1
|
Shams N, Jaydari A, Najafi H, Hataminejad M, Khanizadeh S, Pouladi I. An Overview of the Types of Adjuvants Used in the Vaccination Industry And Their Mechanisms of Action. Viral Immunol 2024; 37:324-336. [PMID: 39172659 DOI: 10.1089/vim.2024.0032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/24/2024] Open
Abstract
The widespread use of efficient vaccines against infectious diseases is regarded as one of the most significant advancements in public health and techniques for preventing and protecting against infectious diseases and cancer. Because the purpose of vaccination is to elicit an appropriate, powerful, and long-lasting immune response against the pathogen, compounds such as adjuvants must be used to enhance these responses. Adjuvants have been widely used since their discovery to boost immune responses, prevent diseases, and activate protective immunity. Today, several types of adjuvants with varying properties are available for specific applications. Adjuvants are supramolecular substances or complexes that strengthen and prolong the immune response to antigens. These compounds have long-term immunological effects and are low in toxicity. They also lower the amount of antigen or the number of immunogenic reactions needed to improve vaccine efficacy and are used in specific populations. This article provides an overview of the adjuvants commonly used in the vaccination industry, their respective mechanisms of action, and discusses how they function to stimulate the immune system. Understanding the mechanisms of action of adjuvants is crucial for the development of effective and safe vaccines.
Collapse
Affiliation(s)
- Nemat Shams
- Department of Microbiology and Food Hygiene, Faculty of Veterinary Medicine, Lorestan University, Khorramabad, Iran
| | - Amin Jaydari
- Department of Microbiology and Food Hygiene, Faculty of Veterinary Medicine, Lorestan University, Khorramabad, Iran
| | - Hamideh Najafi
- Department of Microbiology and Immunology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Maryam Hataminejad
- Department of Parasitology and Mycology, Faculty of Medicine, Mazandaran University of Medical Science, Sari, Iran
| | - Sayyad Khanizadeh
- Hepatitis Research Center, School of Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Iman Pouladi
- Department of Microbiology and Immunology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| |
Collapse
|
2
|
Silveira F, Rivera-Patron M, Deshpande N, Sienra S, Checa J, Moreno M, Chabalgoity JA, Cibulski SP, Baz M. Quillaja brasiliensis nanoparticle adjuvant formulation improves the efficacy of an inactivated trivalent influenza vaccine in mice. Front Immunol 2023; 14:1163858. [PMID: 37197659 PMCID: PMC10183569 DOI: 10.3389/fimmu.2023.1163858] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Accepted: 04/04/2023] [Indexed: 05/19/2023] Open
Abstract
The threat of viral influenza infections has sparked research efforts to develop vaccines that can induce broadly protective immunity with safe adjuvants that trigger robust immune responses. Here, we demonstrate that subcutaneous or intranasal delivery of a seasonal trivalent influenza vaccine (TIV) adjuvanted with the Quillaja brasiliensis saponin-based nanoparticle (IMXQB) increases the potency of TIV. The adjuvanted vaccine (TIV-IMXQB) elicited high levels of IgG2a and IgG1 antibodies with virus-neutralizing capacity and improved serum hemagglutination inhibition titers. The cellular immune response induced by TIV-IMXQB suggests the presence of a mixed Th1/Th2 cytokine profile, antibody-secreting cells (ASCs) skewed toward an IgG2a phenotype, a positive delayed-type hypersensitivity (DTH) response, and effector CD4+ and CD8+ T cells. After challenge, viral titers in the lungs were significantly lower in animals receiving TIV-IMXQB than in those inoculated with TIV alone. Most notably, mice vaccinated intranasally with TIV-IMXQB and challenged with a lethal dose of influenza virus were fully protected against weight loss and lung virus replication, with no mortality, whereas, among animals vaccinated with TIV alone, the mortality rate was 75%. These findings demonstrate that TIV-IMXQB improved the immune responses to TIV, and, unlike the commercial vaccine, conferred full protection against influenza challenge.
Collapse
Affiliation(s)
- Fernando Silveira
- Departamento de Desarrollo Biotecnológico, Instituto de Higiene, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
- *Correspondence: Fernando Silveira, ; Mariana Baz,
| | - Mariana Rivera-Patron
- Departamento de Desarrollo Biotecnológico, Instituto de Higiene, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Nikita Deshpande
- World Health Organization Collaborating Centre for Reference and Research on Influenza at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Soledad Sienra
- Departamento de Desarrollo Biotecnológico, Instituto de Higiene, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Jackeline Checa
- Unidad de Biología Parasitaria, Facultad de Ciencias, Instituto de Higiene, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - María Moreno
- Departamento de Desarrollo Biotecnológico, Instituto de Higiene, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Jose A. Chabalgoity
- Departamento de Desarrollo Biotecnológico, Instituto de Higiene, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Samuel P. Cibulski
- Centro de Biotecnologia – CBiotec, Laboratório de Biotecnologia Celular e Molecular, Universidade Federal da Paraíba, João Pessoa, Paraíba, Brazil
| | - Mariana Baz
- World Health Organization Collaborating Centre for Reference and Research on Influenza at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
- Department of Microbiology, Infectious Disease and Immunology, Faculty of Medicine, Université Laval, Quebec City, QC, Canada
- *Correspondence: Fernando Silveira, ; Mariana Baz,
| |
Collapse
|
3
|
Aoun T, Borrow R, Arkwright PD. Immunogenicity and safety of seasonal influenza vaccines in children under 3 years of age. Expert Rev Vaccines 2023; 22:226-242. [PMID: 36800932 DOI: 10.1080/14760584.2023.2181797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2023]
Abstract
INTRODUCTION Despite children aged 6-35 months developing more severe influenza infections, not all countries include influenza vaccines in their national immunization programs. AREAS COVERED This literature review examines the efficacy, immunogenicity, and safety of seasonal trivalent influenza vaccines (TIVs) and quadrivalent influenzae vaccines (QIVs) in children 6-35 months old to determine if greater valency promotes greater protection while maintaining a similar safety profile. EXPERT OPINION TIVs and QIVs are safe for children under 3 years old. TIVs and QIVs provided good seroprotection, and immunogenicity (GMT, SCR, and SPR) meeting recommended levels set by CHMP (European) and CBER (USA). However, as QIVs carry two influenza B strains and TIVs only one, QIVs has an overall higher seroprotection against particularly influenza B. Vaccines containing adjuncts had better immunogenicity, particularly after the first dose. Seroprotection of all vaccines lasted 12 months. Increasing the dosage from 0.25 mL to 0.5 mL did not cause more systemic or local side-effects. Further comparisons of efficacy, and wider promotion of influenza vaccines in general are required in preschool children.
Collapse
Affiliation(s)
- Tia Aoun
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK
| | - Ray Borrow
- Vaccine Evaluation Unit, UK Health Security Agency, Manchester Royal Infirmary, Manchester, UK
| | - Peter D Arkwright
- Royal Manchester Children's Hospital, Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK
| |
Collapse
|
4
|
Abstract
Current influenza vaccines, while being the best method of managing viral outbreaks, have several major drawbacks that prevent them from being wholly-effective. They need to be updated regularly and require extensive resources to develop. When considering alternatives, the recent deployment of mRNA vaccines for SARS-CoV-2 has created a unique opportunity to evaluate a new platform for seasonal and pandemic influenza vaccines. The mRNA format has previously been examined for application to influenza and promising data suggest it may be a viable format for next-generation influenza vaccines. Here, we discuss the prospect of shifting global influenza vaccination efforts to an mRNA-based system that might allow better control over the product and immune responses and could aid in the development of a universal vaccine.
Collapse
Affiliation(s)
- Jessica R Shartouny
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
| | - Anice C Lowen
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
- Emory Center of Excellence for Influenza Research and Response (Emory-CEIRR), USA
| |
Collapse
|
5
|
Lee S, Ryu JH. Influenza Viruses: Innate Immunity and mRNA Vaccines. Front Immunol 2021; 12:710647. [PMID: 34531860 PMCID: PMC8438292 DOI: 10.3389/fimmu.2021.710647] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 08/13/2021] [Indexed: 12/13/2022] Open
Abstract
The innate immune system represents the first line of defense against influenza viruses, which cause severe inflammation of the respiratory tract and are responsible for more than 650,000 deaths annually worldwide. mRNA vaccines are promising alternatives to traditional vaccine approaches due to their safe dosing, low-cost manufacturing, rapid development capability, and high efficacy. In this review, we provide our current understanding of the innate immune response that uses pattern recognition receptors to detect and respond to mRNA vaccination. We also provide an overview of mRNA vaccines, and discuss the future directions and challenges in advancing this promising therapeutic approach.
Collapse
Affiliation(s)
- SangJoon Lee
- Department of Infection Biology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Jin-Hyeob Ryu
- BIORCHESTRA Co., Ltd, Daejeon, South Korea
- BIORCHESTRA Co., Ltd, Cambridge, MA, United States
| |
Collapse
|
6
|
Intranasal Nanoparticle Vaccination Elicits a Persistent, Polyfunctional CD4 T Cell Response in the Murine Lung Specific for a Highly Conserved Influenza Virus Antigen That Is Sufficient To Mediate Protection from Influenza Virus Challenge. J Virol 2021; 95:e0084121. [PMID: 34076479 DOI: 10.1128/jvi.00841-21] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Lung-localized CD4 T cells play a critical role in the control of influenza virus infection and can provide broadly protective immunity. However, current influenza vaccination strategies primarily target influenza hemagglutinin (HA) and are administered peripherally to induce neutralizing antibodies. We have used an intranasal vaccination strategy targeting the highly conserved influenza nucleoprotein (NP) to elicit broadly protective lung-localized CD4 T cell responses. The vaccine platform consists of a self-assembling nanolipoprotein particle (NLP) linked to NP with an adjuvant. We have evaluated the functionality, in vivo localization, and persistence of the T cells elicited. Our study revealed that intranasal vaccination elicits a polyfunctional subset of lung-localized CD4 T cells that persist long term. A subset of these lung CD4 T cells localize to the airway, where they can act as early responders following encounter with cognate antigen. Polyfunctional CD4 T cells isolated from airway and lung tissue produce significantly more effector cytokines IFN-γ and TNF-α, as well as cytotoxic functionality. When adoptively transferred to naive recipients, CD4 T cells from NLP:NP-immunized lung were sufficient to mediate 100% survival from lethal challenge with H1N1 influenza virus. IMPORTANCE Exploiting new, more efficacious strategies to potentiate influenza virus-specific immune responses is important, particularly for at-risk populations. We have demonstrated the promise of direct intranasal protein vaccination to establish long-lived immunity in the lung with CD4 T cells that possess features and positioning in the lung that are associated with both immediate and long-term immunity, as well as demonstrating direct protective potential.
Collapse
|
7
|
Cortese M, Sherman AC, Rouphael NG, Pulendran B. Systems Biological Analysis of Immune Response to Influenza Vaccination. Cold Spring Harb Perspect Med 2021; 11:cshperspect.a038596. [PMID: 32152245 DOI: 10.1101/cshperspect.a038596] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The last decade has witnessed tremendous progress in immunology and vaccinology, owing to several scientific and technological breakthroughs. Systems vaccinology is a field that has emerged at the forefront of vaccine research and development and provides a unique way to probe immune responses to vaccination in humans. The goals of systems vaccinology are to use systems-based approaches to define signatures that can be used to predict vaccine immunogenicity and efficacy and to delineate the molecular mechanisms driving protective immunity. The application of systems biological approaches in influenza vaccination studies has enabled the discovery of early signatures that predict immunogenicity to vaccination and yielded novel mechanistic insights about vaccine-induced immunity. Here we review the contributions of systems vaccinology to influenza vaccine development and critically examine the potential of systems vaccinology toward enabling the development of a universal influenza vaccine that provides robust and durable immunity against diverse influenza viruses.
