1
|
Hu W, Feng H, Liu Y, Xu X, Zhou P, Sun Z, Tao X, Yang J, Wu J, Qu C, Liu Z. Recent advances in immunotherapy targeting CETP proteins for atherosclerosis prevention. Hum Vaccin Immunother 2025; 21:2462466. [PMID: 39907207 PMCID: PMC11801355 DOI: 10.1080/21645515.2025.2462466] [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: 11/09/2024] [Revised: 01/22/2025] [Accepted: 01/31/2025] [Indexed: 02/06/2025] Open
Abstract
Cholesteryl ester transfer protein (CETP) plays a key role in lipoprotein metabolism, and its activity has been linked to the risk of atherosclerosis (AS). CETP inhibitors, such as obicetrapib, represent a novel approach in immunotherapy to reduce the risk of atherosclerotic cardiovascular disease (ASCVD) by targeting lipid metabolism. In addition, CETP vaccines are being explored as a novel strategy for the prevention and treatment of ASCVD by inducing the body to produce antibodies against CETP, which is expected to reduce CETP activity, thereby increasing high-density lipoproteins (HDL) levels. This paper provides a comprehensive overview of the structure of CETP, the mechanisms of lipid transfer and the progress of immunotherapy in the last decade, which provides possible ideas for future development of novel drugs and optimization of immunization strategies.
Collapse
Affiliation(s)
- Wenhui Hu
- Department of Geriatrics, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, P.R. China
- Key Laboratory for Aging & Disease, Nanjing Medical University, Nanjing, Jiangsu, P.R. China
| | - Han Feng
- Department of Geriatrics, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, P.R. China
- Key Laboratory for Aging & Disease, Nanjing Medical University, Nanjing, Jiangsu, P.R. China
| | - Ying Liu
- Department of Geriatrics, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, P.R. China
- Key Laboratory for Aging & Disease, Nanjing Medical University, Nanjing, Jiangsu, P.R. China
| | - Xiaoshuang Xu
- Key Laboratory for Aging & Disease, Nanjing Medical University, Nanjing, Jiangsu, P.R. China
| | - Ping Zhou
- Department of Geriatrics, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, P.R. China
- Key Laboratory for Aging & Disease, Nanjing Medical University, Nanjing, Jiangsu, P.R. China
| | - Zhonghua Sun
- Department of Geriatrics, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, P.R. China
- Key Laboratory for Aging & Disease, Nanjing Medical University, Nanjing, Jiangsu, P.R. China
| | - Xinyu Tao
- Department of Geriatrics, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, P.R. China
- Key Laboratory for Aging & Disease, Nanjing Medical University, Nanjing, Jiangsu, P.R. China
| | - Jiahui Yang
- Department of Geriatrics, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, P.R. China
- Key Laboratory for Aging & Disease, Nanjing Medical University, Nanjing, Jiangsu, P.R. China
| | - Jun Wu
- Department of Geriatric Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Chen Qu
- Department of Geriatrics, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, P.R. China
- Key Laboratory for Aging & Disease, Nanjing Medical University, Nanjing, Jiangsu, P.R. China
| | - Zhengxia Liu
- Department of Geriatrics, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, P.R. China
- Key Laboratory for Aging & Disease, Nanjing Medical University, Nanjing, Jiangsu, P.R. China
| |
Collapse
|
2
|
Sipos B, Rajab F, Katona G, Csóka I. Current insights into polymeric micelles for nasal drug delivery. Expert Opin Drug Deliv 2025:1-18. [PMID: 40420578 DOI: 10.1080/17425247.2025.2511962] [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: 02/28/2025] [Revised: 05/09/2025] [Accepted: 05/23/2025] [Indexed: 05/28/2025]
Abstract
INTRODUCTION The nasal administration route has gained peak interest in recent literature and as a noninvasive alternative for efficient drug delivery and increasing bioavailability of active substances. Technological challenges arise from the drug's physicochemical properties and the nasal mucosal barrier for which innovative particle engineering techniques must be implemented, such as using polymeric nanocarriers. AREAS COVERED This review deals with the importance of the nasal administration route and its connection to polymeric micelles as innovative nanocarriers. The period between 2015-2025 up to date was chosen to search for original research articles where polymeric micelles were applied nasally. The first part demonstrates the utilization of polymeric micelles, followed by a summary of how drug release and permeability can be achieved in the nasal cavity and through the nasal epithelium. The second part reviews the studies conducted on this matter. EXPERT OPINION The nasal route could be superior to perform as a suitable alternative to conventional routes. Multiple studies have already demonstrated that the main advantages lie in the nose-to-brain drug delivery pathway, which can be conquered via adequately formulated polymeric micelles. As an innovative solution, vaccine delivery is also of great potential by combining the advantages of the delivery route and the polymeric nanocarriers.
Collapse
Affiliation(s)
- Bence Sipos
- Institute of Pharmaceutical Technology and Regulatory Affairs, University of Szeged, Szeged, Hungary
| | - Fatima Rajab
- Institute of Pharmaceutical Technology and Regulatory Affairs, University of Szeged, Szeged, Hungary
| | - Gábor Katona
- Institute of Pharmaceutical Technology and Regulatory Affairs, University of Szeged, Szeged, Hungary
| | - Ildikó Csóka
- Institute of Pharmaceutical Technology and Regulatory Affairs, University of Szeged, Szeged, Hungary
| |
Collapse
|
3
|
Zhang Z, Yang Y, Huang L, Yuan L, Huang S, Zeng Z, Cao Y, Wei X, Wang X, Shi M, Zhong M. Nanotechnology-driven advances in intranasal vaccine delivery systems against infectious diseases. Front Immunol 2025; 16:1573037. [PMID: 40416956 PMCID: PMC12098542 DOI: 10.3389/fimmu.2025.1573037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Accepted: 04/11/2025] [Indexed: 05/27/2025] Open
Abstract
Outbreaks of emerging and re-emerging infectious diseases have consistently threatened human health. Since vaccinations are a powerful tool for preventing infectious illnesses, developing new vaccines is essential. Compared to traditional injectable vaccines, mucosal vaccines have the potential to offer more effective immune protection at mucosal sites. Mucosal immunization strategies include sublingual, oral, intranasal, genital, and rectal routes, in which intranasal immunization being the most efficient and applicable method for mucosal vaccine delivery. Nevertheless, low antigen availability and weak immunogenicity making it challenging to elicit a potent immune response when administered intranasally, necessitating the incorporation of immune delivery systems. However, there is a notable absence of reviews that summarize the intranasal vaccine delivery system against infectious disease. Therefore, this review summarizes the recent advances in intranasal delivery systems, classified by physical and chemical properties, and proposes potential improvement strategies for clinical translation. This review elucidates the potential and current status of intranasal delivery systems, while also serving as a reference point for the future development of intranasal vaccines.
Collapse
Affiliation(s)
- Zhihan Zhang
- Institute of Infection, Immunology and Tumor Microenvironment, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Medical College, Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Yumeng Yang
- Institute of Infection, Immunology and Tumor Microenvironment, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Medical College, Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Liwen Huang
- Institute of Infection, Immunology and Tumor Microenvironment, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Medical College, Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Lei Yuan
- Institute of Infection, Immunology and Tumor Microenvironment, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Medical College, Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Sijian Huang
- Institute of Infection, Immunology and Tumor Microenvironment, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Medical College, Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Zihang Zeng
- Institute of Infection, Immunology and Tumor Microenvironment, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Medical College, Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Yuan Cao
- Institute of Infection, Immunology and Tumor Microenvironment, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Medical College, Wuhan University of Science and Technology, Wuhan, Hubei, China
- Analytical & Testing Center, Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Xianghong Wei
- Institute of Infection, Immunology and Tumor Microenvironment, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Medical College, Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Xiaomei Wang
- Institute of Infection, Immunology and Tumor Microenvironment, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Medical College, Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Mingsong Shi
- NHC Key Laboratory of Nuclear Technology Medical Transformation, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, Sichuan, China
| | - Maohua Zhong
- Institute of Infection, Immunology and Tumor Microenvironment, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Medical College, Wuhan University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
4
|
Raha JR, Kim KH, Le CTT, Bhatnagar N, Pal SS, Liu R, Grovenstein P, Yeasmin M, Racheal F, Shin CH, Wang BZ, Kang SM. Intranasal vaccination with multi-neuraminidase and M2e virus-like particle vaccine results in greater mucosal immunity and protection against influenza than intramuscular injection. Vaccine 2025; 57:127206. [PMID: 40339180 DOI: 10.1016/j.vaccine.2025.127206] [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: 01/19/2025] [Revised: 04/27/2025] [Accepted: 04/29/2025] [Indexed: 05/10/2025]
Abstract
Intramuscular injection of seasonal influenza vaccines provides strain-specific neutralizing antibodies, but not against variants, and no effective mucosal immunity. Here, we report that multi-subtype neuraminidase (NA) and M2 ectodomain repeat (5xM2e) virus-like particle vaccine (NA-M2e) conferred higher efficacy of broad cross-protection after two doses of intranasal delivery than intramuscular injection. The intranasally vaccinated mice displayed high levels of IgA antibodies, IFN-γ+ CD4 and CD8 T cells, germinal center B cells, plasma cells, and early innate immune cells locally in the lungs. In contrast, intramuscular vaccination systemically induced innate and adaptive immune responses in the spleen. Our findings demonstrate that the intranasal delivery of NA-M2e vaccine induces enhanced mucosal immunity and comparable serum IgG antibodies, offering improved efficacy of cross-protection against diverse influenza virus strains compared to the intramuscular injection in a mouse model.
Collapse
Affiliation(s)
- Jannatul Ruhan Raha
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30302, USA
| | - Ki-Hye Kim
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30302, USA
| | - Chau Thuy Tien Le
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30302, USA
| | - Noopur Bhatnagar
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30302, USA
| | - Surya Sekhar Pal
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30302, USA
| | - Rong Liu
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30302, USA
| | - Phillip Grovenstein
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30302, USA
| | - Mahmuda Yeasmin
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30302, USA
| | - Farayola Racheal
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30302, USA
| | - Chong Hyun Shin
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30302, USA
| | - Bao-Zhong Wang
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30302, USA
| | - Sang-Moo Kang
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30302, USA.
| |
Collapse
|
5
|
Song JH, Son SE, Kim HW, Kim SJ, An SH, Lee CY, Kwon HJ, Choi KS. Intranasally administered whole virion inactivated vaccine against clade 2.3.4.4b H5N1 influenza virus with optimized antigen and increased cross-protection. Virol J 2025; 22:131. [PMID: 40320528 PMCID: PMC12051338 DOI: 10.1186/s12985-025-02760-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Accepted: 04/23/2025] [Indexed: 05/08/2025] Open
Abstract
The global spread, frequent antigenic changes, and pandemic potential of clade 2.3.4.4b highly pathogenic avian influenza H5N1 underscore the urgent need for robust cross-protective vaccines. Here, we developed a clade 2.3.4.4b H5N1 whole inactivated virus (WIV) vaccine strain with improved structural stability, productivity, and safety. By analyzing the evolutionary trends of clade 2.3.4.4b H5N1 viruses, we identified a key mutation (R90K) that increases heat stability while preserving antigenicity. Additionally, the PB2 gene of PR8 was replaced with a prototypical avian PB2 gene to increase replication efficiency in embryonated chicken eggs and reduce replication efficiency in mammalian cells, thereby improving productivity and biosafety. We found that our optimized clade 2.3.4.4b H5N1 vaccine strain (22W_KY), inactivated with binary ethylenimine (BEI), had superior antigen internalization into respiratory epithelial cells compared to those inactivated with formaldehyde or beta-propiolactone. Following intranasal administration to mice, the BEI-inactivated 22W_KY also elicited significantly stronger systemic IgG, mucosal IgA, and T-cell responses, especially in the lungs. Protective efficacy studies revealed that the BEI-inactivated 22W_KY vaccine provided complete protection against heterologous viral challenges and significant protection against heterosubtypic viral challenges, with no weight loss and complete suppression of the viral load in the respiratory tract in 2 of 3 mice. These results indicate that the BEI-inactivated 22W_KY vaccine could serve as a promising candidate for a safe, stable, cost-efficient, and broadly protective intranasal influenza vaccine against zoonotic and pandemic threats.
