1
|
Zhang X, Fu Y, Chen S, Liu G, Wang Y, He Q, Wang Q, Li N, Wang Z, Chen L, Wang J, Liang Z, Xu M, Mao Q. Exploring the standardization of human nasal antibody measurements. Emerg Microbes Infect 2025; 14:2475822. [PMID: 40071971 PMCID: PMC11915745 DOI: 10.1080/22221751.2025.2475822] [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: 12/02/2024] [Revised: 01/22/2025] [Accepted: 03/02/2025] [Indexed: 03/18/2025]
Abstract
Mucosal immunity is crucial for preventing the infection and transmission of respiratory viruses. Nasal antibody is inversely correlated with a lower risk of infection with respiratory viruses. However, the current reference standard for nasal antibody assessment is serum-based, mainly consisting of monomeric IgG and IgA. The applicability of serum-derived standards for assessing nasal antibodies, consisting mostly of dimeric or polymeric secretory IgA (sIgA), remains unvalidated. Herein, we first proved that the sera-derived standard was not applicable for assessing nasal antibodies. Using a non-homologous standard as a calibrator introduced systematic error up to 10 times, which did not benefit the understanding of mucosal antibody response. Therefore, we attempted to develop two candidate standards (CS1, CS2) using nasal mucosal lining fluids (NMLFs) collected from SARS-CoV-2 Omicron convalescents or intranasal vaccine recipients, and CS3 using a sIgA monoclonal antibody. CS2 exhibited broad-spectrum binding activity against 12 SARS-CoV-2 strains, including all tested Omicron subvariants. A collaborative study conducted by seven laboratories demonstrated that CS2 improved the harmonization of inter-laboratory variability (pre-standardization geometric coefficients of variance, 14-314%; post-standardization, 3-35%). Using CS2 ensured an accurate assessment of nasal antibodies. Thus, CS2 was established as a national standard for evaluating nasal SARS-CoV-2-specific antibodies (Lot: 300052-202401, 1000 U/mL). Our work provides a benchmark for evaluating mucosal vaccines for SARS-CoV-2 and inspires new avenues for developing new reference standards for other mucosal vaccines.
Collapse
Affiliation(s)
- Xuanxuan Zhang
- State Key Laboratory of Drug Regulatory Science, Evaluation of Biological Products, Key Laboratory of Research on Quality and Standardization of Biotech Products, Institute of Biological Products, National Institutes for Food and Drug Control, Beijing, People's Republic of China
- Research Units of Innovative Vaccine Quality Evaluation and Standardization, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| | - Yulong Fu
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, People's Republic of China
| | - Si Chen
- Drug and Vaccine Research Center, Guangzhou National Laboratory, Guangzhou, People's Republic of China
- Guangzhou Institute of Infectious Disease, Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Guanxing Liu
- Changchun Institute of Biological Products Co., Ltd., Changchun, People's Republic of China
| | - Ying Wang
- State Key Laboratory of Drug Regulatory Science, Evaluation of Biological Products, Key Laboratory of Research on Quality and Standardization of Biotech Products, Institute of Biological Products, National Institutes for Food and Drug Control, Beijing, People's Republic of China
- Research Units of Innovative Vaccine Quality Evaluation and Standardization, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| | - Qian He
- State Key Laboratory of Drug Regulatory Science, Evaluation of Biological Products, Key Laboratory of Research on Quality and Standardization of Biotech Products, Institute of Biological Products, National Institutes for Food and Drug Control, Beijing, People's Republic of China
- Research Units of Innovative Vaccine Quality Evaluation and Standardization, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| | - Qian Wang
- Drug and Vaccine Research Center, Guangzhou National Laboratory, Guangzhou, People's Republic of China
| | - Na Li
- Beijing Minhai Biotechnology Co., Ltd., Beijing, People's Republic of China
| | - Zhongfang Wang
- Drug and Vaccine Research Center, Guangzhou National Laboratory, Guangzhou, People's Republic of China
| | - Ling Chen
- Drug and Vaccine Research Center, Guangzhou National Laboratory, Guangzhou, People's Republic of China
- Guangzhou Institute of Infectious Disease, Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Junzhi Wang
- State Key Laboratory of Drug Regulatory Science, Evaluation of Biological Products, Key Laboratory of Research on Quality and Standardization of Biotech Products, Institute of Biological Products, National Institutes for Food and Drug Control, Beijing, People's Republic of China
- Research Units of Innovative Vaccine Quality Evaluation and Standardization, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| | - Zhenglun Liang
- State Key Laboratory of Drug Regulatory Science, Evaluation of Biological Products, Key Laboratory of Research on Quality and Standardization of Biotech Products, Institute of Biological Products, National Institutes for Food and Drug Control, Beijing, People's Republic of China
- Research Units of Innovative Vaccine Quality Evaluation and Standardization, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| | - Miao Xu
- State Key Laboratory of Drug Regulatory Science, Evaluation of Biological Products, Key Laboratory of Research on Quality and Standardization of Biotech Products, Institute of Biological Products, National Institutes for Food and Drug Control, Beijing, People's Republic of China
- Research Units of Innovative Vaccine Quality Evaluation and Standardization, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| | - Qunying Mao
- State Key Laboratory of Drug Regulatory Science, Evaluation of Biological Products, Key Laboratory of Research on Quality and Standardization of Biotech Products, Institute of Biological Products, National Institutes for Food and Drug Control, Beijing, People's Republic of China
- Research Units of Innovative Vaccine Quality Evaluation and Standardization, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| |
Collapse
|
2
|
Case JB, Jain S, Suthar MS, Diamond MS. SARS-CoV-2: The Interplay Between Evolution and Host Immunity. Annu Rev Immunol 2025; 43:29-55. [PMID: 39705164 DOI: 10.1146/annurev-immunol-083122-043054] [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: 12/22/2024]
Abstract
The persistence of SARS-CoV-2 infections at a global level reflects the repeated emergence of variant strains encoding unique constellations of mutations. These variants have been generated principally because of a dynamic host immune landscape, the countermeasures deployed to combat disease, and selection for enhanced infection of the upper airway and respiratory transmission. The resulting viral diversity creates a challenge for vaccination efforts to maintain efficacy, especially regarding humoral aspects of protection. Here, we review our understanding of how SARS-CoV-2 has evolved during the pandemic, the immune mechanisms that confer protection, and the impact viral evolution has had on transmissibility and adaptive immunity elicited by natural infection and/or vaccination. Evidence suggests that SARS-CoV-2 evolution initially selected variants with increased transmissibility but currently is driven by immune escape. The virus likely will continue to drift to maintain fitness until countermeasures capable of disrupting transmission cycles become widely available.
Collapse
Affiliation(s)
- James Brett Case
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA;
| | - Shilpi Jain
- Emory Vaccine Center, Emory National Primate Research Center, Atlanta, Georgia, USA
- Center for Childhood Infections and Vaccines of Children's Healthcare of Atlanta, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Mehul S Suthar
- Emory Vaccine Center, Emory National Primate Research Center, Atlanta, Georgia, USA
- Center for Childhood Infections and Vaccines of Children's Healthcare of Atlanta, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Michael S Diamond
- Department of Pathology & Immunology; Department of Molecular Microbiology; and Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA;
| |
Collapse
|
3
|
Tian Y, Hu L, Huang Q, Qi J, Shen L, Wang G, Yu W, Hu T. A SARS-CoV-2 mucosal nanovaccine based on assembly of maltodextrin, STING agonist and polyethyleneimine. Int J Biol Macromol 2025; 294:139395. [PMID: 39756748 DOI: 10.1016/j.ijbiomac.2024.139395] [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: 08/27/2024] [Revised: 11/17/2024] [Accepted: 12/29/2024] [Indexed: 01/07/2025]
Abstract
SARS-CoV-2 has the characteristics of strong transmission with severe morbidity and mortality. Protein-based vaccines have the properties of specificity, effectiveness and safety against SARS-CoV-2. Receptor-binding domain (RBD) homotrimer affords high protection efficacy against stringent lethal viral challenge. Mucosal immunity could block the infection that first infect and replicate in the upper airway mucosa. Due to the physical barriers of the mucosa, mucosal vaccines necessitated appropriate adjuvants and delivery system. In the present study, maltodextrin, PEI and 2',3'-cGAMP acted as the mucosal adjuvants and RBD trimer as the antigen. A mucosal nanovaccine was prepared by assembly of adjuvants and the antigen to a nanoparticle. The vaccine elicited strong serum RBD-specific IgG and IgA response, and mild mucosal IgA and IgG response in the respiratory tract. It stimulated strong neutralizing antibody response and high ACE2-blocking activity in the sera. It promoted the RBD-specific CD4+ and CD8+ T cells secreting IFN-γ, IL-4 and IL-17 A. Moreover, it elicited durable RBD-specific memory T and B memory cell response, activated the T and B cells, enhanced the cytotoxic T cell killing effect, and promoted the maturation of DCs. These findings suggested the clinical potential of the vaccine to combat against SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Yu Tian
- College of Chemical Engineering, Shenyang University of Chemical Technology, Shenyang 110142, China; State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
| | - Lijia Hu
- School of International Relations, Beijing Language and Culture University, Beijing 100083, China
| | | | - Jinming Qi
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
| | - Lijuan Shen
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
| | - Guosheng Wang
- College of Chemical Engineering, Shenyang University of Chemical Technology, Shenyang 110142, China
| | - Weili Yu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
| | - Tao Hu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China.
| |
Collapse
|
4
|
Maniyamgama N, Bae KH, Chang ZW, Lee J, Ang MJY, Tan YJ, Ng LFP, Renia L, White KP, Yang YY. Muco-Penetrating Lipid Nanoparticles Having a Liquid Core for Enhanced Intranasal mRNA Delivery. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2407383. [PMID: 39888252 PMCID: PMC11923898 DOI: 10.1002/advs.202407383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 08/15/2024] [Indexed: 02/01/2025]
Abstract
Intranasal delivery of mRNA vaccines offers promising opportunities to combat airborne viruses like SARS-CoV-2 by provoking mucosal immunity, which not only defends against respiratory infection but also prevents contagious transmission. However, the development of nasal mRNA vaccines has been hampered by the lack of effective means to overcome the mucus barrier. Herein, ionizable lipid-incorporated liquid lipid nanoparticles (iLLNs) capable of delivering mRNA cargo across airway mucosa are designed. Adjusting the ratios of ionizable and cationic lipids allows fine-tuning of the pKa of iLLNs to the range of nasal mucosal pH (5.5-6.5), thus facilitating mucus penetration via the formation of near-neutral, PEGylated muco-inert surfaces. When nasally administered to mice, the top candidate iLLN-2/mRNA complexes enable about 60-fold greater reporter gene expression in the nasal cavity, compared to the benchmark mRNA-lipid nanoparticles (ALC-LNP) having the same lipid composition as that of BNT162b2 vaccine. Moreover, a prime-boost intranasal immunization of iLLN-2/mRNA complexes elicits a greater magnitude of SARS-CoV-2 spike-specific mucosal IgA and IgG response than ALC-LNP, without triggering any noticeable inflammatory reactions. Taken together, these results provide useful insights for the design of nasally deliverable mRNA formulations for prophylactic applications.
Collapse
Affiliation(s)
- Nipuni Maniyamgama
- Bioprocessing Technology Institute (BTI)Agency for Science, Technology and Research (A*STAR)20 Biopolis Way, Centros #06‐01Singapore138668Republic of Singapore
| | - Ki Hyun Bae
- Bioprocessing Technology Institute (BTI)Agency for Science, Technology and Research (A*STAR)20 Biopolis Way, Centros #06‐01Singapore138668Republic of Singapore
| | - Zi Wei Chang
- A*STAR Infectious Diseases Labs (A*STAR ID Labs)Agency for Science, Technology and Research (A*STAR)8A Biomedical Grove, Immunos #05‐13Singapore138648Republic of Singapore
| | - Jialing Lee
- Bioprocessing Technology Institute (BTI)Agency for Science, Technology and Research (A*STAR)20 Biopolis Way, Centros #06‐01Singapore138668Republic of Singapore
| | - Melgious J. Y. Ang
- Bioprocessing Technology Institute (BTI)Agency for Science, Technology and Research (A*STAR)20 Biopolis Way, Centros #06‐01Singapore138668Republic of Singapore
| | - Yong Jie Tan
- A*STAR Infectious Diseases Labs (A*STAR ID Labs)Agency for Science, Technology and Research (A*STAR)8A Biomedical Grove, Immunos #05‐13Singapore138648Republic of Singapore
| | - Lisa F. P. Ng
- A*STAR Infectious Diseases Labs (A*STAR ID Labs)Agency for Science, Technology and Research (A*STAR)8A Biomedical Grove, Immunos #05‐13Singapore138648Republic of Singapore
| | - Laurent Renia
- A*STAR Infectious Diseases Labs (A*STAR ID Labs)Agency for Science, Technology and Research (A*STAR)8A Biomedical Grove, Immunos #05‐13Singapore138648Republic of Singapore
- Lee Kong Chian School of MedicineNanyang Technological UniversitySingapore138648Republic of Singapore
- School of Biological SciencesNanyang Technological UniversitySingapore138648Republic of Singapore
| | - Kevin P. White
- Department of Biochemistry and Precision Medicine Translational Research ProgramYong Loo Lin School of MedicineNational University of SingaporeSingapore119228Republic of Singapore
| | - Yi Yan Yang
- Bioprocessing Technology Institute (BTI)Agency for Science, Technology and Research (A*STAR)20 Biopolis Way, Centros #06‐01Singapore138668Republic of Singapore
| |
Collapse
|
5
|
Kumar S, Hazlett K, Bai G. Mucosal immunity elicited by a human-Fcγ receptor-I targeted intranasal vaccine platform enhances resistance against nasopharyngeal colonization of Streptococcus pneumoniae and induces broadly protective immunity against respiratory pathogens. Vaccine 2025; 48:126729. [PMID: 39823848 DOI: 10.1016/j.vaccine.2025.126729] [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: 11/14/2024] [Revised: 12/20/2024] [Accepted: 01/09/2025] [Indexed: 01/20/2025]
Abstract
The development of safe and effective mucosal vaccines are hampered by safety concerns associated with adjuvants or live attenuated microbes. We previously demonstrated that targeting antigens to the human-Fc-gamma-receptor-I (hFcγRI) eliminates the need for adjuvants, thereby mitigating safety concerns associated with the mucosal delivery of adjuvant formulated vaccines. Here we evaluated the role of the route of immunization in the mucosal immunity elicited by the hFcγRI-targeted vaccine approach. To enable Ag targeting, PspA from Streptococcus pneumoniae (Sp) was genetically fused with the hFcγRI-targeting antibody (α-hFcγRI) to generate PspA-FP. Intranasal (IN) immunization with the PspA-FP induced significantly higher IgA, IgG, and memory T cell response in the lung mucosa compared to that of the intramuscular (IM) route, while both routes exhibited similar increase in the systemic IgG response. The IN immunization elicited better resistance against nasal colonization (NC) of Sp compared to the IM immunization. Additionally, the resistance to NC with the IN administered PspA-FP was higher than the PspA-Alum formulation administered by the IM route. While the protection form lethal pulmonary Sp infection correlated with the systemic Ab response, the resistance from NC (of Sp) correlated with the mucosal immune response. Similar to the pneumococcal pneumoniae model, the hFcγRI-targeted vaccine (based on HA as Ag) was equally protective against pulmonary Influenza virus infection via both routes. However, the IN route promoted better protection compared to the IM route against a lethal pulmonary infection with Francisella tularensis (Ft). The enhanced protection against Ft correlated with the superior mucosal immune response elicited by the IN route compared to the IM route. These observations showed a differential requirement for mucosal delivery for protection depending on the type of pathogen. Moreover, this study revealed that the hFcγRI-targeted vaccine platform is broadly-effective as an adjuvant-free mucosal vaccine platform against respiratory pathogens.