Collapse
Affiliation(s)
- Mario Cortese
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, California 94305, USA
| | - Amy C Sherman
- Hope Clinic of the Emory Vaccine Center, Decatur, Georgia 30030, USA
| | - Nadine G Rouphael
- Hope Clinic of the Emory Vaccine Center, Decatur, Georgia 30030, USA
| | - Bali Pulendran
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, California 94305, USA.,Department of Pathology, Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford University, Stanford, California 94305, USA.,Department of Pathology, Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford University, Stanford, California 94305, USA
| |
Collapse
|
8
|
Biswas A, Mandal RS, Chakraborty S, Maiti G. Tapping the immunological imprints to design chimeric SARS-CoV-2 vaccine for elderly population. Int Rev Immunol 2021; 41:448-463. [PMID: 33978550 PMCID: PMC8127164 DOI: 10.1080/08830185.2021.1925267] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 04/14/2021] [Accepted: 04/23/2021] [Indexed: 01/10/2023]
Abstract
The impact of SARS-CoV-2 and COVID-19 disease susceptibility varies depending on the age and health status of an individual. Currently, there are more than 140 COVID-19 vaccines under development. However, the challenge will be to induce an effective immune response in the elderly population. Analysis of B cell epitopes indicates the minor role of the stalk domain of spike protein in viral neutralization due to low surface accessibility. Nevertheless, the accumulation of mutations in the receptor-binding domain (RBD) might reduce the vaccine efficacy in all age groups. We also propose the concept of chimeric vaccines based on the co-expression of SARS-CoV-2 spike and influenza hemagglutinin (HA) and matrix protein 1 (M1) proteins to generate chimeric virus-like particles (VLP). This review discusses the possible approaches by which influenza-specific memory repertoire developed during the lifetime of the elderly populations can converge to mount an effective immune response against the SARS-CoV-2 spike protein with the possibilities of designing single vaccines for COVID-19 and influenza. HighlightsImmunosenescence aggravates COVID-19 symptoms in elderly individuals.Low immunogenicity of SARS-CoV-2 vaccines in elderly population.Tapping the memory T and B cell repertoire in elderly can enhance vaccine efficiency.Chimeric vaccines can mount effective immune response against COVID-19 in elderly.Chimeric vaccines co-express SARS-CoV-2 spike and influenza HA and M1 proteins.
Collapse
Affiliation(s)
- Asim Biswas
- Department of Ophthalmology, New York University Grossman School of Medicine, New York, NY, USA
| | - Rahul Subhra Mandal
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Suparna Chakraborty
- Division of Clinical Medicine, National Institute of Cholera and Enteric Diseases, Kolkata, India
| | | |
Collapse
|
9
|
Cibulski S, Varela APM, Teixeira TF, Cancela MP, Sesterheim P, Souza DO, Roehe PM, Silveira F. Zika Virus Envelope Domain III Recombinant Protein Delivered With Saponin-Based Nanoadjuvant From Quillaja brasiliensis Enhances Anti-Zika Immune Responses, Including Neutralizing Antibodies and Splenocyte Proliferation. Front Immunol 2021; 12:632714. [PMID: 33746970 PMCID: PMC7969523 DOI: 10.3389/fimmu.2021.632714] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 02/09/2021] [Indexed: 11/26/2022] Open
Abstract
Nanoadjuvants that combine immunostimulatory properties and delivery systems reportedly bestow major improvements on the efficacy of recombinant, protein-based vaccines. Among these, self-assembled micellar formulations named ISCOMs (immune stimulating complexes) show a great ability to trigger powerful immunological responses against infectious pathogens. Here, a nanoadjuvant preparation, based on saponins from Quillaja brasiliensis, was evaluated together with an experimental Zika virus (ZIKV) vaccine (IQB80-zEDIII) and compared to an equivalent vaccine with alum as the standard adjuvant. The preparations were administered to mice in two doses (on days zero and 14) and immune responses were evaluated on day 28 post-priming. Serum levels of anti-Zika virus IgG, IgG1, IgG2b, IgG2c, IgG3 were significantly increased by the nanoadjuvant vaccine, compared to the mice that received the alum-adjuvanted vaccine or the unadjuvanted vaccine. In addition, a robust production of neutralizing antibodies and in vitro splenocyte proliferative responses were observed in mice immunized with IQB80-zEDIII nanoformulated vaccine. Therefore, the IQB80-zEDIII recombinant preparation seems to be a suitable candidate vaccine for ZIKV. Overall, this study identified saponin-based delivery systems as an adequate adjuvant for recombinant ZIKV vaccines and has important implications for recombinant protein-based vaccine formulations against other flaviviruses and possibly enveloped viruses.
Collapse
Affiliation(s)
- Samuel Cibulski
- Laboratório de Biotecnologia Celular e Molecular, Centro de Biotecnologia-CBiotec, Universidade Federal da Paraíba, João Pessoa, Brazil
| | - Ana Paula Muterle Varela
- Laboratório de Virologia, Departamento de Microbiologia Imunologia e Parasitologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Thais Fumaco Teixeira
- Laboratório de Virologia, Departamento de Microbiologia Imunologia e Parasitologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Martín Pablo Cancela
- Laboratório de Genômica Estrutural e Funcional, Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Patrícia Sesterheim
- Centro de Cardiologia Experimental, Instituto de Cardiologia/Fundação Universitária de Cardiologia, Porto Alegre, Brazil
| | - Diogo Onofre Souza
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Paulo Michel Roehe
- Laboratório de Virologia, Departamento de Microbiologia Imunologia e Parasitologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Fernando Silveira
- Departamento de Desarrollo Biotecnológico, Instituto de Higiene, Facultad de Medicina, Universidad de la República (UdelaR), Montevideo, Uruguay
| |
Collapse
|
10
|
Abstract
The proportion of the global population aged 65 and older is rapidly increasing. Infections in this age group, most recently with SARS-CoV-2, cause substantial morbidity and mortality. Major improvements have been made in vaccines for older people, either through the addition of novel adjuvants-as in the new recombinant zoster vaccine and an adjuvanted influenza vaccine-or by increasing antigen concentration, as in influenza vaccines. In this article we review improvements in immunization for the three most important vaccine preventable diseases of aging. The recombinant zoster vaccine has an efficacy of 90% that is minimally affected by the age of the person being vaccinated and persists for more than four years. Increasing antigen dose or inclusion of adjuvant has improved the immunogenicity of influenza vaccines in older adults, although the relative effectiveness of the enhanced influenza vaccines and the durability of the immune response are the focus of ongoing clinical trials. Conjugate and polysaccharide pneumococcal vaccines have similar efficacy against invasive pneumococcal disease and pneumococcal pneumonia caused by vaccine serotypes in older adults. Their relative value varies by setting, depending on the prevalence of vaccine serotypes, largely related to conjugate vaccine coverage in children. Improved efficacy will increase public confidence and uptake of these vaccines. Co-administration of these vaccines is feasible and important for maximal uptake in older people. Development of new vaccine platforms has accelerated following the arrival of SARS-CoV-2, and will likely result in new vaccines against other pathogens in the future.
Collapse
Affiliation(s)
- Anthony L Cunningham
- Centre for Virus Research, The Westmead Institute for Medical Research, Faculty of Medicine and Health, University of Sydney, Australia
| | - Peter McIntyre
- Women's and Children's Health, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Kanta Subbarao
- WHO CollaboratingCentre for Reference and Research on Influenza and Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Robert Booy
- National Centre for Immunisation Research and Surveillance of Vaccine Preventable Diseases, The Children's Hospital at Westmead, New South Wales, Australia
- Marie Bashir Institute for Infectious Diseases and Biosecurity, School of Biological Sciences and Sydney Medical School, University of Sydney, Australia
| | - Myron J Levin
- Departments of Pediatrics and Medicine, University of Colorado School of Medicine Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
11
|
Inflammasome-Mediated Immunogenicity of Clinical and Experimental Vaccine Adjuvants. Vaccines (Basel) 2020; 8:vaccines8030554. [PMID: 32971761 PMCID: PMC7565252 DOI: 10.3390/vaccines8030554] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 09/16/2020] [Accepted: 09/18/2020] [Indexed: 02/06/2023] Open
Abstract
In modern vaccines, adjuvants can be sophisticated immunological tools to promote robust and long-lasting protection against prevalent diseases. However, there is an urgent need to improve immunogenicity of vaccines in order to protect mankind from life-threatening diseases such as AIDS, malaria or, most recently, COVID-19. Therefore, it is important to understand the cellular and molecular mechanisms of action of vaccine adjuvants, which generally trigger the innate immune system to enhance signal transition to adaptive immunity, resulting in pathogen-specific protection. Thus, improved understanding of vaccine adjuvant mechanisms may aid in the design of “intelligent” vaccines to provide robust protection from pathogens. Various commonly used clinical adjuvants, such as aluminium salts, saponins or emulsions, have been identified as activators of inflammasomes - multiprotein signalling platforms that drive activation of inflammatory caspases, resulting in secretion of pro-inflammatory cytokines of the IL-1 family. Importantly, these cytokines affect the cellular and humoral arms of adaptive immunity, which indicates that inflammasomes represent a valuable target of vaccine adjuvants. In this review, we highlight the impact of different inflammasomes on vaccine adjuvant-induced immune responses regarding their mechanisms and immunogenicity. In this context, we focus on clinically relevant adjuvants that have been shown to activate the NLRP3 inflammasome and also present various experimental adjuvants that activate the NLRP3-, NLRC4-, AIM2-, pyrin-, or non-canonical inflammasomes and could have the potential to improve future vaccines. Together, we provide a comprehensive overview on vaccine adjuvants that are known, or suggested, to promote immunogenicity through inflammasome-mediated signalling.
Collapse
|
12
|
Recombinant HA-based vaccine outperforms split and subunit vaccines in elicitation of influenza-specific CD4 T cells and CD4 T cell-dependent antibody responses in humans. NPJ Vaccines 2020; 5:77. [PMID: 32884842 PMCID: PMC7450042 DOI: 10.1038/s41541-020-00227-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 07/10/2020] [Indexed: 12/13/2022] Open
Abstract
Although traditional egg-based inactivated influenza vaccines can protect against infection, there have been significant efforts to develop improved formats to overcome disadvantages of this platform. Here, we have assessed human CD4 T cell responses to a traditional egg-based influenza vaccine with recently available cell-derived vaccines and recombinant baculovirus-derived vaccines. Adults were administered either egg-derived Fluzone®, mammalian cell-derived Flucelvax® or recombinant HA (Flublok®). CD4 T cell responses to each HA protein were assessed by cytokine EliSpot and intracellular staining assays. The specificity and magnitude of antibody responses were quantified by ELISA and HAI assays. By all criteria, Flublok vaccine exhibited superior performance in eliciting both CD4 T cell responses and HA-specific antibody responses, whether measured by mean response magnitude or percent of responders. Although the mechanism(s) underlying this advantage is not yet clear, it is likely that both qualitative and quantitative features of the vaccines impact the response.