Collapse
Affiliation(s)
- Jin-Ha Song
- Laboratory of Avian Diseases, College of Veterinary Medicine, Seoul National University, 1, Gwanak-ro, Seoul, 88026, Republic of Korea
| | - Seung-Eun Son
- Laboratory of Avian Diseases, College of Veterinary Medicine, Seoul National University, 1, Gwanak-ro, Seoul, 88026, Republic of Korea
| | - Ho-Won Kim
- Laboratory of Avian Diseases, College of Veterinary Medicine, Seoul National University, 1, Gwanak-ro, Seoul, 88026, Republic of Korea
| | - Seung-Ji Kim
- Laboratory of Avian Diseases, College of Veterinary Medicine, Seoul National University, 1, Gwanak-ro, Seoul, 88026, Republic of Korea
| | - Se-Hee An
- Avian Influenza Research & Diagnostic Division, Animal and Plant Quarantine Agency, Gimcheon-si, Republic of Korea
| | - Chung-Young Lee
- Department of Microbiology, College of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Hyuk-Joon Kwon
- Laboratory of Poultry Medicine, Department of Farm Animal Medicine, College of Veterinary Medicine, BK21 PLUS for Veterinary Science, Seoul National University, 1, Gwanak-ro, Seoul, 88026, Republic of Korea.
- GeNiner Inc, Seoul, Republic of Korea.
| | - Kang-Seuk Choi
- Laboratory of Avian Diseases, College of Veterinary Medicine, Seoul National University, 1, Gwanak-ro, Seoul, 88026, Republic of Korea.
- Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
6
|
Chávez-Valdés S, Marichal-Rodríguez AK, Chacón-Quintero Y, Martínez-Rosales R, Gómez-Hernández N, Ávila-Díaz L, Vázquez-Arteaga A, González-Formental H, Freyre-Corrales G, Coizeau-Rodríguez E, Guillen G, Lemos-Pérez G. Validation and clinical performance of a non-commercial ELISA for SARS-CoV-2 anti-RBD IgA antibodies. Anal Biochem 2025; 700:115787. [PMID: 39894142 DOI: 10.1016/j.ab.2025.115787] [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: 11/11/2024] [Revised: 01/07/2025] [Accepted: 01/24/2025] [Indexed: 02/04/2025]
Abstract
COVID-19 is caused by SARS-CoV-2, first identified in 2019. The Cuban vaccines, Abdala and Mambisa, have demonstrated efficacy in preventing SARS-CoV-2 infection. Immunoglobulin A (IgA) are the main line of defense against pathogens invading the respiratory or digestive tract and its presence in serum can be measured to assess vaccine efficacy. ELISAs are a valuable tool for assessing vaccine immunogenicity. These tests should be validated to ensure their reliability and suitability. The objective of this study was to validate a non-commercial ELISA for the quantification of total anti-RBD IgA in serum samples to support clinical studies. This assay demonstrated high clinical specificity (97.3 %). The accuracy and precision of the assay showed an overall error of less than 20 % at all levels in QCs. Re-evaluation of samples showed a mean difference of less than 30 % in 90.2 % of cases. Anti-RBD IgA titers correlated with viral neutralization titers and percentage inhibition of RBD-ACE2 binding. This assay was found to be highly accurate and reproducible for the quantification of anti-RBD IgA, met the most stringent acceptance criteria and is fit for purpose. It is currently being used to evaluate the immunogenicity of the Abdala and Mambisa vaccines.
Collapse
Affiliation(s)
- Sheila Chávez-Valdés
- Center for Genetic Engineering and Biotechnology, CIGB, Ave. 31 E/ 158 y 190, P.O. Box. 6162, La Habana, 10600, Cuba
| | - Ana K Marichal-Rodríguez
- Center for Genetic Engineering and Biotechnology, CIGB, Ave. 31 E/ 158 y 190, P.O. Box. 6162, La Habana, 10600, Cuba
| | - Yahima Chacón-Quintero
- Center for Genetic Engineering and Biotechnology, CIGB, Ave. 31 E/ 158 y 190, P.O. Box. 6162, La Habana, 10600, Cuba
| | - Ricardo Martínez-Rosales
- Center for Genetic Engineering and Biotechnology, CIGB, Ave. 31 E/ 158 y 190, P.O. Box. 6162, La Habana, 10600, Cuba
| | - Nivaldo Gómez-Hernández
- Center for Genetic Engineering and Biotechnology, CIGB, Ave. 31 E/ 158 y 190, P.O. Box. 6162, La Habana, 10600, Cuba
| | - Lismary Ávila-Díaz
- Center for Genetic Engineering and Biotechnology, CIGB, Ave. 31 E/ 158 y 190, P.O. Box. 6162, La Habana, 10600, Cuba
| | - Amalia Vázquez-Arteaga
- Center for Genetic Engineering and Biotechnology, CIGB, Ave. 31 E/ 158 y 190, P.O. Box. 6162, La Habana, 10600, Cuba
| | - Hany González-Formental
- Center for Genetic Engineering and Biotechnology, CIGB, Ave. 31 E/ 158 y 190, P.O. Box. 6162, La Habana, 10600, Cuba
| | - Giselle Freyre-Corrales
- Center for Genetic Engineering and Biotechnology, CIGB, Ave. 31 E/ 158 y 190, P.O. Box. 6162, La Habana, 10600, Cuba
| | - Edelgis Coizeau-Rodríguez
- Center for Genetic Engineering and Biotechnology, CIGB, Ave. 31 E/ 158 y 190, P.O. Box. 6162, La Habana, 10600, Cuba
| | - Gerardo Guillen
- Center for Genetic Engineering and Biotechnology, CIGB, Ave. 31 E/ 158 y 190, P.O. Box. 6162, La Habana, 10600, Cuba; Latin American School of Medicine (ELAM), Carretera Panamericana Km 3 1/2, Carr. Panamericana, La Habana, 19108, Cuba
| | - Gilda Lemos-Pérez
- Center for Genetic Engineering and Biotechnology, CIGB, Ave. 31 E/ 158 y 190, P.O. Box. 6162, La Habana, 10600, Cuba.
| |
Collapse
|
7
|
Chanda A, Song Y, Nazir J, Lin C, Cheng A, Sargent J, Sikora AE. Bridging Gaps in Antibody Responses and Animal Welfare: Assessing Blood Collection Methods and Vaginal Immunity in Mice Immunized with Intranasal Gonococcal Vaccines. RESEARCH SQUARE 2025:rs.3.rs-6241509. [PMID: 40313749 PMCID: PMC12045373 DOI: 10.21203/rs.3.rs-6241509/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2025]
Abstract
Assessing antibody titers and functional responses is essential for evaluating vaccine efficacy, yet the impact of blood collection methods on these immunological assessments remains unclear. Retro-orbital (RO) blood collection is commonly used but significant complications can occur. Increasingly, investigators have adopted alternative blood collection approaches, such as saphenous vein (SV) sampling to improve laboratory animal welfare. This study compared RO and SV sampling in the development of a Neisseria gonorrhoeae (Ng) vaccine, evaluating Adhesin Complex Protein (ACP) and multiple transferable resistance (Mtr) E protein (MtrE) as antigen candidates. Epitope mapping revealed that ACP and MtrE possess multiple, highly accessible B-cell and T-cell epitope clusters, reinforcing their immunological potential. Following intranasal immunization with rACP, rACP+CpG, and rMtrE+CpG, we assessed the specificity, magnitude, kinetics, and functional quality of immune responses elicited by the immunization regimens. Out of 45 comparisons, only eight significant differences were detected in antibody titers, while the human serum bactericidal assays revealed no differences between RO and SV in antigen-immunized groups. However, antibodies elicited by rACP alone or ACP+CpG in SV samples restored 30.05% and 75.2% of human lysozyme hydrolytic activity compared to 19.3 and 59.9 % in RO, respectively suggesting that SV sampling may be more reliable for assessing functional antibody responses. Beyond its immunological advantages, SV sampling reduces stress, minimizes ocular trauma, and improves animal welfare, making it a viable alternative to RO collection. Given its widespread use in vaccine research, standardizing SV sampling could improve data reliability, ethical compliance, and translational relevance in preclinical studies.
Collapse
Affiliation(s)
- Abhishek Chanda
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, Oregon 97330, United States
| | - Yujuan Song
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, Oregon 97330, United States
| | - Junaid Nazir
- Department of Clinical Biochemistry, Lovely Professional University, Phagwara Punjab, India
| | - Chenwei Lin
- Proteomics Shared Resource, Fred Hutchinson Cancer Research Center, Seattle, United States
| | - Alicia Cheng
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, Oregon 97330, United States
| | - Jennifer Sargent
- Carlson College of Veterinary Medicine, Oregon State University, Corvallis, Oregon 97330, United States
| | - Aleksandra E. Sikora
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, Oregon 97330, United States
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, United States
| |
Collapse
|
8
|
Liu Y, Chen Z, Cheng H, Zheng R, Huang W. Mucosal immunotherapy targeting APC in lung disease. J Inflamm (Lond) 2025; 22:15. [PMID: 40229816 PMCID: PMC11998460 DOI: 10.1186/s12950-025-00432-2] [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: 08/24/2024] [Accepted: 01/28/2025] [Indexed: 04/16/2025] Open
Abstract
Several studies have demonstrated that the pulmonary immune response is primarily facilitated by antigen-presenting cells (APCs), and that both professional and non-professional APCs contribute to overall pulmonary immunity. APCs play unique roles and mechanisms in pathogen elimination and immunomodulation. Mucosal immunity exhibits potential advantages over traditional parenteral immunity in that it stimulates immune defenses in mucosal and systemic tissues, which is important for reducing the burden of lung disease. However, obtaining a comprehensive understanding of the crosstalk between mucosal immunity and APC in the context of various lung diseases remains challenging. This mini-review aimed to elucidate the mechanisms of novel mucosal immunity, targeting APC action during lung infections, allergies, and malignant tumorigenesis. This minreview provides important insights into more effective therapeutic approaches for various lung diseases.
Collapse
Affiliation(s)
- Yangqi Liu
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Zijian Chen
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Hanchang Cheng
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Runzhi Zheng
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Weizhe Huang
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, China.
| |
Collapse
|
9
|
Jüptner A, Scherließ R. Investigation of powder properties and application aspects impacting nasal deposition of spray-dried powders in a nasal cast. Eur J Pharm Biopharm 2025; 209:114666. [PMID: 39954967 DOI: 10.1016/j.ejpb.2025.114666] [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: 10/26/2024] [Revised: 12/30/2024] [Accepted: 02/12/2025] [Indexed: 02/17/2025]
Abstract
In this study, spray-dried formulations differing in morphology (spherical and wrinkled), surface polarity (hydrophilic and hydrophobic), and size (20-30 µm and 3 µm) were evaluated in a nasal cast to assess their deposition profiles. The objective was to identify how formulation properties and application aspects influence the deposition profile. For this purpose, the formulations were administered at different application angles (45° and 60°), fill weights (20 mg and 40 mg), and airflow rates (0 L/min and 15 L/min) in conjunction with a UDS powder device. The results indicate a more posterior deposition profile for 45° compared to 60° due to increased deposition in the turbinate region; conversely, deposition profiles between fill weights were comparable. Application with simultaneous airflow should be avoided because of an increasing postnasal fraction. No influence of morphology could be observed, but for the surface polarity an influence was apparent, if the powder was applied with a simulated inspiration. In these cases, a hydrophobic formulation was better dispersible than a hydrophilic formulation, which led to an increased postnasal fraction. A particle size for pulmonary application demonstrated comparable results to nasal formulations with respect to the turbinate deposition but exhibited a high postnasal fraction for hydrophobic formulations.
