1
|
Sabbaghi A, Malek M, Abdolahi S, Miri SM, Alizadeh L, Samadi M, Mohebbi SR, Ghaemi A. A formulated poly (I:C)/CCL21 as an effective mucosal adjuvant for gamma-irradiated influenza vaccine. Virol J 2021; 18:201. [PMID: 34627297 PMCID: PMC8501930 DOI: 10.1186/s12985-021-01672-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 09/23/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Several studies on gamma-irradiated influenza A virus (γ-Flu) have revealed its superior efficacy for inducing homologous and heterologous virus-specific immunity. However, many inactivated vaccines, notably in nasal delivery, require adjuvants to increase the quality and magnitude of vaccine responses. METHODS To illustrate the impacts of co-administration of the gamma-irradiated H1N1 vaccine with poly (I:C) and recombinant murine CCL21, either alone or in combination with each other, as adjuvants on the vaccine potency, mice were inoculated intranasally 3 times at one-week interval with γ-Flu alone or with any of the three adjuvant combinations and then challenged with a high lethal dose (10 LD50) of A/PR/8/34 (H1N1) influenza virus. Virus-specific humoral, mucosal, and cell-mediated immunity, as well as cytokine profiles in the spleen (IFN-γ, IL-12, and IL-4), and in the lung homogenates (IL-6 and IL-10) were measured by ELISA. The proliferative response of restimulated splenocytes was also determined by MTT assay. RESULTS The findings showed that the co-delivery of the γ-Flu vaccine and CCL21 or Poly (I:C) significantly increased the vaccine immunogenicity compared to the non-adjuvanted vaccine, associated with more potent protection following challenge infection. However, the mice given a combination of CCL21 with poly (I:C) had strong antibody- and cell-mediated immunity, which were considerably higher than responses of mice receiving the γ-Flu vaccine with each adjuvant separately. This combination also reduced inflammatory mediator levels (notably IL-10) in lung homogenate samples. CONCLUSIONS The results indicate that adjuvantation with the CCL21 and poly (I:C) can successfully induce vigorous vaccine-mediated protection, suggesting a robust propensity for CCL21 plus poly (I:C) as a potent mucosal adjuvant.
Collapse
Affiliation(s)
- Ailar Sabbaghi
- Department of Influenza and Other Respiratory Viruses, Pasteur Institute of Iran, P.O.Box: 1316943551, Tehran, Iran
| | - Masoud Malek
- Department of Microbiology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Sara Abdolahi
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran
| | - Seyed Mohammad Miri
- Department of Influenza and Other Respiratory Viruses, Pasteur Institute of Iran, P.O.Box: 1316943551, Tehran, Iran
| | - Leila Alizadeh
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran
| | - Mehdi Samadi
- Department of Medical Virology, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Reza Mohebbi
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amir Ghaemi
- Department of Influenza and Other Respiratory Viruses, Pasteur Institute of Iran, P.O.Box: 1316943551, Tehran, Iran.
| |
Collapse
|
2
|
Behbahani M, Moradi M, Mohabatkar H. In silico design of a multi-epitope peptide construct as a potential vaccine candidate for Influenza A based on neuraminidase protein. In Silico Pharmacol 2021; 9:36. [PMID: 33987075 PMCID: PMC8112742 DOI: 10.1007/s40203-021-00095-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 04/30/2021] [Indexed: 01/25/2023] Open
Abstract
Designing an effective vaccine against different subtypes of Influenza A virus is a critical issue in the field of medical biotechnology. At the current study, a novel potential multi-epitope vaccine candidate based on the neuraminidase proteins for seven subtypes of Influenza virus was designed, using the in silico approach. Potential linear B-cell and T-cell binding epitopes from each neuraminidase protein (N1, N2, N3, N4, N6, N7, N8) were predicted by in silico tools of epitope prediction. The selected epitopes were joined by three different linkers, and physicochemical properties, toxicity, and allergenecity were investigated. The final multi-epitope construct was modeled using GalaxyWEB server, and the molecular interactions with immune receptors were investigated and the immune response simulation assay was performed. A multi-epitope construct with GPGPGPG linker with the lowest allergenicity and highest stability was selected. The molecular docking assay indicated the interactions with immune system receptors, including HLA1, HLA2, and TLR-3. Immune response simulation detected both humoral and cellular response, including the elevated count of B-cells, T-cell, and Nk-cells.
Collapse
Affiliation(s)
- Mandana Behbahani
- Department of Biotechnology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Mohammad Moradi
- Department of Biotechnology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Hassan Mohabatkar
- Department of Biotechnology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| |
Collapse
|
3
|
Boukhvalova MS, Mortensen E, Mbaye A, McKay J, Blanco JCG. Effect of aging on immunogenicity and efficacy of inactivated influenza vaccines in cotton rats Sigmodon hispidus. Hum Vaccin Immunother 2020; 17:133-145. [PMID: 32614696 PMCID: PMC7872023 DOI: 10.1080/21645515.2020.1766334] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Inactivated influenza vaccines are known to be less immunogenic in human elderly in regards to serologic antibody response induced by vaccination. Accumulating evidence, however, points to a comparable effectiveness of influenza vaccines in the young and the elderly individuals. In the current study, we assessed immunogenicity and effectiveness of trivalent inactivated vaccine FluLaval in young and aged cotton rats Sigmodon hispidus and found that while serologic response to immunization was indeed reduced in older animals, comparable protection against influenza infection was afforded by prime-boost vaccination in both young and aged cotton rats. Both hemagglutination inhibition (HAI) titers and seroconversion rates were lower in the aged animals compared to the young ones. Reduction of viral load in the lung and nose, however, was comparable between young and aged animals vaccinated twice. One-time immunization with FluLaval was less efficacious at protecting the nose of aged animals, indicating that boosting of preexisting immunity can be particularly important for nasal protection in the elderly. Coincidentally, a one-time immunization with FluLaval had a detrimental effect on pulmonary pathology in the young animals, suggesting that boosting of immunity is essential for the young as well. Overall, these results suggest that reduced antibody response to and sufficient efficacy of influenza vaccines in the elderly are not two irreconcilable phenomena and that incomplete immunity to influenza can be detrimental at any age.
Collapse
|
4
|
Pushko P, Tretyakova I. Influenza Virus Like Particles (VLPs): Opportunities for H7N9 Vaccine Development. Viruses 2020; 12:v12050518. [PMID: 32397182 PMCID: PMC7291233 DOI: 10.3390/v12050518] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 04/21/2020] [Accepted: 04/27/2020] [Indexed: 01/21/2023] Open
Abstract
In the midst of the ongoing COVID-19 coronavirus pandemic, influenza virus remains a major threat to public health due to its potential to cause epidemics and pandemics with significant human mortality. Cases of H7N9 human infections emerged in eastern China in 2013 and immediately raised pandemic concerns as historically, pandemics were caused by the introduction of new subtypes into immunologically naïve human populations. Highly pathogenic H7N9 cases with severe disease were reported recently, indicating the continuing public health threat and the need for a prophylactic vaccine. Here we review the development of recombinant influenza virus-like particles (VLPs) as vaccines against H7N9 virus. Several approaches to vaccine development are reviewed including the expression of VLPs in mammalian, plant and insect cell expression systems. Although considerable progress has been achieved, including demonstration of safety and immunogenicity of H7N9 VLPs in the human clinical trials, the remaining challenges need to be addressed. These challenges include improvements to the manufacturing processes, as well as enhancements to immunogenicity in order to elicit protective immunity to multiple variants and subtypes of influenza virus.
Collapse
|
5
|
A Bivalent, Spherical Virus-Like Particle Vaccine Enhances Breadth of Immune Responses against Pathogenic Ebola Viruses in Rhesus Macaques. J Virol 2020; 94:JVI.01884-19. [PMID: 32075939 DOI: 10.1128/jvi.01884-19] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 02/13/2020] [Indexed: 12/16/2022] Open
Abstract
The 2013-2016 Ebola outbreak in West Africa led to accelerated efforts to develop vaccines against these highly virulent viruses. A live, recombinant vesicular stomatitis virus-based vaccine has been deployed in outbreak settings and appears highly effective. Vaccines based on replication-deficient adenovirus vectors either alone or in combination with a multivalent modified vaccinia Ankara (MVA) Ebola vaccine also appear promising and are progressing in clinical evaluation. However, the ability of current live vector-based approaches to protect against multiple pathogenic species of Ebola is not yet established, and eliciting durable responses may require additional booster vaccinations. Here, we report the development of a bivalent, spherical Ebola virus-like particle (VLP) vaccine that incorporates glycoproteins (GPs) from Zaire Ebola virus (EBOV) and Sudan Ebola virus (SUDV) and is designed to extend the breadth of immunity beyond EBOV. Immunization of rabbits with bivalent Ebola VLPs produced antibodies that neutralized all four pathogenic species of Ebola viruses and elicited antibody-dependent cell-mediated cytotoxicity (ADCC) responses against EBOV and SUDV. Vaccination of rhesus macaques with bivalent VLPs generated strong humoral immune responses, including high titers of binding, as well as neutralizing antibodies and ADCC responses. VLP vaccination led to a significant increase in the frequency of Ebola GP-specific CD4 and CD8 T cell responses. These results demonstrate that a novel bivalent Ebola VLP vaccine elicits strong humoral and cellular immune responses against pathogenic Ebola viruses and support further evaluation of this approach as a potential addition to Ebola vaccine development efforts.IMPORTANCE Ebola outbreaks result in significant morbidity and mortality in affected countries. Although several leading candidate Ebola vaccines have been developed and advanced in clinical testing, additional vaccine candidates may be needed to provide protection against different Ebola species and to extend the durability of protection. A novel approach demonstrated here is to express two genetically diverse glycoproteins on a spherical core, generating a vaccine that can broaden immune responses against known pathogenic Ebola viruses. This approach provides a new method to broaden and potentially extend protective immune responses against Ebola viruses.
Collapse
|
6
|
Schneider-Ohrum K, Snell Bennett A, Rajani GM, Hostetler L, Maynard SK, Lazzaro M, Cheng LI, O'Day T, Cayatte C. CD4 + T Cells Drive Lung Disease Enhancement Induced by Immunization with Suboptimal Doses of Respiratory Syncytial Virus Fusion Protein in the Mouse Model. J Virol 2019; 93:e00695-19. [PMID: 31092578 PMCID: PMC6639276 DOI: 10.1128/jvi.00695-19] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 05/08/2019] [Indexed: 12/23/2022] Open
Abstract
Respiratory syncytial virus (RSV) infection of seronegative children previously immunized with formalin-inactivated (FI) RSV has been associated with serious enhanced respiratory disease (ERD). The phenomenon was reproduced in the cotton rat and the mouse, and both preclinical models have been routinely used to evaluate the safety of new RSV vaccine candidates. More recently, we demonstrated that immunizations with suboptimal doses of the RSV fusion (F) antigen, in its post- or prefusion conformation, and in the presence of a Th1-biasing adjuvant, unexpectedly led to ERD in the cotton rat model. To assess if those observations are specific to the cotton rat and to elucidate the mechanism by which vaccination with low antigen doses can drive ERD post-RSV challenge, we evaluated RSV post-F antigen dose de-escalation in BALB/c mice in the presence of a Th1-biasing adjuvant. While decreasing antigen doses, we observed an increase in lung inflammation associated with an upregulation of proinflammatory cytokines. The amplitude of the lung histopathology was comparable to that of FI-RSV-induced ERD, confirming the observations made in the cotton rat. Importantly, depletion of CD4+ T cells prior to viral challenge completely abrogated ERD, preventing proinflammatory cytokine upregulation and the infiltration of T cells, neutrophils, eosinophils, and macrophages into the lung. Overall, low-antigen-dose-induced ERD resembles FI-RSV-induced ERD, except that the former appears in the absence of detectable levels of viral replication and in the context of a Th1-biased immune response. Taken together, our observations reinforce the recent concept that vaccines developed for RSV-naïve individuals should be systematically tested under suboptimal dosing conditions.IMPORTANCE RSV poses a significant health care burden and is the leading cause of serious lower-respiratory-tract infections in young children. A formalin-inactivated RSV vaccine developed in the 1960s not only showed a complete lack of efficacy against RSV infection but also induced severe lung disease enhancement in vaccinated children. Since then, establishing safety in preclinical models has been one of the major challenges to RSV vaccine development. We recently observed in the cotton rat model that suboptimal immunizations with RSV fusion protein could induce lung disease enhancement. In the present study, we extended suboptimal dosing evaluation to the mouse model. We confirmed the induction of lung disease enhancement by vaccinations with low antigen doses and dissected the associated immune mechanisms. Our results stress the need to evaluate suboptimal dosing for any new RSV vaccine candidate developed for seronegative infants.
Collapse
Affiliation(s)
| | - Angie Snell Bennett
- Department of Infectious Disease/Vaccines, MedImmune, Gaithersburg, Maryland, USA
| | | | - Leigh Hostetler
- Laboratory Animal Resources, MedImmune, Gaithersburg, Maryland, USA
| | - Sean K Maynard
- Department of Infectious Disease/Vaccines, MedImmune, Gaithersburg, Maryland, USA
| | - Michelle Lazzaro
- Department of Infectious Disease/Vaccines, MedImmune, Gaithersburg, Maryland, USA
| | - Lily I Cheng
- Pathology Department, MedImmune, Gaithersburg, Maryland, USA
| | - Terrence O'Day
- Statistical Sciences, MedImmune, Gaithersburg, Maryland, USA
| | - Corinne Cayatte
- Department of Infectious Disease/Vaccines, MedImmune, Gaithersburg, Maryland, USA
| |
Collapse
|
7
|
Calzas C, Chevalier C. Innovative Mucosal Vaccine Formulations Against Influenza A Virus Infections. Front Immunol 2019; 10:1605. [PMID: 31379823 PMCID: PMC6650573 DOI: 10.3389/fimmu.2019.01605] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 06/27/2019] [Indexed: 12/11/2022] Open
Abstract
Despite efforts made to develop efficient preventive strategies, infections with influenza A viruses (IAV) continue to cause serious clinical and economic problems. Current licensed human vaccines are mainly inactivated whole virus particles or split-virion administered via the parenteral route. These vaccines provide incomplete protection against IAV in high-risk groups and are poorly/not effective against the constant antigenic drift/shift occurring in circulating strains. Advances in mucosal vaccinology and in the understanding of the protective anti-influenza immune mechanisms suggest that intranasal immunization is a promising strategy to fight against IAV. To date, human mucosal anti-influenza vaccines consist of live attenuated strains administered intranasally, which elicit higher local humoral and cellular immune responses than conventional parenteral vaccines. However, because of inconsistent protective efficacy and safety concerns regarding the use of live viral strains, new vaccine candidates are urgently needed. To prime and induce potent and long-lived protective immune responses, mucosal vaccine formulations need to ensure the immunoavailability and the immunostimulating capacity of the vaccine antigen(s) at the mucosal surfaces, while being minimally reactogenic/toxic. The purpose of this review is to compile innovative delivery/adjuvant systems tested for intranasal administration of inactivated influenza vaccines, including micro/nanosized particulate carriers such as lipid-based particles, virus-like particles and polymers associated or not with immunopotentiatory molecules including microorganism-derived toxins, Toll-like receptor ligands and cytokines. The capacity of these vaccines to trigger specific mucosal and systemic humoral and cellular responses against IAV and their (cross)-protective potential are considered.
Collapse
Affiliation(s)
- Cynthia Calzas
- VIM, UR892, Equipe Virus Influenza, INRA, University PARIS-SACLAY, Jouy-en-Josas, France
| | - Christophe Chevalier
- VIM, UR892, Equipe Virus Influenza, INRA, University PARIS-SACLAY, Jouy-en-Josas, France
| |
Collapse
|
8
|
Pattinson DJ, Apte SH, Wibowo N, Chuan YP, Rivera-Hernandez T, Groves PL, Lua LH, Middelberg APJ, Doolan DL. Chimeric Murine Polyomavirus Virus-Like Particles Induce Plasmodium Antigen-Specific CD8 + T Cell and Antibody Responses. Front Cell Infect Microbiol 2019; 9:215. [PMID: 31275867 PMCID: PMC6593135 DOI: 10.3389/fcimb.2019.00215] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 06/03/2019] [Indexed: 12/28/2022] Open
Abstract
An effective vaccine against the Plasmodium parasite is likely to require the induction of robust antibody and T cell responses. Chimeric virus-like particles are an effective vaccine platform for induction of antibody responses, but their capacity to induce robust cellular responses and cell-mediated protection against pathogen challenge has not been established. To evaluate this, we produced chimeric constructs using the murine polyomavirus structural protein with surface-exposed CD8+ or CD4+ T cell or B cell repeat epitopes derived from the Plasmodium yoelii circumsporozoite protein, and assessed immunogenicity and protective capacity in a murine model. Robust CD8+ T cell responses were induced by immunization with the chimeric CD8+ T cell epitope virus-like particles, however CD4+ T cell responses were very low. The B cell chimeric construct induced robust antibody responses but there was no apparent synergy when T cell and B cell constructs were administered as a pool. A heterologous prime/boost regimen using plasmid DNA priming followed by a VLP boost was more effective than homologous VLP immunization for cellular immunity and protection. These data show that chimeric murine polyomavirus virus-like particles are a good platform for induction of CD8+ T cell responses as well as antibody responses.
Collapse
MESH Headings
- Animals
- Antibodies, Protozoan
- Antibody Formation/immunology
- Antigens, Protozoan/immunology
- B-Lymphocytes
- CD4-Positive T-Lymphocytes
- CD8-Positive T-Lymphocytes/immunology
- Disease Models, Animal
- Epitopes, B-Lymphocyte/immunology
- Epitopes, T-Lymphocyte/immunology
- Immunity, Cellular
- Immunization
- Immunization, Secondary
- Malaria Vaccines
- Mice
- Mice, Inbred BALB C
- Plasmodium yoelii
- Polyomavirus/genetics
- Polyomavirus/immunology
- Protozoan Proteins/immunology
- Vaccines, Virus-Like Particle/genetics
- Vaccines, Virus-Like Particle/immunology
Collapse
Affiliation(s)
- David J. Pattinson
- Infectious Diseases Programme, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| | - Simon H. Apte
- Infectious Diseases Programme, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Nani Wibowo
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, QLD, Australia
| | - Yap P. Chuan
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, QLD, Australia
| | - Tania Rivera-Hernandez
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, QLD, Australia
| | - Penny L. Groves
- Infectious Diseases Programme, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Linda H. Lua
- Protein Expression Facility, University of Queensland, Brisbane, QLD, Australia
| | - Anton P. J. Middelberg
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, QLD, Australia
| | - Denise L. Doolan
- Infectious Diseases Programme, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| |
Collapse
|
9
|
Liu J, Feng X, Chen Z, Yang X, Shen Z, Guo M, Deng F, Liu Y, Zhang H, Chen C. The adjuvant effect of C 60(OH) 22 nanoparticles promoting both humoral and cellular immune responses to HCV recombinant proteins. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2018; 97:753-759. [PMID: 30678964 DOI: 10.1016/j.msec.2018.12.088] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 12/13/2018] [Accepted: 12/25/2018] [Indexed: 12/11/2022]
Abstract
Hepatitis c virus (HCV) infection is one of major causes for chronic liver diseases worldwide and could lead to death. Development of effective HCV vaccines is a powerful auxiliary method of existing treatments. Adjuvants are necessary for modern vaccines to promote immune responses. Among the various nanomaterials that have been developed, multihydroxylated fullerene (C60(OH)22) has been proved as an efficient adjuvant for human immunodeficiency virus DNA vaccine. Here, we utilized three types of HCV recombinant proteins as antigens to investigate the activity of C60(OH)22 as a protein vaccine adjuvant. The proteins were carried by C60(OH)22 in a way of surface adsorption and self-assemble encapsulation. C60(OH)22 at a relatively low dose was sufficient to promote both humoral and cellular immune responses to HCV protein antigens and reduce the usage of antigen. These results demonstrated the positive adjuvant properties of C60(OH)22 when applied to protein vaccines.
Collapse
Affiliation(s)
- Jing Liu
- The College of Life Sciences, Northwest University, Xi'an 710069, China; CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
| | - Xiaoyan Feng
- Institute of Military Cognitive and Brain Sciences, Academy of Military Medical Sciences, Beijing 100850, China
| | - Zhiyun Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
| | - Xiqin Yang
- Institute of Military Cognitive and Brain Sciences, Academy of Military Medical Sciences, Beijing 100850, China
| | - Ziyi Shen
- The College of Life Sciences, Northwest University, Xi'an 710069, China
| | - Mengyu Guo
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
| | | | - Ying Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China.
| | - Heqiu Zhang
- Institute of Military Cognitive and Brain Sciences, Academy of Military Medical Sciences, Beijing 100850, China.
| | - Chunying Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China.
| |
Collapse
|
10
|
Martinez-Gil L, Goff PH, Tan GS. The Role of Self-Assembling Lipid Molecules in Vaccination. ADVANCES IN BIOMEMBRANES AND LIPID SELF-ASSEMBLY 2018. [PMCID: PMC7147077 DOI: 10.1016/bs.abl.2017.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The advent of vaccines represents one of the most significant advances in medical history. The protection provided by vaccines has greatly contributed in reducing the number of cases of infections and most notably to the eradication of small pox. A large number of new technologies and approaches in vaccine development are currently being investigated with the goal of providing the basis for the next generation of prophylactics against an ever-expanding list of emerging and reemerging pathogens. In this chapter, we will focus on the role of lipids and lipid self-assembling vesicles in new and promising vaccination approaches. We will start by describing how lipids can induce activation of the innate immune system and focus on some lipid-derived vaccine adjuvants. Next, we will review current lipid-based self-assembling particles used as vaccine platforms, specifically liposomes and virus-like particles, and how virus-like particles have facilitated research of highly pathogenic viruses such as Ebola.
Collapse
|
11
|
Cayatte C, Snell Bennett A, Rajani GM, Hostetler L, Maynard SK, Lazzaro M, McTamney P, Ren K, O’Day T, McCarthy MP, Schneider-Ohrum K. Inferior immunogenicity and efficacy of respiratory syncytial virus fusion protein-based subunit vaccine candidates in aged versus young mice. PLoS One 2017; 12:e0188708. [PMID: 29182682 PMCID: PMC5705161 DOI: 10.1371/journal.pone.0188708] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 11/10/2017] [Indexed: 12/17/2022] Open
Abstract
Respiratory syncytial virus (RSV) is recognized as an important cause of lower and upper respiratory tract infections in older adults, and a successful vaccine would substantially lower morbidity and mortality in this age group. Recently, two vaccine candidates based on soluble purified glycoprotein F (RSV F), either alone or adjuvanted with glucopyranosyl lipid A formulated in a stable emulsion (GLA-SE), failed to reach their primary endpoints in clinical efficacy studies, despite demonstrating the desired immunogenicity profile and efficacy in young rodent models. Here, one of the RSV F vaccine candidates (post-fusion conformation, RSV post-F), and a stabilized pre-fusion form of RSV F (RSV pre-F, DS-Cav1) were evaluated in aged BALB/c mice. Humoral and cellular immunogenicity elicited after immunization of naïve, aged mice was generally lower compared to young animals. In aged mice, RSV post-F vaccination without adjuvant poorly protected the respiratory tract from virus replication, and addition of GLA-SE only improved protection in the lungs, but not in nasal turbinates. RSV pre-F induced higher neutralizing antibody titers compared to RSV post-F (as previously reported) but interestingly, RSV F-specific CD8 T cell responses were lower compared to RSV post-F responses regardless of age. The vaccines were also tested in RSV seropositive aged mice, in which both antigen forms similarly boosted neutralizing antibody titers, although GLA-SE addition boosted neutralizing activity only in RSV pre-F immunized animals. Cell-mediated immune responses in the aged mice were only slightly boosted and well below levels induced in seronegative young mice. Taken together, the findings suggest that the vaccine candidates were not able to induce a strong anti-RSV immune response in recipient mice with an aged immune system, in agreement with recent human clinical trial results. Therefore, the aged mouse model could be a useful tool to evaluate improved vaccine candidates, targeted to prevent RSV disease in older adults.
Collapse
Affiliation(s)
- Corinne Cayatte
- Department of Infectious Diseases/Vaccines, MedImmune, Gaithersburg, Maryland, United States of America
- * E-mail:
| | - Angie Snell Bennett
- Department of Infectious Diseases/Vaccines, MedImmune, Gaithersburg, Maryland, United States of America
| | - Gaurav Manohar Rajani
- Department of Infectious Diseases/Vaccines, MedImmune, Gaithersburg, Maryland, United States of America
| | - Leigh Hostetler
- Laboratory Animal Resources, MedImmune, Gaithersburg, Maryland, United States of America
| | - Sean K. Maynard
- Department of Infectious Diseases/Vaccines, MedImmune, Gaithersburg, Maryland, United States of America
| | - Michelle Lazzaro
- Department of Infectious Diseases/Vaccines, MedImmune, Gaithersburg, Maryland, United States of America
| | - Patrick McTamney
- Department of Infectious Diseases/Vaccines, MedImmune, Gaithersburg, Maryland, United States of America
| | - Kuishu Ren
- Department of Infectious Diseases/Vaccines, MedImmune, Gaithersburg, Maryland, United States of America
| | - Terrence O’Day
- Department of Statistical Sciences, MedImmune, Gaithersburg, Maryland, United States of America
| | - Michael P. McCarthy
- Department of Infectious Diseases/Vaccines, MedImmune, Gaithersburg, Maryland, United States of America
| | - Kirsten Schneider-Ohrum
- Department of Infectious Diseases/Vaccines, MedImmune, Gaithersburg, Maryland, United States of America
| |
Collapse
|
12
|
Li Y, Xu YL, Lai YN, Liao SH, Liu N, Xu PP. Intranasal co-administration of 1,8-cineole with influenza vaccine provide cross-protection against influenza virus infection. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2017; 34:127-135. [PMID: 28899494 DOI: 10.1016/j.phymed.2017.08.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2017] [Revised: 07/05/2017] [Accepted: 08/15/2017] [Indexed: 05/16/2023]
Abstract
BACKGROUND Vaccination is the most efficient means for protection against influenza. However, the various vaccines have low efficacy to protect against pandemic strains because of antigenic drift and recombination of influenza virus. Adjuvant therapy is one of the attempts to improve influenza vaccine effective cross-protection against influenza virus infection. Our previous study confirmed that 1,8-cineole inhibits the NF-κB, reduces pro-inflammatory cytokines, and relieves the pathological changes of viral pneumonia in mice infected with influenza virus. HYPOTHESIS/PURPOSE 1,8-cineole, administered via intranasal (i.n.) route, may also have the capacity to be an adjuvant of the influenza vaccine. This study was designed to investigate the potential use of i.n. co-administration of 1,8-cineole, a major component of the Eucalyptus essential oils, with influenza vaccine and whether could provide cross-protection against influenza virus infection in a mouse model. STUDY DESIGN I.n. co-administration of 1,8-cineole in two doses (6.25 and 12.5 mg/kg) with influenza vaccine was investigated in a mouse model in order to see whether it could provide cross-protection against influenza virus infection. METHODS The mice were intranasally immunized three times at the 0, 7 and 14 day with vaccine containing 0.2 µg hemagglutinin (HA) and/or without 1,8-cineole. Seven days after the 3rd immunization dose, the mice were infected with 50 µl of 15 LD50 (50% mouse lethal dose) influenza virus A/FM/1/47 (H1N1). On day 6 post-infection, 10 mice per group were sacrificed to collect samples, to take the body weight and lung, and detect the viral load, pathological changes in the lungs and antibody, etc. The collected samples included blood serum and nasal lavage fluids. In addition, the survival experiments were carried out to investigate the survival of mice. RESULTS Mice i.n. inoculated with influenza vaccine and 12.5 mg/kg 1,8-cineole increased the production of influenza-specific serum immunoglobulin (Ig) G2a antibodies, stimulated mucosal secretive IgA (s-IgA) responses at the nasal cavity, improved the expression of respiratory tract intraepithelial lymphocytes (IELs) in the upper respiratory tract, and promoted dendritic cell (DC) maturation and the expression of co-stimulatory molecules cluster of differentiation (CD)40, CD80 and CD86 in peripheral blood. Importantly, mice that had received 1,8-cineole-supplemented influenza vaccine showed longer survival time, milder inflammation, less weight loss and mortality rate and lower lung index and viral titers compared to that of mice immunized a non-1,8-cineole-adjuvanted split vaccine. Thus, i.n. immunization with 1,8-cineole-adjuvanted vaccine induces a superior cross-protective immunity against infection with influenza than an inactivated vaccine only. CONCLUSION These results suggest that 1,8-cineole (12.5 mg/kg) has a cross-protection against influenza virus, co-administered with inactivated influenza viral antigen in a mouse model.
Collapse
Affiliation(s)
- Yun Li
- Institute of Tropical Medicine, Guangzhou University of Chinese Medicine, 12 Ji chang Rd., San Yuan li St., Bai Yun Dist., Guangzhou, PR China
| | - Yu-Ling Xu
- Guangzhou Health Vocational and Technical College, Guangzhou, PR China
| | - Yan-Ni Lai
- Institute of Tropical Medicine, Guangzhou University of Chinese Medicine, 12 Ji chang Rd., San Yuan li St., Bai Yun Dist., Guangzhou, PR China
| | - Shang-Hui Liao
- Institute of Tropical Medicine, Guangzhou University of Chinese Medicine, 12 Ji chang Rd., San Yuan li St., Bai Yun Dist., Guangzhou, PR China
| | - Ni Liu
- Institute of Tropical Medicine, Guangzhou University of Chinese Medicine, 12 Ji chang Rd., San Yuan li St., Bai Yun Dist., Guangzhou, PR China
| | - Pei-Ping Xu
- Institute of Tropical Medicine, Guangzhou University of Chinese Medicine, 12 Ji chang Rd., San Yuan li St., Bai Yun Dist., Guangzhou, PR China.
| |
Collapse
|
13
|
How Inflammasomes Inform Adaptive Immunity. J Mol Biol 2017; 430:217-237. [PMID: 28987733 DOI: 10.1016/j.jmb.2017.09.019] [Citation(s) in RCA: 101] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Revised: 09/27/2017] [Accepted: 09/28/2017] [Indexed: 02/07/2023]
Abstract
An immune response consists of a finely orchestrated interplay between initial recognition of potential microbial threats by the innate immune system and subsequent licensed adaptive immune neutralization. The initial recognition integrates environmental cues derived from pathogen-associated molecular patterns and cell-intrinsic damage-associated molecular patterns to contextualize the insult and inform a tailored adaptive response via T and B lymphocytes. While there are much data to support the role of transcriptional responses downstream of pattern recognition receptors in informing the adaptive immune response, markedly less attention has been paid to the role of post-translational responses to pathogen-associated molecular pattern and damage-associated molecular pattern recognition by the innate immune system, and how this may influence adaptive immunity. A well-characterized post-translational consequence of pattern recognition receptor signaling is the assembly of a multimeric signaling platform, termed the inflammasome, by members of the nucleotide-binding oligomerization domain (Nod), leucine-rich repeat-containing receptors (NLRs), and pyrin and HIN domain (PYHIN) families. Inflammasomes assemble in response to cytosolic perturbations, such as mitochondrial dysfunction and aberrant ion fluxes in the case of the canonical NLRP3 inflammasome or the presence of bacterial lipopolysaccharides in the case of the non-canonical inflammasome. Assembly of the inflammasome allows for the cleavage and activation of inflammatory caspases. These activated inflammatory caspases in turn cleave pro-form inflammatory cytokines into their mature bioactive species and lead to unconventional protein secretion and lytic cell death. In this review, we discuss evidence for inflammasome-mediated instruction and contextualization of infectious and sterile agents to the adaptive immune system.
Collapse
|
14
|
Cimica V, Galarza JM. Adjuvant formulations for virus-like particle (VLP) based vaccines. Clin Immunol 2017; 183:99-108. [PMID: 28780375 DOI: 10.1016/j.clim.2017.08.004] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 06/11/2017] [Accepted: 08/01/2017] [Indexed: 12/13/2022]
Abstract
The development of virus-like particle (VLP) technology has had an enormous impact on modern vaccinology. In order to optimize the efficacy and safety of VLP-based vaccines, adjuvants are included in most vaccine formulations. To date, most licensed VLP-based vaccines utilize the classic aluminum adjuvant compositions. Certain challenging pathogens and weak immune responder subjects may require further optimization of the adjuvant formulation to maximize the magnitude and duration of the protective immunity. Indeed, novel classes of adjuvants such as liposomes, agonists of pathogen recognition receptors, polymeric particles, emulsions, cytokines and bacterial toxins, can be used to further improve the immunostimulatory activity of a VLP-based vaccine. This review describes the current advances in adjuvant technology for VLP-based vaccines directed at viral diseases, and discusses the basic principles for designing adjuvant formulations for enhancing the vaccine immunogenicity.
Collapse
Affiliation(s)
- Velasco Cimica
- TechnoVax, Inc., 765 Old Saw Mill River Road, Tarrytown, NY 10591, United States
| | - Jose M Galarza
- TechnoVax, Inc., 765 Old Saw Mill River Road, Tarrytown, NY 10591, United States.
| |
Collapse
|
15
|
Van Hoeven N, Fox CB, Granger B, Evers T, Joshi SW, Nana GI, Evans SC, Lin S, Liang H, Liang L, Nakajima R, Felgner PL, Bowen RA, Marlenee N, Hartwig A, Baldwin SL, Coler RN, Tomai M, Elvecrog J, Reed SG, Carter D. A Formulated TLR7/8 Agonist is a Flexible, Highly Potent and Effective Adjuvant for Pandemic Influenza Vaccines. Sci Rep 2017; 7:46426. [PMID: 28429728 PMCID: PMC5399443 DOI: 10.1038/srep46426] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 03/20/2017] [Indexed: 11/30/2022] Open
Abstract
Since 1997, highly pathogenic avian influenza viruses of the H5N1 subtype have been transmitted from avian hosts to humans. The severity of H5N1 infection in humans, as well as the sporadic nature of H5N1 outbreaks, both geographically and temporally, make generation of an effective vaccine a global public health priority. An effective H5N1 vaccine must ultimately provide protection against viruses from diverse clades. Toll-like receptor (TLR) agonist adjuvant formulations have a demonstrated ability to broaden H5N1 vaccine responses in pre-clinical models. However, many of these agonist molecules have proven difficult to develop clinically. Here, we describe comprehensive adjuvant formulation development of the imidazoquinoline TLR-7/8 agonist 3M-052, in combination with H5N1 hemagglutinin (HA) based antigens. We find that 3M-052 in multiple formulations protects both mice and ferrets from lethal H5N1 homologous virus challenge. Furthermore, we conclusively demonstrate the ability of 3M-052 adjuvant formulations to broaden responses to H5N1 HA based antigens, and show that this broadening is functional using a heterologous lethal virus challenge in ferrets. Given the extensive clinical use of imidazoquinoline TLR agonists for other indications, these studies identify multiple adjuvant formulations which may be rapidly advanced into clinical trials in an H5N1 vaccine.
Collapse
Affiliation(s)
- Neal Van Hoeven
- Infectious Disease Research Institute, 1616 Eastlake Ave E., Seattle WA 98103, USA
| | - Christopher B Fox
- Infectious Disease Research Institute, 1616 Eastlake Ave E., Seattle WA 98103, USA
| | - Brian Granger
- Infectious Disease Research Institute, 1616 Eastlake Ave E., Seattle WA 98103, USA
| | - Tara Evers
- Infectious Disease Research Institute, 1616 Eastlake Ave E., Seattle WA 98103, USA
| | - Sharvari W Joshi
- Infectious Disease Research Institute, 1616 Eastlake Ave E., Seattle WA 98103, USA
| | - Ghislain I Nana
- Infectious Disease Research Institute, 1616 Eastlake Ave E., Seattle WA 98103, USA
| | - Sarah C Evans
- Infectious Disease Research Institute, 1616 Eastlake Ave E., Seattle WA 98103, USA
| | - Susan Lin
- Infectious Disease Research Institute, 1616 Eastlake Ave E., Seattle WA 98103, USA
| | - Hong Liang
- Infectious Disease Research Institute, 1616 Eastlake Ave E., Seattle WA 98103, USA
| | - Li Liang
- University of California Irvine, Department of Medicine, Irvine CA 92697, USA
| | - Rie Nakajima
- University of California Irvine, Department of Medicine, Irvine CA 92697, USA
| | - Philip L Felgner
- University of California Irvine, Department of Medicine, Irvine CA 92697, USA
| | - Richard A Bowen
- Colorado State University Department of Biomedical Sciences, Foothills Campus, Fort Collins, CO 80523, USA
| | - Nicole Marlenee
- Colorado State University Department of Biomedical Sciences, Foothills Campus, Fort Collins, CO 80523, USA
| | - Airn Hartwig
- Colorado State University Department of Biomedical Sciences, Foothills Campus, Fort Collins, CO 80523, USA
| | - Susan L Baldwin
- Infectious Disease Research Institute, 1616 Eastlake Ave E., Seattle WA 98103, USA
| | - Rhea N Coler
- Infectious Disease Research Institute, 1616 Eastlake Ave E., Seattle WA 98103, USA
| | - Mark Tomai
- 3M, Inc., St. Paul, Minnesota 55121, USA
| | | | - Steven G Reed
- Infectious Disease Research Institute, 1616 Eastlake Ave E., Seattle WA 98103, USA
| | - Darrick Carter
- Infectious Disease Research Institute, 1616 Eastlake Ave E., Seattle WA 98103, USA
| |
Collapse
|
16
|
Falkeborn T, Hinkula J, Olliver M, Lindberg A, Maltais AK. The intranasal adjuvant Endocine™ enhances both systemic and mucosal immune responses in aged mice immunized with influenza antigen. Virol J 2017; 14:44. [PMID: 28253901 PMCID: PMC5335733 DOI: 10.1186/s12985-017-0698-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 02/02/2017] [Indexed: 01/17/2023] Open
Abstract
Despite availability of annual influenza vaccines, influenza causes significant morbidity and mortality in the elderly. This is at least in part a result of immunosenescence; the age-dependent decrease in immunological competence that results in greater susceptibility to infections and reduced responses to vaccination. To improve protective immune responses in this age group, new vaccines strategies, such as the use of adjuvants, are needed. Here, we evaluated the mucosal vaccine adjuvant Endocine™, formulated with split influenza antigen and administered intranasally in aged (20-month old) mice. Humoral immune responses were assessed and compared to unadjuvanted intranasal and subcutaneous vaccines. We show that formulation with Endocine™ significantly enhances hemagglutination inhibition (HI) titers, as well as serum IgG and mucosal IgA antibody titers, compared to both types of unadjuvanted vaccines. Thus, our results indicate that intranasal vaccination with Endocine™ is a possible approach for the development of mucosal influenza vaccines for the elderly.
Collapse
Affiliation(s)
- Tina Falkeborn
- Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Jorma Hinkula
- Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Marie Olliver
- Eurocine Vaccines AB, Karolinska Institutet Science Park, Solna, Sweden
| | - Alf Lindberg
- Eurocine Vaccines AB, Karolinska Institutet Science Park, Solna, Sweden
| | | |
Collapse
|
17
|
Immune Responses Induced by Recombinant Bacillus Subtilis Expressing the Hemagglutinin Protein of H5N1 in chickens. Sci Rep 2016; 6:38403. [PMID: 27982050 PMCID: PMC5159790 DOI: 10.1038/srep38403] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 10/17/2016] [Indexed: 11/29/2022] Open
Abstract
To develop an effective, safe, and convenient vaccine for the prevention of highly pathogenic avian influenza (HPAI) H5N1, we have constructed a recombinant Bacillus subtilis strain (B.S.-HA) expressing the hemagglutinin (HA) protein. Then we evaluated the immune function in chicken bone marrow derived dendritic cells (BM-DCs), and the immune response after oral immunization. Our results show that the recombinant Bacillus subtilis B.S.-HA could be sampled by BM-DCs in vitro and increase the BM-DCs major histocompatibility complex (MHC) II phenotype. The weight, height of the small intestine villus, and lymphoid tissue area of the ileum increased significantly in B.S.-HA immunized chickens (P < 0.05 or P < 0.01). B.S.-HA induced the secretion of cytokines and the expression of Toll-like receptors in the trachea and small intestine (P < 0.05 or P < 0.01). In addition, B.S.-HA elevated the specific IgA titers in the trachea, IgG and HI antibody titers in serum (P < 0.05 or P < 0.01). Therefore, B.S.-HA provides a potential novel strategy and approach for developing an H5N1 vaccine.
Collapse
|
18
|
CpG Improves Influenza Vaccine Efficacy in Young Adult but Not Aged Mice. PLoS One 2016; 11:e0150425. [PMID: 26934728 PMCID: PMC4774967 DOI: 10.1371/journal.pone.0150425] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Accepted: 02/13/2016] [Indexed: 11/19/2022] Open
Abstract
Several studies have shown a reduced efficacy of influenza vaccines in the elderly compared to young adults. In this study, we evaluated the immunogenicity and protective efficacy of a commercially available inactivated influenza vaccine (Fluzone®) in young adult and aged mice. C57/BL6 mice were administered a single or double immunization of Fluzone® with or without CpG and challenged intranasally with H1N1 A/California/09 virus. A double immunization of Fluzone® adjuvanted with CpG elicited the highest level of protection in young adult mice which was associated with increases in influenza specific IgG, elevated HAI titres, reduced viral titres and lung inflammation. In contrast, the vaccine schedule which provided fully protective immunity in young adult mice conferred limited protection in aged mice. Antigen presenting cells from aged mice were found to be less responsive to in vitro stimulation by Fluzone and CpG which may partially explain this result. Our data are supportive of studies that have shown limited effectiveness of influenza vaccines in the elderly and provide important information relevant to the design of more immunogenic vaccines in this age group.
Collapse
|
19
|
Yam KK, Gupta J, Allen EK, Burt KR, Beaulieu É, Mallett CP, Burt DS, Ward BJ. Comparison of AS03 and Alum on immune responses elicited by A/H3N2 split influenza vaccine in young, mature and aged BALB/c mice. Vaccine 2016; 34:1444-51. [DOI: 10.1016/j.vaccine.2016.02.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 01/14/2016] [Accepted: 02/01/2016] [Indexed: 12/20/2022]
|
20
|
He F, Leyrer S, Kwang J. Strategies towards universal pandemic influenza vaccines. Expert Rev Vaccines 2015; 15:215-25. [DOI: 10.1586/14760584.2016.1115352] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Fang He
- Animal Health Biotechnology, Temasek Life Sciences Laboratory, Singapore, Singapore
| | - Sonja Leyrer
- Emergent Product Development Germany GmbH, Munich, Germany
| | - Jimmy Kwang
- Animal Health Biotechnology, Temasek Life Sciences Laboratory, Singapore, Singapore
- Department of Microbiology, Faculty of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
21
|
Abstract
Emerflu is an inactivated, split-virion pandemic preparedness vaccine, containing 30 μg of hemagglutinin (HA) and 600 μg of aluminum hydroxide adjuvant. It is administered in two doses, 3 weeks apart. Only moderate immunogenicity was evident from clinical studies with the vaccine in adults, and HA antibody responses were below the criteria established by the EMA and US FDA for licensure. With the exception of Australia, the vaccine remains unlicensed. Further clinical development appears to have been suspended, and newer adjuvants such as MF59 and AS03 have since demonstrated safety and superior immunogenicity with lower HA doses. Emerflu is symbolic of the failure of aluminum salts as an adjuvant for influenza vaccines. Reasons for this failure are unclear, and may reflect problems with the adjuvant-antigen complex or interference in the immune response by heterosubtypic immunity.
Collapse
Affiliation(s)
- Barnaby E Young
- Communicable Diseases Centre, Institute of Infectious Diseases and Epidemiology, Communicable Diseases Centre, 144 Moulmein Road, Singapore, Singapore
| | | | | |
Collapse
|
22
|
Fukuyama Y, Ikeda Y, Ohori J, Sugita G, Aso K, Fujihashi K, Briles DE, McGhee JR, Fujihashi K. A molecular mucosal adjuvant to enhance immunity against pneumococcal infection in the elderly. Immune Netw 2015; 15:9-15. [PMID: 25713504 PMCID: PMC4338268 DOI: 10.4110/in.2015.15.1.9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Revised: 02/10/2015] [Accepted: 02/11/2015] [Indexed: 12/25/2022] Open
Abstract
Streptococcus pneumoniae (the pneumococcus) causes a major upper respiratory tract infection often leading to severe illness and death in the elderly. Thus, it is important to induce safe and effective mucosal immunity against this pathogen in order to prevent pnuemocaccal infection. However, this is a very difficult task to elicit protective mucosal IgA antibody responses in older individuals. A combind nasal adjuvant consisting of a plasmid encoding the Flt3 ligand cDNA (pFL) and CpG oligonucleotide (CpG ODN) successfully enhanced S. pneumoniae-specific mucosal immunity in aged mice. In particular, a pneumococcal surface protein A-based nasal vaccine given with pFL and CpG ODN induced complete protection from S. pneumoniae infection. These results show that nasal delivery of a combined DNA adjuvant offers an attractive potential for protection against the pneumococcus in the elderly.
Collapse
Affiliation(s)
- Yoshiko Fukuyama
- Division of Mucosal Immunology, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Yorihiko Ikeda
- Department of Pediatric Dentistry, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Junichiro Ohori
- Department of Pediatric Dentistry, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Gen Sugita
- Department of Pediatric Dentistry, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Kazuyoshi Aso
- Department of Pediatric Dentistry, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Keiko Fujihashi
- Department of Pediatric Dentistry, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - David E Briles
- Department of Microbiology, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jerry R McGhee
- Department of Pediatric Dentistry, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Kohtaro Fujihashi
- Department of Pediatric Dentistry, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
23
|
Thomas M, Wang Z, Sreenivasan CC, Hause BM, Gourapura J Renukaradhya, Li F, Francis DH, Kaushik RS, Khatri M. Poly I:C adjuvanted inactivated swine influenza vaccine induces heterologous protective immunity in pigs. Vaccine 2014; 33:542-8. [PMID: 25437101 PMCID: PMC7115561 DOI: 10.1016/j.vaccine.2014.11.034] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2014] [Revised: 11/14/2014] [Accepted: 11/19/2014] [Indexed: 11/28/2022]
Abstract
Intranasal administration of Poly I:C adjuvanted bivalent swine influenza vaccine induced challenge virus-specific HI antibodies. Poly I:C adjuvanted vaccine also induced IgA and IgG antibodies in the lungs. Poly I:C adjuvanted vaccine provided protection against antigenic variant and heterologous swine influenza viruses.
Swine influenza is widely prevalent in swine herds in North America and Europe causing enormous economic losses and a public health threat. Pigs can be infected by both avian and mammalian influenza viruses and are sources of generation of reassortant influenza viruses capable of causing pandemics in humans. Current commercial vaccines provide satisfactory immunity against homologous viruses; however, protection against heterologous viruses is not adequate. In this study, we evaluated the protective efficacy of an intranasal Poly I:C adjuvanted UV inactivated bivalent swine influenza vaccine consisting of Swine/OH/24366/07 H1N1 and Swine/CO/99 H3N2, referred as PAV, in maternal antibody positive pigs against an antigenic variant and a heterologous swine influenza virus challenge. Groups of three-week-old commercial-grade pigs were immunized intranasally with PAV or a commercial vaccine (CV) twice at 2 weeks intervals. Three weeks after the second immunization, pigs were challenged with the antigenic variant Swine/MN/08 H1N1 (MN08) and the heterologous Swine/NC/10 H1N2 (NC10) influenza virus. Antibodies in serum and respiratory tract, lung lesions, virus shedding in nasal secretions and virus load in lungs were assessed. Intranasal administration of PAV induced challenge viruses specific-hemagglutination inhibition- and IgG antibodies in the serum and IgA and IgG antibodies in the respiratory tract. Importantly, intranasal administration of PAV provided protection against the antigenic variant MN08 and the heterologous NC10 swine influenza viruses as evidenced by significant reductions in lung virus load, gross lung lesions and significantly reduced shedding of challenge viruses in nasal secretions. These results indicate that Poly I:C or its homologues may be effective as vaccine adjuvants capable of generating cross-protective immunity against antigenic variants/heterologous swine influenza viruses in pigs.
Collapse
Affiliation(s)
- Milton Thomas
- Department of Biology and Microbiology, South Dakota State University, Brookings, SD, USA
| | - Zhao Wang
- Department of Biology and Microbiology, South Dakota State University, Brookings, SD, USA
| | - Chithra C Sreenivasan
- Department of Biology and Microbiology, South Dakota State University, Brookings, SD, USA
| | - Ben M Hause
- Department of Diagnostic Medicine and Pathobiology, Kansas State University, Manhattan, KS, USA
| | - Gourapura J Renukaradhya
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, The Ohio State University, 1680 Madison Avenue, Wooster, OH 44691, USA
| | - Feng Li
- Department of Veterinary and Biomedical Sciences, South Dakota State University, Brookings, SD, USA; Department of Biology and Microbiology, South Dakota State University, Brookings, SD, USA
| | - David H Francis
- Department of Veterinary and Biomedical Sciences, South Dakota State University, Brookings, SD, USA
| | - Radhey S Kaushik
- Department of Veterinary and Biomedical Sciences, South Dakota State University, Brookings, SD, USA; Department of Biology and Microbiology, South Dakota State University, Brookings, SD, USA
| | - Mahesh Khatri
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, The Ohio State University, 1680 Madison Avenue, Wooster, OH 44691, USA.
| |
Collapse
|
24
|
Overcoming barriers in the mucosal delivery of virus-like particle-based vaccines. Ther Deliv 2014; 5:741-4. [DOI: 10.4155/tde.14.52] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
|
25
|
Pérez-Girón JV, Belicha-Villanueva A, Hassan E, Gómez-Medina S, Cruz JLG, Lüdtke A, Ruibal P, Albrecht RA, García-Sastre A, Muñoz-Fontela C. Mucosal polyinosinic-polycytidylic acid improves protection elicited by replicating influenza vaccines via enhanced dendritic cell function and T cell immunity. THE JOURNAL OF IMMUNOLOGY 2014; 193:1324-32. [PMID: 24958904 DOI: 10.4049/jimmunol.1400222] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Live-attenuated influenza vaccines (LAIVs) have the potential to generate CD8 T cell immunity that may limit the virulence of an antigenically shifted influenza strain in a population lacking protective Abs. However, current LAIVs exert limited T cell immunity restricted to the vaccine strains. One approach to improve LAIV-induced T cell responses is the use of specific adjuvants to enhance T cell priming by respiratory dendritic cells, but this hypothesis has not been addressed. In this study, we assessed the effect of the TLR3 ligand polyinosinic-polycytidylic acid (poly IC) on CD8 T cell immunity and protection elicited by LAIVs. Mucosal treatment with poly IC shortly after vaccination enhanced respiratory dendritic cell function, CD8 T cell formation, and production of neutralizing Abs. This adjuvant effect of poly IC was dependent on amplification of TLR3 signaling by nonhematopoietic radioresistant cells and enhanced mouse protection to homosubtypic, as well as heterosubtypic, virus challenge. Our findings indicate that mucosal TLR3 ligation may be used to improve CD8 T cell responses to replicating vaccines, which has implications for protection in the absence of pre-existing Ab immunity.
Collapse
Affiliation(s)
- José V Pérez-Girón
- Heinrich Pette Institute, Leibniz Institute For Experimental Virology, 20251 Hamburg, Germany
| | - Alan Belicha-Villanueva
- Division of Infectious Diseases, Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Ebrahim Hassan
- Heinrich Pette Institute, Leibniz Institute For Experimental Virology, 20251 Hamburg, Germany
| | - Sergio Gómez-Medina
- Heinrich Pette Institute, Leibniz Institute For Experimental Virology, 20251 Hamburg, Germany
| | - Jazmina L G Cruz
- Heinrich Pette Institute, Leibniz Institute For Experimental Virology, 20251 Hamburg, Germany
| | - Anja Lüdtke
- Heinrich Pette Institute, Leibniz Institute For Experimental Virology, 20251 Hamburg, Germany
| | - Paula Ruibal
- Heinrich Pette Institute, Leibniz Institute For Experimental Virology, 20251 Hamburg, Germany
| | - Randy A Albrecht
- Division of Infectious Diseases, Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029; Division of Infectious Diseases, Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029; and
| | - Adolfo García-Sastre
- Division of Infectious Diseases, Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029; Division of Infectious Diseases, Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029; and Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - César Muñoz-Fontela
- Heinrich Pette Institute, Leibniz Institute For Experimental Virology, 20251 Hamburg, Germany;
| |
Collapse
|
26
|
Inactivated influenza vaccine adjuvanted with Bacterium-like particles induce systemic and mucosal influenza A virus specific T-cell and B-cell responses after nasal administration in a TLR2 dependent fashion. Vaccine 2014; 32:2904-10. [DOI: 10.1016/j.vaccine.2014.02.019] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
27
|
Concannon MA, Jiang J. The effect of adjuvants on vaccine-induced antibody response against influenza in aged mice. ACTA ACUST UNITED AC 2014; 9:89-94. [PMID: 32215006 PMCID: PMC7089211 DOI: 10.1007/s11515-014-1301-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2014] [Accepted: 02/21/2014] [Indexed: 11/26/2022]
Abstract
While influenza remains a major threat to public health, researchers continue to search for a universal solution to improving the efficacy of the influenza vaccine. Even though influenza affects people of all different ages, it can be extremely hazardous to people of 65 years of age or older since that is the population that makes up the high majority of the death toll caused by influenza-related diseases. Elderly individuals suffer the effects of immunosenescence as they age, which is the diminishing of the overall immune response. Immunosenescence occurs by specifically affecting the adaptive immune response which controls the establishment of immunity after vaccination or infection. There are many studies under way that are trying to find a resolution to the problem of the influenza vaccine not providing enough protection in the elderly population. One of the possible strategies is to seek the use of an optimal adjuvant, an immunological agent that can enhance immune responses, with the current vaccine formulation. Here, we used the murine model to review the effects of adjuvants on the antibody response to influenza vaccines in aged mice. Since adjuvants can enhance the production of important inflammatory cytokines and activation of dendritic cells, the stimulation of these cells are boosted to increase the effectiveness of the influenza vaccine in aged mice which would hopefully translate to the elderly.
Collapse
Affiliation(s)
- Mark A. Concannon
- Department of Biology, Drexel University, Philadelphia, PA 19104 USA
| | - Jiu Jiang
- Department of Biology, Drexel University, Philadelphia, PA 19104 USA
| |
Collapse
|
28
|
Liu D, Rhebergen AM, Eisenbarth SC. Licensing Adaptive Immunity by NOD-Like Receptors. Front Immunol 2013; 4:486. [PMID: 24409181 PMCID: PMC3873523 DOI: 10.3389/fimmu.2013.00486] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Accepted: 12/10/2013] [Indexed: 12/30/2022] Open
Abstract
The innate immune system is composed of a diverse set of host defense molecules, physical barriers, and specialized leukocytes and is the primary form of immune defense against environmental insults. Another crucial role of innate immunity is to shape the long-lived adaptive immune response mediated by T and B lymphocytes. The activation of pattern recognition receptors (PRRs) from the Toll-like receptor family is now a classic example of innate immune molecules influencing adaptive immunity, resulting in effective antigen presentation to naïve T cells. More recent work suggests that the activation of another family of PRRs, the NOD-like receptors (NLRs), induces a different set of innate immune responses and accordingly, drives different aspects of adaptive immunity. Yet how this unusually diverse family of molecules (some without canonical PRR function) regulates immunity remains incompletely understood. In this review, we discuss the evidence for and against NLR activity orchestrating adaptive immune responses during infectious as well as non-infectious challenges.
Collapse
Affiliation(s)
- Dong Liu
- Department of Laboratory Medicine, Yale University School of Medicine , New Haven, CT , USA ; Department of Immunobiology, Yale University School of Medicine , New Haven, CT , USA ; Department of Internal Medicine, Yale University School of Medicine , New Haven, CT , USA
| | - Anne Marie Rhebergen
- Department of Laboratory Medicine, Yale University School of Medicine , New Haven, CT , USA ; Department of Immunobiology, Yale University School of Medicine , New Haven, CT , USA ; Department of Internal Medicine, Yale University School of Medicine , New Haven, CT , USA
| | - Stephanie C Eisenbarth
- Department of Laboratory Medicine, Yale University School of Medicine , New Haven, CT , USA ; Department of Immunobiology, Yale University School of Medicine , New Haven, CT , USA ; Department of Internal Medicine, Yale University School of Medicine , New Haven, CT , USA
| |
Collapse
|
29
|
Iheagwara UK, Beatty PL, Van PT, Ross TM, Minden JS, Finn OJ. Influenza virus infection elicits protective antibodies and T cells specific for host cell antigens also expressed as tumor-associated antigens: a new view of cancer immunosurveillance. Cancer Immunol Res 2013; 2:263-73. [PMID: 24778322 DOI: 10.1158/2326-6066.cir-13-0125] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Most tumor-associated antigens (TAA) are self-molecules that are abnormally expressed in cancer cells and become targets of antitumor immune responses. Antibodies and T cells specific for some TAAs have been found in healthy individuals and are associated with lowered lifetime risk for developing cancer. Lower risk for cancer has also been associated with a history of febrile viral diseases. We hypothesized that virus infections could lead to transient expression of abnormal forms of self-molecules, some of which are TAAs; facilitated by the adjuvant effects of infection and inflammation, these molecules could elicit specific antibodies, T cells, and lasting immune memory simultaneously with immunity against viral antigens. Such infection-induced immune memory for TAA would be expected to provide life-long immune surveillance of cancer. Using influenza virus infection in mice as a model system, we tested this hypothesis and demonstrated that influenza-experienced mice control 3LL mouse lung tumor challenge better than infection-naive control mice. Using 2D-difference gel electrophoresis and mass spectrometry, we identified numerous molecules, some of which are known TAAs, on the 3LL tumor cells recognized by antibodies elicited by two successive influenza infections. We studied in detail immune responses against glyceraldehyde-3-phosphate dehydrogenase (GAPDH), histone H4, HSP90, malate dehydrogenase 2, and annexin A2, all of which were overexpressed in influenza-infected lungs and in tumor cells. Finally, we show that immune responses generated through vaccination against peptides derived from these antigens correlated with improved tumor control.
Collapse
Affiliation(s)
- Uzoma K Iheagwara
- Authors' Affiliations: Departments of Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, Pennsylvania
| | | | | | | | | | | |
Collapse
|
30
|
Liang J, Fu J, Kang H, Lin J, Yu Q, Yang Q. Comparison of 3 kinds of Toll-like receptor ligands for inactivated avian H5N1 influenza virus intranasal immunization in chicken. Poult Sci 2013; 92:2651-60. [PMID: 24046412 DOI: 10.3382/ps.2013-03193] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
To evaluate the effects of co-administration of inactivated avian influenza H5N1 virus (IAIV) and different Toll-like receptor (TLR) ligands in chickens, 10-d-old chickens were immunized intranasally with IAIV and TLR ligand [Bacillus subtilis spores, polyinosinic-polycytidylic acid, and CpG oligodeoxynucleotides (CpG-ODN), respectively]. The results showed that both anti-avian influenza virus (AIV) specific secretory IgA level in respiratory tract and anti-AIV specific IgG level in serum significantly increased, as well as the expressions of IL-12, interferon-γ, IL-6, and TLR in the nasal cavity and trachea after intranasal immunization with IAIV and TLR ligand. Among the used TLR ligands, B. subtilis spores as the adjuvant for nasal IAIV had the strongest effect on the expression of IL-6 and IL-12 (P < 0.01), whereas the CpG-ODN could present an advantageous effect on the induction of anti-AIV specific IgG and neutralization antibodies (P < 0.01). The chickens that were previously co-administrated with IAIV and B. subtilis spores could survive at an improved rate upon challenge by live AIV H5N1 virus. Our study suggested that B. subtilis spores, polyinosinic-polycytidylic acid, or CpG-ODN all could effectively enhance the local and systemic immune responses to IAIV in chickens. Considering of the effects and cost of these TLR ligands, we prospected that B. subtilis spores might serve as a more affordable and efficacious mucosal adjuvant for nasal IAIV in chickens.
Collapse
Affiliation(s)
- Jinfeng Liang
- Key Lab of Animal Physiology and Biochemistry, Ministry of Agriculture, Nanjing Agricultural University, Weigang 1, Nanjing, Jiangsu, 210095, China
| | | | | | | | | | | |
Collapse
|
31
|
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
|
32
|
Lee N, Wong CK, Hui DSC, Lee SKW, Wong RYK, Ngai KLK, Chan MCW, Chu YJ, Ho AWY, Lui GCY, Wong BCK, Wong SH, Yip SP, Chan PKS. Role of human Toll-like receptors in naturally occurring influenza A infections. Influenza Other Respir Viruses 2013; 7:666-75. [PMID: 23552014 PMCID: PMC5781199 DOI: 10.1111/irv.12109] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/26/2013] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND We investigated the roles of Toll-like receptors (TLRs) in naturally occurring influenza. METHODS A prospective, case - control study was conducted. Adults hospitalized with virologically confirmed influenza A infections (onset <48 hours, before treatment) were compared with age-/gender-matched controls. TLRs (2, 3, 4, 7, 8, 9) expression in monocytes and dendritic cells (DCs - total, myeloid, plasmacytoid) was quantitated using flow cytometry. Gene expression of RLRs (RIG-1, MDA-5) was evaluated using real-time PCR. Concomitant signaling molecules expression, plasma cytokine/chemokine concentrations, and respiratory tract viral loads were measured. PBMCs were cultured and stimulated ex vivo with TLR-specific ligands for cytokine responses. RESULTS Forty two patients with influenza (24 A/H3N2, 18 A/H1N1pdm09) and 20 controls were studied. Patients' mean age was 68 ± 16 years; 81% had respiratory/cardiovascular complications. There were increased cellular expressions of TLR9, TLR8, TLR3, and TLR7 during influenza; TLR2 and TLR4 were suppressed. Results were similar for both virus strains. Higher TLR expression levels at presentation significantly correlated with lower viral loads (Spearman's rho: -0.46 to -0.69 for TLR9, TLR8, and TLR3; P-values <0.05). Multivariate regression models (adjusted for age, comorbidity, disease severity, time from onset) confirmed their independent associations. Increased signaling molecules (phospho-MAPKs, IκB) and inflammatory cytokines (IL-6, sTNFR-1, CCL2/MCP-1; CXCL10/IP-10, IFN-γ) correlated with increased TLR expression. RLRs were upregulated simultaneously. PBMCs of patients with influenza showed significant, dynamic changes in their cytokine responses upon TLR stimulation, compared with controls. CONCLUSIONS Our results suggest that TLRs play an important role in early, innate viral inhibition in naturally occurring influenza. Inflammatory cytokine responses are concomitantly induced. These findings support investigation of TLR targeting as a novel intervention approach for prophylaxis against influenza.
Collapse
Affiliation(s)
- Nelson Lee
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Hepatitis C VLPs delivered to dendritic cells by a TLR2 targeting lipopeptide results in enhanced antibody and cell-mediated responses. PLoS One 2012; 7:e47492. [PMID: 23091628 PMCID: PMC3472981 DOI: 10.1371/journal.pone.0047492] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2012] [Accepted: 09/12/2012] [Indexed: 01/10/2023] Open
Abstract
Although many studies provide strong evidence supporting the development of HCV virus-like particle (VLP)-based vaccines, the fact that heterologous viral vectors and/or multiple dosing regimes are required to induce protective immunity indicates that it is necessary to improve their immunogenicity. In this study, we have evaluated the use of an anionic self-adjuvanting lipopeptide containing the TLR2 agonist Pam2Cys (E8Pam2Cys) to enhance the immunogenicity of VLPs containing the HCV structural proteins (core, E1 and E2) of genotype 1a. While co-formulation of this lipopeptide with VLPs only resulted in marginal improvements in dendritic cell (DC) uptake, its ability to concomitantly induce DC maturation at very small doses is a feature not observed using VLPs alone or in the presence of an aluminium hydroxide-based adjuvant (Alum). Dramatically improved VLP and E2-specific antibody responses were observed in VLP+E8Pam2Cys vaccinated mice where up to 3 doses of non-adjuvanted or traditionally alum-adjuvanted VLPs was required to match the antibody titres obtained with a single dose of VLPs formulated with this lipopeptide. This result also correlated with significantly higher numbers of specific antibody secreting cells that was detected in the spleens of VLP+E8Pam2Cys vaccinated mice and greater ability of sera from these mice to neutralise the binding and uptake of VLPs by Huh7 cells. Moreover, vaccination of HLA-A2 transgenic mice with this formulation also induced better VLP-specific IFN-γ-mediated responses compared to non-adjuvanted VLPs but comparable levels to that achieved when coadministered with complete freund’s adjuvant. These results suggest overall that the immunogenicity of HCV VLPs can be significantly improved by the addition of this novel adjuvant by targeting their delivery to DCs and could therefore constitute a viable vaccine strategy for the treatment of HCV.
Collapse
|
34
|
Blank M, Israeli E, Shoenfeld Y. When APS (Hughes syndrome) met the autoimmune/inflammatory syndrome induced by adjuvants (ASIA). Lupus 2012; 21:711-4. [PMID: 22635209 DOI: 10.1177/0961203312438115] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Vaccination of healthy individuals is the most effective approach to protect the public from infections and prevent the spread of many infectious diseases all over the globe. Licensed vaccines are mostly safe, but in rare cases they may be associated with humoral response to self-antigens due to molecular mimicry, epitope spread, bystander activation or polyclonal triggering. Moreover, the clinical picture of autoimmune conditions following post-vaccination is rarer. Nevertheless, anecdotal case reports on the flare of autoimmune response with clinical manifestations were reported. Herein, we discuss this topic in relation to post-vaccination-induced antiphospholipid antibodies following tetanus toxoid vaccine, HBV and influenza associated in rare cases with antiphospholipid syndrome clinical manifestations. We will discuss the possible mechanisms which pertain to ASIA (Shoenfeld syndrome).
Collapse
Affiliation(s)
- M Blank
- Zabludowitz Center for Autoimmune Diseases, Sheba Medical Center affiliated to Sackler Faculty of Medicine, Tel-Aviv University, Israel
| | | | | |
Collapse
|
35
|
Weldon WC, Zarnitsyn VG, Esser ES, Taherbhai MT, Koutsonanos DG, Vassilieva EV, Skountzou I, Prausnitz MR, Compans RW. Effect of adjuvants on responses to skin immunization by microneedles coated with influenza subunit vaccine. PLoS One 2012; 7:e41501. [PMID: 22848514 PMCID: PMC3405087 DOI: 10.1371/journal.pone.0041501] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2012] [Accepted: 06/21/2012] [Indexed: 11/23/2022] Open
Abstract
Recent studies have demonstrated the effectiveness of vaccine delivery to the skin by vaccine-coated microneedles; however there is little information on the effects of adjuvants using this approach for vaccination. Here we investigate the use of TLR ligands as adjuvants with skin-based delivery of influenza subunit vaccine. BALB/c mice received 1 µg of monovalent H1N1 subunit vaccine alone or with 1 µg of imiquimod or poly(I:C) individually or in combination via coated microneedle patches inserted into the skin. Poly(I:C) adjuvanted subunit influenza vaccine induced similar antigen-specific immune responses compared to vaccine alone when delivered to the skin by microneedles. However, imiquimod-adjuvanted vaccine elicited higher levels of serum IgG2a antibodies and increased hemagglutination inhibition titers compared to vaccine alone, suggesting enhanced induction of functional antibodies. In addition, imiquimod-adjuvanted vaccine induced a robust IFN-γ cellular response. These responses correlated with improved protection compared to influenza subunit vaccine alone, as well as reduced viral replication and production of pro-inflammatory cytokines in the lungs. The finding that microneedle delivery of imiquimod with influenza subunit vaccine induces improved immune responses compared to vaccine alone supports the use of TLR7 ligands as adjuvants for skin-based influenza vaccines.
Collapse
Affiliation(s)
- William C. Weldon
- Department of Microbiology and Immunology and Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Vladimir G. Zarnitsyn
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia, United States of America
| | - E. Stein Esser
- Department of Microbiology and Immunology and Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Misha T. Taherbhai
- Department of Microbiology and Immunology and Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Dimitrios G. Koutsonanos
- Department of Microbiology and Immunology and Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Elena V. Vassilieva
- Department of Microbiology and Immunology and Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Ioanna Skountzou
- Department of Microbiology and Immunology and Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Mark R. Prausnitz
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia, United States of America
| | - Richard W. Compans
- Department of Microbiology and Immunology and Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia, United States of America
- * E-mail:
| |
Collapse
|
36
|
NS1-truncated live attenuated virus vaccine provides robust protection to aged mice from viral challenge. J Virol 2012; 86:10293-301. [PMID: 22787224 DOI: 10.1128/jvi.01131-12] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Immunological changes associated with age contribute to the high rates of influenza virus morbidity and mortality in the elderly. Compounding this problem, aged individuals do not respond to vaccination as well as younger, healthy adults. Efforts to increase protection to this demographic group are of utmost importance, as the proportion of the population above the age of 65 is projected to increase in the coming decade. Using a live influenza virus with a truncated nonstructural protein 1 (NS1), we are able to stimulate cellular and humoral immune responses of aged mice comparable to levels seen in young mice. Impressively, a single vaccination provided protection following stringent lethal challenge in aged mice.
Collapse
|