Collapse
|
13
|
Lopez CE, Legge KL. Influenza A Virus Vaccination: Immunity, Protection, and Recent Advances Toward A Universal Vaccine. Vaccines (Basel) 2020; 8:E434. [PMID: 32756443 PMCID: PMC7565301 DOI: 10.3390/vaccines8030434] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 07/28/2020] [Accepted: 07/31/2020] [Indexed: 12/19/2022] Open
Abstract
Influenza virus infections represent a serious public health threat and account for significant morbidity and mortality worldwide due to seasonal epidemics and periodic pandemics. Despite being an important countermeasure to combat influenza virus and being highly efficacious when matched to circulating influenza viruses, current preventative strategies of vaccination against influenza virus often provide incomplete protection due the continuous antigenic drift/shift of circulating strains of influenza virus. Prevention and control of influenza virus infection with vaccines is dependent on the host immune response induced by vaccination and the various vaccine platforms induce different components of the local and systemic immune response. This review focuses on the immune basis of current (inactivated influenza vaccines (IIV) and live attenuated influenza vaccines (LAIV)) as well as novel vaccine platforms against influenza virus. Particular emphasis will be placed on how each platform induces cross-protection against heterologous influenza viruses, as well as how this immunity compares to and contrasts from the "gold standard" of immunity generated by natural influenza virus infection.
Collapse
Affiliation(s)
- Christopher E. Lopez
- Department of Microbiology and Immunology University of Iowa, Iowa City, IA 52242, USA;
- Department of Pathology, University of Iowa, Iowa City, IA 52242, USA
| | - Kevin L. Legge
- Department of Microbiology and Immunology University of Iowa, Iowa City, IA 52242, USA;
- Department of Pathology, University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
14
|
Adjuvanted H5N1 influenza vaccine enhances both cross-reactive memory B cell and strain-specific naive B cell responses in humans. Proc Natl Acad Sci U S A 2020; 117:17957-17964. [PMID: 32661157 DOI: 10.1073/pnas.1906613117] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
There is a need for improved influenza vaccines. In this study we compared the antibody responses in humans after vaccination with an AS03-adjuvanted versus nonadjuvanted H5N1 avian influenza virus inactivated vaccine. Healthy young adults received two doses of either formulation 3 wk apart. We found that AS03 significantly enhanced H5 hemagglutinin (HA)-specific plasmablast and antibody responses compared to the nonadjuvanted vaccine. Plasmablast response after the first immunization was exclusively directed to the conserved HA stem region and came from memory B cells. Monoclonal antibodies (mAbs) derived from these plasmablasts had high levels of somatic hypermutation (SHM) and recognized the HA stem region of multiple influenza virus subtypes. Second immunization induced a plasmablast response to the highly variable HA head region. mAbs derived from these plasmablasts exhibited minimal SHM (naive B cell origin) and largely recognized the HA head region of the immunizing H5N1 strain. Interestingly, the antibody response to H5 HA stem region was much lower after the second immunization, and this suppression was most likely due to blocking of these epitopes by stem-specific antibodies induced by the first immunization. Taken together, these findings show that an adjuvanted influenza vaccine can substantially increase antibody responses in humans by effectively recruiting preexisting memory B cells as well as naive B cells into the response. In addition, we show that high levels of preexisting antibody can have a negative effect on boosting. These findings have implications toward the development of a universal influenza vaccine.
Collapse
|
15
|
Wei CJ, Crank MC, Shiver J, Graham BS, Mascola JR, Nabel GJ. Next-generation influenza vaccines: opportunities and challenges. Nat Rev Drug Discov 2020; 19:239-252. [PMID: 32060419 PMCID: PMC7223957 DOI: 10.1038/s41573-019-0056-x] [Citation(s) in RCA: 193] [Impact Index Per Article: 48.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/16/2019] [Indexed: 02/07/2023]
Abstract
Seasonal influenza vaccines lack efficacy against drifted or pandemic influenza strains. Developing improved vaccines that elicit broader immunity remains a public health priority. Immune responses to current vaccines focus on the haemagglutinin head domain, whereas next-generation vaccines target less variable virus structures, including the haemagglutinin stem. Strategies employed to improve vaccine efficacy involve using structure-based design and nanoparticle display to optimize the antigenicity and immunogenicity of target antigens; increasing the antigen dose; using novel adjuvants; stimulating cellular immunity; and targeting other viral proteins, including neuraminidase, matrix protein 2 or nucleoprotein. Improved understanding of influenza antigen structure and immunobiology is advancing novel vaccine candidates into human trials.
Collapse
Affiliation(s)
- Chih-Jen Wei
- Sanofi Global Research and Development, Cambridge, MA, USA
| | - Michelle C Crank
- Vaccine Research Center, National Institute for Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | | | - Barney S Graham
- Vaccine Research Center, National Institute for Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - John R Mascola
- Vaccine Research Center, National Institute for Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Gary J Nabel
- Sanofi Global Research and Development, Cambridge, MA, USA.
| |
Collapse
|
16
|
Broecker F, Wegner E, Seco BMS, Kaplonek P, Bräutigam M, Ensser A, Pfister F, Daniel C, Martin CE, Mattner J, Seeberger PH. Synthetic Oligosaccharide-Based Vaccines Protect Mice from Clostridioides difficile Infections. ACS Chem Biol 2019; 14:2720-2728. [PMID: 31692324 PMCID: PMC6929054 DOI: 10.1021/acschembio.9b00642] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
![]()
Infections with Clostridioides difficile (formerly Clostridium difficile) have increased
in incidence, morbidity,
and mortality over the past decade. Preventing infections is becoming
increasingly important, as frontline antibiotics become less effective
and frequently induce recurrence by disrupting intestinal microbiota.
The clinically most advanced vaccine approaches prevent symptoms once C. difficile infection is established by inducing immunity
to secreted clostridial cytotoxins. However, they do not inhibit bacterial
colonization and thereby favor asymptomatic carriage. Synthetic oligosaccharides
resembling the C. difficile surface glycans PS-I,
PS-II, and PS-III are immunogenic and serve as basis for colonization-preventing
vaccines. Here, we demonstrate that glycoconjugate vaccine candidates
based on synthetic oligosaccharides protected mice from infections
with two different C. difficile strains. Four synthetic
antigens, ranging in size from disaccharides to hexasaccharides, were
conjugated to CRM197, which is a carrier protein used in
commercial vaccines. The vaccine candidates induced glycan-specific
antibodies in mice and substantially limited C. difficile colonization and colitis after experimental infection. The glycoconjugates
ameliorated intestinal pathology more substantially than a toxin-targeting
vaccine. Colonization of the gut by C. difficile was
selectively inhibited while intestinal microbiota remained preserved.
Passive transfer experiments with anti-PS-I serum revealed that protection
is mediated by specific antiglycan antibodies; however, cell-mediated
immunity likely also contributed to protection in vivo. Thus, glycoconjugate vaccines against C. difficile are a complementary approach to toxin-targeting strategies and are
advancing through preclinical work.
Collapse
Affiliation(s)
- Felix Broecker
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, Am Mühlenberg 1, 14424 Potsdam, Germany
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Arnimallee 22, 14195 Berlin, Germany
| | - Erik Wegner
- Mikrobiologisches Institut−Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen and Friedrich-Alexander Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Bruna M. S. Seco
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, Am Mühlenberg 1, 14424 Potsdam, Germany
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Arnimallee 22, 14195 Berlin, Germany
| | - Paulina Kaplonek
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, Am Mühlenberg 1, 14424 Potsdam, Germany
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Arnimallee 22, 14195 Berlin, Germany
| | - Maria Bräutigam
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, Am Mühlenberg 1, 14424 Potsdam, Germany
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Arnimallee 22, 14195 Berlin, Germany
| | - Armin Ensser
- Virologisches Institut, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Frederick Pfister
- Department of Nephropathology, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Christoph Daniel
- Department of Nephropathology, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Christopher E. Martin
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, Am Mühlenberg 1, 14424 Potsdam, Germany
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Arnimallee 22, 14195 Berlin, Germany
| | - Jochen Mattner
- Mikrobiologisches Institut−Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen and Friedrich-Alexander Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Peter H. Seeberger
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, Am Mühlenberg 1, 14424 Potsdam, Germany
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Arnimallee 22, 14195 Berlin, Germany
| |
Collapse
|
17
|
Valli E, Harriett AJ, Nowakowska MK, Baudier RL, Provosty WB, McSween Z, Lawson LB, Nakanishi Y, Norton EB. LTA1 is a safe, intranasal enterotoxin-based adjuvant that improves vaccine protection against influenza in young, old and B-cell-depleted (μMT) mice. Sci Rep 2019; 9:15128. [PMID: 31641151 PMCID: PMC6805908 DOI: 10.1038/s41598-019-51356-w] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 09/19/2019] [Indexed: 01/17/2023] Open
Abstract
Enterotoxin-based adjuvants including cholera toxin and heat-labile toxin (LT) are powerful manipulators of mucosal immunity; however, past clinical trials identified unacceptable neurological toxicity when LT or mutant AB5 adjuvant proteins were added to intranasal vaccines. Here, we examined the isolated enzymatic A1 domain of LT (LTA1) for intranasal safety and efficacy in combination with influenza (flu) vaccination. LTA1-treated mice exhibited no neurotoxicity, as measured by olfactory system testing and H&E staining of nasal tissue in contrast with cholera toxin. In vaccination studies, intranasal LTA1 enhanced immune responses to inactivated virus antigen and subsequent protection against H1N1 flu challenge in mice (8-week or 24-months). In addition, lung H1N1 viral titers post-challenge correlated to serum antibody responses; however, enhanced protection was also observed in μMT mice lacking B-cells while activation and recruitment of CD4 T-cells into the lung was apparent. Thus, we report that LTA1 protein is a novel, safe and effective enterotoxin adjuvant that improves protection of an intranasal flu vaccination by a mechanism that does not appear to require B-cells.
Collapse
Affiliation(s)
- E Valli
- Department of Microbiology & Immunology, Tulane University School of Medicine, New Orleans, USA
| | - A J Harriett
- Department of Microbiology & Immunology, Tulane University School of Medicine, New Orleans, USA
| | - M K Nowakowska
- Department of Microbiology & Immunology, Tulane University School of Medicine, New Orleans, USA
| | - R L Baudier
- Department of Microbiology & Immunology, Tulane University School of Medicine, New Orleans, USA
| | - W B Provosty
- Department of Microbiology & Immunology, Tulane University School of Medicine, New Orleans, USA
| | - Z McSween
- Department of Microbiology & Immunology, Tulane University School of Medicine, New Orleans, USA
| | - L B Lawson
- Department of Microbiology & Immunology, Tulane University School of Medicine, New Orleans, USA
| | - Y Nakanishi
- Department of Pathology & Laboratory Medicine, Tulane University School of Medicine, New Orleans, USA
| | - E B Norton
- Department of Microbiology & Immunology, Tulane University School of Medicine, New Orleans, USA.
| |
Collapse
|
18
|
Blanco-Lobo P, Nogales A, Rodríguez L, Martínez-Sobrido L. Novel Approaches for The Development of Live Attenuated Influenza Vaccines. Viruses 2019; 11:E190. [PMID: 30813325 PMCID: PMC6409754 DOI: 10.3390/v11020190] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 02/19/2019] [Accepted: 02/19/2019] [Indexed: 01/04/2023] Open
Abstract
Influenza virus still represents a considerable threat to global public health, despite the advances in the development and wide use of influenza vaccines. Vaccination with traditional inactivate influenza vaccines (IIV) or live-attenuated influenza vaccines (LAIV) remains the main strategy in the control of annual seasonal epidemics, but it does not offer protection against new influenza viruses with pandemic potential, those that have shifted. Moreover, the continual antigenic drift of seasonal circulating influenza viruses, causing an antigenic mismatch that requires yearly reformulation of seasonal influenza vaccines, seriously compromises vaccine efficacy. Therefore, the quick optimization of vaccine production for seasonal influenza and the development of new vaccine approaches for pandemic viruses is still a challenge for the prevention of influenza infections. Moreover, recent reports have questioned the effectiveness of the current LAIV because of limited protection, mainly against the influenza A virus (IAV) component of the vaccine. Although the reasons for the poor protection efficacy of the LAIV have not yet been elucidated, researchers are encouraged to develop new vaccination approaches that overcome the limitations that are associated with the current LAIV. The discovery and implementation of plasmid-based reverse genetics has been a key advance in the rapid generation of recombinant attenuated influenza viruses that can be used for the development of new and most effective LAIV. In this review, we provide an update regarding the progress that has been made during the last five years in the development of new LAIV and the innovative ways that are being explored as alternatives to the currently licensed LAIV. The safety, immunogenicity, and protection efficacy profile of these new LAIVs reveal their possible implementation in combating influenza infections. However, efforts by vaccine companies and government agencies will be needed for controlled testing and approving, respectively, these new vaccine methodologies for the control of influenza infections.
Collapse
Affiliation(s)
- Pilar Blanco-Lobo
- Department of Microbiology and Immunology, School of Medicine and Dentistry, University of Rochester, Rochester, New York, NY 14642, USA.
| | - Aitor Nogales
- Department of Microbiology and Immunology, School of Medicine and Dentistry, University of Rochester, Rochester, New York, NY 14642, USA.
| | - Laura Rodríguez
- Department of Microbiology and Immunology, School of Medicine and Dentistry, University of Rochester, Rochester, New York, NY 14642, USA.
| | - Luis Martínez-Sobrido
- Department of Microbiology and Immunology, School of Medicine and Dentistry, University of Rochester, Rochester, New York, NY 14642, USA.
| |
Collapse
|
19
|
Magnusson SE, Altenburg AF, Bengtsson KL, Bosman F, de Vries RD, Rimmelzwaan GF, Stertman L. Matrix-M™ adjuvant enhances immunogenicity of both protein- and modified vaccinia virus Ankara-based influenza vaccines in mice. Immunol Res 2019; 66:224-233. [PMID: 29594879 PMCID: PMC5899102 DOI: 10.1007/s12026-018-8991-x] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Influenza viruses continuously circulate in the human population and escape recognition by virus neutralizing antibodies induced by prior infection or vaccination through accumulation of mutations in the surface proteins hemagglutinin (HA) and neuraminidase (NA). Various strategies to develop a vaccine that provides broad protection against different influenza A viruses are under investigation, including use of recombinant (r) viral vectors and adjuvants. The replication-deficient modified vaccinia virus Ankara (MVA) is a promising vaccine vector that efficiently induces B and T cell responses specific for the antigen of interest. It is assumed that live vaccine vectors do not require an adjuvant to be immunogenic as the vector already mediates recruitment and activation of immune cells. To address this topic, BALB/c mice were vaccinated with either protein- or rMVA-based HA influenza vaccines, formulated with or without the saponin-based Matrix-M™ adjuvant. Co-formulation with Matrix-M significantly increased HA vaccine immunogenicity, resulting in antigen-specific humoral and cellular immune responses comparable to those induced by unadjuvanted rMVA-HA. Of special interest, rMVA-HA immunogenicity was also enhanced by addition of Matrix-M, demonstrated by enhanced HA inhibition antibody titres and cellular immune responses. Matrix-M added to either protein- or rMVA-based HA vaccines mediated recruitment and activation of antigen-presenting cells and lymphocytes to the draining lymph node 24 and 48 h post-vaccination. Taken together, these results suggest that adjuvants can be used not only with protein-based vaccines but also in combination with rMVA to increase vaccine immunogenicity, which may be a step forward to generate new and more effective influenza vaccines.
Collapse
Affiliation(s)
| | | | | | - Fons Bosman
- Amatsigroup NV, Biologicals Unit, Ghent, Belgium
| | - Rory D de Vries
- Department of Viroscience, Erasmus MC, Rotterdam, The Netherlands
| | - Guus F Rimmelzwaan
- Department of Viroscience, Erasmus MC, Rotterdam, The Netherlands
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine, Hannover, Germany
| | | |
Collapse
|
20
|
Filipić B, Stojić-Vukanić Z. Adjuvants in vaccines registered for human use. ARHIV ZA FARMACIJU 2019. [DOI: 10.5937/arhfarm1906406f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
|
21
|
Silva T, S Salomon P, Hamerski L, Walter J, B Menezes R, Siqueira JE, Santos A, Santos JAM, Ferme N, Guimarães T, O Fistarol G, I Hargreaves P, Thompson C, Thompson F, Souza TM, Siqueira M, Miranda M. Inhibitory effect of microalgae and cyanobacteria extracts on influenza virus replication and neuraminidase activity. PeerJ 2018; 6:e5716. [PMID: 30386690 PMCID: PMC6204821 DOI: 10.7717/peerj.5716] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 09/10/2018] [Indexed: 12/29/2022] Open
Abstract
Background The influenza virus can cause seasonal infections with mild to severe symptoms, circulating worldwide, and it can affect people in any age group. Therefore, this infection is a serious public health problem that causes severe illness and death in high-risk populations. Every year, 0.5% of the world’s population is infected by this pathogen. This percentage can increase up to ten times during pandemics. Influenza vaccination is the most effective way to prevent disease. In addition, anti-influenza drugs are essential for prophylactic and therapeutic interventions. The oseltamivir (OST, a neuraminidase inhibitor) is the primary antiviral used in clinics during outbreaks. However, OST resistant viruses may emerge naturally or due to antiviral pressure, with a prevalence of 1–2% worldwide. Thus, the search for new anti-influenza drugs is extremely important. Currently, several groups have been developing studies describing the biotechnological potential of microalgae and cyanobacteria, including antiviral activity of their extracts. In Brazil, this potential is poorly known and explored. Methods With the aim of increasing the knowledge on this topic, 38 extracts from microalgae and cyanobacteria isolated from marine and freshwater biomes in Brazil were tested against: cellular toxicity; OST-sensitive and resistant influenza replications; and neuraminidase activity. Results For this purpose, Madin-Darby Canine Kidney (MDCK)-infected cells were treated with 200 μg/mL of each extract. A total of 17 extracts (45%) inhibited influenza A replication, with seven of them resulting in more than 80% inhibition. Moreover, functional assays performed with viral neuraminidase revealed two extracts (from Leptolyngbya sp. and Chlorellaceae) with IC50 mean < 210 μg/mL for influenza A and B, and also OST-sensitive and resistant strains. Furthermore, MDCK cells exposed to 1 mg/mL of all the extracts showed viability higher than 80%. Discussion Our results suggest that extracts of microalgae and cyanobacteria have promising anti-influenza properties. Further chemical investigation should be conducted to isolate the active compounds for the development of new anti-influenza drugs. The data generated contribute to the knowledge of the biotechnological potential of Brazilian biomes that are still little explored for this purpose.
Collapse
Affiliation(s)
- Thauane Silva
- Laboratório de Vírus Respiratórios e do Sarampo, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Paulo S Salomon
- Laboratório de Fitoplâncton Marinho, Instituto de Biologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Lidilhone Hamerski
- Instituto de Pesquisas de Produtos Naturais, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Juline Walter
- Laboratório de Microbiologia Marinha, Instituto de Biologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Rafael B Menezes
- Laboratório de Fitoplâncton Marinho, Instituto de Biologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - José Edson Siqueira
- Instituto de Pesquisas de Produtos Naturais, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Aline Santos
- Laboratório de Vírus Respiratórios e do Sarampo, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | | | - Natália Ferme
- Laboratório de Vírus Respiratórios e do Sarampo, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Thaise Guimarães
- Laboratório de Vírus Respiratórios e do Sarampo, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Giovana O Fistarol
- Laboratório de Fitoplâncton Marinho, Instituto de Biologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Paulo I Hargreaves
- Laboratório de Fitoplâncton Marinho, Instituto de Biologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Cristiane Thompson
- Laboratório de Microbiologia Marinha, Instituto de Biologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fabiano Thompson
- Laboratório de Microbiologia Marinha, Instituto de Biologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Thiago Moreno Souza
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil.,Centro de Desenvolvimento Tecnológico em Saúde, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Marilda Siqueira
- Laboratório de Vírus Respiratórios e do Sarampo, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Milene Miranda
- Laboratório de Vírus Respiratórios e do Sarampo, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| |
Collapse
|
22
|
Cao Y, Zhu X, Hossen MN, Kakar P, Zhao Y, Chen X. Augmentation of vaccine-induced humoral and cellular immunity by a physical radiofrequency adjuvant. Nat Commun 2018; 9:3695. [PMID: 30209303 PMCID: PMC6135850 DOI: 10.1038/s41467-018-06151-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 08/17/2018] [Indexed: 11/18/2022] Open
Abstract
Protein/subunit vaccines often require external adjuvants to induce protective immunity. Due to the safety concern of chemical adjuvants, physical adjuvants were recently explored to boost vaccination. Physical adjuvants use physical energies rather than chemicals to stimulate tissue stress and endogenous danger signal release to boost vaccination. Here we present the safety and potency of non-invasive radiofrequency treatment to boost intradermal vaccination in murine models. We show non-invasive radiofrequency can increase protein antigen-induced humoral and cellular immune responses with adjuvant effects comparable to widely used chemical adjuvants. Radiofrequency adjuvant can also safely boost pandemic 2009 H1N1 influenza vaccination with adjuvant effects comparable to MF59-like AddaVax adjuvant. We find radiofrequency adjuvant induces heat shock protein 70 (HSP70) release and activates MyD88 to mediate the adjuvant effects. Physical radiofrequency can potentially be a safe and potent adjuvant to augment protein/subunit vaccine-induced humoral and cellular immune responses.
Collapse
Affiliation(s)
- Yan Cao
- Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, 7 Greenhouse Road, Avedisian Hall, Room 480, Kingston, RI, 02881, USA
| | - Xiaoyue Zhu
- Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, 7 Greenhouse Road, Avedisian Hall, Room 480, Kingston, RI, 02881, USA
| | - Md Nazir Hossen
- Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, 7 Greenhouse Road, Avedisian Hall, Room 480, Kingston, RI, 02881, USA
| | - Prateek Kakar
- Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, 7 Greenhouse Road, Avedisian Hall, Room 480, Kingston, RI, 02881, USA
| | - Yiwen Zhao
- Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, 7 Greenhouse Road, Avedisian Hall, Room 480, Kingston, RI, 02881, USA
| | - Xinyuan Chen
- Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, 7 Greenhouse Road, Avedisian Hall, Room 480, Kingston, RI, 02881, USA.
| |
Collapse
|
23
|
The Future of Influenza Vaccines: A Historical and Clinical Perspective. Vaccines (Basel) 2018; 6:vaccines6030058. [PMID: 30200179 PMCID: PMC6160951 DOI: 10.3390/vaccines6030058] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Revised: 08/21/2018] [Accepted: 08/27/2018] [Indexed: 12/16/2022] Open
Abstract
For centuries, the development of vaccines to prevent infectious disease was an empirical process. From smallpox variolation in Song dynasty China, through the polysaccharide capsule vaccines developed in the 1970s, vaccines were made either from the pathogen itself, treated in some way to render it attenuated or non-infectious, or from a closely related non-pathogenic strain. In recent decades, new scientific knowledge and technologies have enabled rational vaccine design in a way that was unimaginable before. However, vaccines optimal against some infectious diseases, influenza among them, have remained elusive. This review will highlight the challenges that influenza viruses pose for rational vaccine design. In particular, it will consider the clinically beneficial endpoints, beyond complete sterilizing immunity, that have been achieved with vaccines against other infectious diseases, as well as the barriers to achieving similar success against influenza.
Collapse
|
24
|
Valkenburg SA, Leung NHL, Bull MB, Yan LM, Li APY, Poon LLM, Cowling BJ. The Hurdles From Bench to Bedside in the Realization and Implementation of a Universal Influenza Vaccine. Front Immunol 2018; 9:1479. [PMID: 30013557 PMCID: PMC6036122 DOI: 10.3389/fimmu.2018.01479] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 06/14/2018] [Indexed: 12/23/2022] Open
Abstract
Influenza viruses circulate worldwide causing annual epidemics that have a substantial impact on public health. This is despite vaccines being in use for over 70 years and currently being administered to around 500 million people each year. Improvements in vaccine design are needed to increase the strength, breadth, and duration of immunity against diverse strains that circulate during regular epidemics, occasional pandemics, and from animal reservoirs. Universal vaccine strategies that target more conserved regions of the virus, such as the hemagglutinin (HA)-stalk, or recruit other cellular responses, such as T cells and NK cells, have the potential to provide broader immunity. Many pre-pandemic vaccines in clinical development do not utilize new vaccine platforms but use "tried and true" recombinant HA protein or inactivated virus strategies despite substantial leaps in fundamental research on universal vaccines. Significant hurdles exist for universal vaccine development from bench to bedside, so that promising preclinical data is not yet translating to human clinical trials. Few studies have assessed immune correlates derived from asymptomatic influenza virus infections, due to the scale of a study required to identity these cases. The realization and implementation of a universal influenza vaccine requires identification and standardization of set points of protective immune correlates, and consideration of dosage schedule to maximize vaccine uptake.
Collapse
Affiliation(s)
- Sophie A. Valkenburg
- HKU Pasteur Research Pole, The University of Hong Kong, Pokfulam, Hong Kong
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, The University of Hong Kong, Pokfulam, Hong Kong
| | - Nancy H. L. Leung
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, The University of Hong Kong, Pokfulam, Hong Kong
| | - Maireid B. Bull
- HKU Pasteur Research Pole, The University of Hong Kong, Pokfulam, Hong Kong
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, The University of Hong Kong, Pokfulam, Hong Kong
| | - Li-meng Yan
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, The University of Hong Kong, Pokfulam, Hong Kong
| | - Athena P. Y. Li
- HKU Pasteur Research Pole, The University of Hong Kong, Pokfulam, Hong Kong
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, The University of Hong Kong, Pokfulam, Hong Kong
| | - Leo L. M. Poon
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, The University of Hong Kong, Pokfulam, Hong Kong
| | - Benjamin J. Cowling
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, The University of Hong Kong, Pokfulam, Hong Kong
| |
Collapse
|
25
|
Schiffner T, Pallesen J, Russell RA, Dodd J, de Val N, LaBranche CC, Montefiori D, Tomaras GD, Shen X, Harris SL, Moghaddam AE, Kalyuzhniy O, Sanders RW, McCoy LE, Moore JP, Ward AB, Sattentau QJ. Structural and immunologic correlates of chemically stabilized HIV-1 envelope glycoproteins. PLoS Pathog 2018; 14:e1006986. [PMID: 29746590 PMCID: PMC5944921 DOI: 10.1371/journal.ppat.1006986] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 03/21/2018] [Indexed: 11/19/2022] Open
Abstract
Inducing broad spectrum neutralizing antibodies against challenging pathogens such as HIV-1 is a major vaccine design goal, but may be hindered by conformational instability within viral envelope glycoproteins (Env). Chemical cross-linking is widely used for vaccine antigen stabilization, but how this process affects structure, antigenicity and immunogenicity is poorly understood and its use remains entirely empirical. We have solved the first cryo-EM structure of a cross-linked vaccine antigen. The 4.2 Å structure of HIV-1 BG505 SOSIP soluble recombinant Env in complex with a CD4 binding site-specific broadly neutralizing antibody (bNAb) Fab fragment reveals how cross-linking affects key properties of the trimer. We observed density corresponding to highly specific glutaraldehyde (GLA) cross-links between gp120 monomers at the trimer apex and between gp120 and gp41 at the trimer interface that had strikingly little impact on overall trimer conformation, but critically enhanced trimer stability and improved Env antigenicity. Cross-links were also observed within gp120 at sites associated with the N241/N289 glycan hole that locally modified trimer antigenicity. In immunogenicity studies, the neutralizing antibody response to cross-linked trimers showed modest but significantly greater breadth against a global panel of difficult-to-neutralize Tier-2 heterologous viruses. Moreover, the specificity of autologous Tier-2 neutralization was modified away from the N241/N289 glycan hole, implying a novel specificity. Finally, we have investigated for the first time T helper cell responses to next-generation soluble trimers, and report on vaccine-relevant immunodominant responses to epitopes within BG505 that are modified by cross-linking. Elucidation of the structural correlates of a cross-linked viral glycoprotein will allow more rational use of this methodology for vaccine design, and reveals a strategy with promise for eliciting neutralizing antibodies needed for an effective HIV-1 vaccine.
Collapse
MESH Headings
- AIDS Vaccines/chemistry
- AIDS Vaccines/immunology
- Animals
- Antibodies, Neutralizing/immunology
- Antibody Specificity
- Antigen-Antibody Reactions/immunology
- Cross-Linking Reagents
- Cryoelectron Microscopy
- HIV Antibodies/immunology
- HIV Antigens/chemistry
- HIV Antigens/immunology
- HIV Antigens/ultrastructure
- HIV-1/chemistry
- HIV-1/immunology
- Host-Pathogen Interactions/immunology
- Humans
- Immunodominant Epitopes/chemistry
- Immunodominant Epitopes/immunology
- Mice
- Mice, Inbred BALB C
- Models, Molecular
- Protein Conformation
- Protein Stability
- Protein Structure, Quaternary
- Rabbits
- T-Lymphocytes, Helper-Inducer/immunology
- Vaccines, Synthetic/chemistry
- Vaccines, Synthetic/immunology
- env Gene Products, Human Immunodeficiency Virus/chemistry
- env Gene Products, Human Immunodeficiency Virus/immunology
- env Gene Products, Human Immunodeficiency Virus/ultrastructure
Collapse
Affiliation(s)
- Torben Schiffner
- The Sir William Dunn School of Pathology, The University of Oxford, Oxford, United Kingdom
| | - Jesper Pallesen
- Department of Integrative Structural and Computational Biology, IAVI Neutralizing Antibody Center, Collaboration for AIDS Vaccine Discovery and Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery, The Scripps Research Institute, La Jolla, California, United States of America
| | - Rebecca A Russell
- The Sir William Dunn School of Pathology, The University of Oxford, Oxford, United Kingdom
| | - Jonathan Dodd
- The Sir William Dunn School of Pathology, The University of Oxford, Oxford, United Kingdom
| | - Natalia de Val
- Center for Molecular Microscopy (CMM), National Cancer Institute (NCI), Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Celia C LaBranche
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, United States of America
| | - David Montefiori
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Georgia D Tomaras
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, United States of America
- Duke Human Vaccine Institute, Department of Medicine, Duke University School of Medicine, Durham, North Carolina, United States of America
- Departments of Immunology and Molecular Genetics and Microbiology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Xiaoying Shen
- Duke Human Vaccine Institute, Department of Medicine, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Scarlett L Harris
- The Sir William Dunn School of Pathology, The University of Oxford, Oxford, United Kingdom
| | - Amin E Moghaddam
- The Sir William Dunn School of Pathology, The University of Oxford, Oxford, United Kingdom
| | - Oleksandr Kalyuzhniy
- Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery (CHAVI-ID), The Scripps Research Institute, La Jolla, California, United States of America
- IAVI Neutralizing Antibody Center and the Collaboration for AIDS Vaccine Discovery (CAVD), The Scripps Research Institute, La Jolla, California, United States of America
| | - Rogier W Sanders
- Department of Medical Microbiology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Laura E McCoy
- Division of Infection and Immunity, University College London, London, United Kingdom
| | - John P Moore
- Department of Microbiology and Immunology, Weill Medical College of Cornell University, New York, New York, United States of America
| | - Andrew B Ward
- Department of Integrative Structural and Computational Biology, IAVI Neutralizing Antibody Center, Collaboration for AIDS Vaccine Discovery and Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery, The Scripps Research Institute, La Jolla, California, United States of America
| | - Quentin J Sattentau
- The Sir William Dunn School of Pathology, The University of Oxford, Oxford, United Kingdom
| |
Collapse
|
26
|
Vo HTM, Baudner BC, Sammicheli S, Iannacone M, D'Oro U, Piccioli D. Alum/Toll-Like Receptor 7 Adjuvant Enhances the Expansion of Memory B Cell Compartment Within the Draining Lymph Node. Front Immunol 2018; 9:641. [PMID: 29686670 PMCID: PMC5900039 DOI: 10.3389/fimmu.2018.00641] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 03/14/2018] [Indexed: 11/29/2022] Open
Abstract
Vaccination is one of the most cost-effective health interventions and, with the exception of water sanitization, no other action has had such a major effect in mortality reduction. Combined with other approaches, such as clean water, better hygiene, and health education, vaccination contributed to prevent millions of cases of deaths among children under 5 years of age. New or improved vaccines are needed to fight some vaccine-preventable diseases that are still a threat for the public health globally, as reported also in the Global Vaccine Action Plan (GVAP) endorsed by the World Health Assembly in 2012. Adjuvants are substances that enhance the effectiveness of vaccination, but despite their critical role for the development of novel vaccines, very few of them are approved for use in humans. Aluminum hydroxide (Alum) is the most common adjuvant used in vaccines administered in millions of doses around the world to prevent several dangerous diseases. The development of an improved version of Alum can help to design and produce new or better vaccines. Alum/toll-like receptor (TLR)7 is a novel Alum-based adjuvant, currently in phase I clinical development, formed by the attachment of a benzonaphthyridine compound, TLR7 agonist, to Alum. In preclinical studies, Alum/TLR7 showed a superior adjuvant capacity, compared to Alum, in several disease models, such as meningococcal meningitis, anthrax, staphylococcus infections. None of these studies reported the effect of Alum/TLR7 on the generation of the B cell memory compartment, despite this is a critical aspect to achieve a better immunization. In this study, we show, for the first time, that, compared to Alum, Alum/TLR7 enhances the expansion of the memory B cell compartment within the draining lymph node (LN) as result of intranodal sustained proliferation of antigen-engaged B cells and/or accumulation of memory B cells. In addition, we observed that Alum/TLR7 induces a recruitment of naïve antigen-specific B cells within the draining LN that may help to sustain the germinal center reaction. Our data further support Alum/TLR7 as a new promising adjuvant, which might contribute to meet the expectations of the GVAP for 2020 and beyond.
Collapse
Affiliation(s)
| | | | - Stefano Sammicheli
- Dynamics of Immune Responses, Division of Immunology, Transplantation and Infectious Diseases, Experimental Imaging Center, IRCCS San Raffaele Scientific Institute, Milano, Italy.,Vita-Salute San Raffaele University, Milano, Italy
| | - Matteo Iannacone
- Dynamics of Immune Responses, Division of Immunology, Transplantation and Infectious Diseases, Experimental Imaging Center, IRCCS San Raffaele Scientific Institute, Milano, Italy.,Vita-Salute San Raffaele University, Milano, Italy
| | - Ugo D'Oro
- Preclinical Research, GSK Vaccines, Siena, Italy
| | | |
Collapse
|
27
|
Bonanni P, Boccalini S, Zanobini P, Dakka N, Lorini C, Santomauro F, Bechini A. The appropriateness of the use of influenza vaccines: Recommendations from the latest seasons in Italy. Hum Vaccin Immunother 2018; 14:699-705. [PMID: 29059004 PMCID: PMC5861775 DOI: 10.1080/21645515.2017.1388480] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 09/08/2017] [Accepted: 09/30/2017] [Indexed: 02/06/2023] Open
Abstract
Influenza is one of the major infectious causes of excess mortality, hospitalization, and an increase in healthcare expenditure in all countries. In an increasingly ageing population, many members are exposed to flu-related complications. Vaccination coverage rates for the elderly in most European countries, such as Italy, are not satisfactory, and have been decreasing with time due to a sense of skepticism toward vaccination. Nowadays, many types of vaccines are available on the Italian market to prevent influenza illness. Many studies have proven their effectiveness in preventing influenza-related complications in specific risk groups. Any vaccine can be crucial to avoid complications, hospitalizations and death, but use of the most appropriate vaccine could optimize the result at a very modest cost. General practitioners (GPs) should encourage their patients to take the influenza vaccination to prevent complications or deaths. Health authorities should give GPs the opportunity to choose the appropriate vaccines tailored to specific patients.
Collapse
Affiliation(s)
- Paolo Bonanni
- Department of Health Sciences, University of Florence, Florence, Italy
| | - Sara Boccalini
- Department of Health Sciences, University of Florence, Florence, Italy
| | - Patrizio Zanobini
- Specialization Medical School of Hygiene and Preventive Medicine, University of Florence, Florence, Italy
| | - Nawal Dakka
- Specialization Medical School of Hygiene and Preventive Medicine, University of Florence, Florence, Italy
| | - Chiara Lorini
- Department of Health Sciences, University of Florence, Florence, Italy
| | | | - Angela Bechini
- Department of Health Sciences, University of Florence, Florence, Italy
| |
Collapse
|
28
|
Fighting against a protean enemy: immunosenescence, vaccines, and healthy aging. NPJ Aging Mech Dis 2017; 4:1. [PMID: 29285399 PMCID: PMC5740164 DOI: 10.1038/s41514-017-0020-0] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 11/09/2017] [Accepted: 11/27/2017] [Indexed: 12/16/2022] Open
Abstract
The progressive increase of the aged population worldwide mandates new strategies to ensure sustained health and well-being with age. The development of better and/or new vaccines against pathogens that affect older adults is one pivotal intervention in approaching this goal. However, the functional decline of various physiological systems, including the immune system, requires novel approaches to counteract immunosenescence. Although important progress has been made in understanding the mechanisms underlying the age-related decline of the immune response to infections and vaccinations, knowledge gaps remain, both in the areas of basic and translational research. In particular, it will be important to better understand how environmental factors, such as diet, physical activity, co-morbidities, and pharmacological treatments, delay or contribute to the decline of the capability of the aging immune system to appropriately respond to infectious diseases and vaccination. Recent findings suggest that successful approaches specifically targeted to the older population can be developed, such as the high-dose and adjuvanted vaccines against seasonal influenza, the adjuvanted subunit vaccine against herpes zoster, as well as experimental interventions with immune-potentiators or immunostimulants. Learning from these first successes may pave the way to developing novel and improved vaccines for the older adults and immunocompromised. With an integrated, holistic vaccination strategy, society will offer the opportunity for an improved quality of life to the segment of the population that is going to increase most significantly in numbers and proportion over future decades.
Collapse
|
29
|
Cioncada R, Maddaluno M, Vo HTM, Woodruff M, Tavarini S, Sammicheli C, Tortoli M, Pezzicoli A, De Gregorio E, Carroll MC, D’Oro U, Piccioli D. Vaccine adjuvant MF59 promotes the intranodal differentiation of antigen-loaded and activated monocyte-derived dendritic cells. PLoS One 2017; 12:e0185843. [PMID: 29088224 PMCID: PMC5663329 DOI: 10.1371/journal.pone.0185843] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 09/20/2017] [Indexed: 01/09/2023] Open
Abstract
MF59 is an oil-in-water emulsion adjuvant approved for human influenza vaccination in European Union. The mode of action of MF59 is not fully elucidated yet, but results from several years of investigation indicate that MF59 establishes an immunocompetent environment at injection site which promotes recruitment of immune cells, including antigen presenting cells (APCs), that are facilitated to engulf antigen and transport it to draining lymph node (dLN) where the antigen is accumulated. In vitro studies showed that MF59 promotes the differentiation of monocytes to dendritic cells (Mo-DCs). Since after immunization with MF59, monocytes are rapidly recruited both at the injection site and in dLN and appear to have a morphological change toward a DC-like phenotype, we asked whether MF59 could play a role in inducing differentiation of Mo-DC in vivo. To address this question we immunized mice with the auto-fluorescent protein Phycoerythrin (PE) as model antigen, in presence or absence of MF59. We measured the APC phenotype and their antigen uptake within dLNs, the antigen distribution within the dLN compartments and the humoral response to PE. In addition, using Ovalbumin as model antigen, we measured the capacity of dLN APCs to induce antigen-specific CD4 T cell proliferation. Here, we show, for the first time, that MF59 promotes differentiation of Mo-DCs within dLNs from intranodal recruited monocytes and we suggest that this differentiation could take place in the medullary compartment of the LN. In addition we show that the Mo-DC subset represents the major source of antigen-loaded and activated APCs within the dLN when immunizing with MF59. Interestingly, this finding correlates with the enhanced triggering of antigen-specific CD4 T cell response induced by LN APCs. This study therefore demonstrates that MF59 is able to promote an immunocompetent environment also directly within the dLN, offering a novel insight on the mechanism of action of vaccine adjuvants based on emulsions.
Collapse
Affiliation(s)
| | | | | | - Matthew Woodruff
- Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States of America
| | | | | | | | | | | | - Michael C. Carroll
- Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States of America
| | - Ugo D’Oro
- GSK Vaccines, Siena, Italy
- * E-mail: (DP); (UDO)
| | | |
Collapse
|
30
|
Influenza Vaccination in Patients with Common Variable Immunodeficiency (CVID). Curr Allergy Asthma Rep 2017; 17:78. [PMID: 28983790 DOI: 10.1007/s11882-017-0749-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
PURPOSE OF REVIEW Vaccination against influenza in patients with primary antibody deficiency is recommended. Common variable immunodeficiency (CVID) is the most frequent and clinically relevant antibody deficiency disease and is by definition characterized by an impaired vaccination response. The purpose of this review is to present the current knowledge of humoral and cellular vaccine response to influenza in CVID patients. RECENT FINDINGS Studies conducted in CVID patients demonstrated an impaired humoral response upon influenza vaccination. Data on cellular immune response are in part conflicting, with two out of three studies showing responses similar to healthy controls. Available data suggest a benefit from influenza vaccination in CVID patients. Therefore, annual influenza vaccination in patients and their close household contacts is recommended.
Collapse
|
31
|
Forero A, Fenstermacher K, Wohlgemuth N, Nishida A, Carter V, Smith EA, Peng X, Hayes M, Francis D, Treanor J, Morrison J, Klein SL, Lane A, Katze MG, Pekosz A. Evaluation of the innate immune responses to influenza and live-attenuated influenza vaccine infection in primary differentiated human nasal epithelial cells. Vaccine 2017; 35:6112-6121. [PMID: 28967519 DOI: 10.1016/j.vaccine.2017.09.058] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 09/13/2017] [Accepted: 09/19/2017] [Indexed: 01/08/2023]
Abstract
The host innate immune response to influenza virus is a key determinant of pathogenic outcomes and long-term protective immune responses against subsequent exposures. Here, we present a direct contrast of the host responses in primary differentiated human nasal epithelial cell (hNEC) cultures following infection with either a seasonal H3N2 influenza virus (WT) or the antigenically-matched live-attenuated vaccine (LAIV) strain. Comparison of the transcriptional profiles obtained 24 and 36h post-infection showed that the magnitude of gene expression was greater in LAIV infected relative to that observed in WT infected hNEC cultures. Functional enrichment analysis revealed that the antiviral and inflammatory responses were largely driven by type III IFN induction in both WT and LAIV infected cells. However, the enrichment of biological pathways involved in the recruitment of mononuclear leukocytes, antigen-presenting cells, and T lymphocytes was uniquely observed in LAIV infected cells. These observations were reflective of the host innate immune responses observed in individuals acutely infected with influenza viruses. These findings indicate that cell-intrinsic type III IFN-mediated innate immune responses in the nasal epithelium are not only crucial for viral clearance and attenuation, but may also play an important role in the induction of protective immune responses with live-attenuated vaccines.
Collapse
Affiliation(s)
- Adriana Forero
- Department of Microbiology, University of Washington, Seattle, WA, USA
| | - Katherine Fenstermacher
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| | - Nicholas Wohlgemuth
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| | - Andrew Nishida
- Department of Microbiology, University of Washington, Seattle, WA, USA
| | - Victoria Carter
- Department of Microbiology, University of Washington, Seattle, WA, USA
| | - Elise A Smith
- Department of Microbiology, University of Washington, Seattle, WA, USA
| | - Xinxia Peng
- Department of Microbiology, University of Washington, Seattle, WA, USA
| | - Melissa Hayes
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| | - Doreen Francis
- Department of Internal Medicine, University of Rochester, School of Medicine and Dentistry, Rochester, New York, USA
| | - John Treanor
- Department of Internal Medicine, University of Rochester, School of Medicine and Dentistry, Rochester, New York, USA
| | - Juliet Morrison
- Department of Microbiology, University of Washington, Seattle, WA, USA
| | - Sabra L Klein
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| | - Andrew Lane
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Michael G Katze
- Department of Microbiology, University of Washington, Seattle, WA, USA; Washington National Primate Research Center, University of Washington, Seattle, WA, USA
| | - Andrew Pekosz
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA; Department of Environmental Health Sciences, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA.
| |
Collapse
|
32
|
Kumar A, McElhaney JE, Walrond L, Cyr TD, Merani S, Kollmann TR, Halperin SA, Scheifele DW. Cellular immune responses of older adults to four influenza vaccines: Results of a randomized, controlled comparison. Hum Vaccin Immunother 2017. [PMID: 28635557 DOI: 10.1080/21645515.2017.1337615] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Cellular immunity is important for protection against the serious complications of influenza in older adults. As it is unclear if newer influenza vaccines elicit greater cellular responses than standard vaccines, we compared responses to 2 standard and 2 newer licensed trivalent inactivated vaccines (TIVs) in a randomized trial in older adults. Non-frail adults ≥ 65 y old were randomly assigned to receive standard subunit, MF59-adjuvanted subunit, standard split-virus or intradermal split-virus TIV. Peripheral blood mononuclear cells (PBMC) harvested pre- and 3-weeks post-vaccination were stimulated with live A/H3N2 virus. PBMC supernatants were tested for interleukin 10 (IL-10) and interferon gamma (IFN-γ), and lysates for granzyme B (GrB). Flow cytometry identified CD4+ and CD8+ T- cells expressing intracellular IL-2, IL-10, IFN-γ, GrB, or perforin. Differences following immunization were assessed for paired subject samples and among vaccines. 120 seniors participated, 29-31 per group, which were well matched demographically. Virus-stimulated PBMCs were GrB-rich before and after vaccination, with minimal increases evident. Immunization did not increase secretion of IFN-γ or IL-10. However, cytolytic effector T-cells (CD8+GrB+perforin+) increased significantly in percentage post-vaccination in all groups, to similar mean values across groups. CD4+GrB+perforin+ T-cells also increased significantly after each vaccine, to similar mean values among vaccines. Vaccination did not increase the low baseline percentages of CD4+ or CD8+ T-cells expressing IFN-γ, IL-2 or IL-10 . In conclusion, participants had pre-existing cellular immunity to H3N2 virus. All 4 vaccines boosted cellular responses to a similar but limited extent, particularly cytolytic effector CD8+ T-cells associated with clinical protection against influenza.
Collapse
Affiliation(s)
- Arun Kumar
- a Health Sciences North Research Institute , Sudbury , Ontario , Canada
| | - Janet E McElhaney
- a Health Sciences North Research Institute , Sudbury , Ontario , Canada.,b Northern Ontario School of Medicine , Sudbury , Ontario , Canada.,c VITALITY Research Center , Vancouver Coastal Health Research Institute , Vancouver , BC , Canada.,d Public Health Agency of Canada/Canadian Institutes of Health Research Influenza Research Network (PCIRN), Dalhousie University , Halifax , NS , Canada
| | - Lisa Walrond
- e Regulatory Research Division , Biologics and Genetic Therapies Directorate Health Canada , Ottawa , Canada
| | - Terry D Cyr
- e Regulatory Research Division , Biologics and Genetic Therapies Directorate Health Canada , Ottawa , Canada
| | - Shahzma Merani
- a Health Sciences North Research Institute , Sudbury , Ontario , Canada
| | - Tobias R Kollmann
- d Public Health Agency of Canada/Canadian Institutes of Health Research Influenza Research Network (PCIRN), Dalhousie University , Halifax , NS , Canada.,f Vaccine Evaluation Center , University of British Columbia , Vancouver , BC , Canada
| | - Scott A Halperin
- d Public Health Agency of Canada/Canadian Institutes of Health Research Influenza Research Network (PCIRN), Dalhousie University , Halifax , NS , Canada.,g Canadian Center for Vaccinology , Dalhousie University , Halifax , NS , Canada
| | - David W Scheifele
- d Public Health Agency of Canada/Canadian Institutes of Health Research Influenza Research Network (PCIRN), Dalhousie University , Halifax , NS , Canada.,f Vaccine Evaluation Center , University of British Columbia , Vancouver , BC , Canada
| |
Collapse
|
33
|
Andreassen M, Hjertholm H, Cravedi JP, Grob K, Alexander J, Nygaard UC. Effect of dietary pristane and other saturated mineral oils (MOSH) on autoimmune arthritis in rats. Toxicol Rep 2017; 4:104-112. [PMID: 28959631 PMCID: PMC5615100 DOI: 10.1016/j.toxrep.2017.02.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 01/12/2017] [Accepted: 02/13/2017] [Indexed: 12/01/2022] Open
Abstract
Pristane and other adjuvants based on mineral oil saturated hydrocarbons (MOSH) may induce autoimmunity in rodents after intradermal injection; however there is a lack of information on immune effects after oral MOSH exposure. The aim of our study was to determine the impact of dietary exposure to pristane and other MOSH on the development of autoimmune arthritis. Dark Agouti (DA) rats were given feed containing 4000 mg/kg pristane or a broad MOSH mixture in various concentrations (0–4000 mg/kg) for 90 days, or a single intradermal injection of 200 μl pristane (positive control). Arthritis scores, and serum and splenocyte markers previously associated with arthritis development, were determined. All rats injected with pristane displayed arthritis symptoms and higher levels of certain serum markers. None of the rats fed pristane or MOSH developed arthritis symptoms or demonstrated clear changes in any measured arthritis-associated biological markers in serum or splenocytes. The absence of clinical arthritis symptoms or any increase in common arthritis-associated biological markers in sera and spleen following dietary exposure to pristane or a broad MOSH mixture in a sub-chronic rat model of arthritis suggest that dietary MOSH have low capacity to promote development of autoimmunity.
Collapse
Affiliation(s)
- Monica Andreassen
- Norwegian Institute of Public Health, PO Box 4404, NO-0403 Oslo, Norway
| | - Hege Hjertholm
- Norwegian Institute of Public Health, PO Box 4404, NO-0403 Oslo, Norway
| | - Jean-Pierre Cravedi
- Toxalim, INRA, ENVT, INP-EIPurpan, Université de Toulouse, F-31027 Toulouse, France
| | - Koni Grob
- Official Food Control Authority of the Canton of Zurich, P.O. Box 1471, CH-8032 Zurich, Switzerland
| | - Jan Alexander
- Norwegian Institute of Public Health, PO Box 4404, NO-0403 Oslo, Norway
| | - Unni C Nygaard
- Norwegian Institute of Public Health, PO Box 4404, NO-0403 Oslo, Norway
| |
Collapse
|
34
|
Lee I, Il Kim J, Park S, Bae JY, Yoo K, Yun SH, Lee JY, Kim K, Kang C, Park MS. Single PA mutation as a high yield determinant of avian influenza vaccines. Sci Rep 2017; 7:40675. [PMID: 28084423 PMCID: PMC5233958 DOI: 10.1038/srep40675] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Accepted: 12/09/2016] [Indexed: 12/19/2022] Open
Abstract
Human infection with an avian influenza virus persists. To prepare for a potential outbreak of avian influenza, we constructed a candidate vaccine virus (CVV) containing hemagglutinin (HA) and neuraminidase (NA) genes of a H5N1 virus and evaluated its antigenic stability after serial passaging in embryonated chicken eggs. The passaged CVV harbored the four amino acid mutations (R136K in PB2; E31K in PA; A172T in HA; and R80Q in M2) without changing its antigenicity, compared with the parental CVV. Notably, the passaged CVV exhibited much greater replication property both in eggs and in Madin-Darby canine kidney and Vero cells. Of the four mutations, the PA E31K showed the greatest effect on the replication property of reverse genetically-rescued viruses. In a further luciferase reporter, mini-replicon assay, the PA mutation appeared to affect the replication property by increasing viral polymerase activity. When applied to different avian influenza CVVs (H7N9 and H9N2 subtypes), the PA E31K mutation resulted in the increases of viral replication in the Vero cell again. Taken all together, our results suggest the PA E31K mutation as a single, substantial growth determinant of avian influenza CVVs and for the establishment of a high-yield avian influenza vaccine backbone.
Collapse
Affiliation(s)
- Ilseob Lee
- Department of Microbiology, the Institute for Viral Diseases, College of Medicine, Korea University, Seoul 02841, Republic of Korea
| | - Jin Il Kim
- Department of Microbiology, the Institute for Viral Diseases, College of Medicine, Korea University, Seoul 02841, Republic of Korea
| | - Sehee Park
- Department of Microbiology, the Institute for Viral Diseases, College of Medicine, Korea University, Seoul 02841, Republic of Korea
| | - Joon-Yong Bae
- Department of Microbiology, the Institute for Viral Diseases, College of Medicine, Korea University, Seoul 02841, Republic of Korea
| | - Kirim Yoo
- Department of Microbiology, the Institute for Viral Diseases, College of Medicine, Korea University, Seoul 02841, Republic of Korea
| | - Soo-Hyeon Yun
- Department of Microbiology, the Institute for Viral Diseases, College of Medicine, Korea University, Seoul 02841, Republic of Korea
| | - Joo-Yeon Lee
- Division of Influenza Virus, Center for Infectious Diseases, National Institute of Health, Korea Centers for Disease Control and Prevention, Osong 28159, Republic of Korea
| | - Kisoon Kim
- Division of Influenza Virus, Center for Infectious Diseases, National Institute of Health, Korea Centers for Disease Control and Prevention, Osong 28159, Republic of Korea
| | - Chun Kang
- Division of AIDS, Center for Infectious Diseases, National Institute of Health, Korea Centers for Disease Control and Prevention, Osong 28159, Republic of Korea
| | - Man-Seong Park
- Department of Microbiology, the Institute for Viral Diseases, College of Medicine, Korea University, Seoul 02841, Republic of Korea
| |
Collapse
|
35
|
Nogales A, Martínez-Sobrido L. Reverse Genetics Approaches for the Development of Influenza Vaccines. Int J Mol Sci 2016; 18:E20. [PMID: 28025504 PMCID: PMC5297655 DOI: 10.3390/ijms18010020] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 12/15/2016] [Accepted: 12/19/2016] [Indexed: 12/20/2022] Open
Abstract
Influenza viruses cause annual seasonal epidemics and occasional pandemics of human respiratory disease. Influenza virus infections represent a serious public health and economic problem, which are most effectively prevented through vaccination. However, influenza viruses undergo continual antigenic variation, which requires either the annual reformulation of seasonal influenza vaccines or the rapid generation of vaccines against potential pandemic virus strains. The segmented nature of influenza virus allows for the reassortment between two or more viruses within a co-infected cell, and this characteristic has also been harnessed in the laboratory to generate reassortant viruses for their use as either inactivated or live-attenuated influenza vaccines. With the implementation of plasmid-based reverse genetics techniques, it is now possible to engineer recombinant influenza viruses entirely from full-length complementary DNA copies of the viral genome by transfection of susceptible cells. These reverse genetics systems have provided investigators with novel and powerful approaches to answer important questions about the biology of influenza viruses, including the function of viral proteins, their interaction with cellular host factors and the mechanisms of influenza virus transmission and pathogenesis. In addition, reverse genetics techniques have allowed the generation of recombinant influenza viruses, providing a powerful technology to develop both inactivated and live-attenuated influenza vaccines. In this review, we will summarize the current knowledge of state-of-the-art, plasmid-based, influenza reverse genetics approaches and their implementation to provide rapid, convenient, safe and more effective influenza inactivated or live-attenuated vaccines.
Collapse
Affiliation(s)
- Aitor Nogales
- Department of Microbiology and Immunology, University of Rochester, Rochester, NY 14642, USA.
| | - Luis Martínez-Sobrido
- Department of Microbiology and Immunology, University of Rochester, Rochester, NY 14642, USA.
| |
Collapse
|
36
|
Kim C, Fang F, Weyand CM, Goronzy JJ. The life cycle of a T cell after vaccination - where does immune ageing strike? Clin Exp Immunol 2016; 187:71-81. [PMID: 27324743 DOI: 10.1111/cei.12829] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/16/2016] [Indexed: 12/27/2022] Open
Abstract
Vaccination is the optimal intervention to prevent the increased morbidity and mortality from infection in older individuals and to maintain immune health during ageing. To optimize benefits from vaccination, strategies have to be developed that overcome the defects in an adaptive immune response that occur with immune ageing. Most current approaches are concentrated on activating the innate immune system by adjuvants to improve the induction of a T cell response. This review will focus upon T cell-intrinsic mechanisms that control how a T cell is activated, expands rapidly to differentiate into short-lived effector cells and into memory precursor cells, with short-lived effector T cells then mainly undergoing apoptosis and memory precursor cells surviving as long-lived memory T cells. Insights into each step of this longitudinal course of a T cell response that takes place over a period of several weeks is beginning to allow identifying interventions that can improve this process of T cell memory generation and specifically target defects that occur with ageing.
Collapse
Affiliation(s)
- C Kim
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University, Stanford, CA and the Department of Medicine, VAPAHCS, Palo Alto, CA, USA
| | - F Fang
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University, Stanford, CA and the Department of Medicine, VAPAHCS, Palo Alto, CA, USA
| | - C M Weyand
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University, Stanford, CA and the Department of Medicine, VAPAHCS, Palo Alto, CA, USA
| | - J J Goronzy
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University, Stanford, CA and the Department of Medicine, VAPAHCS, Palo Alto, CA, USA
| |
Collapse
|
37
|
Abstract
Obesity is a risk factor for developing severe influenza virus infection, making vaccination of utmost importance for this high-risk population. However, vaccinated obese animals and adults have decreased neutralizing antibody responses. In these studies, we tested the hypothesis that the addition of either alum or a squalene-based adjuvant (AS03) to an influenza vaccine would improve neutralizing antibody responses and protect obese mice from challenge. Our studies demonstrate that adjuvanted vaccine does increase both neutralizing and nonneutralizing antibody levels compared to vaccine alone. Although obese mice mount significantly decreased virus-specific antibody responses, both the breadth and the magnitude of the responses against hemagglutinin (HA) and neuraminidase (NA) are decreased compared to the responses in lean mice. Importantly, even with a greater than fourfold increase in neutralizing antibody levels, obese mice are not protected against influenza virus challenge and viral loads remain elevated in the respiratory tract. Increasing the antigen dose affords no added protection, and a decreasing viral dose did not fully mitigate the increased mortality seen in obese mice. Overall, these studies highlight that, while the use of an adjuvant does improve seroconversion, vaccination does not fully protect obese mice from influenza virus challenge, possibly due to the increased sensitivity of obese animals to infection. Given the continued increase in the global obesity epidemic, our findings have important implications for public health. Vaccination is the most effective strategy for preventing influenza virus infection and is a key component for pandemic preparedness. However, vaccines may fail to provide optimal protection in high-risk groups, including overweight and obese individuals. Given the worldwide obesity epidemic, it is imperative that we understand and improve vaccine efficacy. No work to date has investigated whether adjuvants increase the protective capacity of influenza vaccines in the obese host. In these studies, we show that adjuvants increased the neutralizing and nonneutralizing antibody responses during vaccination of lean and obese mice to levels considered “protective,” and yet, obese mice still succumbed to infection. This vulnerability is likely due to a combination of factors, including the increased susceptibility of obese animals to develop severe and even lethal disease when infected with very low viral titers. Our studies highlight the critical public health need to translate these findings and better understand vaccination in this increasing population.
Collapse
|
38
|
Salyer ACD, Caruso G, Khetani KK, Fox LM, Malladi SS, David SA. Identification of Adjuvantic Activity of Amphotericin B in a Novel, Multiplexed, Poly-TLR/NLR High-Throughput Screen. PLoS One 2016; 11:e0149848. [PMID: 26919709 PMCID: PMC4769227 DOI: 10.1371/journal.pone.0149848] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 02/05/2016] [Indexed: 01/26/2023] Open
Abstract
Small-molecule agonists have been identified for TLR7, TLR8, TLR4 and TLR2 thus far, and chemotypes other than those of canonical ligands are yet to be explored for a number of innate immune receptors. The discovery of novel immunostimulatory molecules would enhance the repertoire of tools available for interrogating innate immune effector mechanisms, and provide additional venues for vaccine adjuvant development. A multiplexed, reporter gene-based high-throughput assay capable of detecting agonists of TLR2, TLR3, TLR4, TLR5, TLR7, TLR8, TLR9, NOD1 and NOD2 was utilized in screening 123,943 compounds, in which amphotericin B (AmpB) and nystatin were identified as prominent hits. The polyene antifungal agents act as TLR2- and TLR4-agonists. The TLR4-stimulatory activity of AmpB was similar to that of monophosphoryl lipid A, suggestive of TRIF-biased signaling. The adjuvantic activity of AmpB, at a dose of 100 micrograms, was comparable to several other candidate adjuvants in rabbit models of immunization. These results point to its potential applicability as a safe and effective adjuvant for human vaccines.
Collapse
Affiliation(s)
- Alex C. D. Salyer
- Department of Medicinal Chemistry, University of Kansas, Lawrence, Kansas, United States of America
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Giuseppe Caruso
- Department of Medicinal Chemistry, University of Kansas, Lawrence, Kansas, United States of America
| | - Karishma K. Khetani
- Department of Medicinal Chemistry, University of Kansas, Lawrence, Kansas, United States of America
| | - Lauren M. Fox
- Department of Medicinal Chemistry, University of Kansas, Lawrence, Kansas, United States of America
| | - Subbalakshmi S. Malladi
- Department of Medicinal Chemistry, University of Kansas, Lawrence, Kansas, United States of America
| | - Sunil A. David
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, Minnesota, United States of America
- * E-mail:
| |
Collapse
|
39
|
Walkowicz WE, Fernández-Tejada A, George C, Corzana F, Jiménez-Barbero J, Ragupathi G, Tan DS, Gin DY. Quillaja Saponin Variants with Central Glycosidic Linkage Modifications Exhibit Distinct Conformations and Adjuvant Activities. Chem Sci 2016; 7:2371-2380. [PMID: 27014435 PMCID: PMC4800320 DOI: 10.1039/c5sc02978c] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
A mouse vaccination model and molecular dynamics studies reveal characteristic conformations of active QS-21 variants.
Immunological adjuvants such as the saponin natural product QS-21 help stimulate the immune response to co-administered antigens and have become increasingly important in the development of prophylactic and therapeutic vaccines. However, clinical use of QS-21 is encumbered by chemical instability, dose-limiting toxicity, and low-yielding purification from the natural source. Previous studies of structure–activity relationships in the four structural domains of QS-21 have led to simplified, chemically stable variants that retain potent adjuvant activity and low toxicity in mouse vaccination models. However, modification of the central glycosyl ester linkage has not yet been explored. Herein, we describe the design, synthesis, immunologic evaluation, and molecular dynamics analysis of a series of novel QS-21 variants with different linker lengths, stereochemistry, and flexibility to investigate the role of this linkage in saponin adjuvant activity and conformation. Despite relatively conservative structural modifications, these variants exhibit striking differences in in vivo adjuvant activity that correlate with specific conformational preferences. These results highlight the junction of the triterpene and linear oligosaccharide domains as playing a critical role in the immunoadjuvant activity of the Quillaja saponins and also suggest a mechanism of action involving interaction with a discrete macromolecular target, in contrast to the non-specific mechanisms of emulsion-based adjuvants.
Collapse
Affiliation(s)
- William E Walkowicz
- Louis V. Gerstner, Jr. Graduate School of Biomedical Sciences, 1275 York Avenue, New York, NY 10065, United States
| | - Alberto Fernández-Tejada
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, United States; Chemical & Physical Biology, Center for Biological Research, CIB-CSIC, 28040 Madrid, Spain
| | - Constantine George
- Melanoma & Immunotherapeutics Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, United States
| | - Francisco Corzana
- Department of Chemistry, University of La Rioja, 26006 Logroño, Spain
| | - Jesús Jiménez-Barbero
- Chemical & Physical Biology, Center for Biological Research, CIB-CSIC, 28040 Madrid, Spain; Structural Biology, Center for Cooperative Research, CIC-bioGUNE, 48160 Derio-Bizkaia, Spain; Ikerbasque, Basque Foundation for Science, 48013 Bilbao, Spain
| | - Govind Ragupathi
- Melanoma & Immunotherapeutics Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, United States
| | - Derek S Tan
- Louis V. Gerstner, Jr. Graduate School of Biomedical Sciences, 1275 York Avenue, New York, NY 10065, United States; Chemical Biology Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, United States; Tri-Institutional Research Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, United States
| | - David Y Gin
- Louis V. Gerstner, Jr. Graduate School of Biomedical Sciences, 1275 York Avenue, New York, NY 10065, United States; Chemical Biology Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, United States; Tri-Institutional Research Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, United States
| |
Collapse
|
40
|
Abstract
In this Minireview, we discuss basic aspects of germinal center biology in the context of immunity to influenza infection and speculate on how the simultaneous evolutionary races of virus and antibody may impact our efforts to design a universal influenza vaccine.
Collapse
Affiliation(s)
- Gabriel D Victora
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA.
| | - Patrick C Wilson
- Department of Medicine, Section of Rheumatology, The Committee on Immunology, The Knapp Center for Lupus and Immunology Research, The University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
41
|
Lofano G, Kumar A, Finco O, Del Giudice G, Bertholet S. B Cells and Functional Antibody Responses to Combat Influenza. Front Immunol 2015; 6:336. [PMID: 26175732 PMCID: PMC4485180 DOI: 10.3389/fimmu.2015.00336] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2015] [Accepted: 06/15/2015] [Indexed: 12/22/2022] Open
Abstract
Vaccination against influenza is the most effective way to protect the population. Current vaccines provide protection by stimulating functional B- and T-cell responses; however, they are poorly immunogenic in particular segments of the population and need to be reformulated almost every year due to the genetic instability of the virus. Next-generation influenza vaccines should be designed to induce cross-reactivity, confer protection against pandemic outbreaks, and promote long-lasting immune responses among individuals at higher risk of infection. Multiple strategies are being developed for the induction of broad functional humoral immunity, including the use of adjuvants, heterologous prime-boost strategies, and epitope-based antigen design. The basic approach is to mimic natural responses to influenza virus infection by promoting cross-reactive neutralizing antibodies that directly prevent the infection. This review provides an overview of the mechanisms underlying humoral responses to influenza vaccination or natural infection, and discusses promising strategies to control influenza virus.
Collapse
Affiliation(s)
- Giuseppe Lofano
- Research Center, Novartis Vaccines and Diagnostics S.r.l. (a GSK Company), Siena, Italy
- Dipartimento di Biologia e Biotecnologie “Charles Darwin”, Università degli Studi di Roma “La Sapienza”, Rome, Italy
| | - Arun Kumar
- Research Center, Novartis Vaccines and Diagnostics S.r.l. (a GSK Company), Siena, Italy
| | - Oretta Finco
- Research Center, Novartis Vaccines and Diagnostics S.r.l. (a GSK Company), Siena, Italy
| | - Giuseppe Del Giudice
- Research Center, Novartis Vaccines and Diagnostics S.r.l. (a GSK Company), Siena, Italy
| | - Sylvie Bertholet
- Research Center, Novartis Vaccines and Diagnostics S.r.l. (a GSK Company), Siena, Italy
| |
Collapse
|
42
|
Abstract
Influenza virus infections are a major public health concern and cause significant morbidity and mortality worldwide. Current influenza virus vaccines are an effective countermeasure against infection but need to be reformulated almost every year owing to antigenic drift. Furthermore, these vaccines do not protect against novel pandemic strains, and the timely production of pandemic vaccines remains problematic because of the limitations of current technology. Several improvements have been made recently to enhance immune protection induced by seasonal and pandemic vaccines, and to speed up production in case of a pandemic. Importantly, vaccine constructs that induce broad or even universal influenza virus protection are currently in preclinical and clinical development.
Collapse
|