Collapse
Affiliation(s)
- Angelika Jüptner
- Department of Pharmaceutics and Biopharmaceutics, Kiel University, 24118 Kiel, Germany; Priority Research Area Kiel Nano, Surface and Interface Sciences (KiNSIS), Kiel University, Kiel, Germany
| | - Regina Scherließ
- Department of Pharmaceutics and Biopharmaceutics, Kiel University, 24118 Kiel, Germany; Priority Research Area Kiel Nano, Surface and Interface Sciences (KiNSIS), Kiel University, Kiel, Germany.
| |
Collapse
|
10
|
Davis MM, Bajrovic I, Croyle MA. Assessment of In Vitro Models of the Human Buccal Mucosa for Vaccine and Adjuvant Development. Mol Pharm 2025. [PMID: 40139941 DOI: 10.1021/acs.molpharmaceut.4c01186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
To understand requirements for immunization via the oral mucosa, an in vitro model that recapitulates the physical barrier of the mouth, allows for quantification of antigen uptake and permeability and mounts an inflammatory response to antigen and adjuvant is needed. The physical structure of 4 models of the human oral mucosa was determined by histochemical staining and transepithelial electrical resistance (TEER) measurements. A TR146 based air-liquid interface (ALI) model most closely mimicked in vivo conditions. This was confirmed by validation studies using dextran and caffeine as diffusant molecules. Apparent permeability coefficients (Papp) of adenovirus (Ad) and adeno-associated virus (AAV) in this model were 4.3 × 10-13 and 2.2 × 10-10 respectively, while 100% of the total dose of H1N1 influenza remained in the epithelial layer. Sodium glycocholate and a hyperosmotic formulation improved the amount of Ad (p = 0.02) and AAV (p = 0.003) that entered the epithelium, respectively. Significant amounts of IL-6 (45.1 pg/mL), GM-CSF (94.7 pg/mL) and IFN-γ (4.3 pg/mL) were produced in response to influenza infection. Treatment with an AS03-like adjuvant induced production of IL-6 (34.9 pg/mL), TNF-∝ (43 pg/mL), GM-CSF (121.2 pg/mL) and IFN-γ (14.1 pg/mL). This highlights the contribution of differentiated epithelial cells to the immune response to vaccines and adjuvants.
Collapse
Affiliation(s)
- Madison M Davis
- College of Pharmacy, Division of Molecular Pharmaceutics and Drug Delivery, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Irnela Bajrovic
- College of Pharmacy, Division of Molecular Pharmaceutics and Drug Delivery, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Maria A Croyle
- College of Pharmacy, Division of Molecular Pharmaceutics and Drug Delivery, The University of Texas at Austin, Austin, Texas 78712, United States
- John R. LaMontagne Center for Infectious Disease, The University of Texas at Austin, Austin, Texas 78712, United States
| |
Collapse
|
11
|
Li HY, Paramanandana A, Kim SY, Granger L, Raimi-Abraham BT, Shattock R, Makatsoris C, Forbes B. Targeted nasal delivery of LNP-mRNAs aerosolised by Rayleigh breakup technology. Int J Pharm 2025; 672:125335. [PMID: 39933606 DOI: 10.1016/j.ijpharm.2025.125335] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2025] [Revised: 01/31/2025] [Accepted: 02/08/2025] [Indexed: 02/13/2025]
Abstract
The nasal delivery of mRNA vaccines attracts great interests in both academia and industry. While the lipid nanoparticle (LNP)-mRNA complexes are vulnerable and need a subtle process for aerosolisation. In this study, a new nasal atomizer, based on the working rationale of Rayleigh breakup, was employed to aerosolise LNP-mRNAs. The data revealed no statistical differences in physiochemical properties before and after aerosolisation, strongly suggesting LNP-mRNAs be well preserved upon aerosolisation by Rayleigh breakup technology. Additionally, these Rayleigh breakup droplets showed a physical size of ∼25 µm in mean with a narrow size distribution (Span: 1.24) and demonstrated a large portion (70-80 % w/w) greater than 10 µm in aerodynamic diameter, strongly suggesting a predominate deposition in the upper airway and designating a great appropriateness for nasal drug delivery. Furthermore, the recently developed Alberta Idealized Nasal Inlet (AINI) was utilized to evaluate the regional nasal deposition of LNP-mRNA aerosols. It was demonstrated that, at the administration angle of 45°, the major deposition of mRNAs (>50 % w/w) was in the target region of turbinates. The inhalation airflow at 7.5 L/min can maximize the targeted delivery of mRNA (∼64 % w/w) and minimize the undesirable depositions in other segments. This study provides a new approach to aerosolise LNP-mRNAs with undisturbed stabilities for targeted nasal vaccine delivery.
Collapse
Affiliation(s)
- Hao-Ying Li
- Institute of Pharmaceutical Sciences, King's College London, London SE1 9NH, United Kingdom.
| | - Abhimata Paramanandana
- Institute of Pharmaceutical Sciences, King's College London, London SE1 9NH, United Kingdom
| | - Sally Yunsun Kim
- Institute of Pharmaceutical Sciences, King's College London, London SE1 9NH, United Kingdom
| | - Luke Granger
- Department of Infectious Diseases, Section of Immunology of Infection, Imperial College London, London W2 1PG, United Kingdom
| | | | - Robin Shattock
- Department of Infectious Diseases, Section of Immunology of Infection, Imperial College London, London W2 1PG, United Kingdom
| | - Charalampos Makatsoris
- Department of Engineering, Faculty of Natural/Mathematical Sciences, King's College London, London WC2R 2LS, United Kingdom
| | - Ben Forbes
- Institute of Pharmaceutical Sciences, King's College London, London SE1 9NH, United Kingdom.
| |
Collapse
|
12
|
Chatzidaki MD, Mitsou E. Advancements in Nanoemulsion-Based Drug Delivery Across Different Administration Routes. Pharmaceutics 2025; 17:337. [PMID: 40143001 PMCID: PMC11945362 DOI: 10.3390/pharmaceutics17030337] [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: 01/02/2025] [Revised: 02/17/2025] [Accepted: 02/24/2025] [Indexed: 03/28/2025] Open
Abstract
Nanoemulsions (NEs) have emerged as effective drug delivery systems over the past few decades due to their multifaceted nature, offering advantages such as enhanced bioavailability, protection of encapsulated compounds, and low toxicity. In the present review, we focus on advancements in drug delivery over the last five years across (trans)dermal, oral, ocular, nasal, and intra-articular administration routes using NEs. Rational selection of components, surface functionalization, incorporation of permeation enhancers, and functionalization with targeting moieties are explored for each route discussed. Additionally, apart from NEs, we explore NE-based drug delivery systems (e.g., NE-based gels) while highlighting emerging approaches such as vaccination and theranostic applications. The growing interest in NEs for drug delivery purposes is reflected in clinical trials, which are also discussed. By summarizing the latest advances, exploring new strategies, and identifying critical challenges, this review focuses on developments for efficient NE-based therapeutic approaches.
Collapse
Affiliation(s)
- Maria D. Chatzidaki
- Institute of Chemical Biology, National Hellenic Research Foundation, 11635 Athens, Greece
- Institute for Bio-Innovation, Biomedical Sciences Research Center “Alexander Fleming”, 16672 Vari, Greece
| | - Evgenia Mitsou
- Institute of Chemical Biology, National Hellenic Research Foundation, 11635 Athens, Greece
- Department of Molecular Chemistry and Materials Science, Weizmann Institute of Science, 6100 Rehovot, Israel
| |
Collapse
|
13
|
Hejazi M, Alshammary AM, Edwards DJ, Golshahi L. Development of a predictive model for pediatric intranasal drug delivery with nasal sprays: Leveraging intersubject variability in anatomical dimensions, administration-related parameters, and airway patency. Comput Biol Med 2025; 187:109746. [PMID: 39879885 DOI: 10.1016/j.compbiomed.2025.109746] [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: 09/12/2024] [Revised: 12/20/2024] [Accepted: 01/22/2025] [Indexed: 01/31/2025]
Abstract
In this study, we examined the correlation between anatomical dimensions, spray administration parameters, pressure drop across 40 pediatric nasal cavities, and in vitro posterior drug delivery (PDD) using Nasacort ALLERGY 24HR and FLONASE SENSIMIST nasal suspensions sprays, with different nozzle and actuation designs. The importance of each parameter and their interaction in the outcome (PDD) was evaluated. To do so, initially we measured anatomical and administration-related parameters, and the pressure drop of each cavity. Afterwards, a stepwise regression method was used to find a combination of parameters that can effectively predict the outcome. Two strong correlations, r2 = 0.802 and 0.895, were found between a combination of three explored parameters and the PDD of Nasacort and Flonase Sensimist, respectively. While, based on the p-values, the insertion depth was found to be the most influential parameter for determining the PDD with Nasacort, and the administration coronal angle was found to be the most important one for Flonase Sensimist, anatomical parameters, or an interaction between anatomical and administration parameters were among the five most important parameters for both sprays. Furthermore, variable importance assessment, based on the variance of the response, demonstrated the tip-to-INV distance as the most important parameter for Nasacort and coronal angle for Flonase Sensimist, while cross sectional area at the end of the anterior piece was the only parameter that found to be in the three most important parameters for both sprays. Having this in mind, as well as the wide range of PDD within the pediatric population, it can be concluded that the performance of targeted intranasal drug delivery, even when using a set of patient-specific administration parameters, is strongly influenced by the anatomical features of the subject. Additionally, comparing the key factors in predicting PDD in pediatric and adult population for Flonase Sensimist demonstrate no similarity between the influential parameters on PDD in the two age groups, which further emphasizes the importance of airway anatomy.
Collapse
Affiliation(s)
- Mohammad Hejazi
- Department of Mechanical and Nuclear Engineering, Virginia Commonwealth University, Richmond, VA, USA
| | - Ahmed M Alshammary
- Department of Mechanical and Nuclear Engineering, Virginia Commonwealth University, Richmond, VA, USA
| | - David J Edwards
- Department of Statistical Sciences and Operations Research, Virginia Commonwealth University, Richmond, VA, USA
| | - Laleh Golshahi
- Department of Mechanical and Nuclear Engineering, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
14
|
Park J, Pho T, Bhatnagar N, Mai LD, Rodriguez-Otero MR, Pal SS, Le CTT, Jenison SE, Li C, May GA, Arioka M, Kang SM, Champion JA. Multilayer Adjuvanted Influenza Protein Nanoparticles Improve Intranasal Delivery and Antigen-Specific Immunity. ACS NANO 2025; 19:7005-7025. [PMID: 39954231 PMCID: PMC11867023 DOI: 10.1021/acsnano.4c14735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 02/06/2025] [Accepted: 02/07/2025] [Indexed: 02/17/2025]
Abstract
Intranasal vaccination is a desired route for protection against influenza viruses by mucosal and systemic immunity. However, the nasal mucosa impedes the intranasal delivery of vaccines. Here, we formulated layer-by-layer (LBL) influenza vaccine nanoparticles for effective intranasal delivery by coating them with alternating mucoadhesive cationic chitosan and muco-inert anionic CpG adjuvants. The nanoparticle cores were formed by desolvating influenza M2e antigen and coating it with hemagglutinin (HA) antigen via biotin-streptavidin conjugation. LBL modification promoted nasal delivery and interaction with the resident immune cells. Intranasal administration with LBL nanoparticles significantly improved cellular and humoral immune responses against HA and M2e including high IgA titers, a hallmark of potent mucosal immunity and persistence of immune responses. Distinct trends for antigen-specific immune responses were observed for different routes of vaccination. The enhanced immune responses conferred mice protection against the influenza challenge and prominently reduced viral titers, demonstrating the effectiveness of intranasal LBL vaccine nanoparticles.
Collapse
Affiliation(s)
- Jaeyoung Park
- School of
Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - Thomas Pho
- School of
Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
- Bioengineering
Program, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - Noopur Bhatnagar
- Center
for
Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia 30302, United States
| | - Linh D. Mai
- School of
Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - Mariela R. Rodriguez-Otero
- School of
Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
- Bioengineering
Program, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - Surya Sekhar Pal
- Center
for
Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia 30302, United States
| | - Chau Thuy Tien Le
- Center
for
Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia 30302, United States
| | - Sarah E. Jenison
- School of
Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - Chenyu Li
- School of
Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - Grace A. May
- School of
Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - Marisa Arioka
- Department
of Chemistry, Tokyo University of Science, Shinjuku-ku, Tokyo 162-8601, Japan
| | - Sang-Moo Kang
- Center
for
Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia 30302, United States
| | - Julie A. Champion
- School of
Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
- Bioengineering
Program, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| |
Collapse
|
15
|
Chanda A, Song Y, Nazir J, Lin C, Cheng A, Sargent J, Sikora AE. Bridging Gaps in Antibody Responses and Animal Welfare: Assessing Blood Collection Methods and Vaginal Immunity in Mice Immunized with Intranasal Gonococcal Vaccines. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.23.639724. [PMID: 40027683 PMCID: PMC11870632 DOI: 10.1101/2025.02.23.639724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Assessing antibody titers and functional responses is essential for evaluating vaccine efficacy, yet the impact of blood collection methods on these immunological assessments remains unclear. Retro-orbital (RO) blood collection is commonly used but significant complications can occur. Increasingly, investigators have adopted alternative blood collection approaches, such as saphenous vein (SV) sampling to improve laboratory animal welfare. This study compared RO and SV sampling in the development of a Neisseria gonorrhoeae (Ng) vaccine, evaluating Adhesin Complex Protein (ACP) and multiple transferable resistance (Mtr) E protein (MtrE) as antigen candidates. Epitope mapping revealed that ACP and MtrE possess multiple, highly accessible B-cell and T-cell epitope clusters, reinforcing their immunological potential. Following intranasal immunization with rACP, rACP+CpG, and rMtrE+CpG, we assessed the specificity, magnitude, kinetics, and functional quality of immune responses elicited by the immunization regimens. Out of 45 comparisons, only eight significant differences were detected in antibody titers, while the human serum bactericidal assays revealed no differences between RO and SV in antigen-immunized groups. However, antibodies elicited by rACP alone or ACP+CpG in SV samples restored 30.05% and 75.2% of human lysozyme hydrolytic activity compared to 19.3 and 59.9 % in RO, respectively suggesting that SV sampling may be more reliable for assessing functional antibody responses. Beyond its immunological advantages, SV sampling reduces stress, minimizes ocular trauma, and improves animal welfare, making it a viable alternative to RO collection. Given its widespread use in vaccine research, standardizing SV sampling could improve data reliability, ethical compliance, and translational relevance in preclinical studies.
Collapse
|
16
|
Angelidou A, Koster JA, Sherman AC, McLoughlin C, Lalwani P, Kelly A, Saeed A, McEnaney K, Baden LR, Brogna M, Weitzman ER, Levy S, Dowling DJ, Levy O. Product and trial design considerations on the path towards a vaccine to combat opioid overdose. NPJ Vaccines 2025; 10:35. [PMID: 39971929 PMCID: PMC11840009 DOI: 10.1038/s41541-025-01083-3] [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: 07/18/2024] [Accepted: 01/23/2025] [Indexed: 02/21/2025] Open
Abstract
Opioid overdose deaths are an evolving public health emergency in the United States. Recent advancements in drug conjugate vaccine design and adjuvantation technologies have re-ignited interest in the potential clinical utility of opioid vaccination. Here we present the concept of fentanyl vaccination as a complementary strategy for opioid overdose prevention with a focus on vaccine safety, efficacy, and considerations for vaccine development and testing in early phase human clinical trials.
Collapse
Affiliation(s)
- Asimenia Angelidou
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
- Department of Neonatology, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Jacob A Koster
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
| | - Amy C Sherman
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA, USA
| | - Caitlyn McLoughlin
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
| | - Pooja Lalwani
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
| | - Aisling Kelly
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
| | - Ahsan Saeed
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
| | - Kerry McEnaney
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
| | - Lindsey R Baden
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA, USA
| | - Melissa Brogna
- Division of Addiction Medicine, Boston Children's Hospital, Boston, MA, USA
| | - Elissa R Weitzman
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Division of Addiction Medicine, Boston Children's Hospital, Boston, MA, USA
| | - Sharon Levy
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Division of Addiction Medicine, Boston Children's Hospital, Boston, MA, USA
| | - David J Dowling
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Ofer Levy
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA.
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA.
- Broad Institute of MIT & Harvard, Cambridge, MA, USA.
| |
Collapse
|
17
|
Mercuri FA, Anderson GP, Miller BE, Demaison C, Tal-Singer R. Discovery and development of INNA-051, a TLR2/6 agonist for the prevention of complications resulting from viral respiratory infections. Antiviral Res 2025; 234:106063. [PMID: 39733845 DOI: 10.1016/j.antiviral.2024.106063] [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: 10/31/2024] [Revised: 12/18/2024] [Accepted: 12/19/2024] [Indexed: 12/31/2024]
Abstract
Viral respiratory infection is associated with significant morbidity and mortality. The diversity of viruses implicated, coupled with their propensity for mutation, ignited an interest in host-directed antiviral therapies effective across a wide range of viral variants. Toll-like receptors (TLRs) are potential targets for the development of broad-spectrum antivirals given their central role in host immune defenses. Synthetic agonists of TLRs have been shown to boost protective innate immune responses against respiratory viruses. However, clinical success was hindered by short duration of benefit and/or induction of systemic adverse effects. INNA-051, a TLR2/6 agonist, is in development as an intranasal innate immune enhancer for prophylactic treatment in individuals at risk of complications resulting from respiratory viral infections. In vivo animal studies demonstrated the efficacy as prophylaxis against multiple viruses including SARS-CoV-2, influenza, and rhinovirus. Early clinical trials demonstrated an acceptable safety and tolerability profile. Intranasal delivery to the primary site of infection in humans induced a local innate host defense response characterized by innate immune cell infiltration into the nasal epithelium and activation and antiviral response genes. Taken together, the preclinical and clinical data on INNA-051 support further investigation of its use in community infection settings.
Collapse
Affiliation(s)
| | - Gary P Anderson
- Department of Biochemistry and Pharmacology, Faculty of Medicine, Dentistry and Health Sciences, the University of Melbourne, Melbourne, VIC, Australia; Lung Health Research Centre, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, the University of Melbourne, Melbourne, VIC, Australia
| | - Bruce E Miller
- ENA Respiratory Pty Ltd, Melbourne, Australia; BEM Consulting LLC, Phoenixville, PA, USA
| | | | | |
Collapse
|
18
|
Patel RS, Duque D, Bavananthasivam J, Hewitt M, Sandhu JK, Kumar R, Tran A, Agrawal B. Mixed lipopeptide-based mucosal vaccine candidate induces cross-variant immunity and protects against SARS-CoV-2 infection in hamsters. Immunohorizons 2025; 9:vlae011. [PMID: 39849995 PMCID: PMC11841972 DOI: 10.1093/immhor/vlae011] [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: 11/07/2024] [Accepted: 11/14/2024] [Indexed: 01/25/2025] Open
Abstract
The global dissemination of SARS-CoV-2 led to a worldwide pandemic in March 2020. Even after the official downgrading of the COVID-19 pandemic, infection with SARS-CoV-2 variants continues. The rapid development and deployment of SARS-CoV-2 vaccines helped to mitigate the pandemic to a great extent. However, the current vaccines are suboptimal; they elicit incomplete and short-lived protection and are ineffective against evolving virus variants. Updating the spike antigen according to the prevailing variant and repeated boosters is not the long-term solution. We have designed a lipopeptide-based, mucosal, pan-coronavirus vaccine candidate, derived from highly conserved and/or functional regions of the SARS-CoV-2 spike, nucleocapsid, and membrane proteins. Our studies demonstrate that the designed lipopeptides (LPMix) induced both cellular and humoral (mucosal and systemic) immune responses upon intranasal immunization in mice. Furthermore, the antibodies bound to the wild-type and mutated S proteins of SARS-CoV-2 variants of concern, including Alpha, Beta, Delta and Omicron, and also led to efficient neutralization in a surrogate viral neutralization assay. Our sequence alignment and 3-dimensional molecular modeling studies demonstrated that spike-derived epitopes, P1 and P2, are sequentially and/or structurally conserved among the SARS-CoV-2 variants. The addition of a novel mucosal adjuvant, heat-killed Caulobacter crescentus, to the lipopeptide vaccine significantly bolstered mucosal antibody responses. Finally, the lipopeptide-based intranasal vaccine demonstrated significant improvement in lung pathologies in a hamster model of SARS-CoV-2 infection. These studies are fundamentally important and open new avenues in the investigation of an innovative, broadly protective intranasal vaccine platform for SARS-CoV-2 and its variants.
Collapse
Affiliation(s)
- Raj S Patel
- Department of Surgery, Faculty of Medicine and Dentistry, College of Health Sciences, University of Alberta, Edmonton, AB, Canada
| | - Diana Duque
- Infectious Diseases, Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada
| | - Jegarubee Bavananthasivam
- Infectious Diseases, Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada
| | - Melissa Hewitt
- Preclinical Imaging, Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada
| | - Jagdeep K Sandhu
- Preclinical Imaging, Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada
| | | | - Anh Tran
- Infectious Diseases, Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - Babita Agrawal
- Department of Surgery, Faculty of Medicine and Dentistry, College of Health Sciences, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
19
|
Sasaki H, Suzuki Y, Morimoto K, Takeda K, Uchida K, Iyoda M, Ishikawa H. Intranasal Immunization with Nasal Immuno-Inducible Sequence-Fused Antigens Elicits Antigen-Specific Antibody Production. Int J Mol Sci 2024; 25:12828. [PMID: 39684539 DOI: 10.3390/ijms252312828] [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: 10/31/2024] [Revised: 11/22/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
Intranasal immunization is one of the most effective methods for eliciting lung mucosal immunity. Multiple intranasal immunization with bacterial polypeptide, termed as a modified PnxIIIA (MP3) protein, is known to elicit production of a specific antibody in mice. In this study, a nasal immuno-inducible sequence (NAIS) was designed to remove the antigenicity of the MP3 protein that can induce mucosal immunity by intranasal immunization, and was examined to induce antigen-specific antibodies against the fused bacterial thioredoxin (Trx) as a model antigen. A NAIS was modified and generated to remove a large number of predicted MHC (Major Histocompatibility Complex)-I and MHC-II binding sites in parent protein PnxIIIA and MP3 in order to reduce the number of antigen epitope sites. For comparative analysis, full-length NAIS291, NAIS230, and NAIS61 fused with Trx and 6× His tag and Trx-fused 6× His tag were used as antigen variants for the intranasal immunization of BALB/c mice every two weeks for three immunizations. Anti-Trx antibody titers in serum and bronchoalveolar lavage fluid (BALF) IgA obtained from NAIS291-fused Trx-immunized mice were significantly higher than those from Trx-immunized mice. The antibody titers against NAIS alone were significantly lower than those against Trx alone in the serum IgG, serum IgA, and BALF IgA. These results indicate that the NAIS contributes to antibody elicitation of the fused antigen as an immunostimulant in intranasal vaccination vaccines. The results indicate that the NAIS and target inactivated antigen fusions can be applied to intranasal vaccine systems.
Collapse
Affiliation(s)
- Hiraku Sasaki
- Graduate School of Health and Sports Science, Juntendo University, Chiba 2701695, Japan
| | - Yoshio Suzuki
- Graduate School of Health and Sports Science, Juntendo University, Chiba 2701695, Japan
| | - Kodai Morimoto
- Center for Immune Therapeutics and Diagnosis, Juntendo University, Tokyo 1138421, Japan
| | - Kazuyoshi Takeda
- Center for Immune Therapeutics and Diagnosis, Juntendo University, Tokyo 1138421, Japan
- Department of Biofunctional Microbiota, Graduate School of Medicine, Juntendo University, Tokyo 1138421, Japan
- Laboratory of Cell Biology, Research Support Center, Graduate School of Medicine, Juntendo University, Tokyo 1138421, Japan
| | - Koichiro Uchida
- Center for Immune Therapeutics and Diagnosis, Juntendo University, Tokyo 1138421, Japan
| | - Masayuki Iyoda
- Department of Microbiology and Immunology, Showa University School of Medicine, Tokyo 1428555, Japan
| | - Hiroki Ishikawa
- Department of Microbiology and Immunology, Showa University School of Medicine, Tokyo 1428555, Japan
| |
Collapse
|
20
|
Kim JK, Zhu W, Dong C, Wei L, Ma Y, Denning T, Kang SM, Wang BZ. Double-layered protein nanoparticles conjugated with truncated flagellin induce improved mucosal and systemic immune responses in mice. NANOSCALE HORIZONS 2024; 9:2016-2030. [PMID: 39240547 PMCID: PMC11493517 DOI: 10.1039/d4nh00287c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/07/2024]
Abstract
Influenza viral infection poses a severe risk to global public health. Considering the suboptimal protection provided by current influenza vaccines against circulating influenza A viruses, it is imperative to develop novel vaccine formulations to combat respiratory infections. Here, we report the development of an intranasally-administered, self-adjuvanted double-layered protein nanoparticle consisting of influenza nucleoprotein (NP) cores coated with hemagglutinin (HA) and a truncated form of bacterial flagellin (tFliC). Intranasal vaccination of these nanoparticles notably amplified both antigen-specific humoral and cellular immune responses in the systematic compartments. Elevated antigen-specific IgA and IgG levels in mucosal washes, along with increased lung-resident memory B cell populations, were observed in the respiratory system of the immunized mice. Furthermore, intranasal vaccination of tFliC-adjuvanted nanoparticles enhanced survival rates against homologous and heterologous H3N2 viral challenges. Intriguingly, mucosal slow delivery of the prime dose (by splitting the dose into 5 applications over 8 days) significantly enhanced germinal center reactions and effector T-cell populations in lung draining lymph nodes, therefore promoting the protective efficacy against heterologous influenza viral challenges compared to single-prime immunization. These findings highlight the potential of intranasal immunization with tFliC-adjuvanted protein nanoparticles to bolster mucosal and systemic immune responses, with a slow-delivery strategy offering a promising approach for combating influenza epidemics.
Collapse
Affiliation(s)
- Joo Kyung Kim
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, 100 Piedmont Ave SE, Atlanta, Georgia 30303, USA.
| | - Wandi Zhu
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, 100 Piedmont Ave SE, Atlanta, Georgia 30303, USA.
| | - Chunhong Dong
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, 100 Piedmont Ave SE, Atlanta, Georgia 30303, USA.
| | - Lai Wei
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, 100 Piedmont Ave SE, Atlanta, Georgia 30303, USA.
| | - Yao Ma
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, 100 Piedmont Ave SE, Atlanta, Georgia 30303, USA.
| | - Timothy Denning
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, 100 Piedmont Ave SE, Atlanta, Georgia 30303, USA.
| | - Sang-Moo Kang
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, 100 Piedmont Ave SE, Atlanta, Georgia 30303, USA.
| | - Bao-Zhong Wang
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, 100 Piedmont Ave SE, Atlanta, Georgia 30303, USA.
| |
Collapse
|
21
|
Colaço M, Cruz MT, de Almeida LP, Borges O. Mannose and Lactobionic Acid in Nasal Vaccination: Enhancing Antigen Delivery via C-Type Lectin Receptors. Pharmaceutics 2024; 16:1308. [PMID: 39458637 PMCID: PMC11510408 DOI: 10.3390/pharmaceutics16101308] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 09/24/2024] [Accepted: 10/06/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES Nasal vaccines are a promising strategy for enhancing mucosal immune responses and preventing diseases at mucosal sites by stimulating the secretion of secretory IgA, which is crucial for early pathogen neutralization. However, designing effective nasal vaccines is challenging due to the complex immunological mechanisms in the nasal mucosa, which must balance protection and tolerance against constant exposure to inhaled pathogens. The nasal route also presents unique formulation and delivery hurdles, such as the mucous layer hindering antigen penetration and immune cell access. METHODS This review focuses on cutting-edge approaches to enhance nasal vaccine delivery, particularly those targeting C-type lectin receptors (CLRs) like the mannose receptor and macrophage galactose-type lectin (MGL) receptor. It elucidates the roles of these receptors in antigen recognition and uptake by antigen-presenting cells (APCs), providing insights into optimizing vaccine delivery. RESULTS While a comprehensive examination of targeted glycoconjugate vaccine development is outside the scope of this study, we provide key examples of glycan-based ligands, such as lactobionic acid and mannose, which can selectively target CLRs in the nasal mucosa. CONCLUSIONS With the rise of new viral infections, this review aims to facilitate the design of innovative vaccines and equip researchers, clinicians, and vaccine developers with the knowledge to enhance immune defenses against respiratory pathogens, ultimately protecting public health.
Collapse
Affiliation(s)
- Mariana Colaço
- CNC-UC—Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; (M.C.); (M.T.C.); (L.P.d.A.)
- CIBB—Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Maria T. Cruz
- CNC-UC—Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; (M.C.); (M.T.C.); (L.P.d.A.)
- CIBB—Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Luís Pereira de Almeida
- CNC-UC—Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; (M.C.); (M.T.C.); (L.P.d.A.)
- CIBB—Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Olga Borges
- CNC-UC—Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; (M.C.); (M.T.C.); (L.P.d.A.)
- CIBB—Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| |
Collapse
|
22
|
Kazakova A, Zhelnov P, Sidorov R, Rogova A, Vasileva O, Ivanov R, Reshetnikov V, Muslimov A. DNA and RNA vaccines against tuberculosis: a scoping review of human and animal studies. Front Immunol 2024; 15:1457327. [PMID: 39421744 PMCID: PMC11483866 DOI: 10.3389/fimmu.2024.1457327] [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: 06/30/2024] [Accepted: 09/02/2024] [Indexed: 10/19/2024] Open
Abstract
Introduction To comprehensively identify and provide an overview of in vivo or clinical studies of nucleic acids (NA)-based vaccines against TB we included human or animal studies of NA vaccines for the prevention or treatment of TB and excluded in vitro or in silico research, studies of microorganisms other than M. tuberculosis, reviews, letters, and low-yield reports. Methods We searched PubMed, Scopus, Embase, selected Web of Science and ProQuest databases, Google Scholar, eLIBRARY.RU, PROSPERO, OSF Registries, Cochrane CENTRAL, EU Clinical Trials Register, clinicaltrials.gov, and others through WHO International Clinical Trials Registry Platform Search Portal, AVMA and CABI databases, bioRxiv, medRxiv, and others through OSF Preprint Archive Search. We searched the same sources and Google for vaccine names (GX-70) and scanned reviews for references. Data on antigenic composition, delivery systems, adjuvants, and vaccine efficacy were charted and summarized descriptively. Results A total of 18,157 records were identified, of which 968 were assessed for eligibility. No clinical studies were identified. 365 reports of 345 animal studies were included in the review. 342 (99.1%) studies involved DNA vaccines, and the remaining three focused on mRNA vaccines. 285 (82.6%) studies used single-antigen vaccines, while 48 (13.9%) used multiple antigens or combinations with adjuvants. Only 12 (3.5%) studies involved multiepitope vaccines. The most frequently used antigens were immunodominant secretory antigens (Ag85A, Ag85B, ESAT6), heat shock proteins, and cell wall proteins. Most studies delivered naked plasmid DNA intramuscularly without additional adjuvants. Only 4 of 17 studies comparing NA vaccines to BCG after M. tuberculosis challenge demonstrated superior protection in terms of bacterial load reduction. Some vaccine variants showed better efficacy compared to BCG. Systematic review registration https://osf.io/, identifier F7P9G.
Collapse
Affiliation(s)
- Alisa Kazakova
- Translational Medicine Research Center, Sirius University of Science and Technology, Sochi, Russia
| | - Pavel Zhelnov
- Zheln, Toronto, ON, Canada
- Institute of Health Policy, Management and Evaluation, Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| | - Roman Sidorov
- Institute of Ecology and Genetics of Microorganisms, Perm Federal Research Center, Russian Academy of Sciences, Ural Branch, Perm, Russia
| | - Anna Rogova
- Saint-Petersburg State Chemical-Pharmaceutical University, St. Petersburg, Russia
- Laboratory of Nano- and Microencapsulation of Biologically Active Compounds, Peter The Great St. Petersburg Polytechnic University, St. Petersburg, Russia
| | - Olga Vasileva
- Translational Medicine Research Center, Sirius University of Science and Technology, Sochi, Russia
| | - Roman Ivanov
- Translational Medicine Research Center, Sirius University of Science and Technology, Sochi, Russia
| | - Vasiliy Reshetnikov
- Translational Medicine Research Center, Sirius University of Science and Technology, Sochi, Russia
| | - Albert Muslimov
- Saint-Petersburg State Chemical-Pharmaceutical University, St. Petersburg, Russia
| |
Collapse
|
23
|
Pekarek MJ, Weaver EA. Influenza B Virus Vaccine Innovation through Computational Design. Pathogens 2024; 13:755. [PMID: 39338946 PMCID: PMC11434669 DOI: 10.3390/pathogens13090755] [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: 07/30/2024] [Revised: 08/26/2024] [Accepted: 08/31/2024] [Indexed: 09/30/2024] Open
Abstract
As respiratory pathogens, influenza B viruses (IBVs) cause a significant socioeconomic burden each year. Vaccine and antiviral development for influenza viruses has historically viewed IBVs as a secondary concern to influenza A viruses (IAVs) due to their lack of animal reservoirs compared to IAVs. However, prior to the global spread of SARS-CoV-2, the seasonal epidemics caused by IBVs were becoming less predictable and inducing more severe disease, especially in high-risk populations. Globally, researchers have begun to recognize the need for improved prevention strategies for IBVs as a primary concern. This review discusses what is known about IBV evolutionary patterns and the effect of the spread of SARS-CoV-2 on these patterns. We also analyze recent advancements in the development of novel vaccines tested against IBVs, highlighting the promise of computational vaccine design strategies when used to target both IBVs and IAVs and explain why these novel strategies can be employed to improve the effectiveness of IBV vaccines.
Collapse
Affiliation(s)
| | - Eric A. Weaver
- Nebraska Center for Virology, School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA;
| |
Collapse
|
24
|
Biswas M, Nurunnabi M, Khatun Z. Understanding Mucosal Physiology and Rationale of Formulation Design for Improved Mucosal Immunity. ACS APPLIED BIO MATERIALS 2024; 7:5037-5056. [PMID: 38787767 DOI: 10.1021/acsabm.4c00395] [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: 05/26/2024]
Abstract
The oral and nasal cavities serve as critical gateways for infectious pathogens, with microorganisms primarily gaining entry through these routes. Our first line of defense against these invaders is the mucosal membrane, a protective barrier that shields the body's internal systems from infection while also contributing to vital functions like air and nutrient intake. One of the key features of this mucosal barrier is its ability to protect the physiological system from pathogens. Additionally, mucosal tolerance plays a crucial role in maintaining homeostasis by regulating the pH and water balance within the body. Recognizing the importance of the mucosal barrier, researchers have developed various mucosal formulations to enhance the immune response. Mucosal vaccines, for example, deliver antigens directly to mucosal tissues, triggering local immune stimulation and ultimately inducing systemic immunity. Studies have shown that lipid-based formulations such as liposomes and virosomes can effectively elicit both local and systemic immune responses. Furthermore, mucoadhesive polymeric particles, with their prolonged delivery to target sites, have demonstrated an enhanced immune response. This Review delves into the critical role of material selection and delivery approaches in optimizing mucosal immunity.
Collapse
Affiliation(s)
- Mila Biswas
- Department of Electrical and Computer Engineering, University of Texas at El Paso, El Paso, Texas 79902, United States
| | - Md Nurunnabi
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, El Paso, Texas 79902, United States
- Department of Biomedical Engineering, College of Engineering, University of Texas at El Paso, El Paso, Texas 79968, United States
| | - Zehedina Khatun
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, El Paso, Texas 79902, United States
| |
Collapse
|
25
|
Kim DH, Lee J, Lee DY, Lee SH, Jeong JH, Kim JY, Kim J, Choi YK, Lee JB, Park SY, Choi IS, Lee SW, Youk S, Song CS. Intranasal Administration of Recombinant Newcastle Disease Virus Expressing SARS-CoV-2 Spike Protein Protects hACE2 TG Mice against Lethal SARS-CoV-2 Infection. Vaccines (Basel) 2024; 12:921. [PMID: 39204044 PMCID: PMC11359043 DOI: 10.3390/vaccines12080921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 08/07/2024] [Accepted: 08/09/2024] [Indexed: 09/03/2024] Open
Abstract
Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), emerged as a global outbreak in 2019, profoundly affecting both human health and the global economy. Various vaccine modalities were developed and commercialized to overcome this challenge, including inactivated vaccines, mRNA vaccines, adenovirus vector-based vaccines, and subunit vaccines. While intramuscular vaccines induce high IgG levels, they often fail to stimulate significant mucosal immunity in the respiratory system. We employed the Newcastle disease virus (NDV) vector expressing the spike protein of the SARS-CoV-2 Beta variant (rK148/beta-S), and evaluated the efficacy of intranasal vaccination with rK148/beta-S in K18-hACE2 transgenic mice. Intranasal vaccination with a low dose (106.0 EID50) resulted in an 86% survival rate after challenge with the SARS-CoV-2 Beta variant. Administration at a high dose (107.0 EID50) led to a reduction in lung viral load and 100% survival against the SARS-CoV-2 Beta and Delta variants. A high level of the SARS-CoV-2 spike-specific IgA was also induced in vaccinated mice lungs following the SARS-CoV-2 challenge. Our findings suggest that rK148/beta-S holds promise as an intranasal vaccine candidate that effectively induces mucosal immunity against SARS-CoV-2.
Collapse
Affiliation(s)
- Deok-Hwan Kim
- Avian Disease Laboratory, College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea; (D.-H.K.)
- KHAV Co., Ltd., 1 Hwayang-dong, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Jiho Lee
- Avian Disease Laboratory, College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea; (D.-H.K.)
- Southeast Poultry Research Laboratory, U.S. National Poultry Research Center, U.S. Department of Agriculture-Agricultural Research Service, 934 College Station Road, Athens, GA 30605, USA
| | - Da-Ye Lee
- KHAV Co., Ltd., 1 Hwayang-dong, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Seung-Hun Lee
- KHAV Co., Ltd., 1 Hwayang-dong, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Jei-Hyun Jeong
- Avian Disease Laboratory, College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea; (D.-H.K.)
- KHAV Co., Ltd., 1 Hwayang-dong, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Ji-Yun Kim
- KHAV Co., Ltd., 1 Hwayang-dong, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Jiwon Kim
- Department of Microbiology, College of Medicine, Chungbuk National University, Cheongju 28160, Republic of Korea
| | - Yang-Kyu Choi
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea;
| | - Joong-Bok Lee
- Avian Disease Laboratory, College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea; (D.-H.K.)
| | - Seung-Young Park
- Avian Disease Laboratory, College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea; (D.-H.K.)
| | - In-Soo Choi
- Avian Disease Laboratory, College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea; (D.-H.K.)
| | - Sang-Won Lee
- Avian Disease Laboratory, College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea; (D.-H.K.)
| | - Sungsu Youk
- Department of Microbiology, College of Medicine, Chungbuk National University, Cheongju 28160, Republic of Korea
- Biomedical Research Institute, Chungbuk National University Hospital, Cheongju 28644, Republic of Korea
| | - Chang-Seon Song
- Avian Disease Laboratory, College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea; (D.-H.K.)
- KHAV Co., Ltd., 1 Hwayang-dong, Gwangjin-gu, Seoul 05029, Republic of Korea
| |
Collapse
|
26
|
Deng H, Li Y, He X, Wang H, Wang S, Zhang H, Zhu J, Gu L, Li R, Wang G. An intranasal attenuated Coxsackievirus B3 vaccine induces strong systemic and mucosal immunity against CVB3 lethal challenge. J Med Virol 2024; 96:e29831. [PMID: 39072815 DOI: 10.1002/jmv.29831] [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: 04/21/2024] [Revised: 06/25/2024] [Accepted: 07/17/2024] [Indexed: 07/30/2024]
Abstract
Coxsackievirus B3 (CVB3) triggers viral myocarditis, with no effective vaccine yet. This fecal-oral transmitted pathogen has prompted interest in mucosal immunization strategies to impede CVB3 spread. We developed a new attenuated vaccine strain, named CVB3(mu). The potential of CVB3(mu) to stimulate mucosal immune protection remains to be elucidated. This study evaluates the attenuation characteristics of CVB3(mu) via a rapid evolution cellular model and RNA sequencing. Its temperature sensitivity and safety were evaluated through in vitro and in vivo experiments. The mucosal immunity protection of CVB3(mu) was assessed via intranasal immunization in Balb/c mice. The results indicate that CVB3(mu) exhibits temperature sensitivity and forms smaller plaques. It sustains fewer genetic mutations and still possesses certain attenuated traits up to the 25th passage, in comparison to CVB3(WT). Intranasal immunization elicited a significant serum neutralizing antibodies, and a substantial sIgA response in nasal washes. In vivo trials revealed CVB3(mu) protection in adult mice and passive protection in suckling mice against lethal CVB3(WT) challenges. In conclusion, CVB3(mu), a live attenuated intranasal vaccine, provides protection involving humoral and mucosal immunity, making it a promising candidate to control CVB3 spread and infection.
Collapse
MESH Headings
- Animals
- Immunity, Mucosal
- Administration, Intranasal
- Mice, Inbred BALB C
- Enterovirus B, Human/immunology
- Vaccines, Attenuated/immunology
- Vaccines, Attenuated/administration & dosage
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- Antibodies, Neutralizing/blood
- Antibodies, Neutralizing/immunology
- Coxsackievirus Infections/immunology
- Coxsackievirus Infections/prevention & control
- Viral Vaccines/immunology
- Viral Vaccines/administration & dosage
- Mice
- Immunoglobulin A, Secretory/immunology
- Humans
- Female
- Disease Models, Animal
Collapse
Affiliation(s)
- Huixiong Deng
- Department of Microbiology and Immunology, Shantou University Medical College, Shantou, China
| | - Yanlei Li
- Department of Microbiology and Immunology, Shantou University Medical College, Shantou, China
| | - Xuanting He
- Department of Microbiology and Immunology, Shantou University Medical College, Shantou, China
| | - Haoyang Wang
- Department of Microbiology and Immunology, Shantou University Medical College, Shantou, China
| | - Shenmiao Wang
- Department of Microbiology and Immunology, Shantou University Medical College, Shantou, China
| | - Hengyao Zhang
- Department of Microbiology and Immunology, Shantou University Medical College, Shantou, China
| | - Jiacheng Zhu
- Department of Microbiology and Immunology, Shantou University Medical College, Shantou, China
| | - Liming Gu
- Department of Microbiology and Immunology, Shantou University Medical College, Shantou, China
| | - Rui Li
- Department of Microbiology and Immunology, Shantou University Medical College, Shantou, China
| | - Gefei Wang
- Department of Microbiology and Immunology, Shantou University Medical College, Shantou, China
| |
Collapse
|
27
|
Hussain W, Chaman S, Koser HN, Aun SM, Bibi Z, Pirzadi AN, Hussain J, Zubaria Z, Nabi G, Ullah MW, Wang S, Perveen I. Nanoparticle-Mediated Mucosal Vaccination: Harnessing Nucleic Acids for Immune Enhancement. Curr Microbiol 2024; 81:279. [PMID: 39031239 DOI: 10.1007/s00284-024-03803-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Accepted: 07/10/2024] [Indexed: 07/22/2024]
Abstract
Recent advancements in in vitro transcribed mRNA (IVT-mRNA) vaccine manufacturing have attracted considerable interest as advanced methods for combating viral infections. The respiratory mucosa is a primary target for pathogen attack, but traditional intramuscular vaccines are not effective in generating protective ion mucosal surfaces. Mucosal immunization can induce both systemic and mucosal immunity by effectively eliminating microorganisms before their growth and development. However, there are several biological and physical obstacles to the administration of genetic payloads, such as IVT-mRNA and DNA, to the pulmonary and nasal mucosa. Nucleic acid vaccine nanocarriers should effectively protect and load genetic payloads to overcome barriers i.e., biological and physical, at the mucosal sites. This may aid in the transfection of specific antigens, epithelial cells, and incorporation of adjuvants. In this review, we address strategies for delivering genetic payloads, such as nucleic acid vaccines, that have been studied in the past and their potential applications.
Collapse
Affiliation(s)
- Wajid Hussain
- Advanced Biomaterials & Tissues Engineering Center, College of Life Sciences and Technology, Huazhong University of Sciences and Technology, Wuhan, 430074, China
| | - Sadia Chaman
- University of Veterinary and Animals Sciences, Lahore, Pakistan
| | | | | | - Zainab Bibi
- University of the Punjab, Lahore, 54590, Pakistan
| | | | - Jawad Hussain
- Department of Biotechnology, College of Life Sciences and Technology, Huazhong University of Sciences and Technology, Wuhan, 430074, China
| | | | - Ghulam Nabi
- Institute of Nature Conservation, Polish Academy of Sciences, Krakow, Poland
| | - Muhammad Wajid Ullah
- Biofuels Institute, School of the Environment and Safety Engineering, Jiangsu University, Zhenjiang, 212013, People's Republic of China
| | - Shenqi Wang
- Advanced Biomaterials & Tissues Engineering Center, College of Life Sciences and Technology, Huazhong University of Sciences and Technology, Wuhan, 430074, China.
| | - Ishrat Perveen
- GenEd and Molecular Biology Labs, Food and Biotechnology Research Centre, Pakistan Council of Scientific and Industrial Research Centre, Lahore, 54000, Pakistan.
| |
Collapse
|
28
|
Lopes Chaves L, Dourado D, Prunache IB, Manuelle Marques da Silva P, Tacyana Dos Santos Lucena G, Cardoso de Souza Z, Muniz Mendes Freire de Moura P, Nunes Bordallo H, Rocha Formiga F, de Souza Rebouças J. Nanocarriers of antigen proteins for vaccine delivery. Int J Pharm 2024; 659:124162. [PMID: 38663646 DOI: 10.1016/j.ijpharm.2024.124162] [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: 12/07/2023] [Revised: 04/17/2024] [Accepted: 04/22/2024] [Indexed: 06/06/2024]
Abstract
Nanoformulations in vaccinology provide antigen stability and enhanced immunogenicity, in addition to providing targeted delivery and controlled release. In the last years, much research has been focused on vaccine development using virus-like particles, liposomes, emulsions, polymeric, lipid, and inorganic nanoparticles. Importantly, nanoparticle interactions with innate and adaptive immune systems must be clearly understood to guide the rational development of nanovaccines. This review provides a recap and updates on different aspects advocating nanoparticles as promising antigen carriers and immune cell activators for vaccination. Moreover, it offers a discussion of how the physicochemical properties of nanoparticles are modified to target specific cells and improve vaccine efficacy.
Collapse
Affiliation(s)
- Luíse Lopes Chaves
- Department of Immunology, Aggeu Magalhães Institute (IAM), Oswaldo Cruz Foundation (FIOCRUZ), 50670-420, Recife, PE, Brazil; Graduate Program in Applied Cellular and Molecular Biology (PGBCMA), Institute of Biological Sciences, University of Pernambuco (UPE), 50100-130, Recife, PE, Brazil
| | - Douglas Dourado
- Department of Immunology, Aggeu Magalhães Institute (IAM), Oswaldo Cruz Foundation (FIOCRUZ), 50670-420, Recife, PE, Brazil
| | - Ioana-Bianca Prunache
- Niels Bohr Institute, University of Copenhagen, Universitetsparken 5, Copenhagen 2100, Denmark
| | - Paloma Manuelle Marques da Silva
- Graduate Program in Applied Cellular and Molecular Biology (PGBCMA), Institute of Biological Sciences, University of Pernambuco (UPE), 50100-130, Recife, PE, Brazil
| | - Gislayne Tacyana Dos Santos Lucena
- Graduate Program in Applied Cellular and Molecular Biology (PGBCMA), Institute of Biological Sciences, University of Pernambuco (UPE), 50100-130, Recife, PE, Brazil
| | - Zilyane Cardoso de Souza
- Graduate Program in Applied Cellular and Molecular Biology (PGBCMA), Institute of Biological Sciences, University of Pernambuco (UPE), 50100-130, Recife, PE, Brazil
| | - Patrícia Muniz Mendes Freire de Moura
- Graduate Program in Applied Cellular and Molecular Biology (PGBCMA), Institute of Biological Sciences, University of Pernambuco (UPE), 50100-130, Recife, PE, Brazil
| | - Heloísa Nunes Bordallo
- Niels Bohr Institute, University of Copenhagen, Universitetsparken 5, Copenhagen 2100, Denmark
| | - Fabio Rocha Formiga
- Department of Immunology, Aggeu Magalhães Institute (IAM), Oswaldo Cruz Foundation (FIOCRUZ), 50670-420, Recife, PE, Brazil; Graduate Program in Applied Cellular and Molecular Biology (PGBCMA), Institute of Biological Sciences, University of Pernambuco (UPE), 50100-130, Recife, PE, Brazil.
| | - Juliana de Souza Rebouças
- Graduate Program in Applied Cellular and Molecular Biology (PGBCMA), Institute of Biological Sciences, University of Pernambuco (UPE), 50100-130, Recife, PE, Brazil
| |
Collapse
|
29
|
Leyva-Grado VH, Marin A, Hlushko R, Yunus AS, Promeneur D, Luckay A, Lazaro GG, Hamm S, Dimitrov AS, Broder CC, Andrianov AK. Nano-Assembled Polyphosphazene Delivery System Enables Effective Intranasal Immunization with Nipah Virus Subunit Vaccine. ACS APPLIED BIO MATERIALS 2024; 7:4133-4141. [PMID: 38812435 PMCID: PMC11321498 DOI: 10.1021/acsabm.4c00441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2024]
Abstract
The ultimate vaccine against infections caused by Nipah virus should be capable of providing protection at the respiratory tract─the most probable port of entry for this pathogen. Intranasally delivered vaccines, which target nasal-associated lymphoid tissue and induce both systemic and mucosal immunity, are attractive candidates for enabling effective vaccination against this lethal disease. Herein, the water-soluble polyphosphazene delivery vehicle assembles into nanoscale supramolecular constructs with the soluble extracellular portion of the Hendra virus attachment glycoprotein─a promising subunit vaccine antigen against both Nipah and Hendra viruses. These supramolecular constructs signal through Toll-like receptor 7/8 and promote binding interactions with mucin─an important feature of effective mucosal adjuvants. High mass contrast of phosphorus-nitrogen backbone of the polymer enables a successful visualization of nanoconstructs in their vitrified state by cryogenic electron microscopy. Here, we characterize the self-assembly of polyphosphazene macromolecule with biologically relevant ligands by asymmetric flow field flow fractionation, dynamic light scattering, fluorescence spectrophotometry, and turbidimetric titration methods. Furthermore, a polyphosphazene-enabled intranasal Nipah vaccine candidate demonstrates the ability to induce immune responses in hamsters and shows superiority in inducing total IgG and neutralizing antibodies when benchmarked against the respective clinical stage alum adjuvanted vaccine. The results highlight the potential of polyphosphazene-enabled nanoassemblies in the development of intranasal vaccines.
Collapse
Affiliation(s)
| | - Alexander Marin
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, MD 20850
| | - Raman Hlushko
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, MD 20850
| | - Abdul S. Yunus
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, MD 20850
| | | | - Amara Luckay
- Auro Vaccines LLC, 401 Middletown Rd. Bldg. 205, Pearl River, NY, 10965
| | - Glorie G. Lazaro
- Auro Vaccines LLC, 401 Middletown Rd. Bldg. 205, Pearl River, NY, 10965
| | - Stefan Hamm
- Auro Vaccines LLC, 401 Middletown Rd. Bldg. 205, Pearl River, NY, 10965
| | - Antony S. Dimitrov
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, MD 20814
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD 20814
| | - Christopher C. Broder
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, MD 20814
| | - Alexander K. Andrianov
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, MD 20850
| |
Collapse
|
30
|
Gulati N, Chellappan DK, MacLoughlin R, Gupta G, Singh SK, Oliver BG, Dua K, Dureja H. Advances in nano-based drug delivery systems for the management of cytokine influx-mediated inflammation in lung diseases. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:3695-3707. [PMID: 38078921 DOI: 10.1007/s00210-023-02882-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 11/30/2023] [Indexed: 05/23/2024]
Abstract
Asthma, lung cancer, cystic fibrosis, tuberculosis, acute respiratory distress syndrome, chronic obstructive pulmonary disease, and COVID-19 are few examples of inflammatory lung conditions that cause cytokine release syndrome. It can initiate a widespread inflammatory response and may activate several inflammatory pathways that cause multiple organ failures leading to increased number of deaths and increased prevalence rates around the world. Nanotechnology-based therapeutic modalities such as nanoparticles, liposomes, nanosuspension, monoclonal antibodies, and vaccines can be used in the effective treatment of inflammatory lung diseases at both cellular and molecular levels. This would also help significantly in the reduction of patient mortality. Therefore, nanotechnology could be a potent platform for repurposing current medications in the treatment of inflammatory lung diseases. The aim and approach of this article are to highlight the clinical manifestations of cytokine storm in inflammatory lung diseases along with the advances and potential applications of nanotechnology-based therapeutics in the management of cytokine storm. Further in-depth studies are required to understand the molecular pathophysiology, and how nanotechnology-based therapeutics can help to effectively combat this problem.
Collapse
Affiliation(s)
- Nisha Gulati
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, 124001, India
| | - Dinesh Kumar Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University, 57000, Bukit Jalil, Kuala Lumpur, Malaysia
| | - Ronan MacLoughlin
- Research and Development, Science and Emerging Technologies, Aerogen Limited, Galway Business Park, Galway, H91 HE94, Ireland
- School of Pharmacy & Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, D02YN77, Ireland
- School of Pharmacy and Pharmaceutical Sciences, Trinity College, Dublin, D02 PN40, Ireland
| | - Gaurav Gupta
- School of Pharmacy, Suresh Gyan Vihar University, Mahal Road, Jagatpura, Jaipur, India
- Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, 144411, Punjab, India
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, 2007, Australia
| | - Brian G Oliver
- Faculty of Science, School of Life Sciences, University of Technology Sydney, Sydney, NSW, 2007, Australia
- Woolcock Institute of Medical Research, University of Sydney, Sydney, NSW, 2037, Australia
| | - Kamal Dua
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, 124001, India.
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, 2007, Australia.
- Faculty of Science, School of Life Sciences, University of Technology Sydney, Sydney, NSW, 2007, Australia.
- Woolcock Institute of Medical Research, University of Sydney, Sydney, NSW, 2037, Australia.
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW, 2007, Australia.
| | - Harish Dureja
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, 124001, India.
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, 2007, Australia.
| |
Collapse
|
31
|
Seifelnasr A, Si X, Xi J. Effects of Nozzle Retraction Elimination on Spray Distribution in Middle-Posterior Turbinate Regions: A Comparative Study. Pharmaceutics 2024; 16:683. [PMID: 38794345 PMCID: PMC11124954 DOI: 10.3390/pharmaceutics16050683] [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: 03/31/2024] [Revised: 05/08/2024] [Accepted: 05/17/2024] [Indexed: 05/26/2024] Open
Abstract
The standard multi-dose nasal spray pump features an integrated actuator and nozzle, which inevitably causes a retraction of the nozzle tip during application. The retraction stroke is around 5.5 mm and drastically reduces the nozzle's insertion depth, which further affects the initial nasal spray deposition and subsequent translocation, potentially increasing drug wastes and dosimetry variability. To address this issue, we designed a new spray pump that separated the nozzle from the actuator and connected them with a flexible tube, thereby eliminating nozzle retraction during application. The objective of this study is to test the new device's performance in comparison to the conventional nasal pump in terms of spray generation, plume development, and dosimetry distribution. For both devices, the spray droplet size distribution was measured using a laser diffraction particle analyzer. Plume development was recorded with a high-definition camera. Nasal dosimetry was characterized in two transparent nasal cavity casts (normal and decongested) under two breathing conditions (breath-holding and constant inhalation). The nasal formulation was a 0.25% w/v methyl cellulose aqueous solution with a fluorescent dye. For each test case, the temporospatial spray translocation in the nasal cavity was recorded, and the final delivered doses were quantified in five nasal regions. The results indicate minor differences in droplet size distribution between the two devices. The nasal plume from the new device presents a narrower plume angle. The head orientation, the depth at which the nozzle is inserted into the nostril, and the administration angle play crucial roles in determining the initial deposition of nasal sprays as well as the subsequent translocation of the liquid film/droplets. Quantitative measurements of deposition distributions in the nasal models were augmented with visualization recordings to evaluate the delivery enhancements introduced by the new device. With an extension tube, the modified device produced a lower spray output and delivered lower doses in the front, middle, and back turbinate than the conventional nasal pump. However, sprays from the new device were observed to penetrate deeper into the nasal passages, predominantly through the middle-upper meatus. This resulted in consistently enhanced dosing in the middle-upper turbinate regions while at the cost of higher drug loss to the pharynx.
Collapse
Affiliation(s)
- Amr Seifelnasr
- Department of Biomedical Engineering, University of Massachusetts, Lowell, MA 01854, USA;
| | - Xiuhua Si
- Department of Aerospace, Industrial and Mechanical Engineering, California Baptist University, Riverside, CA 92504, USA;
| | - Jinxiang Xi
- Department of Biomedical Engineering, University of Massachusetts, Lowell, MA 01854, USA;
| |
Collapse
|
32
|
Song Y, Mehl F, Zeichner SL. Vaccine Strategies to Elicit Mucosal Immunity. Vaccines (Basel) 2024; 12:191. [PMID: 38400174 PMCID: PMC10892965 DOI: 10.3390/vaccines12020191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 01/29/2024] [Accepted: 02/08/2024] [Indexed: 02/25/2024] Open
Abstract
Vaccines are essential tools to prevent infection and control transmission of infectious diseases that threaten public health. Most infectious agents enter their hosts across mucosal surfaces, which make up key first lines of host defense against pathogens. Mucosal immune responses play critical roles in host immune defense to provide durable and better recall responses. Substantial attention has been focused on developing effective mucosal vaccines to elicit robust localized and systemic immune responses by administration via mucosal routes. Mucosal vaccines that elicit effective immune responses yield protection superior to parenterally delivered vaccines. Beyond their valuable immunogenicity, mucosal vaccines can be less expensive and easier to administer without a need for injection materials and more highly trained personnel. However, developing effective mucosal vaccines faces many challenges, and much effort has been directed at their development. In this article, we review the history of mucosal vaccine development and present an overview of mucosal compartment biology and the roles that mucosal immunity plays in defending against infection, knowledge that has helped inform mucosal vaccine development. We explore new progress in mucosal vaccine design and optimization and novel approaches created to improve the efficacy and safety of mucosal vaccines.
Collapse
Affiliation(s)
- Yufeng Song
- Department of Pediatrics, University of Virginia, Charlottesville, VA 22908, USA; (Y.S.)
| | - Frances Mehl
- Department of Pediatrics, University of Virginia, Charlottesville, VA 22908, USA; (Y.S.)
| | - Steven L. Zeichner
- Department of Pediatrics, University of Virginia, Charlottesville, VA 22908, USA; (Y.S.)
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA
| |
Collapse
|
33
|
Ao D, He X, Liu J, Xu L. Strategies for the development and approval of COVID-19 vaccines and therapeutics in the post-pandemic period. Signal Transduct Target Ther 2023; 8:466. [PMID: 38129394 PMCID: PMC10739883 DOI: 10.1038/s41392-023-01724-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 11/24/2023] [Accepted: 12/06/2023] [Indexed: 12/23/2023] Open
Abstract
The spread of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has resulted in significant casualties and put immense strain on public health systems worldwide, leading to economic recession and social unrest. In response, various prevention and control strategies have been implemented globally, including vaccine and drug development and the promotion of preventive measures. Implementing these strategies has effectively curbed the transmission of the virus, reduced infection rates, and gradually restored normal social and economic activities. However, the mutations of SARS-CoV-2 have led to inevitable infections and reinfections, and the number of deaths continues to rise. Therefore, there is still a need to improve existing prevention and control strategies, mainly focusing on developing novel vaccines and drugs, expediting medical authorization processes, and keeping epidemic surveillance. These measures are crucial to combat the Coronavirus disease (COVID-19) pandemic and achieve sustained, long-term prevention, management, and disease control. Here, we summarized the characteristics of existing COVID-19 vaccines and drugs and suggested potential future directions for their development. Furthermore, we discussed the COVID-19-related policies implemented over the past years and presented some strategies for the future.
Collapse
Affiliation(s)
- Danyi Ao
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, 610041, Sichuan, People's Republic of China
| | - Xuemei He
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, 610041, Sichuan, People's Republic of China
| | - Jian Liu
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, 610041, Sichuan, People's Republic of China
| | - Li Xu
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, 610041, Sichuan, People's Republic of China.
| |
Collapse
|
34
|
Vergara EJ, Tran AC, Kim MY, Mussá T, Paul MJ, Harrison T, Reljic R. Mucosal and systemic immune responses after a single intranasal dose of nanoparticle and spore-based subunit vaccines in mice with pre-existing lung mycobacterial immunity. Front Immunol 2023; 14:1306449. [PMID: 38130713 PMCID: PMC10733481 DOI: 10.3389/fimmu.2023.1306449] [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: 10/03/2023] [Accepted: 11/15/2023] [Indexed: 12/23/2023] Open
Abstract
Tuberculosis (TB) is a major global health threat that claims more than one million lives annually. With a quarter of the global population harbouring latent TB, post-exposure vaccination aimed at high-risk populations that could develop active TB disease would be of great public health benefit. Mucosal vaccination is an attractive approach for a predominantly lung disease like TB because it elicits both local and systemic immunity. However, the immunological consequence of mucosal immunisation in the presence of existing lung immunity remains largely unexplored. Using a mycobacterial pre-exposure mouse model, we assessed whether pre-existing mucosal and systemic immune responses can be boosted and/or qualitatively altered by intranasal administration of spore- and nanoparticle-based subunit vaccines. Analysis of lung T cell responses revealed an increasing trend in the frequency of important CD4 and CD8 T cell subsets, and T effector memory cells with a Th1 cytokine (IFNγ and TNFα) signature among immunised mice. Additionally, significantly greater antigen specific Th1, Th17 and IL-10 responses, and antigen-induced T cell proliferation were seen from the spleens of immunised mice. Measurement of antigen-specific IgG and IgA from blood and bronchoalveolar lavage fluid also revealed enhanced systemic and local humoral immune responses among immunised animals. Lastly, peripheral blood mononuclear cells (PBMCs) obtained from the TB-endemic country of Mozambique show that individuals with LTBI showed significantly greater CD4 T cell reactivity to the vaccine candidate as compared to healthy controls. These results support further testing of Spore-FP1 and Nano-FP1 as post-exposure TB vaccines.
Collapse
Affiliation(s)
- Emil Joseph Vergara
- Institute for Infection and Immunity, St. George’s University of London, London, United Kingdom
| | - Andy Cano Tran
- Institute for Infection and Immunity, St. George’s University of London, London, United Kingdom
| | - Mi-Young Kim
- Institute for Infection and Immunity, St. George’s University of London, London, United Kingdom
- Department of Molecular Biology, Jeonbuk National University, Jeonju, Republic of Korea
| | - Tufária Mussá
- Department of Microbiology, Faculty of Medicine, Eduardo Mondlane University, Maputo, Mozambique
| | - Matthew J. Paul
- Institute for Infection and Immunity, St. George’s University of London, London, United Kingdom
| | - Thomas Harrison
- Institute for Infection and Immunity, St. George’s University of London, London, United Kingdom
| | - Rajko Reljic
- Institute for Infection and Immunity, St. George’s University of London, London, United Kingdom
| |
Collapse
|
35
|
Xu Y, Yan X, Wei T, Chen M, Zhu J, Gao J, Liu B, Zhu W, Liu Z. Transmucosal Delivery of Nasal Nanovaccines Enhancing Mucosal and Systemic Immunity. NANO LETTERS 2023; 23:10522-10531. [PMID: 37943583 DOI: 10.1021/acs.nanolett.3c03419] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/10/2023]
Abstract
Intranasal vaccines can induce protective immune responses at the mucosa surface entrance, preventing the invasion of respiratory pathogens. However, the nasal barrier remains a major challenge in the development of intranasal vaccines. Herein, a transmucosal nanovaccine based on cationic fluorocarbon modified chitosan (FCS) is developed to induce mucosal immunity. In our system, FCS can self-assemble with the model antigen ovalbumin and TLR9 agonist CpG, effectively promoting the maturation and cross-presentation of dendritic cells. More importantly, it can enhance the production of secretory immunoglobin A (sIgA) at mucosal surfaces for those intranasally vaccinated mice, which in the meantime showed effective production of immunoglobulin G (IgG) systemically. As a proof-of-concept study, such a mucosal vaccine inhibits ovalbumin-expressing B16-OVA melanoma, especially its lung metastases. Our work presents a unique intranasal delivery system to deliver antigen across mucosal epithelia and promote mucosal and systemic immunity.
Collapse
Affiliation(s)
- Yuchun Xu
- Institute of Functional Nano & Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Xiaoying Yan
- Institute of Functional Nano & Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Ting Wei
- Suzhou InnoBM Pharmaceutics Co. Ltd., Suzhou, Jiangsu 215213, China
| | - Minming Chen
- Institute of Functional Nano & Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Jiafei Zhu
- Institute of Functional Nano & Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Juxin Gao
- Institute of Functional Nano & Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Bo Liu
- Institute of Functional Nano & Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Wenjun Zhu
- Institute of Functional Nano & Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Zhuang Liu
- Institute of Functional Nano & Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices, Soochow University, Suzhou 215123, China
- Suzhou InnoBM Pharmaceutics Co. Ltd., Suzhou, Jiangsu 215213, China
| |
Collapse
|
36
|
Shaw HA, Remmington A, McKenzie G, Winkel C, Mawas F. Mucosal Immunization Has Benefits over Traditional Subcutaneous Immunization with Group A Streptococcus Antigens in a Pilot Study in a Mouse Model. Vaccines (Basel) 2023; 11:1724. [PMID: 38006056 PMCID: PMC10674289 DOI: 10.3390/vaccines11111724] [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: 09/04/2023] [Revised: 10/18/2023] [Accepted: 11/13/2023] [Indexed: 11/26/2023] Open
Abstract
Group A Streptococcus (GAS) is a major human pathogen for which there is no licensed vaccine. To protect against infection, a strong systemic and mucosal immune response is likely to be necessary to prevent initial colonization and any events that might lead to invasive disease. A broad immune response will be necessary to target the varied GAS serotypes and disease presentations. To this end, we designed a representative panel of recombinant proteins to cover the stages of GAS infection and investigated whether mucosal and systemic immunity could be stimulated by these protein antigens. We immunized mice sublingually, intranasally and subcutaneously, then measured IgG and IgA antibody levels and functional activity through in vitro assays. Our results show that both sublingual and intranasal immunization in the presence of adjuvant induced both systemic IgG and mucosal IgA. Meanwhile, subcutaneous immunization generated only a serum IgG response. The antibodies mediated binding and killing of GAS cells and blocked binding of GAS to HaCaT cells, particularly following intranasal and subcutaneous immunizations. Further, antigen-specific assays revealed that immune sera inhibited cleavage of IL-8 by SpyCEP and IgG by Mac/IdeS. These results demonstrate that mucosal immunization can induce effective systemic and mucosal antibody responses. This finding warrants further investigation and optimization of humoral and cellular responses as a viable alternative to subcutaneous immunization for urgently needed GAS vaccines.
Collapse
Affiliation(s)
- Helen Alexandra Shaw
- Vaccines Division, Science, Research & Innovation, Medicines and Healthcare Products Regulatory Agency, Potters Bar EN6 3QG, UK
| | | | | | | | | |
Collapse
|
37
|
Aroffu M, Manca ML, Pedraz JL, Manconi M. Liposome-based vaccines for minimally or noninvasive administration: an update on current advancements. Expert Opin Drug Deliv 2023; 20:1573-1593. [PMID: 38015659 DOI: 10.1080/17425247.2023.2288856] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 11/24/2023] [Indexed: 11/30/2023]
Abstract
INTRODUCTION Vaccination requires innovation to provide effective protection. Traditional vaccines have several drawbacks, which can be overcome with advanced technologies and different administration routes. Over the past 10 years, a significant amount of research has focussed on the delivery of antigens into liposomes due to their dual role as antigen-carrying systems and vaccine adjuvants able to increase the immunogenicity of the carried antigen. AREAS COVERED This review encompasses the progress made over the last 10 years with liposome-based vaccines designed for minimally or noninvasive administration, filling the gaps in previous reviews and providing insights on composition, administration routes, results achieved, and Technology Readiness Level of the most recent formulations. EXPERT OPINION Liposome-based vaccines administered through minimally or noninvasive routes are expected to improve efficacy and complacency of vaccination programs. However, the translation from lab-scale production to large-scale production and collaborations with hospitals, research centers, and companies are needed to allow new products to enter the market and improve the vaccination programs in the future.
Collapse
Affiliation(s)
- Matteo Aroffu
- Department of Scienze della Vita e dell'Ambiente, University of Cagliari, Cagliari, Italy
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain
| | - Maria Letizia Manca
- Department of Scienze della Vita e dell'Ambiente, University of Cagliari, Cagliari, Italy
| | - José Luis Pedraz
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain
- Biomedical Research Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Vitoria-Gasteiz, Spain
- BioAraba, NanoBioCel research Group, Vitoria-Gasteiz, Spain
| | - Maria Manconi
- Department of Scienze della Vita e dell'Ambiente, University of Cagliari, Cagliari, Italy
| |
Collapse
|