. Immunol Lett 2014; 162:346-53. [DOI: 10.1016/j.imlet.2014.06.006] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Revised: 06/09/2014] [Accepted: 06/12/2014] [Indexed: 12/21/2022]
|
84
|
Israely T, Melamed S, Achdout H, Erez N, Politi B, Waner T, Lustig S, Paran N. TLR3 and TLR9 agonists improve postexposure vaccination efficacy of live smallpox vaccines. PLoS One 2014; 9:e110545. [PMID: 25350003 PMCID: PMC4211728 DOI: 10.1371/journal.pone.0110545] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2014] [Accepted: 09/14/2014] [Indexed: 12/14/2022] Open
Abstract
Eradication of smallpox and discontinuation of the vaccination campaign resulted in an increase in the percentage of unvaccinated individuals, highlighting the need for postexposure efficient countermeasures in case of accidental or deliberate viral release. Intranasal infection of mice with ectromelia virus (ECTV), a model for human smallpox, is curable by vaccination with a high vaccine dose given up to 3 days postexposure. To further extend this protective window and to reduce morbidity, mice were vaccinated postexposure with Vaccinia-Lister, the conventional smallpox vaccine or Modified Vaccinia Ankara, a highly attenuated vaccine in conjunction with TLR3 or TLR9 agonists. We show that co-administration of the TLR3 agonist poly(I:C) even 5 days postexposure conferred protection, avoiding the need to increase the vaccination dose. Efficacious treatments prevented death, ameliorated disease symptoms, reduced viral load and maintained tissue integrity of target organs. Protection was associated with significant elevation of serum IFNα and anti-vaccinia IgM antibodies, modulation of IFNγ response, and balanced activation of NK and T cells. TLR9 agonists (CpG ODNs) were less protective than the TLR3 agonist poly(I:C). We show that activation of type 1 IFN by poly(I:C) and protection is achievable even without co-vaccination, requiring sufficient amount of the viral antigens of the infective agent or the vaccine. This study demonstrated the therapeutic potential of postexposure immune modulation by TLR activation, allowing to alleviate the disease symptoms and to further extend the protective window of postexposure vaccination.
Collapse
Affiliation(s)
- Tomer Israely
- Department of Infectious diseases, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Sharon Melamed
- Department of Infectious diseases, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Hagit Achdout
- Department of Infectious diseases, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Noam Erez
- Department of Infectious diseases, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Boaz Politi
- Department of Infectious diseases, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Trevor Waner
- Department of Infectious diseases, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Shlomo Lustig
- Department of Infectious diseases, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Nir Paran
- Department of Infectious diseases, Israel Institute for Biological Research, Ness-Ziona, Israel
- * E-mail:
| |
Collapse
|
85
|
Low JGH, Lee LS, Ooi EE, Ethirajulu K, Yeo P, Matter A, Connolly JE, Skibinski DAG, Saudan P, Bachmann M, Hanson BJ, Lu Q, Maurer-Stroh S, Lim S, Novotny-Diermayr V. Safety and immunogenicity of a virus-like particle pandemic influenza A (H1N1) 2009 vaccine: results from a double-blinded, randomized Phase I clinical trial in healthy Asian volunteers. Vaccine 2014; 32:5041-8. [PMID: 25045806 DOI: 10.1016/j.vaccine.2014.07.011] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Revised: 05/23/2014] [Accepted: 07/08/2014] [Indexed: 02/07/2023]
Abstract
METHODS A novel, fully bacterially produced recombinant virus-like particle (VLP) based influenza vaccine (gH1-Qbeta) against A/California/07/2009(H1N1) was tested in a double-blind, randomized phase I clinical trial at two clinical sites in Singapore. The trial evaluated the immunogenicity and safety of gH1-Qbeta in the presence or absence of alhydrogel adjuvant. Healthy adult volunteers with no or low pre-existing immunity against A/California/07/2009 (H1N1) were randomized to receive two intramuscular injections 21 days apart, with 100μg vaccine, containing 42μg hemagglutinin antigen. Antibody responses were measured before and 21 days after each immunization by hemagglutination inhibition (HAI) assays. The primary endpoint was seroconversion on Day 42, defined as percentage of subjects which reach a HAI titer ≥40 or achieve an at least 4-fold rise in HAI titer (with pre-existing immunity). The co-secondary endpoints were safety and seroconversion on Day 21. RESULTS A total of 84 Asian volunteers were enrolled in this study and randomized to receive the adjuvanted (n=43) or the non-adjuvanted (n=41) vaccine. Of those, 43 and 37 respectively (95%) completed the study. There were no deaths or serious adverse events reported during this trial. A total of 535 adverse events occurred during treatment with 49.5% local solicited symptoms, of mostly (76.4%) mild severity. The most common treatment-related systemic symptom was fatigue. The non-adjuvanted vaccine met all primary and secondary endpoints and showed seroconversion in 62.2% and 70.3% of participants respectively on Day 21 and Day 42. While the adjuvanted vaccine showed an increased seroconversion from 25.5% (Day 21) to 51.2% (Day 42), it did not meet the immunogenicity endpoint. CONCLUSION In summary, non-adjuvanted gH1-Qbeta showed similar antibody mediated immunogenicity and a comparable safety profile in healthy humans to commercially available vaccines. These results warrant the consideration of this VLP vaccine platform for the vaccination against influenza infection (HSA CTC1300092).