Collapse
Affiliation(s)
- Sudeep Kumar
- Department of Immunology and Microbial Disease, Albany Medical College, 47 New Scotland Ave, Albany, NY 12208, United States.
| | - Karsten Hazlett
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, 47 New Scotland Ave, Albany, NY 12208, United States
| | - Guangchun Bai
- Department of Immunology and Microbial Disease, Albany Medical College, 47 New Scotland Ave, Albany, NY 12208, United States
| |
Collapse
|
6
|
Pilapitiya D, Lee WS, Vu MN, Kelly A, Webster RH, Koutsakos M, Kent SJ, Juno JA, Tan HX, Wheatley AK. Mucosal vaccination against SARS-CoV-2 using recombinant influenza viruses delivering self-assembling nanoparticles. Vaccine 2025; 46:126668. [PMID: 39740385 DOI: 10.1016/j.vaccine.2024.126668] [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: 08/30/2024] [Revised: 11/07/2024] [Accepted: 12/21/2024] [Indexed: 01/02/2025]
Abstract
Recombinant influenza viruses are promising vectors that can bolster antibody and resident lymphocyte responses within mucosal sites. This study evaluates recombinant influenza viruses with SARS-CoV-2 RBD genes in eliciting mucosal and systemic responses. Using reverse genetics, we generated replication-competent recombinant influenza viruses carrying heterologous RBD genes in monomeric, trimeric, or ferritin-based nanoparticle forms. Following intranasal immunisation, mice developed potent serological anti-RBD responses, with ferritin nanoparticles superseding monomeric or trimeric RBD responses. While parenteral and mucosal immunisation elicited robust anti-RBD IgG in serum, mucosal immunisation seeded respiratory IgA, RBD-specific lung-resident memory and germinal centre (GC) B cells. In animals with prior intramuscular vaccination, intranasal boosting with recombinant influenza vectors augmented mucosal IgG, IgA, GC and memory B cells, and SARS-CoV-2 lung neutralising titres. Recall of RBD-specific memory B cells via antigen re-exposure in the lung increased antibody-secreting cells in the lung-draining lymph nodes, with maintenance of lung GC B cells. Recombinant influenza-based vaccines effectively deliver highly immunogenic self-assembling nanoparticles, generating antibodies and B cells in the respiratory mucosa. This strategy provides a tractable pathway to augment lung-localised responses against recurrent respiratory viral infections.
Collapse
MESH Headings
- Animals
- Nanoparticles/administration & dosage
- Nanoparticles/chemistry
- SARS-CoV-2/immunology
- COVID-19/prevention & control
- COVID-19/immunology
- Antibodies, Viral/immunology
- Antibodies, Viral/blood
- Mice
- COVID-19 Vaccines/immunology
- COVID-19 Vaccines/administration & dosage
- Influenza Vaccines/immunology
- Influenza Vaccines/administration & dosage
- Female
- Administration, Intranasal
- Immunity, Mucosal
- Immunoglobulin G/immunology
- Vaccines, Synthetic/immunology
- Vaccines, Synthetic/administration & dosage
- Mice, Inbred BALB C
- Antibodies, Neutralizing/immunology
- Immunoglobulin A/immunology
- Spike Glycoprotein, Coronavirus/immunology
- Spike Glycoprotein, Coronavirus/genetics
- Vaccination/methods
- Lung/immunology
- B-Lymphocytes/immunology
- Humans
- Orthomyxoviridae/genetics
- Orthomyxoviridae/immunology
Collapse
Affiliation(s)
- Devaki Pilapitiya
- Department of Microbiology and Immunology, University of Melbourne, at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Wen Shi Lee
- Department of Microbiology and Immunology, University of Melbourne, at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Mai N Vu
- Department of Microbiology and Immunology, University of Melbourne, at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Andrew Kelly
- Department of Microbiology and Immunology, University of Melbourne, at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Rosela H Webster
- Department of Microbiology and Immunology, University of Melbourne, at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Marios Koutsakos
- Department of Microbiology and Immunology, University of Melbourne, at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Stephen J Kent
- Department of Microbiology and Immunology, University of Melbourne, at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia; Melbourne Sexual Health Centre and Department of Infectious Diseases, Alfred Hospital and Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Jennifer A Juno
- Department of Microbiology and Immunology, University of Melbourne, at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Hyon-Xhi Tan
- Department of Microbiology and Immunology, University of Melbourne, at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia.
| | - Adam K Wheatley
- Department of Microbiology and Immunology, University of Melbourne, at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia.
| |
Collapse
|
7
|
Zhou M, Xiao H, Yang X, Cheng T, Yuan L, Xia N. Novel vaccine strategies to induce respiratory mucosal immunity: advances and implications. MedComm (Beijing) 2025; 6:e70056. [PMID: 39830020 PMCID: PMC11739453 DOI: 10.1002/mco2.70056] [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: 08/18/2024] [Revised: 10/31/2024] [Accepted: 12/10/2024] [Indexed: 01/22/2025] Open
Abstract
Rapid advances in vaccine technology are becoming increasingly important in tackling global health crises caused by respiratory virus infections. While traditional vaccines, primarily administered by intramuscular injection, have proven effective, they often fail to provide the broad upper respiratory tract mucosal immunity, which is urgently needed for first-line control of respiratory viral infections. Furthermore, traditional intramuscular vaccines may not adequately address the immune escape of emerging virus variants. In contrast, respiratory mucosal vaccines developed using the body's mucosal immune response mechanism can simultaneously establish both systemic and mucosal immunity. This dual action effectively allows the respiratory mucosal immune system to function as the first line of defense, preventing infections at the entry points. This review highlights the efficacy of respiratory mucosal vaccines, including innovative delivery methods such as nasal and oral formulations, in enhancing local and systemic immune barriers. Notably, respiratory mucosal vaccines offer potential advantages in protecting against emerging virus variants and maintaining long-term and multidimensional immune memory in the upper respiratory tract. In addition, a combination of intramuscular and respiratory mucosal delivery of vaccines largely improves their coverage and effectiveness, providing valuable insights for future vaccine development and public inoculation strategies.
Collapse
Affiliation(s)
- Ming Zhou
- State Key Laboratory of Vaccines for Infectious DiseasesNational Institute of Diagnostics and Vaccine Development in Infectious DiseasesXiang An Biomedicine LaboratorySchool of Life Sciences & School of Public HealthXiamen UniversityXiamenFujianChina
| | - Haiqin Xiao
- State Key Laboratory of Vaccines for Infectious DiseasesNational Institute of Diagnostics and Vaccine Development in Infectious DiseasesXiang An Biomedicine LaboratorySchool of Life Sciences & School of Public HealthXiamen UniversityXiamenFujianChina
| | - Xinyi Yang
- State Key Laboratory of Vaccines for Infectious DiseasesNational Institute of Diagnostics and Vaccine Development in Infectious DiseasesXiang An Biomedicine LaboratorySchool of Life Sciences & School of Public HealthXiamen UniversityXiamenFujianChina
| | - Tong Cheng
- State Key Laboratory of Vaccines for Infectious DiseasesNational Institute of Diagnostics and Vaccine Development in Infectious DiseasesXiang An Biomedicine LaboratorySchool of Life Sciences & School of Public HealthXiamen UniversityXiamenFujianChina
| | - Lunzhi Yuan
- State Key Laboratory of Vaccines for Infectious DiseasesNational Institute of Diagnostics and Vaccine Development in Infectious DiseasesXiang An Biomedicine LaboratorySchool of Life Sciences & School of Public HealthXiamen UniversityXiamenFujianChina
| | - Ningshao Xia
- State Key Laboratory of Vaccines for Infectious DiseasesNational Institute of Diagnostics and Vaccine Development in Infectious DiseasesXiang An Biomedicine LaboratorySchool of Life Sciences & School of Public HealthXiamen UniversityXiamenFujianChina
| |
Collapse
|
8
|
Mambelli F, de Araujo ACVSC, Farias JP, de Andrade KQ, Ferreira LCS, Minoprio P, Leite LCC, Oliveira SC. An Update on Anti-COVID-19 Vaccines and the Challenges to Protect Against New SARS-CoV-2 Variants. Pathogens 2025; 14:23. [PMID: 39860984 PMCID: PMC11768231 DOI: 10.3390/pathogens14010023] [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: 10/04/2024] [Revised: 12/11/2024] [Accepted: 12/20/2024] [Indexed: 01/27/2025] Open
Abstract
The COVID-19 pandemic has posed a significant threat to global health systems, with extensive impacts across many sectors of society. The pandemic has been responsible for millions of deaths worldwide since its first identification in late 2019. Several actions have been taken to prevent the disease, including the unprecedented fast development and global vaccination campaigns, which were pivotal in reducing symptoms and deaths. Given the impact of the pandemic, the continuous changes of the virus, and present vaccine technologies, this review analyzes how, so far, we have met the challenge posed by the emergence of new variants and discusses how next-generation pan-coronavirus vaccines, with enhanced longevity and breadth of immune responses, may be tackled with alternative administration routes and antigen delivery platforms. By addressing these critical aspects, this review aims to contribute to the ongoing efforts to achieve long-term control of COVID-19, stimulating the discussion and work on next-generation vaccines capable of facing future waves of infection.
Collapse
Affiliation(s)
- Fábio Mambelli
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo 05508-000, Brazil; (F.M.); (A.C.V.S.C.d.A.); (K.Q.d.A.)
- Institut Pasteur de São Paulo, São Paulo 05508-020, Brazil; (L.C.S.F.); (P.M.)
| | - Ana Carolina V. S. C. de Araujo
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo 05508-000, Brazil; (F.M.); (A.C.V.S.C.d.A.); (K.Q.d.A.)
| | - Jéssica P. Farias
- Departamento de Microbiologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo 05508-000, Brazil;
| | - Kivia Q. de Andrade
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo 05508-000, Brazil; (F.M.); (A.C.V.S.C.d.A.); (K.Q.d.A.)
| | - Luis C. S. Ferreira
- Institut Pasteur de São Paulo, São Paulo 05508-020, Brazil; (L.C.S.F.); (P.M.)
- Departamento de Microbiologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo 05508-000, Brazil;
| | - Paola Minoprio
- Institut Pasteur de São Paulo, São Paulo 05508-020, Brazil; (L.C.S.F.); (P.M.)
| | - Luciana C. C. Leite
- Laboratório de Desenvolvimento de Vacinas, Instituto Butantan, São Paulo 05503-900, Brazil;
| | - Sergio C. Oliveira
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo 05508-000, Brazil; (F.M.); (A.C.V.S.C.d.A.); (K.Q.d.A.)
- Institut Pasteur de São Paulo, São Paulo 05508-020, Brazil; (L.C.S.F.); (P.M.)
| |
Collapse
|
9
|
Sirota SB, Doxey MC, Dominguez RMV, Bender RG, Vongpradith A, Albertson SB, Novotney A, Burkart K, Carter A, Abdi P, Abdoun M, Abebe AM, Abegaz KH, Aboagye RG, Abolhassani H, Abreu LG, Abualruz H, Abu-Gharbieh E, Aburuz S, Adane MM, Addo IY, Adekanmbi V, Adnani QES, Adzigbli LA, Afzal MS, Afzal S, Ahinkorah BO, Ahmad S, Ahmed A, Ahmed H, Ahmed SA, Akinosoglou K, Akkaif MA, Al Awaidy S, Alalalmeh SO, Albashtawy M, AlBataineh MT, Al-Gheethi AAS, Alhalaiqa FN, Alhassan RK, Ali A, Ali L, Ali MU, Ali SS, Ali W, Almazan JU, Alqahtani JS, Alrawashdeh A, Al-Rifai RH, Alshahrani NZ, Altartoor K, Al-Tawfiq JA, Alvis-Guzman N, Al-Worafi YM, Aly H, Aly S, Alzoubi KH, Al-Zyoud WA, Amhare AF, Amu H, Amusa GA, Anil A, Anvari S, Anyabolo EE, Arabloo J, Arafat M, Areda D, Aregawi BB, Aremu A, Athari SS, Aujayeb A, Aynalem ZB, Azadnajafabad S, Azzam AY, Badar M, Bahrami Taghanaki P, Bahramian S, Baig AA, Bajcetic M, Balakrishnan S, Banach M, Bardhan M, Barqawi HJ, Bastan MM, Batra K, Batra R, Behnoush AH, Beiranvand M, Belete AG, Belete MA, Beloukas A, Beran A, Bhardwaj P, Bhargava A, Bhat AN, Bhuiyan MA, Bitra VR, Bodunrin AO, Bogale EK, Boppana SH, et alSirota SB, Doxey MC, Dominguez RMV, Bender RG, Vongpradith A, Albertson SB, Novotney A, Burkart K, Carter A, Abdi P, Abdoun M, Abebe AM, Abegaz KH, Aboagye RG, Abolhassani H, Abreu LG, Abualruz H, Abu-Gharbieh E, Aburuz S, Adane MM, Addo IY, Adekanmbi V, Adnani QES, Adzigbli LA, Afzal MS, Afzal S, Ahinkorah BO, Ahmad S, Ahmed A, Ahmed H, Ahmed SA, Akinosoglou K, Akkaif MA, Al Awaidy S, Alalalmeh SO, Albashtawy M, AlBataineh MT, Al-Gheethi AAS, Alhalaiqa FN, Alhassan RK, Ali A, Ali L, Ali MU, Ali SS, Ali W, Almazan JU, Alqahtani JS, Alrawashdeh A, Al-Rifai RH, Alshahrani NZ, Altartoor K, Al-Tawfiq JA, Alvis-Guzman N, Al-Worafi YM, Aly H, Aly S, Alzoubi KH, Al-Zyoud WA, Amhare AF, Amu H, Amusa GA, Anil A, Anvari S, Anyabolo EE, Arabloo J, Arafat M, Areda D, Aregawi BB, Aremu A, Athari SS, Aujayeb A, Aynalem ZB, Azadnajafabad S, Azzam AY, Badar M, Bahrami Taghanaki P, Bahramian S, Baig AA, Bajcetic M, Balakrishnan S, Banach M, Bardhan M, Barqawi HJ, Bastan MM, Batra K, Batra R, Behnoush AH, Beiranvand M, Belete AG, Belete MA, Beloukas A, Beran A, Bhardwaj P, Bhargava A, Bhat AN, Bhuiyan MA, Bitra VR, Bodunrin AO, Bogale EK, Boppana SH, Borhany H, Bouaoud S, Brown CS, Buonsenso D, Bustanji Y, Cámera LA, Castañeda-Orjuela CA, Cegolon L, Cenderadewi M, Chakraborty S, Chattu VK, Cheng ETW, Chichagi F, Ching PR, Chopra H, Choudhari SG, Christopher DJ, Chu DT, Chukwu IS, Chung E, Corlateanu A, Cruz-Martins N, Dadana S, Dadras O, Dahiru T, Dai X, Das JK, Dash NR, Dashti M, Dashtkoohi M, De la Hoz FP, Debopadhaya S, Demessa BH, Demis AB, Devanbu VGC, Devegowda D, Dhama K, Dhulipala VR, Diaz D, Diaz MJ, Do TC, Do THP, Dodangeh M, Dorostkar F, Dsouza AC, Dsouza HL, Duraisamy S, Durojaiye OC, Dziedzic AM, Ed-Dra A, Ekholuenetale M, Ekundayo TC, El Sayed I, El-Dahiyat F, Elhadi M, Elshaer M, Eslami M, Eze UA, Fagbamigbe AF, Faramarzi A, Fasina FO, Ferreira N, Fischer F, Fitriana I, Flor LS, Gaihre S, Gajdács M, Galehdar N, Ganiyani MA, Gebregergis MW, Gebrehiwot M, Gebremeskel TG, Getahun GK, Getie M, Ghadiri K, Ghasemzadeh A, Ghorbani M, Goldust M, Golechha M, Goleij P, Gorini G, Goyal A, Guan SY, Guarducci G, Gudeta MD, Gupta R, Gupta S, Gupta VB, Gupta VK, Hadei M, Hadi NR, Haj-Mirzaian A, Halwani R, Hamidi S, Hammoud A, Hanifi N, Hanna F, Haq ZA, Haque MR, Hasan SMM, Hasani H, Hasnain MS, Hassankhani H, Haubold J, Hayat K, Hegazi OE, Hezam K, Holla R, Hoogar P, Horita N, Hostiuc M, Huynh HH, Ibitoye SE, Ilesanmi OS, Ilic IM, Ilic MD, Imam MT, Isa MA, Islam MR, Islam SMS, Ismail NE, Iwagami M, J V, Jafarzadeh A, Jaggi K, Jairoun AA, Jakovljevic M, Jamshidi E, Jayaram S, Jeswani BM, Jha RP, Jose J, Joseph N, Joshua CE, Jozwiak JJ, K V, Kabir Z, Kandel H, Kanmodi KK, Kant S, Kantar RS, Karaye IM, Karimi Behnagh A, Kaur N, Khajuria H, Khalaji A, Khamesipour F, Khan G, Khan MN, Khan M, Khan MJ, Kim MS, Kimokoti RW, Kochhar S, Korshunov VA, Kosen S, Krishan K, Krishna H, Krishnamoorthy V, Kuate Defo B, Kuddus MA, Kuddus M, Kuitunen I, Kulimbet M, Kumar D, Kurmi OP, Kutikuppala LVS, Lahariya C, Lal DK, Lasrado S, Latifinaibin K, Le HH, Le NHH, Le TTT, Le TDT, Lee SW, Lee WC, Li MC, Li P, Lim SS, Liu G, Liu R, Liu W, Liu X, Liu X, Lorenzovici L, Luo L, Majeed A, Malakan Rad E, Malhotra K, Malik I, Manilal A, Mehta B, Mekene Meto T, Mekonnen MM, Meles HN, Memish ZA, Mendez-Lopez MA, Meo SA, Merati M, Mestrovic T, Mettananda S, Minh LHN, Mirrakhimov EM, Misra AK, Mohamed AI, Mohamed NS, Mohammed M, Mohammed M, Mokdad AH, Monasta L, Moni MA, Moodi Ghalibaf A, Moore CE, Morawska L, Motappa R, Mougin V, Mousavi P, Mustafa G, Naghavi P, Naik GR, Nainu F, Najafi MS, Najdaghi S, Najmuldeen HHR, Nargus S, Narimani Davani D, Naser M, Natto ZS, Nayak BP, Nejadghaderi SA, Nguyen DH, Nguyen HTH, Nguyen VT, Nikolouzakis TK, Noman EA, Nri-Ezedi CA, Nuñez-Samudio V, Nwatah VE, Odetokun IA, Okekunle AP, Okonji OC, Okwute PG, Olanipekun TO, Olufadewa II, Olusanya BO, Omer GL, Onyedibe KI, Ordak M, Orish VN, Ortiz-Prado E, Otstavnov N, Ouyahia A, P A MP, Padubidri JR, Pandey A, Pantazopoulos I, Pardhan S, Parija PP, Parikh RR, Park S, Parthasarathi A, Pasovic M, Pathan AR, Patil S, Pawar S, Peprah P, Perianayagam A, Perumal D, Petcu IR, Pham HN, Pham HT, Philip AK, Pigott DM, Piracha ZZ, Poddighe D, Polibin RV, Postma MJ, Pourbabaki R, Prates EJS, Puvvula J, Qazi AS, Qian G, Rafferty Q, Rahim F, Rahimi M, Rahimi-Movaghar V, Rahman MO, Rahman M, Rahman MA, Rahmanian M, Rahmanian N, Rahmanian V, Rahmati M, Rajput P, Ramadan MM, Ramasamy SK, Ramesh PS, Rao IR, Rao M, Rao SJ, Rashedi S, Rashidi MM, Rathish D, Ravikumar N, Rawaf S, Redwan EMM, Reyes LFF, Rezaei N, Rezaei N, Rezahosseini O, Rizvi SMD, Rodriguez JAB, Ronfani L, Roudashti S, Roy P, Ruela GDA, Saddik BA, Saeb MR, Saeed U, Saeedi P, Safari M, Saheb Sharif-Askari F, Saheb Sharif-Askari N, Sahebkar A, Sahu M, Sakshaug JW, Salam N, Salami AA, Saleh MA, Sallam M, Samodra YL, Sanjeev RK, Santric-Milicevic MM, Saravanan A, Sartorius B, Sathyanarayan A, Saulam J, Saxena S, Saya GK, Schaarschmidt BM, Schumacher AE, Sedighi M, Sendekie AK, Senthilkumaran S, Sethi Y, SeyedAlinaghi S, Shafie M, Shahid S, Shaikh MA, Sham S, Shamshirgaran MA, Shanawaz M, Shannawaz M, Sharifan A, Sharifi-Rad J, Shastry RP, Sheikh A, Shigematsu M, Shiri R, Shittu A, Shiue I, Shorofi SA, Siddig EE, Simpson CR, Singh JA, Singh P, Singh S, Sinto R, Solanki R, Soliman SSM, Suleman M, Suliankatchi Abdulkader R, Swain CK, Szarpak L, Tabatabaei SM, Tabish M, Taha ZMA, Taiba J, Talaat IM, Tamuzi JL, Taye BT, Tefera YM, Temsah MH, Terefa DR, Thakur R, Thapar R, Thirunavukkarasu S, Tichopad A, Ticoalu JHV, Tovani-Palone MR, Tran NM, Tran NH, Tran Minh Duc N, Tsegay GM, Tumurkhuu M, Udoakang AJ, Upadhyay E, Vahabi SM, Vaithinathan AG, Valizadeh R, Vasankari TJ, Vinayak M, Waqas M, Weldetinsaa HL, Wickramasinghe ND, Yadollahpour A, Yaghoubi S, Yezli S, Yin D, Yon DK, Yonemoto N, Yu Y, Zakham F, Zandieh GGZ, Zare I, Zarimeidani F, Zastrozhin M, Zhai C, Zhang H, Zhang ZJ, Zhao Y, Zhou J, Zia H, Zielińska M, Zoladl M, Zyoud SH, Aravkin AY, Kassebaum NJ, Naghavi M, Vos T, Hay SI, Murray CJL, Kyu HH. Global, regional, and national burden of upper respiratory infections and otitis media, 1990-2021: a systematic analysis from the Global Burden of Disease Study 2021. THE LANCET. INFECTIOUS DISEASES 2025; 25:36-51. [PMID: 39265593 PMCID: PMC11680489 DOI: 10.1016/s1473-3099(24)00430-4] [Show More Authors] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 06/28/2024] [Accepted: 07/01/2024] [Indexed: 09/14/2024]
Abstract
BACKGROUND Upper respiratory infections (URIs) are the leading cause of acute disease incidence worldwide and contribute to a substantial health-care burden. Although acute otitis media is a common complication of URIs, the combined global burden of URIs and otitis media has not been studied comprehensively. We used results from the Global Burden of Diseases, Injuries, and Risk Factors Study 2021 to explore the fatal and non-fatal burden of the two diseases across all age groups, including a granular analysis of children younger than 5 years, in 204 countries and territories from 1990 to 2021. METHODS Mortality due to URIs and otitis media was estimated with use of vital registration and sample-based vital registration data, which are used as inputs to the Cause of Death Ensemble model to separately model URIs and otitis media mortality by age and sex. Morbidity was modelled with a Bayesian meta-regression tool using data from published studies identified via systematic reviews, population-based survey data, and cause-specific URI and otitis media mortality estimates. Additionally, we assessed and compared the burden of otitis media as it relates to URIs and examined the collective burden and contributing risk factors of both diseases. FINDINGS The global number of new episodes of URIs was 12·8 billion (95% uncertainty interval 11·4 to 14·5) for all ages across males and females in 2021. The global all-age incidence rate of URIs decreased by 10·1% (-12·0 to -8·1) from 1990 to 2019. From 2019 to 2021, the global all-age incidence rate fell by 0·5% (-0·8 to -0·1). Globally, the incidence rate of URIs was 162 484·8 per 100 000 population (144 834·0 to 183 289·4) in 2021, a decrease of 10·5% (-12·4 to -8·4) from 1990, when the incidence rate was 181 552·5 per 100 000 population (160 827·4 to 206 214·7). The highest incidence rates of URIs were seen in children younger than 2 years in 2021, and the largest number of episodes was in children aged 5-9 years. The number of new episodes of otitis media globally for all ages was 391 million (292 to 525) in 2021. The global incidence rate of otitis media was 4958·9 per 100 000 (3705·4 to 6658·6) in 2021, a decrease of 16·3% (-18·1 to -14·0) from 1990, when the incidence rate was 5925·5 per 100 000 (4371·8 to 8097·9). The incidence rate of otitis media in 2021 was highest in children younger than 2 years, and the largest number of episodes was in children aged 2-4 years. The mortality rate of URIs in 2021 was 0·2 per 100 000 (0·1 to 0·5), a decrease of 64·2% (-84·6 to -43·4) from 1990, when the mortality rate was 0·7 per 100 000 (0·2 to 1·1). In both 1990 and 2021, the mortality rate of otitis media was less than 0·1 per 100 000. Together, the combined burden accounted for by URIs and otitis media in 2021 was 6·86 million (4·24 to 10·4) years lived with disability and 8·16 million (4·99 to 12·0) disability-adjusted life-years (DALYs) for all ages across males and females. Globally, the all-age DALY rate of URIs and otitis media combined in 2021 was 103 per 100 000 (63 to 152). Infants aged 1-5 months had the highest combined DALY rate in 2021 (647 per 100 000 [189 to 1412]), followed by early neonates (aged 0-6 days; 582 per 100 000 [176 to 1297]) and late neonates (aged 7-24 days; 482 per 100 000 [161 to 1052]). INTERPRETATION The findings of this study highlight the widespread burden posed by URIs and otitis media across all age groups and both sexes. There is a continued need for surveillance, prevention, and management to better understand and reduce the burden associated with URIs and otitis media, and research is needed to assess their impacts on individuals, communities, economies, and health-care systems worldwide. FUNDING Bill & Melinda Gates Foundation.
Collapse
|
10
|
Park SC, Wiest MJ, Yan V, Wong PT, Schotsaert M. Induction of protective immune responses at respiratory mucosal sites. Hum Vaccin Immunother 2024; 20:2368288. [PMID: 38953250 PMCID: PMC11221474 DOI: 10.1080/21645515.2024.2368288] [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: 04/02/2024] [Accepted: 06/11/2024] [Indexed: 07/03/2024] Open
Abstract
Many pathogens enter the host through mucosal sites. Thus, interfering with pathogen entry through local neutralization at mucosal sites therefore is an effective strategy for preventing disease. Mucosally administered vaccines have the potential to induce protective immune responses at mucosal sites. This manuscript delves into some of the latest developments in mucosal vaccination, particularly focusing on advancements in adjuvant technologies and the role of these adjuvants in enhancing vaccine efficacy against respiratory pathogens. It highlights the anatomical and immunological complexities of the respiratory mucosal immune system, emphasizing the significance of mucosal secretory IgA and tissue-resident memory T cells in local immune responses. We further discuss the differences between immune responses induced through traditional parenteral vaccination approaches vs. mucosal administration strategies, and explore the protective advantages offered by immunization through mucosal routes.
Collapse
Affiliation(s)
- Seok-Chan Park
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Matthew J. Wiest
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
- Michigan Nanotechnology Institute for Medicine and Biological Sciences, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Vivian Yan
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Pamela T. Wong
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
- Michigan Nanotechnology Institute for Medicine and Biological Sciences, University of Michigan Medical School, Ann Arbor, MI, USA
- Mary H. Weiser Food Allergy Center, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Michael Schotsaert
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
11
|
Tang Y, Boribong BP, Swank ZN, Demokritou M, Luban MAF, Fasano A, Du M, Wolf RL, Griffiths J, Shultz J, Borberg E, Chalise S, Gonzalez WI, Walt DR, Yonker LM, Horwitz BH. COVID-19 mRNA Vaccines Induce Robust Levels of IgG but Limited Amounts of IgA Within the Oronasopharynx of Young Children. J Infect Dis 2024; 230:1390-1399. [PMID: 39253950 PMCID: PMC11646609 DOI: 10.1093/infdis/jiae450] [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: 05/02/2024] [Revised: 08/22/2024] [Accepted: 09/06/2024] [Indexed: 09/11/2024] Open
Abstract
BACKGROUND Understanding antibody responses to SARS-CoV-2 vaccination is crucial for refining COVID-19 immunization strategies. Generation of mucosal immune responses, including mucosal IgA, could be of potential benefit to vaccine efficacy; however, limited evidence exists regarding the production of mucosal antibodies following the administration of current mRNA vaccines to young children. METHODS We measured the levels of antibodies against SARS-CoV-2 from a cohort of children under 5 years of age (n = 24) undergoing SARS-CoV-2 mRNA vaccination (serially collected, matched serum and saliva samples) or in a convenience sample of children under 5 years of age presenting to pediatric emergency department (nasal swabs, n = 103). Furthermore, we assessed salivary and nasal samples for the ability to induce SARS-CoV-2 spike-mediated neutrophil extracellular traps (NET) formation. RESULTS Longitudinal analysis of post-vaccine responses in saliva revealed the induction of SARS-CoV-2-specific IgG but not IgA. Similarly, SARS-CoV-2-specific IgA was only observed in nasal samples obtained from previously infected children with or without vaccination, but not in vaccinated children without a history of infection. In addition, oronasopharyngeal samples obtained from children with prior infection were able to trigger enhanced spike-mediated NET formation, and IgA played a key role in driving this process. CONCLUSIONS Despite the induction of specific IgG in the oronasal mucosa, current intramuscular vaccines have limited ability to generate mucosal IgA in young children. These results confirm the independence of mucosal IgA responses from systemic humoral responses following mRNA vaccination and suggest potential future vaccination strategies for enhancing mucosal protection in this young age group.
Collapse
Affiliation(s)
- Ying Tang
- Division of Gastroenterology, Hepatology, and Nutrition, Boston Children's Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Brittany P Boribong
- Harvard Medical School, Boston, Massachusetts, USA
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Zoe N Swank
- Harvard Medical School, Boston, Massachusetts, USA
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts, USA
| | - Melina Demokritou
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Maria A F Luban
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Alessio Fasano
- Harvard Medical School, Boston, Massachusetts, USA
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Michelle Du
- Division of Emergency Medicine, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Rebecca L Wolf
- Division of Emergency Medicine, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Joseph Griffiths
- Division of Emergency Medicine, Boston Children's Hospital, Boston, Massachusetts, USA
| | - John Shultz
- Division of Emergency Medicine, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Ella Borberg
- Harvard Medical School, Boston, Massachusetts, USA
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts, USA
| | - Sujata Chalise
- Harvard Medical School, Boston, Massachusetts, USA
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts, USA
| | - Wanda I Gonzalez
- Harvard Medical School, Boston, Massachusetts, USA
- Department of Pediatrics, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - David R Walt
- Harvard Medical School, Boston, Massachusetts, USA
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts, USA
| | - Lael M Yonker
- Harvard Medical School, Boston, Massachusetts, USA
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Bruce H Horwitz
- Division of Gastroenterology, Hepatology, and Nutrition, Boston Children's Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
- Division of Emergency Medicine, Boston Children's Hospital, Boston, Massachusetts, USA
| |
Collapse
|
12
|
Lykins WR, Pollet J, White JA, Keegan B, Versteeg L, Strych U, Chen WH, Mohamath R, Ramer-Denisoff G, Reed S, Renshaw C, Beaver S, Gerhardt A, Voigt EA, Tomai MA, Sitrin R, Choy RKM, Cassels FJ, Hotez PJ, Bottazzi ME, Fox CB. Optimizing immunogenicity and product presentation of a SARS-CoV-2 subunit vaccine composition: effects of delivery route, heterologous regimens with self-amplifying RNA vaccines, and lyophilization. Front Immunol 2024; 15:1480976. [PMID: 39737197 PMCID: PMC11683073 DOI: 10.3389/fimmu.2024.1480976] [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: 08/14/2024] [Accepted: 11/25/2024] [Indexed: 01/01/2025] Open
Abstract
Introduction Dozens of vaccines have been approved or authorized internationally in response to the ongoing SARS-CoV-2 pandemic, covering a range of modalities and routes of delivery. For example, mucosal delivery of vaccines via the intranasal (i.n.) route has been shown to improve protective mucosal responses in comparison to intramuscular (i.m.) delivery. As we gain knowledge of the limitations of existing vaccines, it is of interest to understand if changes in product presentation or combinations of multiple vaccine modalities can further improve immunological outcomes. Methods We investigated a commercial-stage SARS-CoV-2 receptor binding domain (RBD) antigen adjuvanted with a clinical-stage TLR-7/8 agonist (3M-052) formulated on aluminum oxyhydroxide (Alum). In a murine immunogenicity model, we compared i.n. and i.m. dosing of the RBD-3M-052-Alum vaccine. We measured the magnitude of antibody responses in serum and lungs, the antibody-secreting cell populations in bone marrow, and antigen-specific cytokine-secreting splenocyte populations. Similarly, we compared different heterologous and homologous prime-boost regimens using the RBD-3M-052-Alum vaccine and a clinical-stage self-amplifying RNA (saRNA) vaccine formulated on a nanostructured lipid carrier (NLC) using the i.m. route alone. Finally, we developed a lyophilized presentation of the RBD-3M-052-Alum vaccine and compared it to the liquid presentation and a heterologous regimen including a previously characterized lyophilized form of the saRNA-NLC vaccine. Results and discussion We demonstrate that i.n. dosing of the RBD-3M-052-Alum vaccine increased IgA titers in the lung by more than 1.5 logs, but induced serum IgG titers 0.8 logs lower, in comparison to i.m. dosing of the same vaccine. We also show that the homologous prime-boost RBD-3M-052-Alum regimen led to the highest serum IgG and bronchial IgA titers, whereas the homologous saRNA-NLC regimen led to the highest splenocyte interferon-γ response. We found that priming with the saRNA-NLC vaccine and boosting with the RBD-3M-052-Alum vaccine led to the most desirable immune outcome of all regimens tested. Finally, we show that the lyophilized RBD-3M-052-Alum vaccine retained its immunological characteristics. Our results demonstrate that the route of delivery and the use of heterologous regimens each separately impacts the resulting immune profile, and confirm that multi-product vaccine regimens can be developed with stabilized presentations in mind.
Collapse
MESH Headings
- Animals
- COVID-19 Vaccines/immunology
- COVID-19 Vaccines/administration & dosage
- SARS-CoV-2/immunology
- COVID-19/prevention & control
- COVID-19/immunology
- Mice
- Vaccines, Subunit/immunology
- Vaccines, Subunit/administration & dosage
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- Female
- Immunogenicity, Vaccine
- Administration, Intranasal
- Freeze Drying
- Antibodies, Neutralizing/blood
- Antibodies, Neutralizing/immunology
- Adjuvants, Vaccine
- mRNA Vaccines/immunology
- Mice, Inbred BALB C
- Adjuvants, Immunologic/administration & dosage
- Spike Glycoprotein, Coronavirus/immunology
- Injections, Intramuscular
- Humans
Collapse
Affiliation(s)
| | - Jeroen Pollet
- Texas Children’s Hospital Center for Vaccine Development, Baylor College of Medicine, Houston, TX, United States
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, United States
| | | | - Brian Keegan
- Texas Children’s Hospital Center for Vaccine Development, Baylor College of Medicine, Houston, TX, United States
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, United States
| | - Leroy Versteeg
- Texas Children’s Hospital Center for Vaccine Development, Baylor College of Medicine, Houston, TX, United States
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, United States
| | - Ulrich Strych
- Texas Children’s Hospital Center for Vaccine Development, Baylor College of Medicine, Houston, TX, United States
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, United States
| | - Wen-Hsiang Chen
- Texas Children’s Hospital Center for Vaccine Development, Baylor College of Medicine, Houston, TX, United States
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, United States
| | - Raodoh Mohamath
- Access to Advanced Health Institute, Seattle, WA, United States
| | | | - Sierra Reed
- Access to Advanced Health Institute, Seattle, WA, United States
| | | | - Samuel Beaver
- Access to Advanced Health Institute, Seattle, WA, United States
| | - Alana Gerhardt
- Access to Advanced Health Institute, Seattle, WA, United States
| | - Emily A. Voigt
- Access to Advanced Health Institute, Seattle, WA, United States
| | | | | | | | | | - Peter J. Hotez
- Texas Children’s Hospital Center for Vaccine Development, Baylor College of Medicine, Houston, TX, United States
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, United States
- Department of Biology, Baylor University, Waco, TX, United States
| | - Maria Elena Bottazzi
- Texas Children’s Hospital Center for Vaccine Development, Baylor College of Medicine, Houston, TX, United States
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, United States
| | - Christopher B. Fox
- Access to Advanced Health Institute, Seattle, WA, United States
- Department of Global Health, University of Washington, Seattle, WA, United States
| |
Collapse
|
13
|
Astakhova EA, Baranov KO, Shilova NV, Polyakova SM, Zuev EV, Poteryaev DA, Taranin AV, Filatov AV. Antibody Avidity Maturation Following Booster Vaccination with an Intranasal Adenovirus Salnavac Vaccine. Vaccines (Basel) 2024; 12:1362. [PMID: 39772024 PMCID: PMC11680177 DOI: 10.3390/vaccines12121362] [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: 10/12/2024] [Revised: 11/14/2024] [Accepted: 11/19/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND The COVID-19 pandemic has led to the rapid development of new vaccines and methods of testing vaccine-induced immunity. Despite the extensive research that has been conducted on the level of specific antibodies, less attention has been paid to studying the avidity of these antibodies. The avidity of serum antibodies is associated with a vaccine showing high effectiveness and reflects the process of affinity maturation. In the context of vaccines against SARS-CoV-2, only a limited number of studies have investigated the avidity of antibodies, often solely focusing on the wild-type virus following vaccination. This study provides new insights into the avidity of serum antibodies following adenovirus-based boosters. We focused on the effects of an intranasal Salnavac booster, which is compared, using a single analytical platform, to an intramuscular Sputnik V. METHODS The avidity of RBD-specific IgGs and IgAs was investigated through ELISA using urea and biolayer interferometry. RESULTS The results demonstrated the similar avidities of serum antibodies, which were induced by both vaccines for six months post-booster. However, an increase in antibody avidity was observed for the wild-type and Delta variants, but not for the BA.4/5 variant. CONCLUSIONS Collectively, our data provide the insights into antibody avidity maturation after the adenovirus-based vaccines against SARS-CoV-2.
Collapse
Affiliation(s)
- Ekaterina A. Astakhova
- Laboratory of Immunochemistry, National Research Center Institute of Immunology, Federal Medical Biological Agency of Russia, 115522 Moscow, Russia;
- Department of Immunology, Faculty of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia
- Moscow Center for Advanced Studies, Kulakova Street 20, 123592 Moscow, Russia
| | - Konstantin O. Baranov
- Laboratory of Immunogenetics, Institute of Molecular and Cellular Biology, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia (A.V.T.)
| | - Nadezhda V. Shilova
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - Svetlana M. Polyakova
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | | | | | - Alexander V. Taranin
- Laboratory of Immunogenetics, Institute of Molecular and Cellular Biology, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia (A.V.T.)
| | - Alexander V. Filatov
- Laboratory of Immunochemistry, National Research Center Institute of Immunology, Federal Medical Biological Agency of Russia, 115522 Moscow, Russia;
- Department of Immunology, Faculty of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia
| |
Collapse
|
14
|
Plans-Rubió P. What COVID-19 Vaccination Strategy Should Be Implemented and Which Vaccines Should Be Used in the Post-Pandemic Era? Vaccines (Basel) 2024; 12:1180. [PMID: 39460346 PMCID: PMC11511555 DOI: 10.3390/vaccines12101180] [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: 09/19/2024] [Revised: 10/11/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024] Open
Abstract
COVID-19 vaccines have reduced the negative health and economic impact of the COVID-19 pandemic by preventing severe disease, hospitalizations and deaths. In the new socio-economic normality, the COVID-19 vaccination strategy can be universal or high-risk and seasonal or not seasonal, and different vaccines can be used. The universal vaccination strategy can achieve greater health and herd immunity effects and is associated with greater costs than the high-risk vaccination strategy. In each country, the optimal COVID-19 vaccination strategy must be decided by considering the advantages and disadvantages and assessing the costs, health effects and cost-effectiveness of the universal and high-risk vaccination strategies. The universal vaccination strategy should be implemented when the objective of the vaccination program is to achieve the greatest health benefits from COVID-19 vaccination and when its incremental cost-effectiveness ratio is lower than EUR 30,000-50,000 per QALY or LYG. The use of adapted vaccines targeting currently circulating variants of SARS-CoV-2 is necessary to avoid the immune escape of emerging variants.
Collapse
|
15
|
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: 0] [Impact Index Per Article: 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
|
16
|
O’Reilly S, Byrne J, Feeney ER, Mallon PWG, Gautier V. Navigating the Landscape of B Cell Mediated Immunity and Antibody Monitoring in SARS-CoV-2 Vaccine Efficacy: Tools, Strategies and Clinical Trial Insights. Vaccines (Basel) 2024; 12:1089. [PMID: 39460256 PMCID: PMC11511438 DOI: 10.3390/vaccines12101089] [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: 07/31/2024] [Revised: 09/16/2024] [Accepted: 09/16/2024] [Indexed: 10/28/2024] Open
Abstract
Correlates of Protection (CoP) are biomarkers above a defined threshold that can replace clinical outcomes as primary endpoints, predicting vaccine effectiveness to support the approval of new vaccines or follow up studies. In the context of COVID-19 vaccination, CoPs can help address challenges such as demonstrating vaccine effectiveness in special populations, against emerging SARS-CoV-2 variants or determining the durability of vaccine-elicited immunity. While anti-spike IgG titres and viral neutralising capacity have been characterised as CoPs for COVID-19 vaccination, the contribution of other components of the humoral immune response to immediate and long-term protective immunity is less well characterised. This review examines the evidence supporting the use of CoPs in COVID-19 clinical vaccine trials, and how they can be used to define a protective threshold of immunity. It also highlights alternative humoral immune biomarkers, including Fc effector function, mucosal immunity, and the generation of long-lived plasma and memory B cells and discuss how these can be applied to clinical studies and the tools available to study them.
Collapse
Affiliation(s)
- Sophie O’Reilly
- Centre for Experimental Pathogen Host Research (CEPHR), University College Dublin, Belfield, Dublin 4, Ireland
- School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
| | - Joanne Byrne
- Centre for Experimental Pathogen Host Research (CEPHR), University College Dublin, Belfield, Dublin 4, Ireland
- School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| | - Eoin R. Feeney
- Centre for Experimental Pathogen Host Research (CEPHR), University College Dublin, Belfield, Dublin 4, Ireland
- School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
- Department of Infectious Diseases, St Vincent’s University Hospital, Elm Park, Dublin 4, Ireland
| | - Patrick W. G. Mallon
- Centre for Experimental Pathogen Host Research (CEPHR), University College Dublin, Belfield, Dublin 4, Ireland
- School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
- Department of Infectious Diseases, St Vincent’s University Hospital, Elm Park, Dublin 4, Ireland
| | - Virginie Gautier
- Centre for Experimental Pathogen Host Research (CEPHR), University College Dublin, Belfield, Dublin 4, Ireland
- School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
| |
Collapse
|
17
|
Rai CI, Kuo TH, Chen YC. Novel Administration Routes, Delivery Vectors, and Application of Vaccines Based on Biotechnologies: A Review. Vaccines (Basel) 2024; 12:1002. [PMID: 39340032 PMCID: PMC11436249 DOI: 10.3390/vaccines12091002] [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: 07/25/2024] [Revised: 08/22/2024] [Accepted: 08/29/2024] [Indexed: 09/30/2024] Open
Abstract
Traditional vaccines can be classified into inactivated vaccines, live attenuated vaccines, and subunit vaccines given orally or via intramuscular (IM) injection or subcutaneous (SC) injection for the prevention of infectious diseases. Recently, recombinant protein vaccines, DNA vaccines, mRNA vaccines, and multiple/alternative administering route vaccines (e.g., microneedle or inhalation) have been developed to make vaccines more secure, effective, tolerable, and universal for the public. In addition to preventing infectious diseases, novel vaccines have currently been developed or are being developed to prevent or cure noninfectious diseases, including cancer. These vaccine platforms have been developed using various biotechnologies such as viral vectors, nanoparticles, mRNA, recombination DNA, subunit, novel adjuvants, and other vaccine delivery systems. In this review, we will explore the development of novel vaccines applying biotechnologies, such as vaccines based on novel administration routes, vaccines based on novel vectors, including viruses and nanoparticles, vaccines applied for cancer prevention, and therapeutic vaccines.
Collapse
Affiliation(s)
- Chung-I Rai
- Department of Cosmetic Science, Vanung University, 1, Van Nung Road, Chung-Li City 320676, Taiwan;
| | - Tsu-Hsiang Kuo
- Department of Rehabilitation Science, Jenteh Junior College of Medicine, Nursing and Management, Miaoli County 356006, Taiwan;
- Department of Biotechnology and Pharmaceutical Management, Jenteh Junior College of Medicine, Nursing and Management, Miaoli County 356006, Taiwan
| | - Yuan-Chuan Chen
- Department of Nursing, Jenteh Junior College of Medicine, Nursing and Management, Miaoli County 356006, Taiwan
- Department of Medical Technology, Jenteh Junior College of Medicine, Nursing and Management, Miaoli County 356006, Taiwan
- Program in Comparative Biochemistry, University of California, Berkeley, CA 94720, USA
| |
Collapse
|
18
|
Yamamoto S, Oshiro Y, Inamura N, Nemoto T, Tan T, Horii K, Okudera K, Konishi M, Mizoue T, Sugiyama H, Aoyanagi N, Sugiura W, Ohmagari N. Correlates of Nucleocapsid Antibodies and a Combination of Spike and Nucleocapsid Antibodies Against Protection of SARS-CoV-2 Infection During the Omicron XBB.1.16/EG.5-Predominant Wave. Open Forum Infect Dis 2024; 11:ofae455. [PMID: 39220657 PMCID: PMC11363870 DOI: 10.1093/ofid/ofae455] [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: 04/02/2024] [Accepted: 08/09/2024] [Indexed: 09/04/2024] Open
Abstract
Background We aimed to examine the association among nucleocapsid (N) antibodies, a combination of N and spike (S) antibodies, and protection against SARS-CoV-2 reinfection. Methods We conducted a prospective cohort study among staff at a national medical research center in Tokyo and followed them for the incidence of SARS-CoV-2 infection between June and September 2023 (Omicron XBB.1.16/EG.5 wave). At baseline, participants donated blood samples to measure N- and S-specific antibodies. Cox regression was used to estimate the hazard ratio and protection ([1 - hazard ratio] × 100) against subsequent SARS-CoV-2 infection across these antibody levels. Results Among participants with previous infection, higher pre-reinfection N antibodies were associated with a lower risk of reinfection, even after adjusting S antibody levels (P < .01 for trend). Estimation of the protection matrix for N and S antibodies revealed that high levels in N and S antibodies conferred robust protection (>90%) against subsequent infection. In addition, a pattern of low pre-reinfection N antibodies but high vaccine-enhanced S antibodies showed high protection (>80%). Conclusions Pre-reinfection N antibody levels correlated with protection against reinfection, independent of S antibodies. If the N antibodies were low, vaccine-boosted S antibodies might enhance the reinfection protection.
Collapse
Affiliation(s)
- Shohei Yamamoto
- Department of Epidemiology and Prevention, Center for Clinical Sciences, National Center for Global Health and Medicine, Tokyo, Japan
| | - Yusuke Oshiro
- Department of Laboratory Testing, Center Hospital of the National Center for the Global Health and Medicine, Tokyo, Japan
| | - Natsumi Inamura
- Department of Laboratory Testing, Center Hospital of the National Center for the Global Health and Medicine, Tokyo, Japan
| | - Takashi Nemoto
- Department of Laboratory Testing, Center Hospital of the National Center for the Global Health and Medicine, Tokyo, Japan
| | - Tomofumi Tan
- Department of Laboratory Testing, Center Hospital of the National Center for the Global Health and Medicine, Tokyo, Japan
| | - Kumi Horii
- Infection Control Office, Center Hospital of the National Center for the Global Health and Medicine, Tokyo, Japan
| | - Kaori Okudera
- Infection Control Office, Kohnodai Hospital of the National Center for the Global Health and Medicine, Chiba, Japan
| | - Maki Konishi
- Department of Epidemiology and Prevention, Center for Clinical Sciences, National Center for Global Health and Medicine, Tokyo, Japan
| | - Tetsuya Mizoue
- Department of Epidemiology and Prevention, Center for Clinical Sciences, National Center for Global Health and Medicine, Tokyo, Japan
| | - Haruhito Sugiyama
- Center Hospital of the National Center for the Global Health and Medicine, Tokyo, Japan
| | - Nobuyoshi Aoyanagi
- Kohnodai Hospital of the National Center for the Global Health and Medicine, Chiba, Japan
| | - Wataru Sugiura
- Center for Clinical Sciences, National Center for Global Health and Medicine, Tokyo, Japan
| | - Norio Ohmagari
- Disease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo, Japan
| |
Collapse
|
19
|
Han JP, Nam YR, Chung HY, Lee H, Yeom SC. Polyphenol-Enabled 2D Nanopatch for Enhanced Nasal Mucoadhesion and Immune Activation. NANO LETTERS 2024; 24:10380-10387. [PMID: 39120059 DOI: 10.1021/acs.nanolett.4c03228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/10/2024]
Abstract
The advancement of effective nasal mucoadhesive delivery faces challenges due to rapid mucociliary clearance (MCC). Conventional studies have employed mucoadhesive materials, mainly forming spherical nanoparticles, but these offer limited adhesion to the nasal mucosa. This study hypothesizes that a 2D nanoscale structure utilizing adhesive polyphenols can provide a superior strategy for countering MCC, aligning with the planar mucosal layers. We explore the use of tannic acid (TA), a polyphenolic molecule known for its adhesive properties and ability to form complexes with biomolecules. Our study introduces an unprecedented 2D nanopatch, assembled through the interaction of TA with green fluorescent protein (GFP), and cell-penetrating peptide (CPP). This 2D nanopatch demonstrates robust adhesion to nasal mucosa and significantly enhances immunoglobulin A secretions, suggesting its potential for enhancing nasal vaccine delivery. The promise of a polyphenol-enabled adhesive 2D nanopatch signifies a pivotal shift from conventional spherical nanoparticles, opening new pathways for delivery strategies through respiratory mucoadhesion.
Collapse
Affiliation(s)
- Jeong Pil Han
- Graduate School of International Agricultural Technology and Institute of Green Bio Science and Technology, Seoul National University, Pyeongchang, Gangwon 25354, Republic of Korea
| | - Yu Ri Nam
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Hye Yoon Chung
- Graduate School of International Agricultural Technology and Institute of Green Bio Science and Technology, Seoul National University, Pyeongchang, Gangwon 25354, Republic of Korea
| | - Haeshin Lee
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Su Cheong Yeom
- Graduate School of International Agricultural Technology and Institute of Green Bio Science and Technology, Seoul National University, Pyeongchang, Gangwon 25354, Republic of Korea
| |
Collapse
|
20
|
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
|
21
|
Edo GI, Yousif E, Al-Mashhadani MH. Modified chitosan: Insight on biomedical and industrial applications. Int J Biol Macromol 2024; 275:133526. [PMID: 38960250 DOI: 10.1016/j.ijbiomac.2024.133526] [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: 04/22/2024] [Revised: 06/22/2024] [Accepted: 06/27/2024] [Indexed: 07/05/2024]
Abstract
Chitosan (CS), a by -product of chitin deacetylation can be useful in a broad range of purposes, to mention agriculture, pharmaceuticals, material science, food and nutrition, biotechnology and of recent, in gene therapy. Chitosan is a highly desired biomolecule due to the existence of many sensitive functional groups inside the molecule and also because of its net cationicity. The latter provides flexibility for creating a wide range of derivatives for particular end users across various industries. This overview aims to compile some of the most recent research on the bio-related applications that chitosan and its derivatives can be used for. However, chitosan's reactive functional groups are amendable to chemical reaction. Modifying the material to show enhanced solubility, a greater range of application options and pH-sensitive targeting and others have been a major focus of chitosan research. This review describes the modifications of chitosan that have been made to improve its water solubility, pH sensitivity, and capacity to target chitosan derivatives. Applying the by-products of chitosan as antibacterial, in targeting, extended release and as delivery systems is also covered. The by-products of chitosan will be important and potentially useful in developing new biomedical drugs in time to come.
Collapse
Affiliation(s)
- Great Iruoghene Edo
- Department of Chemistry, College of Science, Al-Nahrain University, Baghdad, Iraq.
| | - Emad Yousif
- Department of Chemistry, College of Science, Al-Nahrain University, Baghdad, Iraq
| | | |
Collapse
|
22
|
Sallard E, Aydin M. Cutting-edge research frontiers in oral cavity vaccines for respiratory diseases: a roadmap for scientific advancement. Front Cell Infect Microbiol 2024; 14:1388222. [PMID: 38988815 PMCID: PMC11234472 DOI: 10.3389/fcimb.2024.1388222] [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: 02/19/2024] [Accepted: 05/13/2024] [Indexed: 07/12/2024] Open
Abstract
Intramuscular vaccines present limitations in eliciting robust mucosal immunity and preventing respiratory pathogens transmission. Sublingual vaccine administration offers promising advantages, including interconnected mucosal protection. Despite these advantages, only a few clinical trials have explored sublingual vaccines, underscoring the necessity of optimizing next-generation vaccine formulas. Critical research priorities include understanding vector behavior in the oral environment, understanding their interactions with mucosal immunity and developing formulations enabling sustained mucosal contact to facilitate efficient transduction. Consequently, tonsil organoids, as representative human mucosal models, could offer critical insights into sublingual immunization. Thus, a multi-disciplinary approach integrating pharmacological, immunological, and manufacturing considerations is pivotal for sublingual vaccines in targeting pathogen-aggravated prevalent respiratory diseases including asthma, COPD and lung cancer, as well as the antimicrobial resistance crisis.
Collapse
Affiliation(s)
- Erwan Sallard
- Virology and Microbiology, Center for Biomedical Education and Research (ZBAF), School of Medicine, Faculty of Health, Witten/Herdecke University, Witten, Germany
| | - Malik Aydin
- Virology and Microbiology, Center for Biomedical Education and Research (ZBAF), School of Medicine, Faculty of Health, Witten/Herdecke University, Witten, Germany
- Laboratory of Experimental Pediatric Pneumology and Allergology, Center for Biomedical Education and Research, School of Life Sciences (ZBAF), Faculty of Health, Witten/Herdecke University, Witten, Germany
- Institute for Medical Laboratory Diagnostics, Center for Clinical and Translational Research, Helios University Hospital Wuppertal, Witten/Herdecke University, Wuppertal, Germany
| |
Collapse
|
23
|
Althaus T, Overton CE, Devaux I, House T, Lapouze A, Troel A, Vanzo B, Laroche M, Bordero A, Jorgensen P, Pebody R, Voiglio EJ. How effective is the BNT162b2 mRNA vaccine against SARS-CoV-2 transmission and infection? A national programme analysis in Monaco, July 2021 to September 2022. BMC Med 2024; 22:227. [PMID: 38840159 PMCID: PMC11155114 DOI: 10.1186/s12916-024-03444-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 05/24/2024] [Indexed: 06/07/2024] Open
Abstract
BACKGROUND We quantified SARS-CoV-2 dynamics in different community settings and the direct and indirect effect of the BNT162b2 mRNA vaccine in Monaco for different variants of concern (VOC). METHODS Between July 2021 and September 2022, we prospectively investigated 20,443 contacts from 6320 index cases using data from the Monaco COVID-19 Public Health Programme. We calculated secondary attack rates (SARs) in households (n = 13,877), schools (n = 2508) and occupational (n = 6499) settings. We used binomial regression with a complementary log-log link function to measure adjusted hazard ratios (aHR) and vaccine effectiveness (aVE) for index cases to infect contacts and contacts to be infected in households. RESULTS In households, the SAR was 55% (95% CI 54-57) and 50% (48-51) among unvaccinated and vaccinated contacts, respectively. The SAR was 32% (28-36) and 12% (10-13) in workplaces, and 7% (6-9) and 6% (3-10) in schools, among unvaccinated and vaccinated contacts respectively. In household, the aHR was lower in contacts than in index cases (aHR 0.68 [0.55-0.83] and 0.93 [0.74-1.1] for delta; aHR 0.73 [0.66-0.81] and 0.89 [0.80-0.99] for omicron BA.1&2, respectively). Vaccination had no significant effect on either direct or indirect aVE for omicron BA.4&5. The direct aVE in contacts was 32% (17, 45) and 27% (19, 34), and for index cases the indirect aVE was 7% (- 17, 26) and 11% (1, 20) for delta and omicron BA.1&2, respectively. The greatest aVE was in contacts with a previous SARS-CoV-2 infection and a single vaccine dose during the omicron BA.1&2 period (45% [27, 59]), while the lowest were found in contacts with either three vaccine doses (aVE - 24% [- 63, 6]) or one single dose and a previous SARS-CoV-2 infection (aVE - 36% [- 198, 38]) during the omicron BA.4&5 period. CONCLUSIONS Protection conferred by the BNT162b2 mRNA vaccine against transmission and infection was low for delta and omicron BA.1&2, regardless of the number of vaccine doses and previous SARS-CoV-2 infection. There was no significant vaccine effect for omicron BA.4&5. Health authorities carrying out vaccination campaigns should bear in mind that the current generation of COVID-19 vaccines may not represent an effective tool in protecting individuals from either transmitting or acquiring SARS-CoV-2 infection.
Collapse
Affiliation(s)
| | - Christopher E Overton
- Department of Mathematical Sciences, University of Liverpool, Liverpool, UK
- United Kingdom Health Security Agency, London, UK
| | - Isabelle Devaux
- World Health Organization Regional Office for Europe, Copenhagen, Denmark
| | - Thomas House
- Department of Mathematics, University of Manchester, Manchester, UK
| | | | | | | | | | | | - Pernille Jorgensen
- World Health Organization Regional Office for Europe, Copenhagen, Denmark
| | - Richard Pebody
- World Health Organization Regional Office for Europe, Copenhagen, Denmark
| | | |
Collapse
|
24
|
Li J, Hsu KS, Howe SE, Hoang T, Xia Z, Berzofsky JA, Sui Y. Sex-biased immunogenicity of a mucosal subunit vaccine against SARS-CoV-2 in mice. Front Immunol 2024; 15:1386243. [PMID: 38835757 PMCID: PMC11148259 DOI: 10.3389/fimmu.2024.1386243] [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: 02/14/2024] [Accepted: 05/06/2024] [Indexed: 06/06/2024] Open
Abstract
Introduction Current vaccines against COVID-19 administered via parenteral route have limited ability to induce mucosal immunity. There is a need for an effective mucosal vaccine to combat SARS-CoV-2 virus replication in the respiratory mucosa. Moreover, sex differences are known to affect systemic antibody responses against vaccines. However, their role in mucosal cellular responses against a vaccine remains unclear and is underappreciated. Methods We evaluated the mucosal immunogenicity of a booster vaccine regimen that is recombinant protein-based and administered intranasally in mice to explore sex differences in mucosal humoral and cellular responses. Results Our results showed that vaccinated mice elicited strong systemic antibody (Ab), nasal, and bronchiole alveolar lavage (BAL) IgA responses, and local T cell immune responses in the lung in a sex-biased manner irrespective of mouse genetic background. Monocytes, alveolar macrophages, and CD103+ resident dendritic cells (DCs) in the lungs are correlated with robust mucosal Ab and T cell responses induced by the mucosal vaccine. Discussion Our findings provide novel insights into optimizing next-generation booster vaccines against SARS-CoV-2 by inducing spike-specific lung T cell responses, as well as optimizing mucosal immunity for other respiratory infections, and a rationale for considering sex differences in future vaccine research and vaccination practice.
Collapse
MESH Headings
- Animals
- Female
- Mice
- SARS-CoV-2/immunology
- COVID-19 Vaccines/immunology
- COVID-19/prevention & control
- COVID-19/immunology
- COVID-19/virology
- Vaccines, Subunit/immunology
- Vaccines, Subunit/administration & dosage
- Male
- Immunity, Mucosal
- Immunogenicity, Vaccine
- Antibodies, Viral/immunology
- Antibodies, Viral/blood
- Lung/immunology
- Lung/virology
- T-Lymphocytes/immunology
- Spike Glycoprotein, Coronavirus/immunology
- Mice, Inbred C57BL
- Administration, Intranasal
- Sex Factors
- Immunoglobulin A/immunology
- Dendritic Cells/immunology
- Immunization, Secondary
- Immunity, Humoral
Collapse
Affiliation(s)
- Jianping Li
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Kevin S Hsu
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Savannah E Howe
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Tanya Hoang
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Zheng Xia
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Jay A Berzofsky
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Yongjun Sui
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD, United States
| |
Collapse
|
25
|
Ongun M, Lokras AG, Baghel S, Shi Z, Schmidt ST, Franzyk H, Rades T, Sebastiani F, Thakur A, Foged C. Lipid nanoparticles for local delivery of mRNA to the respiratory tract: Effect of PEG-lipid content and administration route. Eur J Pharm Biopharm 2024; 198:114266. [PMID: 38499255 DOI: 10.1016/j.ejpb.2024.114266] [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: 12/18/2023] [Revised: 03/11/2024] [Accepted: 03/13/2024] [Indexed: 03/20/2024]
Abstract
Design of inhalable mRNA therapeutics is promising because local administration in the respiratory tract is minimally invasive and induces a local response. However, several challenges related to administration via inhalation and respiratory tract barriers have so far prevented the progress of inhaled mRNA therapeutics. Here, we investigated factors of importance for lipid nanoparticle (LNP)-mediated delivery of mRNA to the respiratory tract. We hypothesized that: (i) the PEG-lipid content is important for providing colloidal stability during aerosolization and for mucosal delivery, (ii) the PEG-lipid contentinfluences the expression of mRNA-encoded protein in the lungs, and (iii) the route of administration (nasal versus pulmonary) affects mRNA delivery in the lungs. In this study, we aimed to optimize the PEG-lipid content for mucosal delivery and to investigatethe effect of administration route on the kinetics of protein expression. Our results show that increasing the PEG-lipid content improves the colloidal stability during the aerosolization process, but has a negative impact on the transfection efficiencyin vitro. The kinetics of protein expressionin vivois dependent on the route of administration, and we found that pulmonaryadministration of mRNA-LNPs to mice results inmore durable protein expression than nasaladministration. These results demonstrate that the design of the delivery system and the route of administration are importantfor achieving high mRNA transfection efficiency in the respiratory tract.
Collapse
Affiliation(s)
- Melike Ongun
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen Ø, Denmark
| | - Abhijeet Girish Lokras
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen Ø, Denmark
| | - Saahil Baghel
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen Ø, Denmark
| | - Zhenning Shi
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen Ø, Denmark
| | - Signe Tandrup Schmidt
- Department of Infectious Disease Immunology, Statens Serum Institut, Artillerivej 5, 2300 Copenhagen S, Denmark
| | - Henrik Franzyk
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Jagtvej 162, DK-2100 Copenhagen Ø, Denmark
| | - Thomas Rades
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen Ø, Denmark
| | - Federica Sebastiani
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen Ø, Denmark; Division of Physical Chemistry, Department of Chemistry, Lund University, 22100 Lund, Sweden
| | - Aneesh Thakur
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen Ø, Denmark
| | - Camilla Foged
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen Ø, Denmark.
| |
Collapse
|
26
|
Yahalom-Ronen Y, Melamed S, Politi B, Erez N, Tamir H, Bar-On L, Ryvkin J, Leshkowitz D, Israeli O, Weiss S, Ben-Shmuel A, Barlev-Gross M, Cherry Mimran L, Achdout H, Paran N, Israely T. Induction of Superior Systemic and Mucosal Protective Immunity to SARS-CoV-2 by Nasal Administration of a VSV-ΔG-Spike Vaccine. Vaccines (Basel) 2024; 12:491. [PMID: 38793742 PMCID: PMC11125831 DOI: 10.3390/vaccines12050491] [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: 03/20/2024] [Revised: 04/21/2024] [Accepted: 04/25/2024] [Indexed: 05/26/2024] Open
Abstract
The emergence of rapidly spreading variants of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) poses a major challenge to vaccines' protective efficacy. Intramuscular (IM) vaccine administration induces short-lived immunity but does not prevent infection and transmission. New vaccination strategies are needed to extend the longevity of vaccine protection, induce mucosal and systemic immunity and prevent viral transmission. The intranasal (IN) administration of the VSV-ΔG-spike vaccine candidate directly to mucosal surfaces yielded superior mucosal and systemic immunity at lower vaccine doses. Compared to IM vaccination in the K18-hACE2 model, IN vaccination preferentially induced mucosal IgA and T-cells, reduced the viral load at the site of infection, and ameliorated disease-associated brain gene expression. IN vaccination was protective even one year after administration. As most of the world population has been vaccinated by IM injection, we demonstrate the potential of a heterologous IM + IN vaccination regimen to induce mucosal immunity while maintaining systemic immunity. Furthermore, the IM + IN regimen prevented virus transmission in a golden Syrian hamster co-caging model. Taken together, we show that IN vaccination with VSV-ΔG-spike, either as a homologous IN + IN regimen or as a boost following IM vaccination, has a favorable potential over IM vaccination in inducing efficient mucosal immunity, long-term protection and preventing virus transmission.
Collapse
Affiliation(s)
- Yfat Yahalom-Ronen
- Department of Infectious Diseases, Israel Institute for Biological Research, Ness Ziona 74100, Israel; (Y.Y.-R.); (S.M.); (B.P.); (N.E.); (H.T.); (S.W.); (A.B.-S.); (M.B.-G.); (L.C.M.); (H.A.)
| | - Sharon Melamed
- Department of Infectious Diseases, Israel Institute for Biological Research, Ness Ziona 74100, Israel; (Y.Y.-R.); (S.M.); (B.P.); (N.E.); (H.T.); (S.W.); (A.B.-S.); (M.B.-G.); (L.C.M.); (H.A.)
| | - Boaz Politi
- Department of Infectious Diseases, Israel Institute for Biological Research, Ness Ziona 74100, Israel; (Y.Y.-R.); (S.M.); (B.P.); (N.E.); (H.T.); (S.W.); (A.B.-S.); (M.B.-G.); (L.C.M.); (H.A.)
| | - Noam Erez
- Department of Infectious Diseases, Israel Institute for Biological Research, Ness Ziona 74100, Israel; (Y.Y.-R.); (S.M.); (B.P.); (N.E.); (H.T.); (S.W.); (A.B.-S.); (M.B.-G.); (L.C.M.); (H.A.)
| | - Hadas Tamir
- Department of Infectious Diseases, Israel Institute for Biological Research, Ness Ziona 74100, Israel; (Y.Y.-R.); (S.M.); (B.P.); (N.E.); (H.T.); (S.W.); (A.B.-S.); (M.B.-G.); (L.C.M.); (H.A.)
| | - Liat Bar-On
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness Ziona 74100, Israel; (L.B.-O.); (O.I.)
| | - Julia Ryvkin
- Bioinformatics Unit, Life Science Core Facilities, Weizmann Institute of Science, Rehovot 52621, Israel; (J.R.); (D.L.)
| | - Dena Leshkowitz
- Bioinformatics Unit, Life Science Core Facilities, Weizmann Institute of Science, Rehovot 52621, Israel; (J.R.); (D.L.)
| | - Ofir Israeli
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness Ziona 74100, Israel; (L.B.-O.); (O.I.)
| | - Shay Weiss
- Department of Infectious Diseases, Israel Institute for Biological Research, Ness Ziona 74100, Israel; (Y.Y.-R.); (S.M.); (B.P.); (N.E.); (H.T.); (S.W.); (A.B.-S.); (M.B.-G.); (L.C.M.); (H.A.)
| | - Amir Ben-Shmuel
- Department of Infectious Diseases, Israel Institute for Biological Research, Ness Ziona 74100, Israel; (Y.Y.-R.); (S.M.); (B.P.); (N.E.); (H.T.); (S.W.); (A.B.-S.); (M.B.-G.); (L.C.M.); (H.A.)
| | - Moria Barlev-Gross
- Department of Infectious Diseases, Israel Institute for Biological Research, Ness Ziona 74100, Israel; (Y.Y.-R.); (S.M.); (B.P.); (N.E.); (H.T.); (S.W.); (A.B.-S.); (M.B.-G.); (L.C.M.); (H.A.)
| | - Lilach Cherry Mimran
- Department of Infectious Diseases, Israel Institute for Biological Research, Ness Ziona 74100, Israel; (Y.Y.-R.); (S.M.); (B.P.); (N.E.); (H.T.); (S.W.); (A.B.-S.); (M.B.-G.); (L.C.M.); (H.A.)
| | - Hagit Achdout
- Department of Infectious Diseases, Israel Institute for Biological Research, Ness Ziona 74100, Israel; (Y.Y.-R.); (S.M.); (B.P.); (N.E.); (H.T.); (S.W.); (A.B.-S.); (M.B.-G.); (L.C.M.); (H.A.)
| | - Nir Paran
- Department of Infectious Diseases, Israel Institute for Biological Research, Ness Ziona 74100, Israel; (Y.Y.-R.); (S.M.); (B.P.); (N.E.); (H.T.); (S.W.); (A.B.-S.); (M.B.-G.); (L.C.M.); (H.A.)
| | - Tomer Israely
- Department of Infectious Diseases, Israel Institute for Biological Research, Ness Ziona 74100, Israel; (Y.Y.-R.); (S.M.); (B.P.); (N.E.); (H.T.); (S.W.); (A.B.-S.); (M.B.-G.); (L.C.M.); (H.A.)
| |
Collapse
|
27
|
Ma B, Tao M, Li Z, Zheng Q, Wu H, Chen P. Mucosal vaccines for viral diseases: Status and prospects. Virology 2024; 593:110026. [PMID: 38373360 DOI: 10.1016/j.virol.2024.110026] [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: 09/19/2023] [Revised: 02/08/2024] [Accepted: 02/12/2024] [Indexed: 02/21/2024]
Abstract
Virus-associated infectious diseases are highly detrimental to human health and animal husbandry. Among all countermeasures against infectious diseases, prophylactic vaccines, which developed through traditional or novel approaches, offer potential benefits. More recently, mucosal vaccines attract attention for their extraordinary characteristics compared to conventional parenteral vaccines, particularly for mucosal-related pathogens. Representatively, coronavirus disease 2019 (COVID-19), a respiratory disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), further accelerated the research and development efforts for mucosal vaccines by thoroughly investigating existing strategies or involving novel techniques. While several vaccine candidates achieved positive progresses, thus far, part of the current COVID-19 mucosal vaccines have shown poor performance, which underline the need for next-generation mucosal vaccines and corresponding platforms. In this review, we summarized the typical mucosal vaccines approved for humans or animals and sought to elucidate the underlying mechanisms of these successful cases. In addition, mucosal vaccines against COVID-19 that are in human clinical trials were reviewed in detail since this public health event mobilized all advanced technologies for possible solutions. Finally, the gaps in developing mucosal vaccines, potential solutions and prospects were discussed. Overall, rational application of mucosal vaccines would facilitate the establishing of mucosal immunity and block the transmission of viral diseases.
Collapse
Affiliation(s)
- Bingjie Ma
- College of Animal Science and Technology, Xinyang Agriculture and Forestry University, Xinyang, China
| | - Mengxiao Tao
- College of Animal Science and Technology, Xinyang Agriculture and Forestry University, Xinyang, China
| | - Zhili Li
- College of Animal Science and Technology, Xinyang Agriculture and Forestry University, Xinyang, China
| | - Quanfang Zheng
- College of Animal Science and Technology, Xinyang Agriculture and Forestry University, Xinyang, China
| | - Haigang Wu
- College of Animal Science and Technology, Xinyang Agriculture and Forestry University, Xinyang, China
| | - Peirong Chen
- College of Animal Science and Technology, Xinyang Agriculture and Forestry University, Xinyang, China.
| |
Collapse
|
28
|
Kumar A, Tripathi P, Kumar P, Shekhar R, Pathak R. From Detection to Protection: Antibodies and Their Crucial Role in Diagnosing and Combatting SARS-CoV-2. Vaccines (Basel) 2024; 12:459. [PMID: 38793710 PMCID: PMC11125746 DOI: 10.3390/vaccines12050459] [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: 03/13/2024] [Revised: 04/20/2024] [Accepted: 04/22/2024] [Indexed: 05/26/2024] Open
Abstract
Understanding the antibody response to SARS-CoV-2, the virus responsible for COVID-19, is crucial to comprehending disease progression and the significance of vaccine and therapeutic development. The emergence of highly contagious variants poses a significant challenge to humoral immunity, underscoring the necessity of grasping the intricacies of specific antibodies. This review emphasizes the pivotal role of antibodies in shaping immune responses and their implications for diagnosing, preventing, and treating SARS-CoV-2 infection. It delves into the kinetics and characteristics of the antibody response to SARS-CoV-2 and explores current antibody-based diagnostics, discussing their strengths, clinical utility, and limitations. Furthermore, we underscore the therapeutic potential of SARS-CoV-2-specific antibodies, discussing various antibody-based therapies such as monoclonal antibodies, polyclonal antibodies, anti-cytokines, convalescent plasma, and hyperimmunoglobulin-based therapies. Moreover, we offer insights into antibody responses to SARS-CoV-2 vaccines, emphasizing the significance of neutralizing antibodies in order to confer immunity to SARS-CoV-2, along with emerging variants of concern (VOCs) and circulating Omicron subvariants. We also highlight challenges in the field, such as the risks of antibody-dependent enhancement (ADE) for SARS-CoV-2 antibodies, and shed light on the challenges associated with the original antigenic sin (OAS) effect and long COVID. Overall, this review intends to provide valuable insights, which are crucial to advancing sensitive diagnostic tools, identifying efficient antibody-based therapeutics, and developing effective vaccines to combat the evolving threat of SARS-CoV-2 variants on a global scale.
Collapse
Affiliation(s)
- Anoop Kumar
- Molecular Diagnostic Laboratory, National Institute of Biologicals, Noida 201309, India
| | - Prajna Tripathi
- Department of Microbiology and Immunology, Weill Cornell Medical College, New York, NY 10021, USA;
| | - Prashant Kumar
- R. Ken Coit College of Pharmacy, University of Arizona, Tucson, AZ 85721, USA
| | - Ritu Shekhar
- Department of Molecular Genetics and Microbiology, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Rajiv Pathak
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA
| |
Collapse
|
29
|
Hill-Batorski L, Bowen R, Bielefeldt-Ohmann H, Moser MJ, Matejka SM, Marshall D, Kawaoka Y, Neumann G, Bilsel P. Mucosal immunization with dual influenza/COVID-19 single-replication virus vector protects hamsters from SARS-CoV-2 challenge. Vaccine 2024; 42:2770-2780. [PMID: 38508930 DOI: 10.1016/j.vaccine.2024.03.040] [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: 12/13/2023] [Revised: 03/15/2024] [Accepted: 03/16/2024] [Indexed: 03/22/2024]
Abstract
The COVID-19 pandemic has highlighted the need for mucosal vaccines as breakthrough infections, short-lived immune responses and emergence of new variants have challenged the efficacy provided by the first generation of vaccines against SARS-CoV-2 viruses. M2SR SARS-CoV-2, an M2-deleted single-replication influenza virus vector modified to encode the SARS-CoV-2 receptor binding domain, was evaluated following intranasal delivery in a hamster challenge model for protection against Wuhan SARS-CoV-2. An adjuvanted inactivated SARS-CoV-2 whole virus vaccine administered intramuscularly was also evaluated. The intranasal M2SR SARS-CoV-2 was more effective than the intramuscular adjuvanted inactivated whole virus vaccine in providing protection against SARS-CoV-2 challenge. M2SR SARS-CoV-2 elicited neutralizing serum antibodies against Wuhan and Omicron SARS-CoV-2 viruses in addition to cross-reactive mucosal antibodies. Furthermore, M2SR SARS-CoV-2 generated serum HAI and mucosal antibody responses against influenza similar to an H3N2 M2SR influenza vaccine. The intranasal dual influenza/COVID M2SR SARS-CoV-2 vaccine has the potential to provide protection against both influenza and COVID.
Collapse
|
30
|
Tang Y, Boribong BP, Swank ZN, Demokritou M, Luban MA, Fasano A, Du M, Wolf RL, Griffiths J, Shultz J, Borberg E, Chalise S, Gonzalez WI, Walt DR, Yonker LM, Horwitz BH. COVID-19 mRNA vaccines induce robust levels of IgG but limited amounts of IgA within the oronasopharynx of young children. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.04.15.24305767. [PMID: 38699375 PMCID: PMC11065043 DOI: 10.1101/2024.04.15.24305767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2024]
Abstract
Background Understanding antibody responses to SARS-CoV-2 vaccination is crucial for refining COVID-19 immunization strategies. Generation of mucosal immune responses, including mucosal IgA, could be of potential benefit to vaccine efficacy, yet limited evidence exists regarding the production of mucosal antibodies following the administration of current mRNA vaccines to young children. Methods We measured the levels of antibodies against SARS-CoV-2 from a cohort of children under 5 years of age undergoing SARS-CoV-2 mRNA vaccination (serially collected, matched serum and saliva samples, N=116) or on convenience samples of children under 5 years of age presenting to a pediatric emergency department (nasal swabs, N=103). Further, we assessed salivary and nasal samples for the ability to induce SARS-CoV-2 spike-mediated neutrophil extracellular traps (NET) formation. Results Longitudinal analysis of post-vaccine responses in saliva revealed the induction of SARS-CoV-2 specific IgG but not IgA. Similarly, SARS-CoV-2 specific IgA was only observed in nasal samples obtained from previously infected children with or without vaccination, but not in vaccinated children without a history of infection. In addition, oronasopharyngeal samples obtained from children with prior infection were able to trigger enhanced spike-mediated NET formation, and IgA played a key role in driving this process. Conclusions Despite the induction of specific IgG in the oronasal mucosa, current intramuscular vaccines have limited ability to generate mucosal IgA in young children. These results confirm the independence of mucosal IgA responses from systemic humoral responses following mRNA vaccination and suggest potential future vaccination strategies for enhancing mucosal protection in this young age group.
Collapse
Affiliation(s)
- Ying Tang
- Division of Gastroenterology, Hepatology, and Nutrition, Boston Children’s Hospital, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Brittany P. Boribong
- Harvard Medical School, Boston, MA 02115, USA
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Pediatrics, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Zoe N. Swank
- Harvard Medical School, Boston, MA 02115, USA
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02215, USA
| | - Melina Demokritou
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Maria A.F. Luban
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Alessio Fasano
- Harvard Medical School, Boston, MA 02115, USA
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Pediatrics, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Michelle Du
- Division of Emergency Medicine, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Rebecca L. Wolf
- Division of Emergency Medicine, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Joseph Griffiths
- Division of Emergency Medicine, Boston Children’s Hospital, Boston, MA 02115, USA
| | - John Shultz
- Division of Emergency Medicine, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Ella Borberg
- Harvard Medical School, Boston, MA 02115, USA
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02215, USA
| | - Sujata Chalise
- Harvard Medical School, Boston, MA 02115, USA
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02215, USA
| | - Wanda I. Gonzalez
- Harvard Medical School, Boston, MA 02115, USA
- Department of Pediatrics, Massachusetts General Hospital, Boston, MA 02114, USA
| | - David R. Walt
- Harvard Medical School, Boston, MA 02115, USA
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02215, USA
| | - Lael M. Yonker
- Harvard Medical School, Boston, MA 02115, USA
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Pediatrics, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Bruce H. Horwitz
- Division of Gastroenterology, Hepatology, and Nutrition, Boston Children’s Hospital, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
- Division of Emergency Medicine, Boston Children’s Hospital, Boston, MA 02115, USA
| |
Collapse
|
31
|
Song G, Li R, Cheng MQ. Safety, immunogenicity, and protective effective of inhaled COVID-19 vaccines: A systematic review and meta-analysis. J Med Virol 2024; 96:e29625. [PMID: 38650361 DOI: 10.1002/jmv.29625] [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: 02/07/2024] [Revised: 03/27/2024] [Accepted: 04/12/2024] [Indexed: 04/25/2024]
Abstract
This study aimed to examine the safety, immunogenicity and protective effective of inhaled COVID-19 vaccines (ICVs). Literature research was done through EMBASE, Cochrane, PubMed, and Web of Science up to 10 March 2024. Pooled estimates with corresponding 95% confidence intervals (CI) were computed and compared using the random effects and common effects model. Of the 15 studies, 11 analyzed safety, 13 analyzed immunogenicity, and 3 analyzed protective effective. The results showed a favorable safety profile of ICVs for primary vaccination series, however it does not always seem to produce the expected immune response and protective effective. Meta-analysis of ICVs booster vaccinations (BVs) showed that the levels of neutralizing antibody Geometric mean titer (nAb-GMT) with aerosolised Ad5-nCoV (AAd5-nCoV) were all higher than those with inactivated vaccine (INA-nCoV) (standard mean difference (SMD) = 2.32; 95% CI: 1.96-2.69) and intramuscular Ad5-nCoV (IMAd5-nCoV) (SMD = 0.31; 95% CI: 0.14-0.48) against the original strain of SARS-CoV-2. Importantly, we also observed similar results in the omicron variant. In addition, ICV in BVs has high mucosal immunity to IgA antibodies. The risk of adverse events was comparable or lower for AAd5-nCoV compared to INA-nCoV or IMAd5-nCoV. Current evidence shows that the safety profile of ICVs were well. The booster dose of AAd5-nCoV had a high immune response (including mucosal immunity) and provided protection against COVID-19 caused by the SARS-CoV-2 omicron variant. Further studies are needed to investigate the long-term safety of intranasal vaccine booster protection and various types of ICVs.
Collapse
Affiliation(s)
- Gao Song
- Department of Pharmacy, Puer People's Hospital, Pu'er, China
| | - Rong Li
- Department of Pharmacy, Puer People's Hospital, Pu'er, China
| | - Meng-Qun Cheng
- Department of Reproductive Medicine, Puer People's Hospital, Pu'er, China
| |
Collapse
|
32
|
Bladh O, Aguilera K, Marking U, Kihlgren M, Greilert Norin N, Smed-Sörensen A, Sällberg Chen M, Klingström J, Blom K, Russell MW, Havervall S, Thålin C, Åberg M. Comparison of SARS-CoV-2 spike-specific IgA and IgG in nasal secretions, saliva and serum. Front Immunol 2024; 15:1346749. [PMID: 38558811 PMCID: PMC10978617 DOI: 10.3389/fimmu.2024.1346749] [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: 11/29/2023] [Accepted: 01/30/2024] [Indexed: 04/04/2024] Open
Abstract
Introduction Several novel vaccine platforms aim at mucosal immunity in the respiratory tract to block SARS-CoV-2 transmission. Standardized methods for mucosal sample collection and quantification of mucosal antibodies are therefore urgently needed for harmonized comparisons and interpretations across mucosal vaccine trials and real-world data. Methods Using commercial electrochemiluminescence antibody panels, we compared SARS-CoV-2 spike-specific IgA and IgG in paired saliva, nasal secretions, and serum from 1048 healthcare workers with and without prior infection. Results Spike-specific IgA correlated well in nasal secretions and saliva (r>0.65, p<0.0001), but the levels were more than three-fold higher in nasal secretions as compared to in saliva (p<0.01). Correlations between the total population of spike-specific IgA and spike-specific secretory IgA (SIgA) were significantly stronger (p<0.0001) in nasal secretions (r=0.96, p<0.0001) as opposed to in saliva (r=0.77, p<0.0001), and spike-specific IgA correlated stronger (p<0.0001) between serum and saliva (r=0.73, p<0.001) as opposed to between serum and nasal secretions (r=0.54, p<0.001), suggesting transudation of monomeric spike specific IgA from the circulation to saliva. Notably, spike-specific SIgA had a markedly higher SARS-CoV-2 variant cross-binding capacity as compared to the total population of spike specific IgA and IgG in both nasal secretions, saliva and serum, (all p<0.0001), which emphasizes the importance of taking potential serum derived monomeric IgA into consideration when investigating mucosal immune responses. Discussion Taken together, although spike-specific IgA can be reliably measured in both nasal secretions and saliva, our findings imply an advantage of higher levels and likely also a larger proportion of SIgA in nasal secretions as compared to in saliva. We further corroborate the superior variant cross-binding capacity of SIgA in mucosal secretions, highlighting the potential protective benefits of a vaccine targeting the upper respiratory tract.
Collapse
Affiliation(s)
- Oscar Bladh
- Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Katherina Aguilera
- Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Ulrika Marking
- Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
- Public Health Agency of Sweden, Solna, Sweden
| | - Martha Kihlgren
- Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Nina Greilert Norin
- Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Anna Smed-Sörensen
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Margaret Sällberg Chen
- Department of Dental Medicine, Karolinska Institutet, Stockholm, Sweden
- Division of Pathology, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Jonas Klingström
- Public Health Agency of Sweden, Solna, Sweden
- Department of Biomedical and Clinical Sciences (BKV), Linköping University, Linköping, Sweden
| | - Kim Blom
- Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
- Public Health Agency of Sweden, Solna, Sweden
| | - Michael W. Russell
- Department of Microbiology and Immunology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, United States
| | - Sebastian Havervall
- Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Charlotte Thålin
- Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Mikael Åberg
- Department of Medical Sciences, Clinical Chemistry and SciLifeLab Affinity Proteomics, Uppsala University, Uppsala, Sweden
| |
Collapse
|
33
|
Arevalo-Romero JA, Chingaté-López SM, Camacho BA, Alméciga-Díaz CJ, Ramirez-Segura CA. Next-generation treatments: Immunotherapy and advanced therapies for COVID-19. Heliyon 2024; 10:e26423. [PMID: 38434363 PMCID: PMC10907543 DOI: 10.1016/j.heliyon.2024.e26423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 02/12/2024] [Accepted: 02/13/2024] [Indexed: 03/05/2024] Open
Abstract
The COVID-19 pandemic, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), emerged in 2019 following prior outbreaks of coronaviruses like SARS and MERS in recent decades, underscoring their high potential of infectivity in humans. Insights from previous outbreaks of SARS and MERS have played a significant role in developing effective strategies to mitigate the global impact of SARS-CoV-2. As of January 7, 2024, there have been 774,075,242 confirmed cases of COVID-19 worldwide. To date, 13.59 billion vaccine doses have been administered, and there have been 7,012,986 documented fatalities (https://www.who.int/) Despite significant progress in addressing the COVID-19 pandemic, the rapid evolution of SARS-CoV-2 challenges human defenses, presenting ongoing global challenges. The emergence of new SARS-CoV-2 lineages, shaped by mutation and recombination processes, has led to successive waves of infections. This scenario reveals the need for next-generation vaccines as a crucial requirement for ensuring ongoing protection against SARS-CoV-2. This demand calls for formulations that trigger a robust adaptive immune response without leading the acute inflammation linked with the infection. Key mutations detected in the Spike protein, a critical target for neutralizing antibodies and vaccine design -specifically within the Receptor Binding Domain region of Omicron variant lineages (B.1.1.529), currently dominant worldwide, have intensified concerns due to their association with immunity evasion from prior vaccinations and infections. As the world deals with this evolving threat, the narrative extends to the realm of emerging variants, each displaying new mutations with implications that remain largely misunderstood. Notably, the JN.1 Omicron lineage is gaining global prevalence, and early findings suggest it stands among the immune-evading variants, a characteristic attributed to its mutation L455S. Moreover, the detrimental consequences of the novel emergence of SARS-CoV-2 lineages bear a particularly critical impact on immunocompromised individuals and older adults. Immunocompromised individuals face challenges such as suboptimal responses to COVID-19 vaccines, rendering them more susceptible to severe disease. Similarly, older adults have an increased risk of severe disease and the presence of comorbid conditions, find themselves at a heightened vulnerability to develop COVID-19 disease. Thus, recognizing these intricate factors is crucial for effectively tailoring public health strategies to protect these vulnerable populations. In this context, this review aims to describe, analyze, and discuss the current progress of the next-generation treatments encompassing immunotherapeutic approaches and advanced therapies emerging as complements that will offer solutions to counter the disadvantages of the existing options. Preliminary outcomes show that these strategies target the virus and address the immunomodulatory responses associated with COVID-19. Furthermore, the capacity to promote tissue repair has been demonstrated, which can be particularly noteworthy for immunocompromised individuals who stand as vulnerable actors in the global landscape of coronavirus infections. The emerging next-generation treatments possess broader potential, offering protection against a wide range of variants and enhancing the ability to counter the impact of the constant evolution of the virus. Furthermore, advanced therapies are projected as potential treatment alternatives for managing Chronic Post-COVID-19 syndromeand addressing its associated long-term complications.
Collapse
Affiliation(s)
- Jenny Andrea Arevalo-Romero
- Laboratorio de Investigación en Ingeniería Celular y Molecular, Instituto Distrital de Ciencia, Biotecnología e Innovación en Salud, IDCBIS, 111611, Bogotá, DC, Colombia
- Instituto de Errores Innatos del Metabolismo, Facultad de Ciencias, Pontificia Universidad Javeriana, 110231, Bogotá, D.C., Colombia
| | - Sandra M. Chingaté-López
- Laboratorio de Investigación en Ingeniería Celular y Molecular, Instituto Distrital de Ciencia, Biotecnología e Innovación en Salud, IDCBIS, 111611, Bogotá, DC, Colombia
| | - Bernardo Armando Camacho
- Laboratorio de Investigación en Ingeniería Celular y Molecular, Instituto Distrital de Ciencia, Biotecnología e Innovación en Salud, IDCBIS, 111611, Bogotá, DC, Colombia
| | - Carlos Javier Alméciga-Díaz
- Instituto de Errores Innatos del Metabolismo, Facultad de Ciencias, Pontificia Universidad Javeriana, 110231, Bogotá, D.C., Colombia
| | - Cesar A. Ramirez-Segura
- Laboratorio de Investigación en Ingeniería Celular y Molecular, Instituto Distrital de Ciencia, Biotecnología e Innovación en Salud, IDCBIS, 111611, Bogotá, DC, Colombia
| |
Collapse
|
34
|
Zhang H, Liu Z, Lihe H, Lu L, Zhang Z, Yang S, Meng N, Xiong Y, Fan X, Chen Z, Lu W, Xie C, Liu M. Intranasal G5-BGG/pDNA Vaccine Elicits Protective Systemic and Mucosal Immunity against SARS-CoV-2 by Transfecting Mucosal Dendritic Cells. Adv Healthc Mater 2024; 13:e2303261. [PMID: 37961920 DOI: 10.1002/adhm.202303261] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 10/18/2023] [Indexed: 11/15/2023]
Abstract
Infectious disease pandemics, including the coronavirus disease 2019 pandemic, have heightened the demand for vaccines. Although parenteral vaccines induce robust systemic immunity, their effectiveness in respiratory mucosae is limited. Considering the crucial role of nasal-associated lymphoid tissue (NALT) in mucosal immune responses, in this study, the intranasal complex composed of G5-BGG and antigen-expressing plasmid DNA (pSP), named G5-BGG/pSP complex, is developed to activate NALT and to promote both systemic and mucosal immune defense. G5-BGG/pSP could traverse mucosal barriers and deliver DNA to the target cells because of its superior nasal retention and permeability characteristics. The intranasal G5-BGG/pSP complex elicits robust antigen-specific immune responses, such as the notable production of IgG antibody against several virus variants. More importantly, it induces elevated levels of antigen-specific IgA antibody and a significant expansion of the lung-resident T lymphocyte population. Notably, the intranasal G5-BGG/pSP complex results in antigen expression and maturation of dendritic cells in nasal mucosae. These findings exhibit the potential of G5-BGG, a novel cationic material, as an effective gene carrier for intranasal vaccines to obtain robust systemic and mucosal immunity.
Collapse
Affiliation(s)
- Han Zhang
- Department of Pharmaceutics and the Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Zezhong Liu
- Department of Pharmacology and the Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Hongye Lihe
- Department of Pharmaceutics and the Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Linwei Lu
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, 201203, China
- Institutes of Integrative Medicine, Fudan University, Shanghai, 201203, China
| | - Zongxu Zhang
- Department of Pharmaceutics and the Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Shengmin Yang
- Department of Pharmaceutics and the Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Nana Meng
- Department of Pharmaceutics and the Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Yin Xiong
- Department of Pharmaceutics and the Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Xingyan Fan
- Department of Pharmaceutics and the Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Zhikai Chen
- Department of Pharmacology and the Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Weiyue Lu
- Department of Pharmaceutics and the Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, China
- Shanghai Engineering Technology Research Center for Pharmaceutica Intelligent Equipment, Shanghai Frontiers Science Center for Druggability of Cardiovascular non-coding RNA Institute for Frontier Medical Technology Shanghai University of Engineering Science, Shanghai, 201203, China
- Shanghai Tayzen Pharmlab Co., Ltd., Shanghai, 201203, China
| | - Cao Xie
- Shanghai Tayzen Pharmlab Co., Ltd., Shanghai, 201203, China
| | - Min Liu
- Department of Pharmaceutics and the Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, China
| |
Collapse
|
35
|
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
|
36
|
Ellis S, Way R, Nel M, Burleigh A, Doykov I, Kembou-Ringert J, Woodall M, Masonou T, Case KM, Ortez AT, McHugh TD, Casal A, McCoy LE, Murdan S, Hynds RE, Gilmour KC, Grandjean L, Cortina-Borja M, Heywood WE, Mills K, Smith CM. Salivary IgA and vimentin differentiate in vitro SARS-CoV-2 infection: A study of 290 convalescent COVID-19 patients. Mucosal Immunol 2024; 17:124-136. [PMID: 38007005 PMCID: PMC11139657 DOI: 10.1016/j.mucimm.2023.11.007] [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: 06/26/2023] [Revised: 11/07/2023] [Accepted: 11/20/2023] [Indexed: 11/27/2023]
Abstract
SARS-CoV-2 initially infects cells in the nasopharynx and oral cavity. The immune system at these mucosal sites plays a crucial role in minimizing viral transmission and infection. To develop new strategies for preventing SARS-CoV-2 infection, this study aimed to identify proteins that protect against viral infection in saliva. We collected 551 saliva samples from 290 healthcare workers who had tested positive for COVID-19, before vaccination, between June and December 2020. The samples were categorized based on their ability to block or enhance infection using in vitro assays. Mass spectrometry and enzyme-linked immunosorbent assay experiments were used to identify and measure the abundance of proteins that specifically bind to SARS-CoV-2 antigens. Immunoglobulin (Ig)A specific to SARS-CoV-2 antigens was detectable in over 83% of the convalescent saliva samples. We found that concentrations of anti-receptor-binding domain IgA >500 pg/µg total protein in saliva correlate with reduced viral infectivity in vitro. However, there is a dissociation between the salivary IgA response to SARS-CoV-2, and systemic IgG titers in convalescent COVID-19 patients. Then, using an innovative technique known as spike-baited mass spectrometry, we identified novel spike-binding proteins in saliva, most notably vimentin, which correlated with increased viral infectivity in vitro and could serve as a therapeutic target against COVID-19.
Collapse
Affiliation(s)
- Samuel Ellis
- UCL Great Ormond Street Institute of Child Health, London, UK
| | - Rosie Way
- UCL Great Ormond Street Institute of Child Health, London, UK
| | - Miranda Nel
- UCL Great Ormond Street Institute of Child Health, London, UK
| | - Alice Burleigh
- UCL Great Ormond Street Institute of Child Health, London, UK; Centre for Adolescent Rheumatology, University College London, London, UK
| | - Ivan Doykov
- UCL Great Ormond Street Institute of Child Health, London, UK
| | | | | | - Tereza Masonou
- UCL Great Ormond Street Institute of Child Health, London, UK
| | | | | | - Timothy D McHugh
- UCL Centre for Clinical Microbiology, Royal Free Hospital, London, UK
| | - Antonio Casal
- Department of Pharmaceutics, UCL School of Pharmacy, London, UK
| | - Laura E McCoy
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, London, UK
| | | | - Robert E Hynds
- Epithelial Cell Biology in ENT Research (EpiCENTR) Group, Developmental Biology and Cancer Department, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Kimberly C Gilmour
- UCL Great Ormond Street Institute of Child Health, London, UK; Great Ormond Street Hospital NHS Foundation Trust, London, UK
| | - Louis Grandjean
- UCL Great Ormond Street Institute of Child Health, London, UK; Great Ormond Street Hospital NHS Foundation Trust, London, UK
| | | | - Wendy E Heywood
- UCL Great Ormond Street Institute of Child Health, London, UK
| | - Kevin Mills
- UCL Great Ormond Street Institute of Child Health, London, UK
| | - Claire M Smith
- UCL Great Ormond Street Institute of Child Health, London, UK.
| |
Collapse
|
37
|
Son YM, Cheon IS, Li C, Sun J. Persistent B Cell-Derived MHC Class II Signaling Is Required for the Optimal Maintenance of Tissue-Resident Helper T Cells. Immunohorizons 2024; 8:163-171. [PMID: 38345472 PMCID: PMC10916357 DOI: 10.4049/immunohorizons.2300093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 01/17/2024] [Indexed: 02/15/2024] Open
Abstract
Emerging studies have identified the critical roles of tissue-resident memory CD8+ T (TRM) and B (BRM) cells in the protection against mucosal viral infections, but the underlying mechanisms regulating robust development of TRM and BRM cells remain incompletely understood. We have recently shown that tissue-resident helper CD4+ T (TRH) cells, developed following influenza virus infection, function to sustain the optimal maintenance of TRM and BRM cells at the mucosal surface. In this study, we have explored the cellular and molecular cues modulating lung TRH persistence after influenza infection in C57BL/6 mice. We found that TRH cells were colocalized in tertiary lymphoid structures (TLSs) with local B cells. Abolishing TLSs or the depletion of B cells impaired lung TRH cell numbers. Of note, we found that persistent TCR signaling is needed for the maintenance of TRH cells after the clearance of infectious influenza virus. Furthermore, selective ablation of B cell-derived MHC class II resulted in partial reduction of lung TRH cell number after influenza infection. Our findings suggest that the interaction between lung-resident TRH cells and B cells, along with persistent Ag stimulation, is required to maintain TRH cells after respiratory viral infection.
Collapse
Affiliation(s)
- Young Min Son
- Department of Systems Biotechnology, Chung-Ang University, Anseong, Republic of Korea
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic College of Medicine and Science, Rochester, MN
| | - In Su Cheon
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic College of Medicine and Science, Rochester, MN
- Carter Immunology Center, University of Virginia, Charlottesville, VA
- Division of Infectious Disease and International Health, Department of Medicine, University of Virginia, Charlottesville, VA
| | - Chaofan Li
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic College of Medicine and Science, Rochester, MN
- Carter Immunology Center, University of Virginia, Charlottesville, VA
- Division of Infectious Disease and International Health, Department of Medicine, University of Virginia, Charlottesville, VA
| | - Jie Sun
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic College of Medicine and Science, Rochester, MN
- Carter Immunology Center, University of Virginia, Charlottesville, VA
- Division of Infectious Disease and International Health, Department of Medicine, University of Virginia, Charlottesville, VA
| |
Collapse
|
38
|
Mi H, Chen Q, Lin H, He T, Zhang R, Ren S, Liu L, Wang J, Huang H, Wang M, Guo Z, Su C. Short-term effectiveness of single-dose intranasal spray COVID-19 vaccine against symptomatic SARS-CoV-2 Omicron infection in healthcare workers: a prospective cohort study. EClinicalMedicine 2024; 67:102374. [PMID: 38169940 PMCID: PMC10758709 DOI: 10.1016/j.eclinm.2023.102374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 11/28/2023] [Accepted: 11/28/2023] [Indexed: 01/05/2024] Open
Abstract
Background The pivotal phase 3 efficacy clinical trial has demonstrated that a two-dose regimen of dNS1-RBD (Beijing Wantai Biological Pharmacy Enterprise, Beijing, China) is well-tolerated and provides wide protection against SARS-CoV-2 infection. However, the effectiveness of a single-dose regimen is still unknown. We aimed to estimate the effectiveness of one-dose of dNS1-RBD against symptomatic Omicron infections in real-world conditions. Methods This prospective cohort study was conducted during an Omicron outbreak among healthcare workers in Xiamen, China, from December 22, 2022 to January 16, 2023. Participants chose to receive single-dose of dNS1-RBD or remain unvaccinated based on personal preference. Healthcare workers daily validated their SARS-CoV-2 infection status, using either RT-PCR or rapid antigen test. A survey questionnaire was conducted to gather information on acute symptoms from individuals infected with SARS-CoV-2. The primary outcome was the symptomatic SARS-CoV-2 infections after enrollment in the dNS1-RBD recipients or the control group among all participants and by prior COVID-19 vaccination status. Findings On December 22, 2022, a total of 1391 eligible participants without a history of prior SARS-CoV-2 infection were enrolled. Among them, 550 received single-dose of dNS1-RBD, while 841 remained unvaccinated. In the total cohort, the range of follow-up time was 1∼26 days. During the study period, a total of 880 symptomatic SARS-CoV-2 infections were identified in the total cohort. The adjusted vaccine effectiveness against symptomatic SARS-CoV-2 infections and the infections requiring medical attention were 19.0% (95% CI: 6.7, 29.7, P = 0.004) and 59.4% (95% CI: 25.1, 78.0, P = 0.004) in the total cohort, 11.6% (95% CI: -2.4, 23.7, P = 0.100) and 55.3% (95% CI: 15.3, 76.4, P = 0.014) in the participants with inactivated COVID-19 vaccination history, as well as 87.0% (95% CI: 72.6, 93.9, P < 0.001) and 84.2% (95% CI: -41.8, 98.2, P = 0.099) in the naïve participants, respectively. Interpretation When administered as a booster to individuals with a history of inactivated COVID-19 vaccination, a single-dose of dNS1-RBD provides protection against infections requiring medical attention at least in the short-term after vaccination. The data also showed that a single-dose of dNS1-RBD is protective against symptomatic SARS-CoV-2 infections as a primary immunization for individuals without prior exposure, but due to the limited sample size of naïve participants, further research with a larger sample size is needed to make a solid conclusion. Funding Xiamen Science and Technology Bureau 2022 General Science and Technology Plan Project and the Bill & Melinda Gates Foundation.
Collapse
Affiliation(s)
- Hongfei Mi
- Zhongshan Hospital, Fudan University (Xiamen Branch), Xiamen, 361015, China
- Xiamen Clinical Research Center for Cancer Therapy, Xiamen, 361015, China
| | - Qi Chen
- School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Hongyan Lin
- School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Tingjuan He
- Zhongshan Hospital, Fudan University (Xiamen Branch), Xiamen, 361015, China
- Xiamen Clinical Research Center for Cancer Therapy, Xiamen, 361015, China
| | - Ruixin Zhang
- Zhongshan Hospital, Fudan University (Xiamen Branch), Xiamen, 361015, China
- Xiamen Clinical Research Center for Cancer Therapy, Xiamen, 361015, China
- School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Shuhao Ren
- Zhongshan Hospital, Fudan University (Xiamen Branch), Xiamen, 361015, China
- Xiamen Clinical Research Center for Cancer Therapy, