1
|
Adediran E, Arte T, Pasupuleti D, Vijayanand S, Singh R, Patel P, Gulani M, Ferguson A, Uddin M, Zughaier SM, D’Souza MJ. Delivery of PLGA-Loaded Influenza Vaccine Microparticles Using Dissolving Microneedles Induces a Robust Immune Response. Pharmaceutics 2025; 17:510. [PMID: 40284505 PMCID: PMC12030082 DOI: 10.3390/pharmaceutics17040510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Revised: 04/04/2025] [Accepted: 04/09/2025] [Indexed: 04/29/2025] Open
Abstract
Background: Influenza virus is one of the major respiratory virus infections that is a global health concern. Although there are already approved vaccines, most are administered via the intramuscular route, which is usually painful, leading to vaccine hesitancy. To this end, exploring the non-invasive, transdermal vaccination route using dissolving microneedles would significantly improve vaccine compliance. Research on innovative vaccine delivery systems, such as antigen-loaded PLGA microparticles, has the potential to pave the way for a broader range of vaccine candidates. Methods: In this proof-of-concept study, a combination of the inactivated influenza A H1N1 virus and inactivated influenza A H3N2 virus were encapsulated in a biodegradable poly (lactic-co-glycolic acid) (PLGA) polymeric matrix within microparticles, which enhanced antigen presentation. The antigen PLGA microparticles were prepared separately using a double emulsion (w/o/w), lyophilized, and characterized. Next, the vaccine microparticles were assessed in vitro in dendritic cells (DC 2.4) for immunogenicity. To explore pain-free transdermal vaccination, the vaccine microparticles were loaded into dissolving microneedles and administered in mice (n = 5). Results: Our vaccination study demonstrated that the microneedle-based vaccine elicited strong humoral responses as demonstrated by high antigen-specific IgA, IgG, IgG1, and IgG2a antibodies in serum samples and IgA in lung supernatant. Further, the vaccine also elicited a strong cellular response as evidenced by high levels of CD4+ and CD8a+ T cells in lymphoid organs such as the lymph nodes and spleen. Conclusion: The delivery of influenza vaccine-loaded PLGA microparticles using microneedles would be beneficial to individuals experiencing needle-phobia, as well as the geriatric and pediatric population.
Collapse
Affiliation(s)
- Emmanuel Adediran
- Center for Drug Delivery Research, Vaccine Nanotechnology Laboratory, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA; (E.A.); (T.A.); (D.P.); (S.V.); (R.S.); (P.P.); (M.G.); (A.F.); (M.U.)
| | - Tanisha Arte
- Center for Drug Delivery Research, Vaccine Nanotechnology Laboratory, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA; (E.A.); (T.A.); (D.P.); (S.V.); (R.S.); (P.P.); (M.G.); (A.F.); (M.U.)
| | - Dedeepya Pasupuleti
- Center for Drug Delivery Research, Vaccine Nanotechnology Laboratory, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA; (E.A.); (T.A.); (D.P.); (S.V.); (R.S.); (P.P.); (M.G.); (A.F.); (M.U.)
| | - Sharon Vijayanand
- Center for Drug Delivery Research, Vaccine Nanotechnology Laboratory, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA; (E.A.); (T.A.); (D.P.); (S.V.); (R.S.); (P.P.); (M.G.); (A.F.); (M.U.)
| | - Revanth Singh
- Center for Drug Delivery Research, Vaccine Nanotechnology Laboratory, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA; (E.A.); (T.A.); (D.P.); (S.V.); (R.S.); (P.P.); (M.G.); (A.F.); (M.U.)
| | - Parth Patel
- Center for Drug Delivery Research, Vaccine Nanotechnology Laboratory, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA; (E.A.); (T.A.); (D.P.); (S.V.); (R.S.); (P.P.); (M.G.); (A.F.); (M.U.)
| | - Mahek Gulani
- Center for Drug Delivery Research, Vaccine Nanotechnology Laboratory, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA; (E.A.); (T.A.); (D.P.); (S.V.); (R.S.); (P.P.); (M.G.); (A.F.); (M.U.)
| | - Amarae Ferguson
- Center for Drug Delivery Research, Vaccine Nanotechnology Laboratory, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA; (E.A.); (T.A.); (D.P.); (S.V.); (R.S.); (P.P.); (M.G.); (A.F.); (M.U.)
| | - Mohammad Uddin
- Center for Drug Delivery Research, Vaccine Nanotechnology Laboratory, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA; (E.A.); (T.A.); (D.P.); (S.V.); (R.S.); (P.P.); (M.G.); (A.F.); (M.U.)
| | - Susu M. Zughaier
- College of Medicine, QU Health, Qatar University, Doha P.O. Box 2713, Qatar;
| | - Martin J. D’Souza
- Center for Drug Delivery Research, Vaccine Nanotechnology Laboratory, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA; (E.A.); (T.A.); (D.P.); (S.V.); (R.S.); (P.P.); (M.G.); (A.F.); (M.U.)
| |
Collapse
|
2
|
Taaffe J, Goldin S, Lambach P, Sparrow E. Global production capacity of seasonal and pandemic influenza vaccines in 2023. Vaccine 2025; 51:126839. [PMID: 39970592 PMCID: PMC11895838 DOI: 10.1016/j.vaccine.2025.126839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 01/26/2025] [Accepted: 01/29/2025] [Indexed: 02/21/2025]
Abstract
INTRODUCTION Vaccination is a critical part of the response to an influenza pandemic. Future influenza pandemics will likely leverage existing production processes and manufacturing facilities for seasonal influenza to make pandemic vaccines. Therefore, pandemic influenza vaccine response is heavily dependent on seasonal influenza vaccine production capacity. METHODS WHO monitors global vaccine production to inform pandemic preparedness by regularly surveying influenza vaccine manufacturers to estimate both seasonal and potential pandemic vaccine production capacity overall and by region, vaccine type, and manufacturing process. The last survey estimates were for 2019; here, we report updated estimates based on data from the 2023 survey and compare to estimates from previous surveys. RESULTS Our analysis estimates that annual seasonal influenza vaccine production capacity has remained relatively stable since 2019 at 1.53 billion doses and pandemic vaccine capacity at 4.13 and 8.26 billion doses for moderate and best case scenarios, respectively. Over 80 % of seasonal and pandemic vaccine production capacity relies on embryonated eggs, and inactivated influenza virus vaccines comprise the majority of vaccine supply. There is influenza vaccine manufacturing capacity in all WHO regions, except for the African Region, though influenza vaccine production is concentrated in high and upper-middle income countries. The ability to achieve maximum production capacity could be hindered by access to eggs and other ancillary supplies. CONCLUSIONS While influenza vaccine production capacity has been sustained since 2019, significant gaps persist in its distribution, especially in low and lower-middle income countries, and most notably in the African region. This imbalance in production could result in unequal access to vaccines in the event of a pandemic. Strengthening local vaccine manufacturing, promoting seasonal vaccination programmes, and investing in research and development of next-generation influenza vaccines or improved production platforms are essential to improve pandemic preparedness, sustain the influenza vaccine market, and enable more robust local responses.
Collapse
|
3
|
Khalifa AZ, Perrie Y, Shahiwala A. Subunit antigen delivery: emulsion and liposomal adjuvants for next-generation vaccines. Expert Opin Drug Deliv 2025; 22:583-597. [PMID: 40021342 DOI: 10.1080/17425247.2025.2474088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 02/26/2025] [Indexed: 03/03/2025]
Abstract
INTRODUCTION Developing new vaccines to combat emerging infectious diseases has gained more significance after the COVID-19 pandemic. Vaccination is the most cost-effective method for preventing infectious diseases, and subunit antigens are a safer alternative to traditional live, attenuated, and inactivated vaccines. AREAS COVERED Challenges in delivering subunit antigens and the status of different vaccine adjuvants. Recent research developments involving emulsion and liposomal adjuvants and their compositions and properties affecting their adjuvancy. EXPERT OPINION Lipid-based adjuvants, e.g. emulsions and liposomes, represent a paradigm shift in vaccine technology by enabling robust humoral and cellular immune responses with lower antigen doses, a property that is particularly critical during pandemics or in resource-limited settings. These adjuvants can optimize vaccine administration strategies by potentially reducing the frequency of booster doses, thereby improving patient compliance and lowering healthcare costs. While emulsions excel in dose-sparing and broadening immune responses, liposomes offer customization and precision in antigen delivery. However, the broader clinical application of these technologies is not without challenges. Stability issues, e.g. the susceptibility of emulsion-based adjuvants to freezing and their reliance on cold-chain logistics, pose significant barriers to their use in remote/underserved regions. Future developments will likely focus on improving manufacturing scalability and cost-effectiveness.
Collapse
Affiliation(s)
- Al Zahraa Khalifa
- Department of Pharmaceutical Sciences, Dubai Pharmacy College for Girls, Dubai Medical University, Dubai, United Arab Emirates
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Yvonne Perrie
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Aliasgar Shahiwala
- Department of Pharmaceutical Sciences, Dubai Pharmacy College for Girls, Dubai Medical University, Dubai, United Arab Emirates
| |
Collapse
|
4
|
Chen Y, Feng D, Cheng Y, Jiang X, Qiu L, Zhang L, Shi D, Wang J. Research progress of metal-CpG composite nanoadjuvants in tumor immunotherapy. Biomater Sci 2025; 13:1605-1623. [PMID: 39998438 DOI: 10.1039/d4bm01399a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/26/2025]
Abstract
The practical benefits and therapeutic potential of tumor vaccines in immunotherapy have drawn significant attention in the field of cancer treatment. Among the available vaccines, nanovaccines that utilize nanoparticles as carriers or adjuvants have demonstrated considerable effectiveness in combating cancer. Cytosine-phosphate-guanine oligodeoxynucleotide (CpG ODN), a common adjuvant in tumor nanovaccines, activates both humoral and cellular immunity by recognizing toll-like receptor 9 (TLR9), thereby aiding in the prevention and treatment of cancer. Metal nanoparticles hold great promise in tumor immunotherapy due to their adjustable size, surface functionalization, ability to regulate innate immunity, and capacity for controlled delivery of antigens or immunomodulators. Consequently, composite nanoadjuvants, formed by combining metal nanoparticles with CpG ODNs, can be customized to meet the specific performance requirements of different application scenarios, effectively overcoming the limitations of conventional immunotherapy approaches. This review provides a comprehensive analysis of the critical role of metal-CpG composite nanoadjuvants in advancing vaccine adjuvants for cancer therapy and prevention, highlighting their efficacy in preclinical settings.
Collapse
Affiliation(s)
- Yifan Chen
- School of Pharmacy, Changzhou University, Changzhou 213164, China.
| | - Danna Feng
- School of Pharmacy, Changzhou University, Changzhou 213164, China.
| | - Yilin Cheng
- School of Pharmacy, Changzhou University, Changzhou 213164, China.
| | - Xianmeng Jiang
- School of Pharmacy, Changzhou University, Changzhou 213164, China.
| | - Lin Qiu
- School of Pharmacy, Changzhou University, Changzhou 213164, China.
| | - Li Zhang
- School of Pharmacy, Changzhou University, Changzhou 213164, China.
- Key Laboratory of Synthetic and Biological Colloids, Ministry of Education, Jiangnan University, Wuxi 214122, China
| | - Dongjian Shi
- Key Laboratory of Synthetic and Biological Colloids, Ministry of Education, Jiangnan University, Wuxi 214122, China
| | - Jianhao Wang
- School of Pharmacy, Changzhou University, Changzhou 213164, China.
| |
Collapse
|
5
|
Ridelfi M, Pierleoni G, Zucconi Galli Fonseca V, Batani G, Rappuoli R, Sala C. State of the Art and Emerging Technologies in Vaccine Design for Respiratory Pathogens. Semin Respir Crit Care Med 2025. [PMID: 39870103 DOI: 10.1055/a-2500-1878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2025]
Abstract
In this review, we present the efforts made so far in developing effective solutions to prevent infections caused by seven major respiratory pathogens: influenza virus, respiratory syncytial virus (RSV), the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), Bordetella pertussis, Streptococcus pneumoniae (pneumococcus), Mycobacterium tuberculosis, and Pseudomonas aeruginosa. Advancements driven by the recent coronavirus disease 2019 (COVID-19) crisis have largely focused on viruses, but effective prophylactic solutions for bacterial pathogens are also needed, especially in light of the antimicrobial resistance (AMR) phenomenon. Here, we discuss various innovative key technologies that can help address this critical need, such as (a) the development of Lung-on-Chip ex vivo models to gain a better understanding of the pathogenesis process and the host-microbe interactions; (b) a more thorough investigation of the mechanisms behind mucosal immunity as the first line of defense against pathogens; (c) the identification of correlates of protection (CoPs) which, in conjunction with the Reverse Vaccinology 2.0 approach, can push a more rational and targeted design of vaccines. By focusing on these critical areas, we expect substantial progress in the development of new vaccines against respiratory bacterial pathogens, thereby enhancing global health protection in the framework of the increasingly concerning AMR emergence.
Collapse
Affiliation(s)
- Matteo Ridelfi
- Monoclonal Antibody Discovery (MAD) Lab, Fondazione Toscana Life Sciences, Siena, Italy
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Giulio Pierleoni
- Monoclonal Antibody Discovery (MAD) Lab, Fondazione Toscana Life Sciences, Siena, Italy
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | | | - Giampiero Batani
- Monoclonal Antibody Discovery (MAD) Lab, Fondazione Toscana Life Sciences, Siena, Italy
| | | | - Claudia Sala
- Monoclonal Antibody Discovery (MAD) Lab, Fondazione Toscana Life Sciences, Siena, Italy
| |
Collapse
|
6
|
Wells TJ, Esposito T, Henderson IR, Labzin LI. Mechanisms of antibody-dependent enhancement of infectious disease. Nat Rev Immunol 2025; 25:6-21. [PMID: 39122820 DOI: 10.1038/s41577-024-01067-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/04/2024] [Indexed: 08/12/2024]
Abstract
Antibody-dependent enhancement (ADE) of infectious disease is a phenomenon whereby host antibodies increase the severity of an infection. It is well established in viral infections but ADE also has an underappreciated role during bacterial, fungal and parasitic infections. ADE can occur during both primary infections and re-infections with the same or a related pathogen; therefore, understanding the underlying mechanisms of ADE is critical for understanding the pathogenesis and progression of many infectious diseases. Here, we review the four distinct mechanisms by which antibodies increase disease severity during an infection. We discuss the most established mechanistic explanation for ADE, where cross-reactive, disease-enhancing antibodies bound to pathogens interact with Fc receptors, thereby enhancing pathogen entry or replication, ultimately increasing the total pathogen load. Additionally, we explore how some pathogenic antibodies can shield bacteria from complement-dependent killing, thereby enhancing bacterial survival. We interrogate the molecular mechanisms by which antibodies can amplify inflammation to drive severe disease, even in the absence of increased pathogen replication. We also examine emerging roles for autoantibodies in enhancing the pathogenesis of infectious diseases. Finally, we discuss how we can leverage these insights to improve vaccine design and future treatments for infectious diseases.
Collapse
Affiliation(s)
- Timothy J Wells
- Frazer Institute, The University of Queensland, Brisbane, Queensland, Australia.
| | - Tyron Esposito
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Ian R Henderson
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Larisa I Labzin
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia.
| |
Collapse
|
7
|
Zhang Y, Gao J, Xu W, Huo X, Wang J, Xu Y, Ding W, Guo Z, Liu R. Advances in protein subunit vaccines against H1N1/09 influenza. Front Immunol 2024; 15:1499754. [PMID: 39650643 PMCID: PMC11621219 DOI: 10.3389/fimmu.2024.1499754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Accepted: 11/05/2024] [Indexed: 12/11/2024] Open
Abstract
The A/H1N1pdm09 influenza virus, which caused the 2009 pandemic, has since become a recurring strain in seasonal influenza outbreaks. Given the ongoing threat of influenza, protein subunit vaccines have garnered significant attention for their safety and effectiveness. This review seeks to highlight the latest developments in protein subunit vaccines that specifically target the A/H1N1pdm09 virus. It will also examine the structure and replication cycle of influenza A viruses and compare different types of influenza vaccines. Additionally, the review will address key aspects of H1N1 protein subunit vaccine development, such as antigen selection, protein expression systems, and the use of adjuvants. The role of animal models in evaluating these vaccines will also be discussed. Despite challenges like antigenic variability and the complexities of vaccine production and distribution, protein subunit vaccines remain a promising option for future influenza prevention efforts.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Immunology, College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, China
- Department of Medical Imaging, School of Medicine, Zhoukou Vocational and Technical College, Zhoukou, China
| | - Jingyao Gao
- Department of Immunology, College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, China
| | - Wenqi Xu
- Department of Immunology, College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, China
| | - Xingyu Huo
- Department of Immunology, College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, China
| | - Jingyan Wang
- Department of Immunology, College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, China
| | - Yirui Xu
- Department of Immunology, College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, China
| | - Wenting Ding
- Department of Immunology, College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, China
| | - Zeliang Guo
- Department of Immunology, College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, China
| | - Rongzeng Liu
- Department of Immunology, College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, China
| |
Collapse
|
8
|
Singh A, Boggiano C, Yin DE, Polakowski L, Majji SP, Leitner WW, Levy O, De Paris K. Precision adjuvants for pediatric vaccines. Sci Transl Med 2024; 16:eabq7378. [PMID: 39231242 PMCID: PMC11911902 DOI: 10.1126/scitranslmed.abq7378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 08/06/2024] [Indexed: 09/06/2024]
Abstract
Elucidating optimal vaccine adjuvants for harnessing age-specific immune pathways to enhance magnitude, breadth, and durability of immunogenicity remains a key gap area in pediatric vaccine design. A better understanding of age-specific adjuvants will inform precision discovery and development of safe and effective vaccines for protecting children from preventable infectious diseases.
Collapse
Affiliation(s)
- Anjali Singh
- Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20852, USA
| | - César Boggiano
- Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20852, USA
| | - Dwight E. Yin
- Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20852, USA
| | - Laura Polakowski
- Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20852, USA
| | - Sai P. Majji
- Maternal and Pediatric Infectious Disease Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20817, USA
| | - Wolfgang W. Leitner
- Division of Allergy, Immunology, and Transplantation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20852, USA
| | - Ofer Levy
- Precision Vaccines Program, Department of Pediatrics, Boston Children’s Hospital, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Kristina De Paris
- Department of Microbiology and Immunology, School of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
| |
Collapse
|
9
|
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
|
10
|
Coelho CH, Marquez S, Nguemwo Tentokam BC, Berhe AD, Miura K, Rao VN, Long CA, Doumbo OK, Sagara I, Healy S, Kleinstein SH, Duffy PE. Antibody gene features associated with binding and functional activity in malaria vaccine-derived human mAbs. NPJ Vaccines 2024; 9:144. [PMID: 39127706 PMCID: PMC11316794 DOI: 10.1038/s41541-024-00929-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 07/17/2024] [Indexed: 08/12/2024] Open
Abstract
The impact of adjuvants on malaria vaccine-induced antibody repertoire is poorly understood. Here, we characterize the impact of two adjuvants, Alhydrogel® and AS01, on antibody clonotype diversity, binding and function, post malaria vaccination. We expressed 132 recombinant anti-Pfs230D1 human monoclonal antibodies (mAbs) from participants immunized with malaria transmission-blocking vaccine Pfs230D1, formulated with either Alhydrogel® or AS01. Anti-Pfs230D1 mAbs generated by Alhydrogel® formulation showed higher binding frequency to Pfs230D1 compared to AS01 formulation, although the frequency of functional mAbs was similar between adjuvant groups. Overall, the AS01 formulation induced anti-Pfs230D1 functional antibodies from a broader array of germline sequences versus the Alhydrogel® formulation. All mAbs using IGHV1-69 gene from the Alhydrogel® cohort bound to recombinant Pfs230D1, but did not block parasite transmission to mosquitoes, similar to the IGHV1-69 mAbs isolated from the AS01 cohort. These findings may help inform vaccine design and adjuvant selection for immunization with Plasmodium antigens.
Collapse
Affiliation(s)
- Camila H Coelho
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- C-VARPP- Center for Vaccine Research and Pandemic Preparedness, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Immunology Precision Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Susanna Marquez
- Department of Pathology, Yale School of Medicine, New Haven, CT, 06520, USA
| | - Bergeline C Nguemwo Tentokam
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Anne D Berhe
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Kazutoyo Miura
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, NIH, Rockville, MD, USA
| | - Vishal N Rao
- C-VARPP- Center for Vaccine Research and Pandemic Preparedness, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Carole A Long
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, NIH, Rockville, MD, USA
| | - Ogobara K Doumbo
- Malaria Research and Training Center, University of Sciences, Techniques and Technologies, Bamako, Mali
| | - Issaka Sagara
- Malaria Research and Training Center, University of Sciences, Techniques and Technologies, Bamako, Mali
| | - Sara Healy
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Steven H Kleinstein
- Department of Pathology, Yale School of Medicine, New Haven, CT, 06520, USA
- Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, 06511, USA
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, 06520, USA
| | - Patrick E Duffy
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
11
|
Zimna M, Brzuska G, Salát J, Růžek D, Krol E. Influence of adjuvant type and route of administration on the immunogenicity of Leishmania-derived tick-borne encephalitis virus-like particles - A recombinant vaccine candidate. Antiviral Res 2024; 228:105941. [PMID: 38901737 DOI: 10.1016/j.antiviral.2024.105941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 05/16/2024] [Accepted: 06/17/2024] [Indexed: 06/22/2024]
Abstract
Tick-borne encephalitis virus (TBEV) is a tick-borne flavivirus that induces severe central nervous system disorders. It has recently raised concerns due to an expanding geographical range and increasing infection rates. Existing vaccines, though effective, face low coverage rates in numerous TBEV endemic regions. Our previous work demonstrated the immunogenicity and full protection afforded by a TBEV vaccine based on virus-like particles (VLPs) produced in Leishmania tarentolae cells in immunization studies in a mouse model. In the present study, we explored the impact of adjuvants (AddaS03™, Alhydrogel®+MPLA) and administration routes (subcutaneous, intramuscular) on the immune response. Adjuvanted groups exhibited significantly enhanced antibody responses, higher avidity, and more balanced Th1/Th2 response. IFN-γ responses depended on the adjuvant type, while antibody levels were influenced by both adjuvant and administration routes. The combination of Leishmania-derived TBEV VLPs with Alhydrogel® and MPLA via intramuscular administration emerged as a highly promising prophylactic vaccine candidate, eliciting a robust, balanced immune response with substantial neutralization potential.
Collapse
MESH Headings
- Animals
- Encephalitis Viruses, Tick-Borne/immunology
- Mice
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- Adjuvants, Immunologic/administration & dosage
- Vaccines, Synthetic/immunology
- Vaccines, Synthetic/administration & dosage
- Encephalitis, Tick-Borne/prevention & control
- Encephalitis, Tick-Borne/immunology
- Viral Vaccines/immunology
- Viral Vaccines/administration & dosage
- Vaccines, Virus-Like Particle/immunology
- Vaccines, Virus-Like Particle/administration & dosage
- Leishmania/immunology
- Female
- Adjuvants, Vaccine/administration & dosage
- Antibodies, Neutralizing/blood
- Antibodies, Neutralizing/immunology
- Immunogenicity, Vaccine
- Injections, Intramuscular
- Mice, Inbred BALB C
- Interferon-gamma/immunology
- Th1 Cells/immunology
Collapse
Affiliation(s)
- Marta Zimna
- Department of Recombinant Vaccines, Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, Abrahama 58, 80-307, Gdansk, Poland.
| | - Gabriela Brzuska
- Department of Recombinant Vaccines, Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, Abrahama 58, 80-307, Gdansk, Poland.
| | - Jiří Salát
- Laboratory of Emerging Viral Infections, Veterinary Research Institute, Hudcova 70, CZ-62100, Brno, Czech Republic; Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Branisovska 31, CZ-37005, Ceske Budejovice, Czech Republic.
| | - Daniel Růžek
- Laboratory of Emerging Viral Infections, Veterinary Research Institute, Hudcova 70, CZ-62100, Brno, Czech Republic; Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Branisovska 31, CZ-37005, Ceske Budejovice, Czech Republic; Department of Experimental Biology, Faculty of Science, Masaryk University, Kamenice 735/5, CZ-62500, Brno, Czech Republic.
| | - Ewelina Krol
- Department of Recombinant Vaccines, Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, Abrahama 58, 80-307, Gdansk, Poland.
| |
Collapse
|
12
|
Li Y, Tian S, Ai Y, Hu Z, Ma C, Fu M, Xu Z, Li Y, Liu S, Zou Y, Zhou Y, Jin J. A nanoparticle vaccine displaying varicella-zoster virus gE antigen induces a superior cellular immune response than a licensed vaccine in mice and non-human primates. Front Immunol 2024; 15:1419634. [PMID: 39081325 PMCID: PMC11286566 DOI: 10.3389/fimmu.2024.1419634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 07/01/2024] [Indexed: 08/02/2024] Open
Abstract
Herpes zoster (HZ), also known as shingles, remains a significant global health issue and most commonly seen in elderly individuals with an early exposure history to varicella-zoster virus (VZV). Currently, the licensed vaccine Shingrix, which comprises a recombinant VZV glycoprotein E (gE) formulated with a potent adjuvant AS01B, is the most effective shingles vaccine on the market. However, undesired reactogenicity and increasing global demand causing vaccine shortage, prompting the development of novel shingles vaccines. Here, we developed novel vaccine candidates utilising multiple nanoparticle (NP) platforms to display the recombinant gE antigen, formulated in an MF59-biosimilar adjuvant. In naïve mice, all tested NP vaccines induced higher humoral and cellular immune responses than Shingrix, among which, the gEM candidate induced the highest cellular response. In live attenuated VZV (VZV LAV)-primed mouse and rhesus macaque models, the gEM candidate elicited superior cell-mediated immunity (CMI) over Shingrix. Collectively, we demonstrated that NP technology remains a suitable tool for developing shingles vaccine, and the reported gEM construct is a highly promising candidate in the next-generation shingles vaccine development.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Jing Jin
- Patronus Biotech Co. Ltd., Guangzhou, China
| |
Collapse
|
13
|
Soni D, Borriello F, Scott DA, Feru F, DeLeon M, Brightman SE, Cheng WK, Melhem G, Smith JA, Ramirez JC, Barman S, Cameron M, Kelly A, Walker K, Nanishi E, van Haren SD, Phan T, Qi Y, Kinsey R, Raczy MM, Ozonoff A, Pettengill MA, Hubbell JA, Fox CB, Dowling DJ, Levy O. From hit to vial: Precision discovery and development of an imidazopyrimidine TLR7/8 agonist adjuvant formulation. SCIENCE ADVANCES 2024; 10:eadg3747. [PMID: 38959314 PMCID: PMC11221515 DOI: 10.1126/sciadv.adg3747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Accepted: 05/29/2024] [Indexed: 07/05/2024]
Abstract
Vaccination can help prevent infection and can also be used to treat cancer, allergy, and potentially even drug overdose. Adjuvants enhance vaccine responses, but currently, the path to their advancement and development is incremental. We used a phenotypic small-molecule screen using THP-1 cells to identify nuclear factor-κB (NF-κB)-activating molecules followed by counterscreening lead target libraries with a quantitative tumor necrosis factor immunoassay using primary human peripheral blood mononuclear cells. Screening on primary cells identified an imidazopyrimidine, dubbed PVP-037. Moreover, while PVP-037 did not overtly activate THP-1 cells, it demonstrated broad innate immune activation, including NF-κB and cytokine induction from primary human leukocytes in vitro as well as enhancement of influenza and SARS-CoV-2 antigen-specific humoral responses in mice. Several de novo synthesis structural enhancements iteratively improved PVP-037's in vitro efficacy, potency, species-specific activity, and in vivo adjuvanticity. Overall, we identified imidazopyrimidine Toll-like receptor-7/8 adjuvants that act in synergy with oil-in-water emulsion to enhance immune responses.
Collapse
Affiliation(s)
- Dheeraj Soni
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Francesco Borriello
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - David A. Scott
- Harvard Medical School, Boston, MA, USA
- Dana-Farber Cancer Institute, Boston, MA, USA
| | - Frederic Feru
- Harvard Medical School, Boston, MA, USA
- Dana-Farber Cancer Institute, Boston, MA, USA
| | - Maria DeLeon
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Spencer E. Brightman
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
| | - Wing Ki Cheng
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Gandolina Melhem
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | | | - Juan C. Ramirez
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Soumik Barman
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Michael Cameron
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL, USA
| | - Aisling Kelly
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
| | - Kristina Walker
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
| | - Etsuro Nanishi
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Simon Daniel van Haren
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Tony Phan
- Access to Advanced Health Institute (AAHI), Seattle, WA, USA
| | - Yizhi Qi
- Access to Advanced Health Institute (AAHI), Seattle, WA, USA
| | - Robert Kinsey
- Access to Advanced Health Institute (AAHI), Seattle, WA, USA
| | - Michal M. Raczy
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Al Ozonoff
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT & Harvard, Cambridge, MA, USA
| | - Matthew A. Pettengill
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Jeffery A. Hubbell
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Christopher B. Fox
- Access to Advanced Health Institute (AAHI), Seattle, WA, USA
- Department of Global Health, University of Washington, 3980 15th Ave NE, Seattle, WA 98195, USA
| | - David J. Dowling
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Ofer Levy
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT & Harvard, Cambridge, MA, USA
| |
Collapse
|
14
|
Han R, Wang T, Cheng X, Bing J, Li J, Deng Y, Shan X, Zhang X, Wang D, Sun S, Tan W. Immune Responses and Protection Profiles in Mice Induced by Subunit Vaccine Candidates Based on the Extracellular Domain Antigen of Respiratory Syncytial Virus G Protein Combined with Different Adjuvants. Vaccines (Basel) 2024; 12:686. [PMID: 38932414 PMCID: PMC11209252 DOI: 10.3390/vaccines12060686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 06/11/2024] [Accepted: 06/17/2024] [Indexed: 06/28/2024] Open
Abstract
Respiratory syncytial virus (RSV) is a leading cause of severe lower respiratory tract disease of infants and older people. There is an urgent need for safe and effective vaccines against RSV infection. In this study, we analyzed the effects of the immune response and protection with the RSV recombinant G protein extracellular domain (Gecto) combined with various adjuvants as novel subunit vaccines in mice. All groups receiving RSV Gecto combined with adjuvants exhibited robust humoral and cellular immunity compared to those receiving an adjuvant alone or inactivated RSV vaccine. The greatest effect was observed in mice receiving Gecto combined with a CpG ODN + Alum salt adjuvant, resulting in the highest production of neutralizing antibodies against both RSV A and B subtypes, G-specific IgG and IFN-γ production in splenocytes, and interleukin-2 and interferon-γ expression in CD4+ T cells. Significant humoral and cellular immune responses were observed in mice immunized with Gecto combined with AddaS03™ or cyclosporin A adjuvants. The vaccine containing the AddaS03™ adjuvant showed significantly high expression of interleukin-4 in CD4+ T cells. Cross-protection against a challenge with either RSV A or B subtypes was observed in the Gecto plus adjuvant groups, resulting in a significant decrease in viral load and reduced pathological damage in the mouse lungs. These findings offer valuable insights into the development and application of recombinant RSV G-subunit vaccines with adjuvants.
Collapse
Affiliation(s)
- Ruiwen Han
- Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, China; (R.H.); (T.W.); (J.L.)
| | - Tangqi Wang
- Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, China; (R.H.); (T.W.); (J.L.)
| | - Xueting Cheng
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Key Laboratory of Biosafety, National Health Commissions, National Institute for Viral Disease Control and Prevention, China CDC, 155 Changbai Road, Beijing 102206, China; (X.C.); (Y.D.)
| | - Jialuo Bing
- School of Public Health, Xinxiang Medical University, Xinxiang 453003, China; (J.B.); (X.S.); (X.Z.)
| | - Jia Li
- Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, China; (R.H.); (T.W.); (J.L.)
| | - Yao Deng
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Key Laboratory of Biosafety, National Health Commissions, National Institute for Viral Disease Control and Prevention, China CDC, 155 Changbai Road, Beijing 102206, China; (X.C.); (Y.D.)
| | - Xuchang Shan
- School of Public Health, Xinxiang Medical University, Xinxiang 453003, China; (J.B.); (X.S.); (X.Z.)
| | - Xuejie Zhang
- School of Public Health, Xinxiang Medical University, Xinxiang 453003, China; (J.B.); (X.S.); (X.Z.)
| | - Donghong Wang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Key Laboratory of Biosafety, National Health Commissions, National Institute for Viral Disease Control and Prevention, China CDC, 155 Changbai Road, Beijing 102206, China; (X.C.); (Y.D.)
| | - Shucai Sun
- Department of Nuclear Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang 050004, China;
| | - Wenjie Tan
- Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, China; (R.H.); (T.W.); (J.L.)
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Key Laboratory of Biosafety, National Health Commissions, National Institute for Viral Disease Control and Prevention, China CDC, 155 Changbai Road, Beijing 102206, China; (X.C.); (Y.D.)
- School of Public Health, Xinxiang Medical University, Xinxiang 453003, China; (J.B.); (X.S.); (X.Z.)
- Department of Nuclear Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang 050004, China;
| |
Collapse
|
15
|
Sanchez PL, Staats HF, Abraham SN, Ross TM. Mastoparan-7 adjuvanted COBRA H1 and H3 hemagglutinin influenza vaccines. Sci Rep 2024; 14:13800. [PMID: 38877101 PMCID: PMC11178843 DOI: 10.1038/s41598-024-64351-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 06/07/2024] [Indexed: 06/16/2024] Open
Abstract
Adjuvants enhance, prolong, and modulate immune responses by vaccine antigens to maximize protective immunity and enable more effective immunization in the young and elderly. Most adjuvants are formulated with injectable vaccines. However, an intranasal route of vaccination may induce mucosal and systemic immune responses for enhancing protective immunity in individuals and be easier to administer compared to injectable vaccines. In this study, a next generation of broadly-reactive influenza hemagglutinin (HA) vaccines were developed using the Computationally Optimized Broadly Reactive Antigen (COBRA) methodology. These HA vaccines were formulated with Mastoparan 7 (M7-NH2) mast cell degranulating peptide adjuvant and administered intranasally to determine vaccine-induced seroconversion of antibodies against a panel of influenza viruses and protection following infection with H1N1 and H3N2 viruses in mice. Mice vaccinated intranasally with M7-NH2-adjuvanted COBRA HA vaccines had high HAIs against a panel of H1N1 and H3N2 influenza viruses and were protected against both morbidity and mortality, with reduced viral lung titers, following challenge with an H1N1 influenza virus. Additionally, M7-NH2 adjuvanted COBRA HA vaccines induced Th2 skewed immune responses with robust IgG and isotype antibodies in the serum and mucosal lung lavages. Overall, this intranasally delivered M7-NH2 -adjuvanted COBRA HA vaccine provides effective protection against drifted H1N1 and H3N2 viruses.
Collapse
Affiliation(s)
- Pedro L Sanchez
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, USA
- Department of Infectious Diseases, University of Georgia, Athens, GA, USA
- Florida Research and Innovation Center, Cleveland Clinic, Port Saint Lucie, FL, USA
| | - Herman F Staats
- Pathology Department, School of Medicine, Duke University Medical Center, Durham, NC, USA
- Department of Immunology, School of Medicine, Duke University Medical Center, Durham, NC, USA
- Duke Human Vaccine Institute, Duke University, Duke University Medical Center, Durham, NC, USA
| | - Soman N Abraham
- Pathology Department, School of Medicine, Duke University Medical Center, Durham, NC, USA
- Department of Immunology, School of Medicine, Duke University Medical Center, Durham, NC, USA
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC, USA
- Program in Emerging Infectious Diseases, Duke-National University of Singapore, Singapore, Singapore
| | - Ted M Ross
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, USA.
- Department of Infectious Diseases, University of Georgia, Athens, GA, USA.
- Florida Research and Innovation Center, Cleveland Clinic, Port Saint Lucie, FL, USA.
- Department of Infection Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.
| |
Collapse
|
16
|
Oladejo M, Tijani AO, Puri A, Chablani L. Adjuvants in cutaneous vaccination: A comprehensive analysis. J Control Release 2024; 369:475-492. [PMID: 38569943 DOI: 10.1016/j.jconrel.2024.03.045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 03/15/2024] [Accepted: 03/26/2024] [Indexed: 04/05/2024]
Abstract
Skin is the body's largest organ and serves as a protective barrier from physical, thermal, and mechanical environmental challenges. Alongside, the skin hosts key immune system players, such as the professional antigen-presenting cells (APCs) like the Langerhans cells in the epidermis and circulating macrophages in the blood. Further, the literature supports that the APCs can be activated by antigen or vaccine delivery via multiple routes of administration through the skin. Once activated, the stimulated APCs drain to the associated lymph nodes and gain access to the lymphatic system. This further allows the APCs to engage with the adaptive immune system and activate cellular and humoral immune responses. Thus, vaccine delivery via skin offers advantages such as reliable antigen delivery, superior immunogenicity, and convenient delivery. Several preclinical and clinical studies have demonstrated the significance of vaccine delivery using various routes of administration via skin. However, such vaccines often employ adjuvant/(s), along with the antigen of interest. Adjuvants augment the immune response to a vaccine antigen and improve the therapeutic efficacy. Due to these reasons, adjuvants have been successfully used with infectious disease vaccines, cancer immunotherapy, and immune-mediated diseases. To capture these developments, this review will summarize preclinical and clinical study results of vaccine delivery via skin in the presence of adjuvants. A focused discussion regarding the FDA-approved adjuvants will address the experiences of using such adjuvant-containing vaccines. In addition, the challenges and regulatory concerns with these adjuvants will be discussed. Finally, the review will share the prospects of adjuvant-containing vaccines delivered via skin.
Collapse
Affiliation(s)
- Mariam Oladejo
- Department of Immunotherapeutics and Biotechnology, Jerry H Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX 79601, USA
| | - Akeemat O Tijani
- Department of Pharmaceutical Sciences, Bill Gatton College of Pharmacy, East Tennessee State University, Johnson City, TN, USA
| | - Ashana Puri
- Department of Pharmaceutical Sciences, Bill Gatton College of Pharmacy, East Tennessee State University, Johnson City, TN, USA.
| | - Lipika Chablani
- Wegmans School of Pharmacy, St. John Fisher University, 3690 East Ave, Rochester, NY 14618, USA.
| |
Collapse
|
17
|
Sanchez PL, Andre G, Antipov A, Petrovsky N, Ross TM. Advax-SM™-Adjuvanted COBRA (H1/H3) Hemagglutinin Influenza Vaccines. Vaccines (Basel) 2024; 12:455. [PMID: 38793706 PMCID: PMC11125990 DOI: 10.3390/vaccines12050455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 03/25/2024] [Accepted: 04/22/2024] [Indexed: 05/26/2024] Open
Abstract
Adjuvants enhance immune responses stimulated by vaccines. To date, many seasonal influenza vaccines are not formulated with an adjuvant. In the present study, the adjuvant Advax-SM™ was combined with next generation, broadly reactive influenza hemagglutinin (HA) vaccines that were designed using a computationally optimized broadly reactive antigen (COBRA) methodology. Advax-SM™ is a novel adjuvant comprising inulin polysaccharide and CpG55.2, a TLR9 agonist. COBRA HA vaccines were combined with Advax-SM™ or a comparator squalene emulsion (SE) adjuvant and administered to mice intramuscularly. Mice vaccinated with Advax-SM™ adjuvanted COBRA HA vaccines had increased serum levels of anti-influenza IgG and IgA, high hemagglutination inhibition activity against a panel of H1N1 and H3N2 influenza viruses, and increased anti-influenza antibody secreting cells isolated from spleens. COBRA HA plus Advax-SM™ immunized mice were protected against both morbidity and mortality following viral challenge and, at postmortem, had no detectable lung viral titers or lung inflammation. Overall, the Advax-SM™-adjuvanted COBRA HA formulation provided effective protection against drifted H1N1 and H3N2 influenza viruses.
Collapse
Affiliation(s)
- Pedro L. Sanchez
- Center for Vaccines and Immunology, University of Georgia, Athens, GA 30602, USA;
- Department of Infectious Diseases, University of Georgia, Athens, GA 30602, USA
- Florida Research and Innovation Center, Cleveland Clinic, Port Saint Lucie, FL 34987, USA
| | - Greiciely Andre
- Vaxine Pty Ltd., Adelaide, SA 5046, Australia; (G.A.); (A.A.); (N.P.)
| | - Anna Antipov
- Vaxine Pty Ltd., Adelaide, SA 5046, Australia; (G.A.); (A.A.); (N.P.)
| | - Nikolai Petrovsky
- Vaxine Pty Ltd., Adelaide, SA 5046, Australia; (G.A.); (A.A.); (N.P.)
| | - Ted M. Ross
- Center for Vaccines and Immunology, University of Georgia, Athens, GA 30602, USA;
- Department of Infectious Diseases, University of Georgia, Athens, GA 30602, USA
- Florida Research and Innovation Center, Cleveland Clinic, Port Saint Lucie, FL 34987, USA
- Department of Infection Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| |
Collapse
|
18
|
Pagh-Berendtsen N, Pavlovskyi A, Flores Téllez D, Egebjerg C, Kolmos MG, Justinussen J, Kornum BR. Downregulation of hypocretin/orexin after H1N1 Pandemrix vaccination of adolescent mice. Sleep 2024; 47:zsae014. [PMID: 38227834 DOI: 10.1093/sleep/zsae014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 11/07/2023] [Indexed: 01/18/2024] Open
Abstract
Narcolepsy type 1 (NT1), characterized by the loss of hypocretin/orexin (HCRT) production in the lateral hypothalamus, has been linked to Pandemrix vaccination during the 2009 H1N1 pandemic, especially in children and adolescents. It is still unknown why this vaccination increased the risk of developing NT1. This study investigated the effects of Pandemrix vaccination during adolescence on Hcrt mRNA expression in mice. Mice received a primary vaccination (50 µL i.m.) during prepubescence and a booster vaccination during peri-adolescence. Hcrt expression was measured at three-time points after the vaccinations. Control groups included both a saline group and an undisturbed group of mice. Hcrt expression was decreased after both Pandemrix and saline injections, but 21 days after the second injection, the saline group no longer showed decreased Hcrt expression, while the Pandemrix group still exhibited a significant reduction of about 60% compared to the undisturbed control group. This finding suggests that Pandemrix vaccination during adolescence influences Hcrt expression in mice into early adulthood. The Hcrt mRNA level did not reach the low levels known to induce NT1 symptoms, instead, our finding supports the multiple-hit hypothesis of NT1 that states that several insults to the HCRT system may be needed to induce NT1 and that Pandemrix could be one such insult.
Collapse
Affiliation(s)
- Nicolai Pagh-Berendtsen
- Faculty of Health and Medical Sciences, Department of Neuroscience, University of Copenhagen, Denmark
| | - Artem Pavlovskyi
- Faculty of Health and Medical Sciences, Department of Neuroscience, University of Copenhagen, Denmark
| | - Daniel Flores Téllez
- Faculty of Health and Medical Sciences, Department of Neuroscience, University of Copenhagen, Denmark
| | - Christine Egebjerg
- Faculty of Health and Medical Sciences, Department of Neuroscience, University of Copenhagen, Denmark
| | - Mie Gunni Kolmos
- Faculty of Health and Medical Sciences, Department of Neuroscience, University of Copenhagen, Denmark
| | - Jessica Justinussen
- Faculty of Health and Medical Sciences, Department of Neuroscience, University of Copenhagen, Denmark
| | - Birgitte Rahbek Kornum
- Faculty of Health and Medical Sciences, Department of Neuroscience, University of Copenhagen, Denmark
| |
Collapse
|
19
|
Lin YJ, Zimmermann J, Schülke S. Novel adjuvants in allergen-specific immunotherapy: where do we stand? Front Immunol 2024; 15:1348305. [PMID: 38464539 PMCID: PMC10920236 DOI: 10.3389/fimmu.2024.1348305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 02/05/2024] [Indexed: 03/12/2024] Open
Abstract
Type I hypersensitivity, or so-called type I allergy, is caused by Th2-mediated immune responses directed against otherwise harmless environmental antigens. Currently, allergen-specific immunotherapy (AIT) is the only disease-modifying treatment with the potential to re-establish clinical tolerance towards the corresponding allergen(s). However, conventional AIT has certain drawbacks, including long treatment durations, the risk of inducing allergic side effects, and the fact that allergens by themselves have a rather low immunogenicity. To improve AIT, adjuvants can be a powerful tool not only to increase the immunogenicity of co-applied allergens but also to induce the desired immune activation, such as promoting allergen-specific Th1- or regulatory responses. This review summarizes the knowledge on adjuvants currently approved for use in human AIT: aluminum hydroxide, calcium phosphate, microcrystalline tyrosine, and MPLA, as well as novel adjuvants that have been studied in recent years: oil-in-water emulsions, virus-like particles, viral components, carbohydrate-based adjuvants (QS-21, glucans, and mannan) and TLR-ligands (flagellin and CpG-ODN). The investigated adjuvants show distinct properties, such as prolonging allergen release at the injection site, inducing allergen-specific IgG production while also reducing IgE levels, as well as promoting differentiation and activation of different immune cells. In the future, better understanding of the immunological mechanisms underlying the effects of these adjuvants in clinical settings may help us to improve AIT.
Collapse
Affiliation(s)
- Yen-Ju Lin
- Section Molecular Allergology, Paul-Ehrlich-Institut, Langen, Germany
| | | | - Stefan Schülke
- Section Molecular Allergology, Paul-Ehrlich-Institut, Langen, Germany
- Section Research Allergology (ALG 5), Division of Allergology, Paul-Ehrlich-Institut, Langen, Germany
| |
Collapse
|
20
|
Huang Z, Gong H, Sun Q, Yang J, Yan X, Xu F. Research progress on emulsion vaccine adjuvants. Heliyon 2024; 10:e24662. [PMID: 38317888 PMCID: PMC10839794 DOI: 10.1016/j.heliyon.2024.e24662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 01/11/2024] [Accepted: 01/11/2024] [Indexed: 02/07/2024] Open
Abstract
Vaccination is the most cost-effective method for preventing various infectious diseases. Compared with conventional vaccines, new-generation vaccines, especially recombinant protein or synthetic peptide vaccines, are safer but less immunogenic than crude inactivated microbial vaccines. The immunogenicity of these vaccines can be enhanced using suitable adjuvants. This is the main reason why adjuvants are of great importance in vaccine development. Several novel human emulsion-based vaccine adjuvants (MF59, AS03) have been approved for clinical use. This paper reviews the research progress on emulsion-based adjuvants and focuses on their mechanism of action. An outlook can be provided for the development of emulsion-based vaccine adjuvants.
Collapse
Affiliation(s)
- Zhuanqing Huang
- Department of Ophthalmology, The No. 944 Hospital of Joint Logistic Support Force of PLA, Gansu 735000, China
- Pharmaceutical Sciences Research Division, Department of Pharmacy, Medical Supplies Centre, PLA General Hospital, Beijing 100853, China
| | - Hui Gong
- Medical School of Chinese PLA, Beijing 100853, China
| | - Qi Sun
- Pharmaceutical Sciences Research Division, Department of Pharmacy, Medical Supplies Centre, PLA General Hospital, Beijing 100853, China
| | - Jinjin Yang
- The Fifth medical center of Chinese PLA General Hospital, Beijing 100071, China
| | - Xiaochuan Yan
- Department of Ophthalmology, The No. 944 Hospital of Joint Logistic Support Force of PLA, Gansu 735000, China
| | - Fenghua Xu
- Pharmaceutical Sciences Research Division, Department of Pharmacy, Medical Supplies Centre, PLA General Hospital, Beijing 100853, China
| |
Collapse
|
21
|
Hoover AR, More S, Liu K, West CL, Valerio TI, Furrer CL, Adams JP, Yu N, Villalva C, Kumar A, Alleruzzo L, Lam SSK, Hode T, Papin JF, Chen WR. N-dihydrogalactochitosan serves as an effective mucosal adjuvant for intranasal vaccine in combination with recombinant viral proteins against respiratory infection. Acta Biomater 2024; 175:279-292. [PMID: 38160856 DOI: 10.1016/j.actbio.2023.12.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 12/21/2023] [Accepted: 12/22/2023] [Indexed: 01/03/2024]
Abstract
Mucosal vaccinations for respiratory pathogens provide effective protection as they stimulate localized cellular and humoral immunities at the site of infection. Currently, the major limitation of intranasal vaccination is using effective adjuvants capable of withstanding the harsh environment imposed by the mucosa. Herein, we describe the efficacy of using a unique biopolymer, N-dihydrogalactochitosan (GC), as a nasal mucosal vaccine adjuvant against respiratory infections. Specifically, we mixed GC with recombinant SARS-CoV-2 trimeric spike (S) and nucleocapsid (NC) proteins to intranasally vaccinate K18-hACE2 transgenic mice, in comparison with Addavax (AV), an MF-59 equivalent. In contrast to AV, intranasal application of GC induces a robust, systemic antigen-specific antibody response and increases the number of T cells in the cervical lymph nodes. Moreover, GC+S+NC-vaccinated animals were largely resistant to the lethal SARS-CoV-2 challenge and experienced drastically reduced morbidity and mortality, with animal weights and behavior returning to normal 22 days post-infection. In contrast, animals intranasally vaccinated with AV+S+NC experienced severe weight loss, mortality, and respiratory distress, with none surviving beyond 6 days post-infection. Our findings demonstrate that GC can serve as a potent mucosal vaccine adjuvant against SARS-CoV-2 and potentially other respiratory viruses. STATEMENT OF SIGNIFICANCE: We demonstrated that a unique biopolymer, N-dihydrogalactochitosan (GC), was an effective nasal mucosal vaccine adjuvant against respiratory infections. Specifically, we mixed GC with recombinant SARS-CoV-2 trimeric spike (S) and nucleocapsid (NC) proteins to intranasally vaccinate K18-hACE2 transgenic mice, in comparison with Addavax (AV). In contrast to AV, GC induces a robust, systemic antigen-specific antibody response and increases the number of T cells in the cervical lymph nodes. About 90 % of the GC+S+NC-vaccinated animals survived the lethal SARS-CoV-2 challenge and remained healthy 22 days post-infection, while the AV+S+NC-vaccinated animals experienced severe weight loss and respiratory distress, and all died within 6 days post-infection. Our findings demonstrate that GC is a potent mucosal vaccine adjuvant against SARS-CoV-2 and potentially other respiratory viruses.
Collapse
Affiliation(s)
- Ashley R Hoover
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA; Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Sunil More
- Veterinary Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK USA
| | - Kaili Liu
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA
| | - Connor L West
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA
| | - Trisha I Valerio
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA
| | - Coline L Furrer
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA
| | - Jacob P Adams
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA
| | - Ningli Yu
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA
| | - Crystal Villalva
- Veterinary Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK USA
| | - Amit Kumar
- Biogen Inc., 225 Bnney Street, Cambridge, MA, USA
| | - Lu Alleruzzo
- Immunophotonics, Inc., 4340 Duncan Avenue, Suite 212, Saint Louis, MO, USA
| | - Samuel S K Lam
- Immunophotonics, Inc., 4340 Duncan Avenue, Suite 212, Saint Louis, MO, USA
| | - Tomas Hode
- Immunophotonics, Inc., 4340 Duncan Avenue, Suite 212, Saint Louis, MO, USA
| | - James F Papin
- Department Pathology and Division of Comparative Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, USA
| | - Wei R Chen
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA.
| |
Collapse
|
22
|
Albalawi ARS, Alhassun JAS, Almarshud RK, Almejali HA, Alharbi SM, Shaybah AM, Alshehab ZMA, Alzahrani SM, Abomelha LS, Almalki AA, Alkhurayyif AO, Alalawi MS, Alnass AJ, Alzibali KF, Alabdulrahim JM. Unlocking the Power of Influenza Vaccines for Pediatric Population: A Narrative Review. Cureus 2024; 16:e55119. [PMID: 38558642 PMCID: PMC10979318 DOI: 10.7759/cureus.55119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/27/2024] [Indexed: 04/04/2024] Open
Abstract
The flu, often known as influenza, is a dangerous public health hazard for the pediatric population. Immunization is essential for decreasing the burden of the disease and avoiding complications related to influenza. However, the immunogenicity, efficacy, and safety of different influenza vaccines in children warrant careful evaluation. The purpose of this narrative review is to give a summary of the existing literature on the immunogenicity, efficacy, and safety of several vaccinations against influenza viruses in children. The review incorporates evidence from a range of studies focusing on the outcomes of interest. Immunogenicity studies have shown that influenza vaccines induce a robust immune response in children, primarily through neutralizing antibodies' formation. However, variations in vaccine composition influence the duration and magnitude of immune responses. Safety is a crucial consideration in pediatric vaccination. In children, influenza vaccinations have generally shown a high safety profile, with mild and temporary side effects being the most common. Vaccinations against influenza have shown a modest level of efficacy in avoiding hospitalizations linked to influenza, laboratory-confirmed influenza infections, and serious consequences in children. Live attenuated vaccines have shown higher effectiveness against matched strains compared to inactivated vaccines. In conclusion, this narrative review highlights that receiving influenza vaccination in children aged six to 47 months is very important. While different vaccines exhibit varying immunogenicity, safety profiles, and effectiveness, they all contribute to reducing the burden of influenza among children. Future research should focus on optimizing vaccine strategies, improving vaccine coverage, and evaluating long-term protection.
Collapse
Affiliation(s)
| | | | - Raghad K Almarshud
- Family Medicine, Unaizah College of Medicine and Medical Sciences, Qassim University, Al Qassim, SAU
| | - Hamad A Almejali
- General Practice, General Administration of Prison Health, Riyadh, SAU
| | | | - Amal M Shaybah
- General Practice, Primary Health Care Center, Jazan, SAU
| | | | | | - Lama S Abomelha
- General Practice, Almahalh Primary Health Care Center, Abha, SAU
| | | | | | | | - Anwar J Alnass
- General Practice, Ibn Sina Primary Health Care Center, Jubail, SAU
| | | | | |
Collapse
|
23
|
Margaroni M, Tsanaktsidou E, Agallou M, Kiparissides C, Kammona O, Karagouni E. Development of a novel squalene/α-tocopherol-based self-emulsified nanoemulsion incorporating Leishmania peptides for induction of antigen-specific immune responses. Int J Pharm 2024; 649:123621. [PMID: 38000650 DOI: 10.1016/j.ijpharm.2023.123621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 11/02/2023] [Accepted: 11/15/2023] [Indexed: 11/26/2023]
Abstract
Vaccination has emerged as the most effective strategy to confront infectious diseases, among which is leishmaniasis, that threat public health. Despite laborious efforts there is still no vaccine for humans to confront leishmaniasis. Multi-epitope protein/peptide vaccines present a number of advantages, however their use along with appropriate adjuvants that may also act as antigen carriers is considered essential to overcome subunit vaccines' low immunogenicity. In the present study, a stable self-emulsified nanoemulsion was developed and double-adjuvanted with squalene and α-tocopherol. The prepared nanoemulsion droplets exhibited low cytotoxicity in a certain range of concentrations, while they were efficiently taken up by macrophages and dendritic cells in vitro as well as in vivo in secondary lymphoid organs. To further characterize nanoformulation's potent antigen delivery capability, three multi-epitope Leishmania peptides were incorporated into the nanoemulsion. Peptide encapsulation resulted in dendritic cells' functional differentiation characterized by elevated levels of maturation markers and intracellular cytokine production. Intramuscular administration of the nanoemulsion incorporating Leishmania peptides induced antigen-specific spleen cell proliferation as well as elicitation of CD4+ central memory cells, supporting the potential of the developed nanoformulation to successfully act also as an antigen delivery vehicle and thus encouraging further preclinical studies on its vaccine candidate potency.
Collapse
Affiliation(s)
- Maritsa Margaroni
- Immunology of Infection Laboratory, Hellenic Pasteur Institute, 125 21 Athens, Greece.
| | - Evgenia Tsanaktsidou
- Chemical Process & Energy Resources Institute, Centre for Research and Technology Hellas, P.O. Box 60361, 57 001 Thessaloniki, Greece.
| | - Maria Agallou
- Immunology of Infection Laboratory, Hellenic Pasteur Institute, 125 21 Athens, Greece.
| | - Costas Kiparissides
- Chemical Process & Energy Resources Institute, Centre for Research and Technology Hellas, P.O. Box 60361, 57 001 Thessaloniki, Greece; Department of Chemical Engineering, Aristotle University of Thessaloniki, P.O. Box 472, 54 124 Thessaloniki, Greece.
| | - Olga Kammona
- Chemical Process & Energy Resources Institute, Centre for Research and Technology Hellas, P.O. Box 60361, 57 001 Thessaloniki, Greece.
| | - Evdokia Karagouni
- Immunology of Infection Laboratory, Hellenic Pasteur Institute, 125 21 Athens, Greece.
| |
Collapse
|
24
|
Jang H, Matsuoka M, Freire M. Oral mucosa immunity: ultimate strategy to stop spreading of pandemic viruses. Front Immunol 2023; 14:1220610. [PMID: 37928529 PMCID: PMC10622784 DOI: 10.3389/fimmu.2023.1220610] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 09/11/2023] [Indexed: 11/07/2023] Open
Abstract
Global pandemics are most likely initiated via zoonotic transmission to humans in which respiratory viruses infect airways with relevance to mucosal systems. Out of the known pandemics, five were initiated by respiratory viruses including current ongoing coronavirus disease 2019 (COVID-19). Striking progress in vaccine development and therapeutics has helped ameliorate the mortality and morbidity by infectious agents. Yet, organism replication and virus spread through mucosal tissues cannot be directly controlled by parenteral vaccines. A novel mitigation strategy is needed to elicit robust mucosal protection and broadly neutralizing activities to hamper virus entry mechanisms and inhibit transmission. This review focuses on the oral mucosa, which is a critical site of viral transmission and promising target to elicit sterile immunity. In addition to reviewing historic pandemics initiated by the zoonotic respiratory RNA viruses and the oral mucosal tissues, we discuss unique features of the oral immune responses. We address barriers and new prospects related to developing novel therapeutics to elicit protective immunity at the mucosal level to ultimately control transmission.
Collapse
Affiliation(s)
- Hyesun Jang
- Genomic Medicine and Infectious Diseases, J. Craig Venter Institute, La Jolla, CA, United States
| | - Michele Matsuoka
- Genomic Medicine and Infectious Diseases, J. Craig Venter Institute, La Jolla, CA, United States
| | - Marcelo Freire
- Genomic Medicine and Infectious Diseases, J. Craig Venter Institute, La Jolla, CA, United States
- Division of Infectious Diseases and Global Public Health Department of Medicine, University of California San Diego, La Jolla, CA, United States
| |
Collapse
|
25
|
Cao L, Qian W, Li W, Ma Z, Xie S. Type III interferon exerts thymic stromal lymphopoietin in mediating adaptive antiviral immune response. Front Immunol 2023; 14:1250541. [PMID: 37809098 PMCID: PMC10556530 DOI: 10.3389/fimmu.2023.1250541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 09/07/2023] [Indexed: 10/10/2023] Open
Abstract
Previously, it was believed that type III interferon (IFN-III) has functions similar to those of type I interferon (IFN-I). However, recently, emerging findings have increasingly indicated the non-redundant role of IFN-III in innate antiviral immune responses. Still, the regulatory activity of IFN-III in adaptive immune response has not been clearly reported yet due to the low expression of IFN-III receptors on most immune cells. In the present study, we reviewed the adjuvant, antiviral, antitumor, and disease-moderating activities of IFN-III in adaptive immunity; moreover, we further elucidated the mechanisms of IFN-III in mediating the adaptive antiviral immune response in a thymic stromal lymphopoietin (TSLP)-dependent manner, a pleiotropic cytokine involved in mucosal adaptive immunity. Research has shown that IFN-III can enhance the antiviral immunogenic response in mouse species by activating germinal center B (GC B) cell responses after stimulating TSLP production by microfold (M) cells, while in human species, TSLP exerts OX40L for regulating GC B cell immune responses, which may also depend on IFN-III. In conclusion, our review highlights the unique role of the IFN-III/TSLP axis in mediating host adaptive immunity, which is mechanically different from IFN-I. Therefore, the IFN-III/TSLP axis may provide novel insights for clinical immunotherapy.
Collapse
Affiliation(s)
- Luhong Cao
- Department of Otolaryngology Head and Neck Surgery Surgery, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Weiwei Qian
- Department of Emergency Medicine, Laboratory of Emergency Medicine, West China Hospital, and Disaster Medical Center, Sichuan University, Chengdu, Sichuan, China
| | - Wanlin Li
- National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, Shenzhen, China
| | - Zhiyue Ma
- Department of Otolaryngology Head and Neck Surgery Surgery, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Shenglong Xie
- Department of Thoracic Surgery, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
26
|
Desai DN, Mahal A, Varshney R, Obaidullah AJ, Gupta B, Mohanty P, Pattnaik P, Mohapatra NC, Mishra S, Kandi V, Rabaan AA, Mohapatra RK. Nanoadjuvants: Promising Bioinspired and Biomimetic Approaches in Vaccine Innovation. ACS OMEGA 2023; 8:27953-27968. [PMID: 37576639 PMCID: PMC10413842 DOI: 10.1021/acsomega.3c02030] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Accepted: 07/13/2023] [Indexed: 08/15/2023]
Abstract
Adjuvants are the important part of vaccine manufacturing as they elicit the vaccination effect and enhance the durability of the immune response through controlled release. In light of this, nanoadjuvants have shown unique broad spectrum advantages. As nanoparticles (NPs) based vaccines are fast-acting and better in terms of safety and usability parameters as compared to traditional vaccines, they have attracted the attention of researchers. A vaccine nanocarrier is another interesting and promising area for the development of next-generation vaccines for prophylaxis. This review looks at the various nanoadjuvants and their structure-function relationships. It compiles the state-of-art literature on numerous nanoadjuvants to help domain researchers orient their understanding and extend their endeavors in vaccines research and development.
Collapse
Affiliation(s)
- Dhruv N. Desai
- Department
of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Ahmed Mahal
- Department
of Medical Biochemical Analysis, College of Health Technology, Cihan University−Erbil, Erbil, Kurdistan Region, Iraq
| | - Rajat Varshney
- Department
of Veterinary Microbiology, FVAS, Banaras
Hindu University, Mirzapur 231001, India
| | - Ahmad J. Obaidullah
- Department
of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Bhawna Gupta
- School
of Biotechnology, KIIT Deemed-to-be University, Bhubaneswar 751024, Odisha, India
| | - Pratikhya Mohanty
- Bioenergy
Lab, BDTC, School of Biotechnology, KIIT
Deemed-to-be University, Bhubaneswar 751024, Odisha, India
| | | | | | - Snehasish Mishra
- Bioenergy
Lab, BDTC, School of Biotechnology, KIIT
Deemed-to-be University, Bhubaneswar 751024, Odisha, India
| | - Venkataramana Kandi
- Department
of Microbiology, Prathima Institute of Medical
Sciences, Karimnagar 505 417, Telangana, India
| | - Ali A. Rabaan
- Molecular
Diagnostic Laboratory, Johns Hopkins Aramco
Healthcare, Dhahran 31311, Saudi Arabia
- College
of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
- Department
of Public Health and Nutrition, The University
of Haripur, Haripur 22610, Pakistan
| | - Ranjan K. Mohapatra
- Department
of Chemistry, Government College of Engineering, Keonjhar 758002, Odisha, India
| |
Collapse
|
27
|
Coelho CH, Marquez S, Tentokam BCN, Berhe AD, Miura K, Long CA, Sagara I, Healy S, Kleinstein SH, Duffy PE. Antibody gene features associated with binding and functional activity in vaccine-derived human mAbs targeting malaria parasites. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.01.551554. [PMID: 37781572 PMCID: PMC10541113 DOI: 10.1101/2023.08.01.551554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/03/2023]
Abstract
Adjuvants have been essential to malaria vaccine development, but their impact on the vaccine-induced antibody repertoire is poorly understood. Here, we used cDNA sequences from antigen-specific single memory B cells to express 132 recombinant human anti-Pfs230 monoclonal antibodies (mAbs). Alhydrogel®-induced mAbs demonstrated higher binding to Pfs230D1, although functional activity was similar between adjuvants. All Alhydrogel® mAbs using IGHV1-69 gene bound to recombinant Pfs230D1, but none blocked parasite transmission to mosquitoes; similarly, no AS01 mAb using IGHV1-69 blocked transmission. Functional mAbs from both Alhydrogel® and AS01 vaccines used IGHV3-21 and IGHV3-30 genes. Antibodies with the longest CDR3 sequences were associated with binding but not functional activity. This study assesses adjuvant effects on antibody clonotype diversity during malaria vaccination.
Collapse
Affiliation(s)
- Camila H. Coelho
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, 10029, NY
| | - Susanna Marquez
- Department of Pathology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Bergeline C. Nguemwo Tentokam
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Anne D. Berhe
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kazutoyo Miura
- Laboratory of Malaria and Vector and Research, National Institute of Allergy and Infectious Diseases, NIH, Rockville, Maryland, USA
| | - Carole A. Long
- Laboratory of Malaria and Vector and Research, National Institute of Allergy and Infectious Diseases, NIH, Rockville, Maryland, USA
| | - Issaka Sagara
- Malaria Research and Training Center, University of Sciences, Techniques, and Technology, Bamako, Mali
| | - Sara Healy
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Steven H. Kleinstein
- Department of Pathology, Yale School of Medicine, New Haven, CT 06520, USA
- Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT 06511, USA
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Patrick E. Duffy
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
28
|
Kumru OS, Bajoria S, Kaur K, Hickey JM, Van Slyke G, Doering J, Berman K, Richardson C, Lien H, Kleanthous H, Mantis NJ, Joshi SB, Volkin DB. Effects of aluminum-salt, CpG and emulsion adjuvants on the stability and immunogenicity of a virus-like particle displaying the SARS-CoV-2 receptor-binding domain (RBD). Hum Vaccin Immunother 2023; 19:2264594. [PMID: 37932241 PMCID: PMC10760504 DOI: 10.1080/21645515.2023.2264594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 09/25/2023] [Indexed: 11/08/2023] Open
Abstract
Second-generation COVID-19 vaccines with improved immunogenicity (e.g., breadth, duration) and availability (e.g., lower costs, refrigerator stable) are needed to enhance global coverage. In this work, we formulated a clinical-stage SARS-CoV-2 receptor-binding domain (RBD) virus-like particle (VLP) vaccine candidate (IVX-411) with widely available adjuvants. Specifically, we assessed the in vitro storage stability and in vivo mouse immunogenicity of IVX-411 formulated with aluminum-salt adjuvants (Alhydrogel™, AH and Adjuphos™, AP), without or with the TLR-9 agonist CpG-1018™ (CpG), and compared these profiles to IVX-411 adjuvanted with an oil-in-water nano-emulsion (AddaVax™, AV). Although IVX-411 bound both AH and AP, lower binding strength of antigen to AP was observed by Langmuir binding isotherms. Interestingly, AH- and AP-adsorbed IVX-411 had similar storage stability profiles as measured by antigen-binding assays (competitive ELISAs), but the latter displayed higher pseudovirus neutralizing titers (pNT) in mice, at levels comparable to titers elicited by AV-adjuvanted IVX-411. CpG addition to alum (AP or AH) resulted in a marginal trend of improved pNTs in stressed samples only, yet did not impact the storage stability profiles of IVX-411. In contrast, previous work with AH-formulations of a monomeric RBD antigen showed greatly improved immunogenicity and decreased stability upon CpG addition to alum. At elevated temperatures (25, 37°C), IVX-411 formulated with AH or AP displayed decreased in vitro stability compared to AV-formulated IVX-411and this rank-ordering correlated with in vivo performance (mouse pNT values). This case study highlights the importance of characterizing antigen-adjuvant interactions to develop low cost, aluminum-salt adjuvanted recombinant subunit vaccine candidates.
Collapse
Affiliation(s)
- Ozan S. Kumru
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS, USA
| | - Sakshi Bajoria
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS, USA
| | - Kawaljit Kaur
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS, USA
| | - John M. Hickey
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS, USA
| | - Greta Van Slyke
- Division of Infectious Disease, Wadsworth Center, New York State Department of Health, Albany, NY, USA
| | - Jennifer Doering
- Division of Infectious Disease, Wadsworth Center, New York State Department of Health, Albany, NY, USA
| | - Katherine Berman
- Division of Infectious Disease, Wadsworth Center, New York State Department of Health, Albany, NY, USA
| | | | | | - Harry Kleanthous
- Discovery & Translational Sciences, Global Health, Bill and Melinda Gates Foundation, Seattle, WA, USA
| | - Nicholas J. Mantis
- Division of Infectious Disease, Wadsworth Center, New York State Department of Health, Albany, NY, USA
| | - Sangeeta B. Joshi
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS, USA
| | - David B. Volkin
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS, USA
| |
Collapse
|
29
|
Coirada FC, Fernandes ER, Mello LRD, Schuch V, Soares Campos G, Braconi CT, Boscardin SB, Santoro Rosa D. Heterologous DNA Prime- Subunit Protein Boost with Chikungunya Virus E2 Induces Neutralizing Antibodies and Cellular-Mediated Immunity. Int J Mol Sci 2023; 24:10517. [PMID: 37445695 DOI: 10.3390/ijms241310517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 06/07/2023] [Accepted: 06/08/2023] [Indexed: 07/15/2023] Open
Abstract
Chikungunya virus (CHIKV) has become a significant public health concern due to the increasing number of outbreaks worldwide and the associated comorbidities. Despite substantial efforts, there is no specific treatment or licensed vaccine against CHIKV to date. The E2 glycoprotein of CHIKV is a promising vaccine candidate as it is a major target of neutralizing antibodies during infection. In this study, we evaluated the immunogenicity of two DNA vaccines (a non-targeted and a dendritic cell-targeted vaccine) encoding a consensus sequence of E2CHIKV and a recombinant protein (E2*CHIKV). Mice were immunized with different homologous and heterologous DNAprime-E2* protein boost strategies, and the specific humoral and cellular immune responses were accessed. We found that mice immunized with heterologous non-targeted DNA prime- E2*CHIKV protein boost developed high levels of neutralizing antibodies, as well as specific IFN-γ producing cells and polyfunctional CD4+ and CD8+ T cells. We also identified 14 potential epitopes along the E2CHIKV protein. Furthermore, immunization with recombinant E2*CHIKV combined with the adjuvant AS03 presented the highest humoral response with neutralizing capacity. Finally, we show that the heterologous prime-boost strategy with the non-targeted pVAX-E2 DNA vaccine as the prime followed by E2* protein + AS03 boost is a promising combination to elicit a broad humoral and cellular immune response. Together, our data highlights the importance of E2CHIKV for the development of a CHIKV vaccine.
Collapse
Affiliation(s)
- Fernanda Caroline Coirada
- Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de São Paulo (UNIFESP/EPM), São Paulo 04023-062, Brazil
| | - Edgar Ruz Fernandes
- Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de São Paulo (UNIFESP/EPM), São Paulo 04023-062, Brazil
| | - Lucas Rodrigues de Mello
- Departamento de Biofísica, Universidade Federal de São Paulo (UNIFESP/EPM), São Paulo 04044-020, Brazil
| | - Viviane Schuch
- Departamento de Análises Clínicas e Toxicológicas, Universidade de São Paulo (USP), São Paulo 05508-000, Brazil
| | - Gúbio Soares Campos
- Laboratório de Virologia, Universidade Federal da Bahia (UFBA), Salvador 40110-909, Brazil
| | - Carla Torres Braconi
- Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de São Paulo (UNIFESP/EPM), São Paulo 04023-062, Brazil
| | - Silvia Beatriz Boscardin
- Departamento de Parasitologia, Universidade de São Paulo (USP), São Paulo 05508-000, Brazil
- Instituto Nacional de Ciência e Tecnologia de Investigação em Imunologia-INCT (III), São Paulo 05403-900, Brazil
| | - Daniela Santoro Rosa
- Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de São Paulo (UNIFESP/EPM), São Paulo 04023-062, Brazil
- Instituto Nacional de Ciência e Tecnologia de Investigação em Imunologia-INCT (III), São Paulo 05403-900, Brazil
| |
Collapse
|
30
|
Xu S, Duan H, An Y, Jin X, Duan M, Dubois PM, Huang Y, Xu K, Du H, Kleanthous H, Dai L, Gao GF. Effect of adjuvanting RBD-dimer-based subunit COVID-19 vaccines with Sepivac SWE™. Vaccine 2023; 41:2793-2803. [PMID: 36967286 PMCID: PMC10028357 DOI: 10.1016/j.vaccine.2023.03.035] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 03/12/2023] [Accepted: 03/16/2023] [Indexed: 03/24/2023]
Abstract
Protein subunit vaccines have been widely used to combat infectious diseases, including the current COVID-19 pandemic. Adjuvants play the key role in shaping the quality and magnitude of the immune response to protein and inactivated vaccines. We previously developed a protein subunit COVID-19 vaccine, termed ZF2001, based on an aluminium hydroxide-adjuvanted tandem-repeat dimeric receptor-binding domain (RBD) of the viral spike (S) protein. Here, we described the use of a squalene-based oil-in-water adjuvant, Sepivac SWE™ (abbreviated to SWE), to further improve the immunogenicity of this RBD-dimer-based subunit vaccines. Compared with ZF2001, SWE adjuvant enhanced the antibody and CD4+ T-cell responses in mice with at least 10 fold of dose sparing compared with ZF2001 adjuvanted with aluminium hydroxide. SWE-adjuvanted vaccine protected mice against SARS-CoV-2 challenge. To ensure adequate protection against the currently circulating Omicron variant, we evaluated this adjuvant in combination with Delta-Omicron chimeric RBD-dimer. SWE significantly increased antibody responses compared with aluminium hydroxide adjuvant and afforded greater neutralization breadth. These data highlight the advantage of emulsion-based adjuvants to elevate the protective immune response of protein subunit COVID-19 vaccines.
Collapse
Affiliation(s)
- Senyu Xu
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China; Research Network of Immunity and Health (RNIH), Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing 100101, China
| | - Huixin Duan
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China; CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Yaling An
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China; Research Network of Immunity and Health (RNIH), Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing 100101, China
| | - Xiyue Jin
- School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Minrun Duan
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China; School of Life Sciences, Yunnan University, Kunming 650091, China
| | - Patrice M Dubois
- Vaccine Formulation Institute, 1228Plan-Les-Ouates, Geneva, Switzerland
| | - Yan Huang
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China; Beijing Jingdu Children's Hospital, Beijing, China
| | - Kun Xu
- Research Network of Immunity and Health (RNIH), Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing 100101, China
| | - Heng Du
- Bill & Melinda Gates Foundation, Beijing 100027, China
| | - Harry Kleanthous
- Bill & Melinda Gates Foundation, PO Box 23350, Seattle, WA 98102, USA.
| | - Lianpan Dai
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China.
| | - George F Gao
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China; Research Network of Immunity and Health (RNIH), Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing 100101, China; CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
31
|
Zhao L, Shu M, Chen H, Shi K, Li Z. Preparation of graphene oxide-stabilized Pickering emulsion adjuvant for Pgp3 recombinant vaccine and enhanced immunoprotection against Chlamydia Trachomatis infection. Front Immunol 2023; 14:1148253. [PMID: 37143655 PMCID: PMC10152066 DOI: 10.3389/fimmu.2023.1148253] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 03/28/2023] [Indexed: 05/06/2023] Open
Abstract
Background Traditional emulsion adjuvants are limited in clinical application because of their surfactant dependence. Graphene oxide (GO) has unique amphiphilic properties and therefore has potential to be used as a surfactant substitute to stabilize Pickering emulsions. Methods In this study, GO-stabilized Pickering emulsion (GPE) was prepared and used as an adjuvant to facilitate an enhanced immune response to the Chlamydia trachomatis (Ct) Pgp3 recombinant vaccine. Firstly, GPE was prepared by optimizing the sonication conditions, pH, salinity, GO concentration, and water/oil ratio. GPE with small-size droplets was characterized and chosen as the candidate. Subsequently, controlled-release antigen delivery by GPE was explored. Cellular uptake behaviors, M1 polarization, and cytokine stimulation by GPE + Pgp3 was considered in terms of the production of macrophages. Finally, GPE's adjuvant effect was evaluated by vaccination with Pgp3 recombinant in BALB/c mouse models. Results GPE with the smallest droplet sizes was prepared by sonication under 163 W for 2 min at 1 mg/mL GO in natural salinity with a pH of 2 when the water/oil ratio was 10:1 (w/w). The optimized average GPE droplet size was 1.8 μm and the zeta potential was -25.0 ± 1.3 mv. GPE delivered antigens by adsorption onto the droplet surface, demonstrating the controlled release of antigens both in vitro and in vivo. In addition, GPE promoted antigen uptake, which stimulated proinflammatory tumor necrosis factor alpha (TNF-α), enhancing the M1 polarization of macrophages in vitro. Macrophage recruitment was also significantly promoted by GPE at the injection site. In the GPE + Pgp3 treatment group, higher levels of immunoglobin (IgG), immunoglobin G1 (IgG1), immunoglobin G2a (IgG2a) sera, and immunoglobin A (IgA) were detected in vaginal fluid, and higher levels of IFN-γ and IL-2 secretion were stimulated, than in the Pgp3 group, showing a significant type 1 T helper (Th1)-type cellular immune response. Chlamydia muridarum challenging showed that GPE enhanced Pgp3's immunoprotection through its advanced clearance of bacterial burden and alleviation of chronic pathological damage in the genital tract. Conclusion This study enabled the rational design of small-size GPE, shedding light on antigen adsorption and control release, macrophage uptake, polarization and recruitment, which enhanced augmented humoral and cellular immunity and ameliorated chlamydial-induced tissue damage in the genital tract.
Collapse
Affiliation(s)
- Lanhua Zhao
- Institute of Pathogenic Biology, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan, China
| | - Mingyi Shu
- Institute of Pathogenic Biology, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan, China
| | - Hongliang Chen
- ILaboratory Department of Chenzhou First People's Hospital, Chenzhou, Hunan, China
| | - Keliang Shi
- Institute of Pathogenic Biology, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan, China
| | - Zhongyu Li
- Institute of Pathogenic Biology, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan, China
- Institute of Pathogenic Biology, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, The School of Nursing, University of South China, Hengyang, Hunan, China
| |
Collapse
|
32
|
Vono M, Mastelic-Gavillet B, Mohr E, Östensson M, Persson J, Olafsdottir TA, Lemeille S, Pejoski D, Hartley O, Christensen D, Andersen P, Didierlaurent AM, Harandi AM, Lambert PH, Siegrist CA. C-type lectin receptor agonists elicit functional IL21-expressing Tfh cells and induce primary B cell responses in neonates. Front Immunol 2023; 14:1155200. [PMID: 37063899 PMCID: PMC10102809 DOI: 10.3389/fimmu.2023.1155200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 03/16/2023] [Indexed: 04/03/2023] Open
Abstract
IntroductionC-type lectin receptor (CLR) agonists emerged as superior inducers of primary B cell responses in early life compared with Toll-like receptor (TLR) agonists, while both types of adjuvants are potent in adults.MethodsHere, we explored the mechanisms accounting for the differences in neonatal adjuvanticity between a CLR-based (CAF®01) and a TLR4-based (GLA-SE) adjuvant administered with influenza hemagglutinin (HA) in neonatal mice, by using transcriptomics and systems biology analyses.ResultsOn day 7 after immunization, HA/CAF01 increased IL6 and IL21 levels in the draining lymph nodes, while HA/GLA-SE increased IL10. CAF01 induced mixed Th1/Th17 neonatal responses while T cell responses induced by GLA-SE had a more pronounced Th2-profile. Only CAF01 induced T follicular helper (Tfh) cells expressing high levels of IL21 similar to levels induced in adult mice, which is essential for germinal center (GC) formation. Accordingly, only CAF01- induced neonatal Tfh cells activated adoptively transferred hen egg lysozyme (HEL)-specific B cells to form HEL+ GC B cells in neonatal mice upon vaccination with HEL-OVA.DiscussionCollectively, the data show that CLR-based adjuvants are promising neonatal and infant adjuvants due to their ability to harness Tfh responses in early life.
Collapse
Affiliation(s)
- Maria Vono
- Center for Vaccine Immunology, Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
- *Correspondence: Maria Vono,
| | - Beatris Mastelic-Gavillet
- Center for Vaccine Immunology, Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| | - Elodie Mohr
- Center for Vaccine Immunology, Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| | - Malin Östensson
- Department of Microbiology and Immunology, University of Gothenburg, Gothenburg, Sweden
| | - Josefine Persson
- Department of Microbiology and Immunology, University of Gothenburg, Gothenburg, Sweden
| | | | - Sylvain Lemeille
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| | - David Pejoski
- Center for Vaccine Immunology, Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| | - Oliver Hartley
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| | - Dennis Christensen
- Vaccine Adjuvant Research, Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Peter Andersen
- Vaccine Adjuvant Research, Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Arnaud M. Didierlaurent
- Center for Vaccine Immunology, Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| | - Ali M. Harandi
- Department of Microbiology and Immunology, University of Gothenburg, Gothenburg, Sweden
- Vaccine Evaluation Center, British Columbia (BC) Children’s Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Paul-Henri Lambert
- Center for Vaccine Immunology, Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| | - Claire-Anne Siegrist
- Center for Vaccine Immunology, Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| |
Collapse
|
33
|
Myers ML, Gallagher JR, Kim AJ, Payne WH, Maldonado-Puga S, Assimakopoulos H, Bock KW, Torian U, Moore IN, Harris AK. Commercial influenza vaccines vary in HA-complex structure and in induction of cross-reactive HA antibodies. Nat Commun 2023; 14:1763. [PMID: 36997521 PMCID: PMC10060936 DOI: 10.1038/s41467-023-37162-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Accepted: 03/03/2023] [Indexed: 04/04/2023] Open
Abstract
Influenza virus infects millions of people annually and can cause global pandemics. Hemagglutinin (HA) is the primary component of commercial influenza vaccines (CIV), and antibody titer to HA is a primary correlate of protection. Continual antigenic variation of HA requires that CIVs are reformulated yearly. Structural organization of HA complexes have not previously been correlated with induction of broadly reactive antibodies, yet CIV formulations vary in how HA is organized. Using electron microscopy to study four current CIVs, we find structures including: individual HAs, starfish structures with up to 12 HA molecules, and novel spiked-nanodisc structures that display over 50 HA molecules along the complex's perimeter. CIV containing these spiked nanodiscs elicit the highest levels of heterosubtypic cross-reactive antibodies in female mice. Here, we report that HA structural organization can be an important CIV parameter and can be associated with the induction of cross-reactive antibodies to conserved HA epitopes.
Collapse
Affiliation(s)
- Mallory L Myers
- Structural Informatics Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 50 South Drive, Room 6351, Bethesda, MD, 20892, USA
| | - John R Gallagher
- Structural Informatics Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 50 South Drive, Room 6351, Bethesda, MD, 20892, USA
| | - Alexander J Kim
- Structural Informatics Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 50 South Drive, Room 6351, Bethesda, MD, 20892, USA
| | - Walker H Payne
- Structural Informatics Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 50 South Drive, Room 6351, Bethesda, MD, 20892, USA
| | - Samantha Maldonado-Puga
- Structural Informatics Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 50 South Drive, Room 6351, Bethesda, MD, 20892, USA
| | - Haralabos Assimakopoulos
- Structural Informatics Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 50 South Drive, Room 6351, Bethesda, MD, 20892, USA
| | - Kevin W Bock
- Infectious Disease Pathogenesis Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 33 North Drive, Room BN25, Bethesda, MD, 20892, USA
| | - Udana Torian
- Structural Informatics Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 50 South Drive, Room 6351, Bethesda, MD, 20892, USA
- Laboratory of Human Carcinogenesis, National Cancer Institute, 37 Convent Drive, Room 306C, Bethesda, MD, 20892, USA
| | - Ian N Moore
- Infectious Disease Pathogenesis Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 33 North Drive, Room BN25, Bethesda, MD, 20892, USA
- Yerkes National Primate Research Center, Emory University, 954 Gatewood Rd NE, Atlanta, GA, 30329 37, USA
| | - Audray K Harris
- Structural Informatics Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 50 South Drive, Room 6351, Bethesda, MD, 20892, USA.
| |
Collapse
|
34
|
Zhao L, Shu M, Shi K, Tang S, Li Z. Novel use of graphene oxide quantum dots in a pickering emulsion as a Chlamydia trachomatis vaccine adjuvant. Int Immunopharmacol 2023; 118:110035. [PMID: 36958212 DOI: 10.1016/j.intimp.2023.110035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 03/01/2023] [Accepted: 03/10/2023] [Indexed: 03/25/2023]
Abstract
Graphene oxide quantum dots (GOQDs), which are graphene-based nanoparticles, are potential surfactant substitutes for stabilizing Pickering emulsions, due to their high surface area, biodegradability, and reasonable biocompatibility. In the present study, GOQDs stabilized Pickering emulsion (GQPE) was prepared by simple sonication and then used as an adjuvant to enhance immune responses to the Chlamydia trachomatis Pgp3 recombinant vaccine. Immunization of mice showed that GQPE robustly activates adaptive immunity by efficiently stimulating IgG, sIgA, IFN-γ, IL-4, and TNF-α production. Controlled release repository of antigens both in vivo and in vitro prolonged the immune response. In addition, GQPE enhanced dendritic cell recruitment at the injection site, ensuring rapid and efficient innate immunity. Safety assessment revealed that GQPE does not cause liver, kidney, and myocardial damage in mice, suggesting its favorable biocompatibility. This study provides evidence for the use of GOPE as a facile, effective, and safe strategy to enhance the immune response to Pgp3 recombinant vaccines.
Collapse
Affiliation(s)
- Lanhua Zhao
- Institute of Pathogenic Biology, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, the School of Nuring, University of South China, Hengyang, 421001 Hunan, PR China
| | - Mingyi Shu
- Institute of Pathogenic Biology, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, the School of Nuring, University of South China, Hengyang, 421001 Hunan, PR China
| | - Keliang Shi
- Institute of Pathogenic Biology, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, the School of Nuring, University of South China, Hengyang, 421001 Hunan, PR China
| | - Shuangyang Tang
- Institute of Pathogenic Biology, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, the School of Nuring, University of South China, Hengyang, 421001 Hunan, PR China
| | - Zhongyu Li
- Institute of Pathogenic Biology, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, the School of Nuring, University of South China, Hengyang, 421001 Hunan, PR China.
| |
Collapse
|
35
|
Vijayanand S, Patil S, Menon I, Braz Gomes K, Kale A, Bagwe P, Uddin MN, Zughaier SM, D’Souza MJ. An Adjuvanted Inactivated SARS-CoV-2 Microparticulate Vaccine Delivered Using Microneedles Induces a Robust Immune Response in Vaccinated Mice. Pharmaceutics 2023; 15:pharmaceutics15030895. [PMID: 36986756 PMCID: PMC10058898 DOI: 10.3390/pharmaceutics15030895] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 02/21/2023] [Accepted: 03/07/2023] [Indexed: 03/12/2023] Open
Abstract
SARS-CoV-2, the causal agent of COVID-19, is a contagious respiratory virus that frequently mutates, giving rise to variant strains and leading to reduced vaccine efficacy against the variants. Frequent vaccination against the emerging variants may be necessary; thus, an efficient vaccination system is needed. A microneedle (MN) vaccine delivery system is non-invasive, patient-friendly, and can be self-administered. Here, we tested the immune response produced by an adjuvanted inactivated SARS-CoV-2 microparticulate vaccine administered via the transdermal route using a dissolving MN. The inactivated SARS-CoV-2 vaccine antigen and adjuvants (Alhydrogel® and AddaVax™) were encapsulated in poly(lactic-co-glycolic acid) (PLGA) polymer matrices. The resulting MP were approximately 910 nm in size, with a high percentage yield and percent encapsulation efficiency of 90.4%. In vitro, the vaccine MP was non-cytotoxic and increased the immunostimulatory activity measured as nitric oxide release from dendritic cells. The adjuvant MP potentiated the immune response of the vaccine MP in vitro. In vivo, the adjuvanted SARS-CoV-2 MP vaccine induced high levels of IgM, IgG, IgA, IgG1, and IgG2a antibodies and CD4+ and CD8+ T-cell responses in immunized mice. In conclusion, the adjuvanted inactivated SARS-CoV-2 MP vaccine delivered using MN induced a robust immune response in vaccinated mice.
Collapse
Affiliation(s)
- Sharon Vijayanand
- Vaccine Nanotechnology Laboratory, Center for Drug Delivery and Research, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA
| | - Smital Patil
- Vaccine Nanotechnology Laboratory, Center for Drug Delivery and Research, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA
| | - Ipshita Menon
- Vaccine Nanotechnology Laboratory, Center for Drug Delivery and Research, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA
| | - Keegan Braz Gomes
- Vaccine Nanotechnology Laboratory, Center for Drug Delivery and Research, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA
| | - Akanksha Kale
- Vaccine Nanotechnology Laboratory, Center for Drug Delivery and Research, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA
| | - Priyal Bagwe
- Vaccine Nanotechnology Laboratory, Center for Drug Delivery and Research, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA
| | - Mohammad N. Uddin
- Vaccine Nanotechnology Laboratory, Center for Drug Delivery and Research, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA
| | - Susu M. Zughaier
- College of Medicine, QU Health, Qatar University, Doha P.O. Box 2713, Qatar
- Correspondence:
| | - Martin J. D’Souza
- Vaccine Nanotechnology Laboratory, Center for Drug Delivery and Research, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA
| |
Collapse
|
36
|
Verma SK, Mahajan P, Singh NK, Gupta A, Aggarwal R, Rappuoli R, Johri AK. New-age vaccine adjuvants, their development, and future perspective. Front Immunol 2023; 14:1043109. [PMID: 36911719 PMCID: PMC9998920 DOI: 10.3389/fimmu.2023.1043109] [Citation(s) in RCA: 67] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 01/26/2023] [Indexed: 02/26/2023] Open
Abstract
In the present scenario, immunization is of utmost importance as it keeps us safe and protects us from infectious agents. Despite the great success in the field of vaccinology, there is a need to not only develop safe and ideal vaccines to fight deadly infections but also improve the quality of existing vaccines in terms of partial or inconsistent protection. Generally, subunit vaccines are known to be safe in nature, but they are mostly found to be incapable of generating the optimum immune response. Hence, there is a great possibility of improving the potential of a vaccine in formulation with novel adjuvants, which can effectively impart superior immunity. The vaccine(s) in formulation with novel adjuvants may also be helpful in fighting pathogens of high antigenic diversity. However, due to the limitations of safety and toxicity, very few human-compatible adjuvants have been approved. In this review, we mainly focus on the need for new and improved vaccines; the definition of and the need for adjuvants; the characteristics and mechanisms of human-compatible adjuvants; the current status of vaccine adjuvants, mucosal vaccine adjuvants, and adjuvants in clinical development; and future directions.
Collapse
Affiliation(s)
| | - Pooja Mahajan
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Nikhlesh K. Singh
- Integrative Biosciences Center, Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University, School of Medicine, Detroit, MI, United States
| | - Ankit Gupta
- Microbiology Division, Defence Research and Development Establishment, Gwalior, India
| | - Rupesh Aggarwal
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | | | - Atul Kumar Johri
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| |
Collapse
|
37
|
Molina Estupiñan JL, Aradottir Pind AA, Foroutan Pajoohian P, Jonsdottir I, Bjarnarson SP. The adjuvants dmLT and mmCT enhance humoral immune responses to a pneumococcal conjugate vaccine after both parenteral or mucosal immunization of neonatal mice. Front Immunol 2023; 13:1078904. [PMID: 36741402 PMCID: PMC9896006 DOI: 10.3389/fimmu.2022.1078904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 12/12/2022] [Indexed: 01/21/2023] Open
Abstract
Immaturity of the neonatal immune system contributes to increased susceptibility to infectious diseases and poor vaccine responses. Therefore, better strategies for early life vaccination are needed. Adjuvants can enhance the magnitude and duration of immune responses. In this study we assessed the effects of the adjuvants dmLT and mmCT and different immunization routes, subcutaneous (s.c.) and intranasal (i.n.), on neonatal immune response to a pneumococcal conjugate vaccine Pn1-CRM197. Pn1-specific antibody (Ab) levels of neonatal mice immunized with Pn1-CRM197 alone were low. The adjuvants enhanced IgG Ab responses up to 8 weeks after immunization, more after s.c. than i.n. immunization. On the contrary, i.n. immunization with either adjuvant enhanced serum and salivary IgA levels more than s.c. immunization. In addition, both dmLT and mmCT enhanced germinal center formation and accordingly, dmLT and mmCT enhanced the induction and persistence of Pn1-specific IgG+ Ab-secreting cells (ASCs) in spleen and bone marrow (BM), irrespective of the immunization route. Furthermore, i.n. immunization enhanced Pn1-specific IgA+ ASCs in BM more than s.c. immunizatiofimmu.2022.1078904n. However, a higher i.n. dose of the Pn1-CRM197 was needed to achieve IgG response comparable to that elicited by s.c. immunization with either adjuvant. We conclude that dmLT and mmCT enhance both induction and persistence of the neonatal immune response to the vaccine Pn1-CRM197, following mucosal or parenteral immunization. This indicates that dmLT and mmCT are promising adjuvants for developing safe and effective early life vaccination strategies.
Collapse
Affiliation(s)
- Jenny Lorena Molina Estupiñan
- Department of Immunology, Landspitali, The National University Hospital of Iceland, Reykjavik, Iceland,Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Audur Anna Aradottir Pind
- Department of Immunology, Landspitali, The National University Hospital of Iceland, Reykjavik, Iceland,Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Poorya Foroutan Pajoohian
- Department of Immunology, Landspitali, The National University Hospital of Iceland, Reykjavik, Iceland,Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Ingileif Jonsdottir
- Department of Immunology, Landspitali, The National University Hospital of Iceland, Reykjavik, Iceland,Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Stefania P. Bjarnarson
- Department of Immunology, Landspitali, The National University Hospital of Iceland, Reykjavik, Iceland,Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland,*Correspondence: Stefania P. Bjarnarson,
| |
Collapse
|
38
|
Yang J, Dong X, Li B, Chen T, Yu B, Wang X, Dou X, Peng B, Hu Q. Poria cocos polysaccharide-functionalized graphene oxide nanosheet induces efficient cancer immunotherapy in mice. Front Bioeng Biotechnol 2023; 10:1050077. [PMID: 36727039 PMCID: PMC9885324 DOI: 10.3389/fbioe.2022.1050077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 12/27/2022] [Indexed: 01/18/2023] Open
Abstract
Introduction: Tumor vaccines that induce robust humoral and cellular immune responses have attracted tremendous interest for cancer immunotherapy. Despite the tremendous potential of tumor vaccines as an effective approach for cancer treatment and prevention, a major challenge in achieving sustained antitumor immunity is inefficient antigen delivery to secondary lymphoid organs, even with adjuvant aid. Methods: Herein, we present antigen/adjuvant integrated nanocomplexes termed nsGO/PCP/OVA by employing graphene oxide nanosheet (nsGO) as antigen nanocarriers loaded with model antigen ovalbumin (OVA) and adjuvant, Poria cocos polysaccharides (PCP). We evaluated the efficacy of nsGO/PCP/OVA in activating antigen-specific humoral as well as cellular immune responses and consequent tumor prevention and rejection in vivo. Results: The optimally formed nsGO/PCP/OVA was approximately 120-150 nm in diameter with a uniform size distribution. Nanoparticles can be effectively engulfed by dendritic cells (DCs) through receptor-mediated endocytosis, induced the maturation of DCs and improved the delivery efficiency both in vitro and in vivo. The nsGO/PCP/OVA nanoparticles also induced a significant enhancement of OVA antigen-specific Th1 and Th2 immune responses in vivo. In addition, vaccination with nsGO/PCP/OVA not only significantly suppressed tumor growth in prophylactic treatments, but also achieved a therapeutic effect in inhibiting the growth of already-established tumors. Conclusion: Therefore, this potent nanovaccine platform with nanocarrier nsGO and PCP as adjuvants provides a promising strategy for boosting anti-tumor immunity for cancer immunotherapy.
Collapse
Affiliation(s)
- Jinning Yang
- The Faculty of Environment and Life, Beijing University of Technology, Beijing, China,Beijing International Science and Technology Cooperation Base of Antivirus Drug, Beijing University of Technology, Beijing, China
| | - Xiaoxiao Dong
- Institute of Medical Biotechnology, Chinese Academy of Medical Sciences, Beijing, China
| | - Boye Li
- Civil Aviation Medicine Center, Civil Aviation Administration of China, Beijing, China
| | - Tian Chen
- The Faculty of Environment and Life, Beijing University of Technology, Beijing, China,Beijing International Science and Technology Cooperation Base of Antivirus Drug, Beijing University of Technology, Beijing, China
| | - Boyang Yu
- The Faculty of Environment and Life, Beijing University of Technology, Beijing, China,Beijing International Science and Technology Cooperation Base of Antivirus Drug, Beijing University of Technology, Beijing, China
| | - Xiaoli Wang
- The Faculty of Environment and Life, Beijing University of Technology, Beijing, China,Beijing International Science and Technology Cooperation Base of Antivirus Drug, Beijing University of Technology, Beijing, China,*Correspondence: Xiaoli Wang, ; Xiangnan Dou, ; Bo Peng, ; Qin Hu,
| | - Xiangnan Dou
- The Faculty of Environment and Life, Beijing University of Technology, Beijing, China,*Correspondence: Xiaoli Wang, ; Xiangnan Dou, ; Bo Peng, ; Qin Hu,
| | - Bo Peng
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China,*Correspondence: Xiaoli Wang, ; Xiangnan Dou, ; Bo Peng, ; Qin Hu,
| | - Qin Hu
- The Faculty of Environment and Life, Beijing University of Technology, Beijing, China,Beijing International Science and Technology Cooperation Base of Antivirus Drug, Beijing University of Technology, Beijing, China,*Correspondence: Xiaoli Wang, ; Xiangnan Dou, ; Bo Peng, ; Qin Hu,
| |
Collapse
|
39
|
Ko KH, Cha SB, Lee SH, Bae HS, Ham CS, Lee MG, Kim DH, Han SH. A novel defined TLR3 agonist as an effective vaccine adjuvant. Front Immunol 2023; 14:1075291. [PMID: 36761735 PMCID: PMC9902914 DOI: 10.3389/fimmu.2023.1075291] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 01/06/2023] [Indexed: 01/25/2023] Open
Abstract
Synthetic double-stranded RNA analogs recognized by Toll-like receptor 3 (TLR3) are an attractive adjuvant candidate for vaccines, especially against intracellular pathogens or tumors, because of their ability to enhance T cell and antibody responses. Although poly(I:C) is a representative dsRNA with potent adjuvanticity, its clinical application has been limited due to heterogeneous molecular size, inconsistent activity, poor stability, and toxicity. To overcome these limitations, we developed a novel dsRNA-based TLR3 agonist named NexaVant (NVT) by using PCR-coupled bidirectional in vitro transcription. Agarose gel electrophoresis and reverse phase-HPLC analysis demonstrated that NVT is a single 275-kDa homogeneous molecule. NVT appears to be stable since its appearance, concentration, and molecular size were unaffected under 6 months of accelerated storage conditions. Moreover, preclinical evaluation of toxicity under good laboratory practices showed that NVT is a safe substance without any signs of serious toxicity. NVT stimulated TLR3 and increased the expression of viral nucleic acid sensors TLR3, MDA-5, and RIG-1. When intramuscularly injected into C57BL/6 mice, ovalbumin (OVA) plus NVT highly increased the migration of dendritic cells (DCs), macrophages, and neutrophils into inguinal lymph node (iLN) compared with OVA alone. In addition, NVT substantially induced the phenotypic markers of DC maturation and activation including MHC-II, CD40, CD80, and CD86 together with IFN-β production. Furthermore, NVT exhibited an appropriate adjuvanticity because it elevated OVA-specific IgG, in particular, higher levels of IgG2c (Th1-type) but lower IgG1 (Th2-type). Concomitantly, NVT increased the levels of Th1-type T cells such as IFN-γ+CD4+ and IFN-γ+CD8+ cells in response to OVA stimulation. Collectively, we suggest that NVT with appropriate safety and effectiveness is a novel and promising adjuvant for vaccines, especially those requiring T cell mediated immunity such as viral and cancer vaccines.
Collapse
Affiliation(s)
- Kwang Hyun Ko
- Research and Development Center, NA Vaccine Institute, Seoul, Republic of Korea.,Interdisciplinary Program in Genetic Engineering, College of Natural Sciences, Seoul National University, Seoul, Republic of Korea
| | - Seung Bin Cha
- Research and Development Center, NA Vaccine Institute, Seoul, Republic of Korea
| | - Seung-Hwan Lee
- Research and Development Center, NA Vaccine Institute, Seoul, Republic of Korea
| | - Hyun Shik Bae
- Research and Development Center, NA Vaccine Institute, Seoul, Republic of Korea
| | - Chul Soo Ham
- Research and Development Center, NA Vaccine Institute, Seoul, Republic of Korea
| | - Min-Gyu Lee
- Research and Development Center, NA Vaccine Institute, Seoul, Republic of Korea
| | - Dong-Ho Kim
- Research and Development Center, NA Vaccine Institute, Seoul, Republic of Korea
| | - Seung Hyun Han
- Interdisciplinary Program in Genetic Engineering, College of Natural Sciences, Seoul National University, Seoul, Republic of Korea.,Department of Oral Microbiology and Immunology, and Dental Research Institute, School of Dentistry, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
40
|
Microneedle Delivery of an Adjuvanted Microparticulate Vaccine Induces High Antibody Levels in Mice Vaccinated against Coronavirus. Vaccines (Basel) 2022; 10:vaccines10091491. [PMID: 36146568 PMCID: PMC9503342 DOI: 10.3390/vaccines10091491] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/01/2022] [Accepted: 09/05/2022] [Indexed: 11/26/2022] Open
Abstract
This ‘proof-of-concept’ study aimed to test the microparticulate vaccine delivery system and a transdermal vaccine administration strategy using dissolving microneedles (MN). For this purpose, we formulated poly(lactic-co-glycolic) acid (PLGA) microparticles (MP) encapsulating the inactivated canine coronavirus (iCCoV), as a model antigen, along with adjuvant MP encapsulating Alhydrogel® and AddaVax. We characterized the vaccine MP for size, surface charge, morphology, and encapsulation efficiency. Further, we evaluated the in vitro immunogenicity, cytotoxicity, and antigen-presentation of vaccine/adjuvant MP in murine dendritic cells (DCs). Additionally, we tested the in vivo immunogenicity of the MP vaccine in mice through MN administration. We evaluated the serum IgG, IgA, IgG1, and IgG2a responses using an enzyme-linked immunosorbent assay. The results indicate that the particulate form of the vaccine is more immunogenic than the antigen suspension in vitro. We found the vaccine/adjuvant MP to be non-cytotoxic to DCs. The expression of antigen-presenting molecules, MHC I/II, and their costimulatory molecules, CD80/40, increased with the addition of the adjuvants. Moreover, the results suggest that the MP vaccine is cross presented by the DCs. In vivo, the adjuvanted MP vaccine induced increased antibody levels in mice following vaccination and will further be assessed for its cell-mediated responses.
Collapse
|
41
|
Aradottir Pind AA, Thorsdottir S, Magnusdottir G, Meinke A, Del Giudice G, Jonsdottir I, Bjarnarson SP. A comparative study of adjuvants effects on neonatal plasma cell survival niche in bone marrow and persistence of humoral immune responses. Front Immunol 2022; 13:904415. [PMID: 35990686 PMCID: PMC9381929 DOI: 10.3389/fimmu.2022.904415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 07/07/2022] [Indexed: 11/13/2022] Open
Abstract
The neonatal immune system is distinct from the immune system of older individuals rendering neonates vulnerable to infections and poor responders to vaccination. Adjuvants can be used as tools to enhance immune responses to co-administered antigens. Antibody (Ab) persistence is mediated by long-lived plasma cells that reside in specialized survival niches in the bone marrow, and transient Ab responses in early life have been associated with decreased survival of plasma cells, possibly due to lack of survival factors. Various cells can secrete these factors and which cells are the main producers is still up for debate, especially in early life where this has not been fully addressed. The receptor BCMA and its ligand APRIL have been shown to be important in the maintenance of plasma cells and Abs. Herein, we assessed age-dependent maturation of a broad range of bone marrow accessory cells and their expression of the survival factors APRIL and IL-6. Furthermore, we performed a comparative analysis of the potential of 5 different adjuvants; LT-K63, mmCT, MF59, IC31 and alum, to enhance expression of survival factors and BCMA following immunization of neonatal mice with tetanus toxoid (TT) vaccine. We found that APRIL expression was reduced in the bone marrow of young mice whereas IL-6 expression was higher. Eosinophils, macrophages, megakaryocytes, monocytes and lymphocytes were important secretors of survival factors in early life but undefined cells also constituted a large fraction of secretors. Immunization and adjuvants enhanced APRIL expression but decreased IL-6 expression in bone marrow cells early after immunization. Furthermore, neonatal immunization with adjuvants enhanced the proportion of plasmablasts and plasma cells that expressed BCMA both in spleen and bone marrow. Enhanced BCMA expression correlated with enhanced vaccine-specific humoral responses, even though the effect of alum on BCMA was less pronounced than those of the other adjuvants at later time points. We propose that low APRIL expression in bone marrow as well as low BCMA expression of plasmablasts/plasma cells in early life together cause transient Ab responses and could represent targets to be triggered by vaccine adjuvants to induce persistent humoral immune responses in this age group.
Collapse
Affiliation(s)
- Audur Anna Aradottir Pind
- Department of Immunology, Landspitali, The National University Hospital of Iceland, Reykjavik, Iceland
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Sigrun Thorsdottir
- Department of Immunology, Landspitali, The National University Hospital of Iceland, Reykjavik, Iceland
| | - Gudbjorg Julia Magnusdottir
- Department of Immunology, Landspitali, The National University Hospital of Iceland, Reykjavik, Iceland
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | | | | | - Ingileif Jonsdottir
- Department of Immunology, Landspitali, The National University Hospital of Iceland, Reykjavik, Iceland
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Stefania P. Bjarnarson
- Department of Immunology, Landspitali, The National University Hospital of Iceland, Reykjavik, Iceland
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
- *Correspondence: Stefania P. Bjarnarson,
| |
Collapse
|
42
|
Tabynov K, Babayeva M, Nurpeisov T, Fomin G, Nurpeisov T, Saltabayeva U, Renu S, Renukaradhya GJ, Petrovsky N, Tabynov K. Evaluation of a Novel Adjuvanted Vaccine for Ultrashort Regimen Therapy of Artemisia Pollen-Induced Allergic Bronchial Asthma in a Mouse Model. Front Immunol 2022; 13:828690. [PMID: 35371056 PMCID: PMC8965083 DOI: 10.3389/fimmu.2022.828690] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 02/15/2022] [Indexed: 12/25/2022] Open
Abstract
Wormwood (Artemisia) pollen is among the top 10 aeroallergens globally that cause allergic rhinitis and bronchial asthma. Allergen-specific immunotherapy (ASIT) is the gold standard for treating patients with allergic rhinitis, conjunctivitis, and asthma. A significant disadvantage of today's ASIT methods is the long duration of therapy and multiplicity of allergen administrations. The goal of this study was to undertake a pilot study in mice of a novel ultrashort vaccine immunotherapy regimen incorporating various adjuvants to assess its ability to treat allergic bronchial asthma caused by wormwood pollen. We evaluated in a mouse model of wormwood pollen allergy candidates comprising recombinant Art v 1 wormwood pollen protein formulated with either newer (Advax, Advax-CpG, ISA-51) or more traditional [aluminum hydroxide, squalene water emulsion (SWE)] adjuvants administered by the intramuscular or subcutaneous route vs. intranasal administration of a mucosal vaccine formulation using chitosan-mannose nanoparticle entrapped with Art v 1 protein. The vaccine formulations were administered to previously wormwood pollen-sensitized animals, four times at weekly intervals. Desensitization was determined by measuring decreases in immunoglobulin E (IgE), cellular immunity, ear swelling test, and pathological changes in the lungs of animals after aeroallergen challenge. Art v 1 protein formulation with Advax, Advax-CpG, SWE, or ISA-51 adjuvants induced a significant decrease in both total and Art v 1-specific IgE with a concurrent increase in Art v 1-specific IgG compared to the positive control group. There was a shift in T-cell cytokine secretion toward a Th1 (Advax-CpG, ISA-51, and Advax) or a balanced Th1/Th2 (SWE) pattern. Protection against lung inflammatory reaction after challenge was seen with ISA-51, Advax, and SWE Art v 1 formulations. Overall, the ISA-51-adjuvanted vaccine group induced the largest reduction of allergic ear swelling and protection against type 2 and non-type 2 lung inflammation in challenged animals. This pilot study shows the potential to develop an ultrashort ASIT regimen for wormwood pollen-induced bronchial asthma using appropriately adjuvanted recombinant Art v 1 protein. The data support further preclinical studies with the ultimate goal of advancing this therapy to human clinical trials.
Collapse
Affiliation(s)
- Kairat Tabynov
- International Center for Vaccinology, Kazakh National Agrarian Research University (KazNARU), Almaty, Kazakhstan.,Preclinical Research Laboratory With Vivarium, M. Aikimbayev National Research Center for Especially Dangerous Infections, Almaty, Kazakhstan.,T&TvaX LLC, Almaty, Kazakhstan
| | - Meruert Babayeva
- International Center for Vaccinology, Kazakh National Agrarian Research University (KazNARU), Almaty, Kazakhstan.,Department of General Immunology, Asfendiyarov Kazakh National Medical University (KazNMU), Almaty, Kazakhstan
| | - Tair Nurpeisov
- Department of General Immunology, Asfendiyarov Kazakh National Medical University (KazNMU), Almaty, Kazakhstan.,Republican Allergy Center, Research Institute of Cardiology and Internal Medicine, Almaty, Kazakhstan
| | - Gleb Fomin
- International Center for Vaccinology, Kazakh National Agrarian Research University (KazNARU), Almaty, Kazakhstan
| | - Temirzhan Nurpeisov
- Department of General Immunology, Asfendiyarov Kazakh National Medical University (KazNMU), Almaty, Kazakhstan
| | | | - Sankar Renu
- Center for Food Animal Health, Ohio Agricultural Research and Development Center, The Ohio State University (OSU), Wooster, OH, United States
| | - Gourapura J Renukaradhya
- Center for Food Animal Health, Ohio Agricultural Research and Development Center, The Ohio State University (OSU), Wooster, OH, United States
| | | | - Kaissar Tabynov
- International Center for Vaccinology, Kazakh National Agrarian Research University (KazNARU), Almaty, Kazakhstan.,T&TvaX LLC, Almaty, Kazakhstan.,Republican Allergy Center, Research Institute of Cardiology and Internal Medicine, Almaty, Kazakhstan
| |
Collapse
|
43
|
Mendes A, Azevedo-Silva J, Fernandes JC. From Sharks to Yeasts: Squalene in the Development of Vaccine Adjuvants. Pharmaceuticals (Basel) 2022; 15:265. [PMID: 35337064 PMCID: PMC8951290 DOI: 10.3390/ph15030265] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 02/11/2022] [Accepted: 02/16/2022] [Indexed: 02/04/2023] Open
Abstract
Squalene is a natural linear triterpene that can be found in high amounts in certain fish liver oils, especially from deep-sea sharks, and to a lesser extent in a wide variety of vegeTable oils. It is currently used for numerous vaccine and drug delivery emulsions due to its stability-enhancing properties and biocompatibility. Squalene-based vaccine adjuvants, such as MF59 (Novartis), AS03 (GlaxoSmithKline Biologicals), or AF03 (Sanofi) are included in seasonal vaccines against influenza viruses and are presently being considered for inclusion in several vaccines against SARS-CoV-2 and future pandemic threats. However, harvesting sharks for this purpose raises serious ecological concerns that the exceptional demand of the pandemic has exacerbated. In this line, the use of plants to obtain phytosqualene has been seen as a more sustainable alternative, yet the lower yields and the need for huge investments in infrastructures and equipment makes this solution economically ineffective. More recently, the enormous advances in the field of synthetic biology provided innovative approaches to make squalene production more sustainable, flexible, and cheaper by using genetically modified microbes to produce pharmaceutical-grade squalene. Here, we review the biological mechanisms by which squalene-based vaccine adjuvants boost the immune response, and further compare the existing sources of squalene and their environmental impact. We propose that genetically engineered microbes are a sustainable alternative to produce squalene at industrial scale, which are likely to become the sole source of pharmaceutical-grade squalene in the foreseeable future.
Collapse
Affiliation(s)
- Adélia Mendes
- Centro de Biotecnologia e Química Fina (CBQF), Escola Superior de Biotecnologia, Universidade Católica Portuguesa, Amyris Bio Products Portugal, 4169-005 Porto, Portugal; (J.A.-S.); (J.C.F.)
| | | | | |
Collapse
|
44
|
Yang JX, Tseng JC, Yu GY, Luo Y, Huang CYF, Hong YR, Chuang TH. Recent Advances in the Development of Toll-like Receptor Agonist-Based Vaccine Adjuvants for Infectious Diseases. Pharmaceutics 2022; 14:pharmaceutics14020423. [PMID: 35214155 PMCID: PMC8878135 DOI: 10.3390/pharmaceutics14020423] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 02/11/2022] [Accepted: 02/14/2022] [Indexed: 02/06/2023] Open
Abstract
Vaccines are powerful tools for controlling microbial infections and preventing epidemic diseases. Efficient inactive, subunit, or viral-like particle vaccines usually rely on a safe and potent adjuvant to boost the immune response to the antigen. After a slow start, over the last decade there has been increased developments on adjuvants for human vaccines. The development of adjuvants has paralleled our increased understanding of the molecular mechanisms for the pattern recognition receptor (PRR)-mediated activation of immune responses. Toll-like receptors (TLRs) are a group of PRRs that recognize microbial pathogens to initiate a host’s response to infection. Activation of TLRs triggers potent and immediate innate immune responses, which leads to subsequent adaptive immune responses. Therefore, these TLRs are ideal targets for the development of effective adjuvants. To date, TLR agonists such as monophosphoryl lipid A (MPL) and CpG-1018 have been formulated in licensed vaccines for their adjuvant activity, and other TLR agonists are being developed for this purpose. The COVID-19 pandemic has also accelerated clinical research of vaccines containing TLR agonist-based adjuvants. In this paper, we reviewed the agonists for TLR activation and the molecular mechanisms associated with the adjuvants’ effects on TLR activation, emphasizing recent advances in the development of TLR agonist-based vaccine adjuvants for infectious diseases.
Collapse
Affiliation(s)
- Jing-Xing Yang
- Immunology Research Center, National Health Research Institutes, Miaoli 35053, Taiwan; (J.-X.Y.); (J.-C.T.)
| | - Jen-Chih Tseng
- Immunology Research Center, National Health Research Institutes, Miaoli 35053, Taiwan; (J.-X.Y.); (J.-C.T.)
| | - Guann-Yi Yu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli 35053, Taiwan;
| | - Yunping Luo
- Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing 100005, China;
| | - Chi-Ying F. Huang
- Institute of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan;
| | - Yi-Ren Hong
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
| | - Tsung-Hsien Chuang
- Immunology Research Center, National Health Research Institutes, Miaoli 35053, Taiwan; (J.-X.Y.); (J.-C.T.)
- Department of Life Sciences, National Central University, Taoyuan City 32001, Taiwan
- Program in Environmental and Occupational Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Correspondence: ; Tel.: +886-37-246166 (ext. 37611)
| |
Collapse
|
45
|
Abstract
Prevention of emerging infections in children is a dynamic arena where substantial medical advances have enabled intervention and prevention of infection outbreaks. This article discusses 5 infections causing significant morbidity and mortality across Asia, Latin America, and Africa. Avian influenza and the Middle East respiratory syndrome are highly contagious zoonoses spread through aerosol and droplets, affecting predominantly Asia. Dengue infection and chikungunya are endemic mosquito-borne viruses in tropical regions across Asia, Latin America, and Africa. Ebola is a highly contagious virus spread through human-to-human contact. The latest information in clinical manifestations, infection, prevention control, chemoprophylaxis, vaccination, and public health measures is reviewed.
Collapse
Affiliation(s)
- Thanyawee Puthanakit
- Division of Pediatric Infectious Diseases, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, King Chulalongkorn Memorial Hospital, 9th Floor, Sor Kor Building, Rama 4 Road, Patumwan, Bangkok 10330, Thailand.
| | | | - Watsamon Jantarabenjakul
- Center of Excellence for Pediatric Infectious Diseases, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Bangkok, Thailand
| |
Collapse
|
46
|
Mangla B, Javed S, Sultan MH, Ahsan W, Aggarwal G, Kohli K. Nanocarriers-Assisted Needle-Free Vaccine Delivery Through Oral and Intranasal Transmucosal Routes: A Novel Therapeutic Conduit. Front Pharmacol 2022; 12:757761. [PMID: 35087403 PMCID: PMC8787087 DOI: 10.3389/fphar.2021.757761] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 12/21/2021] [Indexed: 01/01/2023] Open
Abstract
Drug delivery using oral route is the most popular, convenient, safest and least expensive approach. It includes oral transmucosal delivery of bioactive compounds as the mucosal cavity offers an intriguing approach for systemic drug distribution. Owing to the dense vascular architecture and high blood flow, oral mucosal layers are easily permeable and can be an ideal site for drug administration. Recently, the transmucosal route is being investigated for other therapeutic candidates such as vaccines for their efficient delivery. Vaccines have the potential to trigger immune reactions and can act as both prophylactic and therapeutic conduit to a variety of diseases. Administration of vaccines using transmucosal route offers multiple advantages, the most important one being the needle-free (non-invasive) delivery. Development of needle-free devices are the most recent and pioneering breakthrough in the delivery of drugs and vaccines, enabling patients to avoid needles, reducing anxiety, pain and fear as well as improving compliance. Oral, nasal and aerosol vaccination is a novel immunization approach that utilizes a nanocarrier to administer the vaccine. Nanocarriers improve the bioavailability and serve as adjuvants to elicit a stronger immune response, resulting in increased effectiveness of vaccination. Drugs and vaccines with lower penetration abilities can also be delivered transmucosally while maintaining their biological function. The development of micro/nanocarriers for transmucosal delivery of macromolecules, vaccines and other substances is currently drawing much attention and a number of studies were performed recently. This comprehensive review is aimed to summarize the most recent investigations on needle-free and non-invasive approaches for the delivery of vaccines using oral transmucosal route, their strengths and associated challenges. The oral transmucosal vaccine delivery by nanocarriers is the most upcoming advancement in efficient vaccine delivery and this review would help further research and trials in this field.
Collapse
Affiliation(s)
- Bharti Mangla
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University (DPSRU), New Delhi, India
| | - Shamama Javed
- Department of Pharmaceutics, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
| | - Muhammad H. Sultan
- Department of Pharmaceutics, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
| | - Waquar Ahsan
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
| | - Geeta Aggarwal
- Department of Pharmaceutics, Delhi Pharmaceutical Sciences and Research University, New Delhi, India
| | - Kanchan Kohli
- Director Research and Publication, Lloyd Institute of Management and Technology (Pharm.), Greater Noida, India
| |
Collapse
|
47
|
O'Hagan DT, van der Most R, Lodaya RN, Coccia M, Lofano G. "World in motion" - emulsion adjuvants rising to meet the pandemic challenges. NPJ Vaccines 2021; 6:158. [PMID: 34934069 PMCID: PMC8692316 DOI: 10.1038/s41541-021-00418-0] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Accepted: 11/23/2021] [Indexed: 02/06/2023] Open
Abstract
Emulsion adjuvants such as MF59 and AS03 have been used for more than two decades as key components of licensed vaccines, with over 100 million doses administered to diverse populations in more than 30 countries. Substantial clinical experience of effectiveness and a well-established safety profile, along with the ease of manufacturing have established emulsion adjuvants as one of the leading platforms for the development of pandemic vaccines. Emulsion adjuvants allow for antigen dose sparing, more rapid immune responses, and enhanced quality and quantity of adaptive immune responses. The mechanisms of enhancement of immune responses are well defined and typically characterized by the creation of an "immunocompetent environment" at the site of injection, followed by the induction of strong and long-lasting germinal center responses in the draining lymph nodes. As a result, emulsion adjuvants induce distinct immunological responses, with a mixed Th1/Th2 T cell response, long-lived plasma cells, an expanded repertoire of memory B cells, and high titers of cross-neutralizing polyfunctional antibodies against viral variants. Because of these various properties, emulsion adjuvants were included in pandemic influenza vaccines deployed during the 2009 H1N1 influenza pandemic, are still included in seasonal influenza vaccines, and are currently at the forefront of the development of vaccines against emerging SARS-CoV-2 pandemic variants. Here, we comprehensively review emulsion adjuvants, discuss their mechanism of action, and highlight their profile as a benchmark for the development of additional vaccine adjuvants and as a valuable tool to allow further investigations of the general principles of human immunity.
Collapse
|
48
|
Du X, Tan D, Gong Y, Zhang Y, Han J, Lv W, Xie T, He P, Hou Z, Xu K, Tan J, Zhu B. A new poly(I:C)-decorated PLGA-PEG nanoparticle promotes Mycobacterium tuberculosis fusion protein to induce comprehensive immune responses in mice intranasally. Microb Pathog 2021; 162:105335. [PMID: 34861347 DOI: 10.1016/j.micpath.2021.105335] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 11/03/2021] [Accepted: 11/27/2021] [Indexed: 11/25/2022]
Abstract
Protein-based subunit vaccine against tuberculosis (TB) is regarded as safer but with lower immunogenicity. To investigate effective adjuvant to improve the immunogenicity of TB subunit vaccine, we modified ploy(I:C) onto PLGA-PEG copolymer nanoparticle with polydopamine to produce a new nanoparticle adjuvant named "PLGA-PEG-poly(I:C)" (NP). M. tuberculosis fusion proteins Mtb10.4-HspX and ESAT-6-Rv2626c (M4) were encapsulated in the nanoparticles to produce the NP/M4 subunit vaccine. The PLGA-PEG/M4 nanoparticle was 200.21 ± 1.07 nm in diameter, and the polydispersity index (PDI) was 0.127 ± 0.02. Following modification with poly(I:C) by polydopamine, the NP/M4 was administered to C57BL/6 female mice intranasally and the immune responses were evaluated. The NP/M4 significantly induced antigen-specific CD4+ T cells proliferation, IL-2 and IFN-γ production. In addition, the NP/M4 could promote the production of antigen-specific IgG, IgG1, IgG2c in serum, and sIgA in lung washings. Overall, our results indicated that the NP would be a potential TB subunit vaccine adjuvant with the ability to induce strong Th1-type cell-mediated immunity and humoral immune responses.
Collapse
Affiliation(s)
- Xiufen Du
- Gansu Provincial Key Laboratory of Evidence Based Medicine and Clinical Translation and Lanzhou Center for Tuberculosis Research, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China; Department of Immunology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Daquan Tan
- Gansu Provincial Key Laboratory of Evidence Based Medicine and Clinical Translation and Lanzhou Center for Tuberculosis Research, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China; Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Yang Gong
- Gansu Provincial Key Laboratory of Evidence Based Medicine and Clinical Translation and Lanzhou Center for Tuberculosis Research, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China; Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Yifan Zhang
- Gansu Provincial Key Laboratory of Evidence Based Medicine and Clinical Translation and Lanzhou Center for Tuberculosis Research, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China; Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Jiangyuan Han
- Gansu Provincial Key Laboratory of Evidence Based Medicine and Clinical Translation and Lanzhou Center for Tuberculosis Research, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China; Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Wei Lv
- Gansu Provincial Key Laboratory of Evidence Based Medicine and Clinical Translation and Lanzhou Center for Tuberculosis Research, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China; Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Tao Xie
- Gansu Provincial Key Laboratory of Evidence Based Medicine and Clinical Translation and Lanzhou Center for Tuberculosis Research, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China; Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Pu He
- Gansu Provincial Key Laboratory of Evidence Based Medicine and Clinical Translation and Lanzhou Center for Tuberculosis Research, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China; Department of Immunology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Zongjie Hou
- Department of Immunology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Kun Xu
- Gansu Provincial Key Laboratory of Evidence Based Medicine and Clinical Translation and Lanzhou Center for Tuberculosis Research, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China; Department of Immunology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Jiying Tan
- Gansu Provincial Key Laboratory of Evidence Based Medicine and Clinical Translation and Lanzhou Center for Tuberculosis Research, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China; Department of Immunology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China.
| | - Bingdong Zhu
- Gansu Provincial Key Laboratory of Evidence Based Medicine and Clinical Translation and Lanzhou Center for Tuberculosis Research, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China; Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China.
| |
Collapse
|
49
|
Xu H, Cai L, Hufnagel S, Cui Z. Intranasal vaccine: Factors to consider in research and development. Int J Pharm 2021; 609:121180. [PMID: 34637935 DOI: 10.1016/j.ijpharm.2021.121180] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 10/04/2021] [Accepted: 10/07/2021] [Indexed: 01/01/2023]
Abstract
Most existing vaccines for human use are administered by needle-based injection. Administering vaccines needle-free intranasally has numerous advantages over by needle-based injection, but there are only a few intranasal vaccines that are currently approved for human use, and all of them are live attenuated influenza virus vaccines. Clearly, there are immunological as well as non-immunological challenges that prevent vaccine developers from choosing the intranasal route of administration. We reviewed current approved intranasal vaccines and pipelines and described the target of intranasal vaccines, i.e. nose and lymphoid tissues in the nasal cavity. We then analyzed factors unique to intranasal vaccines that need to be considered when researching and developing new intranasal vaccines. We concluded that while the choice of vaccine formulations, mucoadhesives, mucosal and epithelial permeation enhancers, and ligands that target M-cells are important, safe and effective intranasal mucosal vaccine adjuvants are needed to successfully develop an intranasal vaccine that is not based on live-attenuated viruses or bacteria. Moreover, more effective intranasal vaccine application devices that can efficiently target a vaccine to lymphoid tissues in the nasal cavity as well as preclinical animal models that can better predict intranasal vaccine performance in clinical trials are needed to increase the success rate of intranasal vaccines in clinical trials.
Collapse
Affiliation(s)
- Haiyue Xu
- The University of Texas at Austin, College of Pharmacy, Division of Molecular Pharmaceutics and Drug Delivery, Austin, TX, United States
| | - Lucy Cai
- University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Stephanie Hufnagel
- The University of Texas at Austin, College of Pharmacy, Division of Molecular Pharmaceutics and Drug Delivery, Austin, TX, United States
| | - Zhengrong Cui
- The University of Texas at Austin, College of Pharmacy, Division of Molecular Pharmaceutics and Drug Delivery, Austin, TX, United States.
| |
Collapse
|
50
|
Fiege JK, Block KE, Pierson MJ, Nanda H, Shepherd FK, Mickelson CK, Stolley JM, Matchett WE, Wijeyesinghe S, Meyerholz DK, Vezys V, Shen SS, Hamilton SE, Masopust D, Langlois RA. Mice with diverse microbial exposure histories as a model for preclinical vaccine testing. Cell Host Microbe 2021; 29:1815-1827.e6. [PMID: 34731647 DOI: 10.1016/j.chom.2021.10.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 08/30/2021] [Accepted: 10/08/2021] [Indexed: 12/13/2022]
Abstract
Laboratory mice comprise an expeditious model for preclinical vaccine testing; however, vaccine immunogenicity in these models often inadequately translates to humans. Reconstituting physiologic microbial experience to specific pathogen-free (SPF) mice induces durable immunological changes that better recapitulate human immunity. We examined whether mice with diverse microbial experience better model human responses post vaccination. We co-housed laboratory mice with pet-store mice, which have varied microbial exposures, and then assessed immune responses to influenza vaccines. Human transcriptional responses to influenza vaccination are better recapitulated in co-housed mice. Although SPF and co-housed mice were comparably susceptible to acute influenza infection, vaccine-induced humoral responses were dampened in co-housed mice, resulting in poor control upon challenge. Additionally, protective heterosubtypic T cell immunity was compromised in co-housed mice. Because SPF mice exaggerated humoral and T cell protection upon influenza vaccination, reconstituting microbial experience in laboratory mice through co-housing may better inform preclinical vaccine testing.
Collapse
Affiliation(s)
- Jessica K Fiege
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55455, USA; Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Katharine E Block
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA; Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Mark J Pierson
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA; Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Hezkiel Nanda
- Institute for Health Informatics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Frances K Shepherd
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55455, USA; Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Clayton K Mickelson
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55455, USA; Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA
| | - J Michael Stolley
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55455, USA; Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA
| | - William E Matchett
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55455, USA; Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Sathi Wijeyesinghe
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55455, USA; Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA
| | - David K Meyerholz
- Department of Pathology, University of Iowa, Iowa City, IA 52242, USA
| | - Vaiva Vezys
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55455, USA; Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Steven S Shen
- Institute for Health Informatics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Sara E Hamilton
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA; Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA.
| | - David Masopust
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55455, USA; Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Ryan A Langlois
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55455, USA; Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|