Collapse
Affiliation(s)
- Jenny G H Low
- SingHealth Investigational Medicine Unit, Singapore General Hospital, Block 7, Outram Rd., Singapore 169610, Singapore; Singapore General Hospital, 20 College Road, Singapore 169856, Singapore
| | - Lawrence S Lee
- Clinical Trials Research Unit, Changi General Hospital, 2 Simei St. 3, Singapore 529889, Singapore; National University of Singapore, Department of Medicine, 1E Kent Ridge Road, Singapore 119228, Singapore
| | - Eng Eong Ooi
- Duke-NUS Graduate Medical School, Program in Emerging Infectious Diseases, 8 College Rd., Singapore 169857, Singapore
| | - Kantharaj Ethirajulu
- D3 (Drug Discovery and Development), 31 Biopolis Way, #01-02a Nanos, Singapore 138669, Singapore
| | - Pauline Yeo
- D3 (Drug Discovery and Development), 31 Biopolis Way, #01-02a Nanos, Singapore 138669, Singapore
| | - Alex Matter
- D3 (Drug Discovery and Development), 31 Biopolis Way, #01-02a Nanos, Singapore 138669, Singapore
| | - John E Connolly
- Institute for Molecular and Cell Biology (IMCB) and Singapore Immunology Network (SIgN), A*STAR Program in Translational Research on Infectious Disease, Agency for Science, Technology and Research (A*STAR), Singapore; 61 Biopolis Drive, Proteos, Singapore 138673, Singapore
| | - David A G Skibinski
- Institute for Molecular and Cell Biology (IMCB) and Singapore Immunology Network (SIgN), A*STAR Program in Translational Research on Infectious Disease, Agency for Science, Technology and Research (A*STAR), Singapore; 61 Biopolis Drive, Proteos, Singapore 138673, Singapore
| | - Philippe Saudan
- Cytos Biotechnology AG, Wagistr. 25, Zürich-Schlieren CH-8952, Switzerland
| | - Martin Bachmann
- Cytos Biotechnology AG, Wagistr. 25, Zürich-Schlieren CH-8952, Switzerland
| | - Brendon J Hanson
- DSO National Laboratories, Bio-Defense Therapeutics Lab, 27 Medical Drive, Singapore 117510, Singapore
| | - Qingshu Lu
- Singapore Clinical Research Institute, 31 Biopolis Way, Level 2, Singapore 138669, Singapore
| | - Sebastian Maurer-Stroh
- Bioinformatics Institute, 30 Biopolis Street, #07-01 Matrix, Singapore 138671, Singapore
| | - Sam Lim
- D3 (Drug Discovery and Development), 31 Biopolis Way, #01-02a Nanos, Singapore 138669, Singapore
| | | |
Collapse
|
86
|
Kalnin K, Tibbitts T, Yan Y, Stegalkina S, Shen L, Costa V, Sabharwal R, Anderson SF, Day PM, Christensen N, Schiller JT, Jagu S, Roden RBS, Almond J, Kleanthous H. Low doses of flagellin-L2 multimer vaccines protect against challenge with diverse papillomavirus genotypes. Vaccine 2014; 32:3540-7. [PMID: 24780250 DOI: 10.1016/j.vaccine.2014.04.032] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Revised: 03/16/2014] [Accepted: 04/14/2014] [Indexed: 12/31/2022]
Abstract
Genetically modified bacterial flagellin (Fla), a Toll-like receptor-5 (TLR5) ligand, was evaluated as a fusion partner for human papillomavirus (HPV) L2-based immunogens in two animal challenge models; either cutaneous inoculation of rabbits with HPV 'quasivirions' containing cottontail rabbit papillomavirus (CRPV) genomes that induce warts, or intra-vaginal inoculation of mice with HPV 'pseudovirions' encapsidating a luciferase reporter plasmid and measurement of bioluminescence to determine infectivity. An Escherichia coli production system was developed for flagellin-L2 (Fla-L2) fusions containing either monomeric HPV-16 L2 a.a. 11(×11-200) or oligomeric L2 comprising a fusion of the a.a. 11-88 peptides of five (Fla∼5×11-88) or eight (Fla∼8×11-88) genital HPV types. Immunogenicity and bioactivity of Fla-L2 constructs were assessed using an in vitro neutralization and cell-based TLR-5 binding assay, respectively. Efficacy was evaluated following active immunization of rabbits or mice administered 3 intramuscular doses of Fla-L2 recombinants without exogenous adjuvant, followed by challenge. In addition, passive immunization studies of naïve rabbits with serial dilutions of pooled immune sera were used to determine End-Point Protection Titers (EPPT) for each formulation against a broader spectrum of HPV quasivirions. Efficacy was assessed for up to 10 weeks on the basis of wart volume induced following challenge and results compared to licensed L1-VLP vaccines (Gardasil and Cervarix). Following active immunization at doses as low as 1 μg, Fla-L2 fusions afforded complete protection against infection (mice) and disease (rabbits) following either homologous or heterologous HPV challenge. Passive immunization with anti-L2 immune sera discriminated between the different vaccine candidates under evaluation, demonstrated the protective role of antibody and suggested the superiority of this oligomeric L2-TLR5 agonist fusion approach compared to L1-based vaccines in its ability to cross-protect against non-vaccine HPV types.
Collapse
Affiliation(s)
- Kirill Kalnin
- Discovery, Sanofi Pasteur, 38 Sidney Street, Cambridge, MA, USA.
| | | | - Yanhua Yan
- Discovery, Sanofi Pasteur, 38 Sidney Street, Cambridge, MA, USA
| | | | - Lihua Shen
- Discovery, Sanofi Pasteur, 38 Sidney Street, Cambridge, MA, USA
| | - Victor Costa
- Discovery, Sanofi Pasteur, 38 Sidney Street, Cambridge, MA, USA
| | | | | | - Patricia M Day
- Laboratory of Cellular Oncology, National Cancer Institute, Bethesda, MD, USA
| | - Neil Christensen
- Department of Pathology, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - John T Schiller
- Laboratory of Cellular Oncology, National Cancer Institute, Bethesda, MD, USA
| | - Subhashini Jagu
- Department of Pathology, Johns Hopkins University, Baltimore, MD, USA
| | - Richard B S Roden
- Department of Pathology, Johns Hopkins University, Baltimore, MD, USA
| | - Jeffrey Almond
- Discovery, Sanofi Pasteur, 38 Sidney Street, Cambridge, MA, USA
| | | |
Collapse
|
87
|
Imaging murine NALT following intranasal immunization with flagellin-modified circumsporozoite protein malaria vaccines. Mucosal Immunol 2014; 7:304-14. [PMID: 23820750 PMCID: PMC3884030 DOI: 10.1038/mi.2013.48] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2013] [Accepted: 06/04/2013] [Indexed: 02/04/2023]
Abstract
Intranasal (IN) immunization with a Plasmodium circumsporozoite (CS) protein conjugated to flagellin, a Toll-like receptor 5 agonist, was found to elicit antibody-mediated protective immunity in our previous murine studies. To better understand IN-elicited immune responses, we examined the nasopharynx-associated lymphoid tissue (NALT) in immunized mice and the interaction of flagellin-modified CS with murine dendritic cells (DCs) in vitro. NALT of immunized mice contained a predominance of germinal center (GC) B cells and increased numbers of CD11c+ DCs localized beneath the epithelium and within the GC T-cell area. We detected microfold cells distributed throughout the NALT epithelial cell layer and DC dendrites extending into the nasal cavity, which could potentially function in luminal CS antigen uptake. Flagellin-modified CS taken up by DCs in vitro was initially localized within intracellular vesicles followed by a cytosolic distribution. Vaccine modifications to enhance delivery to the NALT and specifically target NALT antigen-presenting cell populations will advance development of an efficacious needle-free vaccine for the 40% of the world's population at risk of malaria.
Collapse
|
88
|
López-Yglesias AH, Zhao X, Quarles EK, Lai MA, VandenBos T, Strong RK, Smith KD. Flagellin induces antibody responses through a TLR5- and inflammasome-independent pathway. THE JOURNAL OF IMMUNOLOGY 2014; 192:1587-96. [PMID: 24442437 DOI: 10.4049/jimmunol.1301893] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Flagellin is a potent immunogen that activates the innate immune system via TLR5 and Naip5/6, and generates strong T and B cell responses. The adaptor protein MyD88 is critical for signaling by TLR5, as well as IL-1Rs and IL-18Rs, major downstream mediators of the Naip5/6 Nlrc4-inflammasome. In this study, we define roles of known flagellin receptors and MyD88 in Ab responses generated toward flagellin. We used mice genetically deficient in flagellin recognition pathways to characterize innate immune components that regulate isotype-specific Ab responses. Using purified flagellin from Salmonella, we dissected the contribution of innate flagellin recognition pathways to promote Ab responses toward flagellin and coadministered OVA in C57BL/6 mice. We demonstrate IgG2c responses toward flagellin were TLR5 and inflammasome dependent; IgG1 was the dominant isotype and partially TLR5 and inflammasome dependent. Our data indicate a substantial flagellin-specific IgG1 response was induced through a TLR5-, inflammasome-, and MyD88-independent pathway. IgA anti-FliC responses were TLR5 and MyD88 dependent and caspase-1 independent. Unlike C57BL/6 mice, flagellin-immunized A/J mice induced codominant IgG1 and IgG2a responses. Furthermore, MyD88-independent, flagellin-induced Ab responses were even more pronounced in A/J MyD88(-/-) mice, and IgA anti-FliC responses were suppressed by MyD88. Flagellin also worked as an adjuvant toward coadministered OVA, but it only promoted IgG1 anti-OVA responses. Our results demonstrate that a novel pathway for flagellin recognition contributes to Ab production. Characterization of this pathway will be useful for understanding immunity to flagellin and the rationale design of flagellin-based vaccines.
Collapse
|
89
|
Abstract
The challenges in successful vaccination against influenza using conventional approaches lie in their variable efficacy in different age populations, the antigenic variability of the circulating virus, and the production and manufacturing limitations to ensure safe, timely, and adequate supply of vaccine. The conventional influenza vaccine platform is based on stimulating immunity against the major neutralizing antibody target, hemagglutinin (HA), by virus attenuation or inactivation. Improvements to this conventional system have focused primarily on improving production and immunogenicity. Cell culture, reverse genetics, and baculovirus expression technology allow for safe and scalable production, while adjuvants, dose variation, and alternate routes of delivery aim to improve vaccine immunogenicity. Fundamentally different approaches that are currently under development hope to signal new generations of influenza vaccines. Such approaches target nonvariable regions of antigenic proteins, with the idea of stimulating cross-protective antibodies and thus creating a "universal" influenza vaccine. While such approaches have obvious benefits, there are many hurdles yet to clear. Here, we discuss the process and challenges of the current influenza vaccine platform as well as new approaches that are being investigated based on the same antigenic target and newer technologies based on different antigenic targets.
Collapse
|
90
|
Atif SM, Uematsu S, Akira S, McSorley SJ. CD103-CD11b+ dendritic cells regulate the sensitivity of CD4 T-cell responses to bacterial flagellin. Mucosal Immunol 2014; 7:68-77. [PMID: 23632327 PMCID: PMC4032068 DOI: 10.1038/mi.2013.25] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2012] [Accepted: 03/22/2013] [Indexed: 02/04/2023]
Abstract
Toll-like receptor 5 (TLR5) has been widely studied in an inflammatory context, but the effect of TLR5 on the adaptive response to bacterial flagellin has received considerably less attention. Here, we demonstrate that TLR5 expression by dendritic cells (DCs) allows a 1,000-fold enhancement of T-cell sensitivity to flagellin, and this enhancement did not require the expression of NLRC4 or Myd88. The effect of TLR5 on CD4 T-cell sensitivity was independent of the adjuvant effect of flagellin and TLR5 ligation did not alter the sensitivity of ovalbumin (OVA)-specific T cells to OVA. In the spleen, the exquisite T-cell sensitivity to flagellin was regulated by CD4-CD8α- DCs and was blocked by a monoclonal antibody to TLR5. In the mesenteric lymph nodes, flagellin-specific T-cell activation was regulated by a population of CD103-CD11b+ DCs. Thus, TLR5 expression by mucosal and systemic DC subsets controls the sensitivity of the adaptive immune response to flagellated pathogens.
Collapse
Affiliation(s)
- Shaikh M. Atif
- Center for Comparative Medicine, Department of Anatomy, Physiology, and Cell Biology, UC Davis, Davis, CA 95616.,Correspondence should be addressed to: Shaikh M. Atif, Tel: (530) 752 5032, Fax: (530) 752 7914 and Stephen McSorley, Tel: (530) 752 3414, Fax: (530) 752 7914,
| | - Satoshi Uematsu
- Laboratory of Host Defense, WPI Immunology Frontier Research Center, Osaka University Suita Osaka, 565-0871, Japan
| | - Shizuo Akira
- Laboratory of Host Defense, WPI Immunology Frontier Research Center, Osaka University Suita Osaka, 565-0871, Japan
| | - Stephen J. McSorley
- Center for Comparative Medicine, Department of Anatomy, Physiology, and Cell Biology, UC Davis, Davis, CA 95616.,Correspondence should be addressed to: Shaikh M. Atif, Tel: (530) 752 5032, Fax: (530) 752 7914 and Stephen McSorley, Tel: (530) 752 3414, Fax: (530) 752 7914,
| |
Collapse
|
91
|
van Els C, Mjaaland S, Næss L, Sarkadi J, Gonczol E, Smith Korsholm K, Hansen J, de Jonge J, Kersten G, Warner J, Semper A, Kruiswijk C, Oftung F. Fast vaccine design and development based on correlates of protection (COPs). Hum Vaccin Immunother 2014; 10:1935-48. [PMID: 25424803 PMCID: PMC4186026 DOI: 10.4161/hv.28639] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Revised: 03/14/2014] [Accepted: 03/24/2014] [Indexed: 01/02/2023] Open
Abstract
New and reemerging infectious diseases call for innovative and efficient control strategies of which fast vaccine design and development represent an important element. In emergency situations, when time is limited, identification and use of correlates of protection (COPs) may play a key role as a strategic tool for accelerated vaccine design, testing, and licensure. We propose that general rules for COP-based vaccine design can be extracted from the existing knowledge of protective immune responses against a large spectrum of relevant viral and bacterial pathogens. Herein, we focus on the applicability of this approach by reviewing the established and up-coming COPs for influenza in the context of traditional and a wide array of new vaccine concepts. The lessons learnt from this field may be applied more generally to COP-based accelerated vaccine design for emerging infections.
Collapse
Affiliation(s)
- Cécile van Els
- National Institute for Public Health and the Environment; Bilthoven, the Netherlands
| | | | - Lisbeth Næss
- Norwegian Institute of Public Health; Oslo, Norway
| | - Julia Sarkadi
- National Center for Epidemiology (NCE); Budapest, Hungary
| | - Eva Gonczol
- National Center for Epidemiology (NCE); Budapest, Hungary
| | | | - Jon Hansen
- Statens Serum Institut; Copenhagen, Denmark
| | - Jørgen de Jonge
- National Institute for Public Health and the Environment; Bilthoven, the Netherlands
| | - Gideon Kersten
- Institute for Translational Vaccinology; Bilthoven, the Netherlands
- Leiden Academic Center for Drug Research; University of Leiden; The Netherlands
| | | | | | - Corine Kruiswijk
- Institute for Translational Vaccinology; Bilthoven, the Netherlands
| | | |
Collapse
|
92
|
Lee YT, Kim KH, Ko EJ, Lee YN, Kim MC, Kwon YM, Tang Y, Cho MK, Lee YJ, Kang SM. New vaccines against influenza virus. Clin Exp Vaccine Res 2013; 3:12-28. [PMID: 24427759 PMCID: PMC3890446 DOI: 10.7774/cevr.2014.3.1.12] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2013] [Revised: 11/15/2013] [Accepted: 11/20/2013] [Indexed: 12/23/2022] Open
Abstract
Vaccination is one of the most effective and cost-benefit interventions that prevent the mortality and reduce morbidity from infectious pathogens. However, the licensed influenza vaccine induces strain-specific immunity and must be updated annually based on predicted strains that will circulate in the upcoming season. Influenza virus still causes significant health problems worldwide due to the low vaccine efficacy from unexpected outbreaks of next epidemic strains or the emergence of pandemic viruses. Current influenza vaccines are based on immunity to the hemagglutinin antigen that is highly variable among different influenza viruses circulating in humans and animals. Several scientific advances have been endeavored to develop universal vaccines that will induce broad protection. Universal vaccines have been focused on regions of viral proteins that are highly conserved across different virus subtypes. The strategies of universal vaccines include the matrix 2 protein, the hemagglutinin HA2 stalk domain, and T cell-based multivalent antigens. Supplemented and/or adjuvanted vaccination in combination with universal target antigenic vaccines would have much promise. This review summarizes encouraging scientific advances in the field with a focus on novel vaccine designs.
Collapse
Affiliation(s)
- Young-Tae Lee
- Center for Inflammation, Immunity & Infection, and Department of Biology, Georgia State University, Atlanta, GA, USA
| | - Ki-Hye Kim
- Center for Inflammation, Immunity & Infection, and Department of Biology, Georgia State University, Atlanta, GA, USA
| | - Eun-Ju Ko
- Center for Inflammation, Immunity & Infection, and Department of Biology, Georgia State University, Atlanta, GA, USA
| | - Yu-Na Lee
- Center for Inflammation, Immunity & Infection, and Department of Biology, Georgia State University, Atlanta, GA, USA
| | - Min-Chul Kim
- Animal and Plant Quarantine Agency, Anyang, Korea
| | - Young-Man Kwon
- Center for Inflammation, Immunity & Infection, and Department of Biology, Georgia State University, Atlanta, GA, USA
| | - Yinghua Tang
- Center for Inflammation, Immunity & Infection, and Department of Biology, Georgia State University, Atlanta, GA, USA
| | - Min-Kyoung Cho
- Center for Inflammation, Immunity & Infection, and Department of Biology, Georgia State University, Atlanta, GA, USA
| | | | - Sang-Moo Kang
- Center for Inflammation, Immunity & Infection, and Department of Biology, Georgia State University, Atlanta, GA, USA
| |
Collapse
|
93
|
Carapau D, Mitchell R, Nacer A, Shaw A, Othoro C, Frevert U, Nardin E. Protective humoral immunity elicited by a needle-free malaria vaccine comprised of a chimeric Plasmodium falciparum circumsporozoite protein and a Toll-like receptor 5 agonist, flagellin. Infect Immun 2013; 81:4350-62. [PMID: 24042110 PMCID: PMC3837993 DOI: 10.1128/iai.00263-13] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Accepted: 09/05/2013] [Indexed: 12/31/2022] Open
Abstract
Immunization with Plasmodium sporozoites can elicit high levels of sterile immunity, and neutralizing antibodies from protected hosts are known to target the repeat region of the circumsporozoite (CS) protein on the parasite surface. CS-based subunit vaccines have been hampered by suboptimal immunogenicity and the requirement for strong adjuvants to elicit effective humoral immunity. Pathogen-associated molecular patterns (PAMPs) that signal through Toll-like receptors (TLRs) can function as potent adjuvants for innate and adaptive immunity. We examined the immunogenicity of recombinant proteins containing a TLR5 agonist, flagellin, and either full-length or selected epitopes of the Plasmodium falciparum CS protein. Mice immunized with either of the flagellin-modified CS constructs, administered intranasally (i.n.) or subcutaneously (s.c.), developed similar levels of malaria-specific IgG1 antibody and interleukin-5 (IL-5)-producing T cells. Importantly, immunization via the i.n. but not the s.c. route elicited sporozoite neutralizing antibodies capable of inhibiting >90% of sporozoite invasion in vitro and in vivo, as measured using a transgenic rodent parasite expressing P. falciparum CS repeats. These findings demonstrate that functional sporozoite neutralizing antibody can be elicited by i.n. immunization with a flagellin-modified P. falciparum CS protein and raise the potential of a scalable, safe, needle-free vaccine for the 40% of the world's population at risk of malaria.
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Adjuvants, Immunologic/pharmacology
- Administration, Intranasal
- Animals
- Antibodies, Protozoan/immunology
- Cells, Cultured
- Dendritic Cells/immunology
- Epitopes, T-Lymphocyte/immunology
- Flagellin/immunology
- Humans
- Immunity, Humoral/immunology
- Immunization
- Immunoglobulin G/biosynthesis
- Immunoglobulin G/immunology
- Interleukin-5/biosynthesis
- Malaria Vaccines/administration & dosage
- Malaria Vaccines/immunology
- Malaria, Falciparum/immunology
- Malaria, Falciparum/parasitology
- Malaria, Falciparum/prevention & control
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Plasmodium falciparum/immunology
- Protozoan Proteins/administration & dosage
- Protozoan Proteins/immunology
- Recombinant Proteins/immunology
- Sporozoites/immunology
- Toll-Like Receptor 5/agonists
- Vaccines, Subunit/immunology
Collapse
Affiliation(s)
- Daniel Carapau
- Department of Microbiology, Division of Medical Parasitology, New York University School of Medicine, New York, New York, USA
| | - Robert Mitchell
- Department of Microbiology, Division of Medical Parasitology, New York University School of Medicine, New York, New York, USA
| | - Adéla Nacer
- Department of Microbiology, Division of Medical Parasitology, New York University School of Medicine, New York, New York, USA
| | - Alan Shaw
- Vaxinnate Corporation, Cranbury, New Jersey, USA
| | - Caroline Othoro
- Department of Microbiology, Division of Medical Parasitology, New York University School of Medicine, New York, New York, USA
| | - Ute Frevert
- Department of Microbiology, Division of Medical Parasitology, New York University School of Medicine, New York, New York, USA
| | - Elizabeth Nardin
- Department of Microbiology, Division of Medical Parasitology, New York University School of Medicine, New York, New York, USA
| |
Collapse
|
94
|
Shaw AC, Goldstein DR, Montgomery RR. Age-dependent dysregulation of innate immunity. Nat Rev Immunol 2013; 13:875-87. [PMID: 24157572 DOI: 10.1038/nri3547] [Citation(s) in RCA: 737] [Impact Index Per Article: 67.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
As we age, the innate immune system becomes dysregulated and is characterized by persistent inflammatory responses that involve multiple immune and non-immune cell types and that vary depending on the cell activation state and tissue context. This ageing-associated basal inflammation, particularly in humans, is thought to be induced by several factors, including the reactivation of latent viral infections and the release of endogenous damage-associated ligands of pattern recognition receptors (PRRs). Innate immune cell functions that are required to respond to pathogens or vaccines, such as cell migration and PRR signalling, are also impaired in aged individuals. This immune dysregulation may affect conditions associated with chronic inflammation, such as atherosclerosis and Alzheimer's disease.
Collapse
Affiliation(s)
- Albert C Shaw
- Section of Infectious Diseases, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut 06520, USA
| | | | | |
Collapse
|
95
|
De Gregorio E, Caproni E, Ulmer JB. Vaccine adjuvants: mode of action. Front Immunol 2013; 4:214. [PMID: 23914187 PMCID: PMC3728558 DOI: 10.3389/fimmu.2013.00214] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Accepted: 07/12/2013] [Indexed: 12/12/2022] Open
Abstract
Vaccines were first introduced more than 200 years ago and have since played a key role in the reduction of morbidity and mortality caused by infectious diseases. Many of the safest and most effective vaccines in use today are based on attenuated live viruses, as they mimic a live infection without causing disease. However, it is not always practical to take this approach, such as when it may not be safe to do so (e.g., for viruses that cause chronic infections such as HIV) or may not be feasible to manufacture (e.g., for viruses that do not grow well in cell culture such as HCV). In addition, it may preferable in some cases to target immune responses toward specific antigens from the pathogen, rather than the entirety of the genome. In these cases, subunit vaccines consisting of antigens purified from the pathogen or produced by recombinant DNA technology are being developed. However, highly purified proteins are typically not inherently immunogenic, as they usually lack the means to directly stimulate the innate immune system, and often require the addition of adjuvants to enhance vaccine potency. Despite more than a century of human use, only a few adjuvants are licensed today. However many adjuvants have been tested in humans and are in advanced stages of development. Much of the early work on adjuvants discovery and development was empirical producing safe and effective products, but without a clear understanding of how they worked. Recent insight into the functioning of the innate immune system has demonstrated its important role in triggering and shaping the adaptive immune response to vaccines.
Collapse
|
96
|
Knudsen ML, Johansson DX, Kostic L, Nordström EKL, Tegerstedt K, Pasetto A, Applequist SE, Ljungberg K, Sirard JC, Liljeström P. The adjuvant activity of alphavirus replicons is enhanced by incorporating the microbial molecule flagellin into the replicon. PLoS One 2013; 8:e65964. [PMID: 23785460 PMCID: PMC3681802 DOI: 10.1371/journal.pone.0065964] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2013] [Accepted: 04/30/2013] [Indexed: 11/23/2022] Open
Abstract
Ligands of pattern recognition receptors (PRRs) including Toll-like receptors (TLRs) stimulate innate and adaptive immune responses and are considered as potent adjuvants. Combinations of ligands might act in synergy to induce stronger and broader immune responses compared to stand-alone ligands. Alphaviruses stimulate endosomal TLRs 3, 7 and 8 as well as the cytoplasmic PRR MDA-5, resulting in induction of a strong type I interferon (IFN) response. Bacterial flagellin stimulates TLR5 and when delivered intracellularly the cytosolic PRR NLRC4, leading to secretion of proinflammatory cytokines. Both alphaviruses and flagellin have independently been shown to act as adjuvants for antigen-specific antibody responses. Here, we hypothesized that alphavirus and flagellin would act in synergy when combined. We therefore cloned the Salmonella Typhimurium flagellin (FliC) gene into an alphavirus replicon and assessed its adjuvant activity on the antibody response against co-administered antigen. In mice immunized with recombinant alphavirus, antibody responses were greatly enhanced compared to soluble FliC or control alphavirus. Both IgG1 and IgG2a/c responses were increased, indicating an enhancement of both Th1 and Th2 type responses. The adjuvant activity of FliC-expressing alphavirus was diminished but not abolished in the absence of TLR5 or type I IFN signaling, suggesting the contribution of several signaling pathways and some synergistic and redundant activity of its components. Thus, we have created a recombinant adjuvant that stimulates multiple signaling pathways of innate immunity resulting in a strong and broad antibody response.
Collapse
Affiliation(s)
- Maria L Knudsen
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
97
|
Lefebvre JS, Haynes L. Vaccine strategies to enhance immune responses in the aged. Curr Opin Immunol 2013; 25:523-8. [PMID: 23764092 DOI: 10.1016/j.coi.2013.05.014] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Revised: 05/20/2013] [Accepted: 05/21/2013] [Indexed: 12/20/2022]
Abstract
The elderly population is more susceptible to infections with higher risks of morbidity and mortality. This is caused by the accumulation of immune defects with aging. The best way to protect people against infections is vaccination. Unfortunately, the same immune defects that render the elderly susceptible to infectious diseases also prevent the development of protective immunity following immunization. A good example of this is the influenza vaccine that only protects between 40 and 60% of the vaccinees over 65 years. In the past decade, tremendous efforts have been put toward improving the influenza vaccine for the elderly. We therefore use this example to present various strategies employed to overcome these age-associated immune defects and hence make vaccines more efficacious for the aged.
Collapse
Affiliation(s)
- Julie S Lefebvre
- Trudeau Institute, 154 Algonquin Avenue, Saranac Lake, NY 12983, USA
| | | |
Collapse
|
98
|
Noah DL, Noah JW. Adapting global influenza management strategies to address emerging viruses. Am J Physiol Lung Cell Mol Physiol 2013; 305:L108-17. [PMID: 23709619 DOI: 10.1152/ajplung.00105.2013] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Death by respiratory complications from influenza infections continues to be a major global health concern. Antiviral drugs are widely available for therapy and prophylaxis, but viral mutations have resulted in resistance that threatens to reduce the long-term utility of approved antivirals. Vaccination is the best method for controlling influenza, but vaccine strategies are blunted by virus antigenic drift and shift. Genetic shift in particular has led to four pandemics in the last century, which have prompted the development of efficient global surveillance and vaccination programs. Although the influenza pandemic of 2009 emphasized the need for the rapid standardization of global surveillance methods and the preparation and dissemination of global assay standards for improved reporting and diagnostic tools, outbreaks of novel influenza strains continue to occur, and current efforts must be enhanced by aggressive public education programs to promote increased vaccination rates in the global population. Recently, a novel H7N9 avian influenza virus with potential to become a pandemic strain emerged in China and was transmitted from animals to humans with a demonstrated >20% mortality rate. Sporadic outbreaks of highly lethal avian virus strains have already increased public awareness and altered annual vaccine production strategies to prevent the natural adaption of this virus to human-to-human transmission. Additional strategies for combating influenza include advancement of new antivirals for unexploited viral or host cellular targets; novel adjuvants and alternate vaccine delivery systems; and development of universal protein, DNA, or multivalent vaccines designed to increase immune responsiveness and enhance public health response times.
Collapse
Affiliation(s)
- Diana L Noah
- Southern Research Institute, Birmingham, AL 35205, USA
| | | |
Collapse
|
99
|
Yan H. Salivary IgA enhancement strategy for development of a nasal-spray anti-caries mucosal vaccine. SCIENCE CHINA-LIFE SCIENCES 2013; 56:406-13. [PMID: 23633072 DOI: 10.1007/s11427-013-4473-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2013] [Accepted: 03/21/2013] [Indexed: 01/09/2023]
Abstract
Dental caries remains one of the most common global chronic diseases caused by Streptococcus mutans, which is prevalent all over the world. The caries prevalence of children aged between 5-6 years old in China is still in very high rate. A potent and effective anti-caries vaccine has long been expected for caries prevention but no vaccines have been brought to market till now mainly due to the low ability to induce and maintain protective antibody in oral fluids. This review will give a brief historical retrospect on study of dental caries and pathogenesis, effective targets for anti-caries vaccines, oral immune system and immunization against dental caries. Then, salivary IgA antibodies and the protective responses are discussed in the context of the ontogeny of mucosal immunity to indigenous oral streptococcal. The methods and advancement for induction of specific anti-caries salivary sIgA antibodies and enhancement of specific anti-caries salivary sIgA antibodies by intranasal immunization with a safe effective mucosal adjuvant are described. The progress in the enhancement of salivary sIgA antibodies and anti-caries protection by intranasal immunization with flagellin-PAc fusion protein will be highlighted. Finally, some of the main strategies that have been used for successful mucosal vaccination of caries vaccine are reviewed, followed by discussion of the mucosal adjuvant choice for achieving protective immunity at oral mucosal membranes for development of a nasal-spray or nasal-drop anti-caries vaccine for human.
Collapse
Affiliation(s)
- Huimin Yan
- Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China.
| |
Collapse
|
100
|
Kang SM, Kim MC, Compans RW. Virus-like particles as universal influenza vaccines. Expert Rev Vaccines 2013; 11:995-1007. [PMID: 23002980 DOI: 10.1586/erv.12.70] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Current influenza vaccines are primarily targeted to induce immunity to the influenza virus strain-specific hemagglutinin antigen and are not effective in controlling outbreaks of new pandemic viruses. An approach for developing universal vaccines is to present highly conserved antigenic epitopes in an immunogenic conformation such as virus-like particles (VLPs) together with an adjuvant to enhance the vaccine immunogenicity. In this review, the authors focus on conserved antigenic targets and molecular adjuvants that were presented in VLPs. Conserved antigenic targets that include the hemagglutinin stalk domain, the external domain of influenza M2 and neuraminidase are discussed in addition to molecular adjuvants that are engineered to be incorporated into VLPs in a membrane-anchored form.
Collapse
Affiliation(s)
- Sang-Moo Kang
- Center for Inflammation, Immunity & Infection, and Department of Biology, Georgia State University, Atlanta, GA 30303, USA.
| | | | | |
Collapse
|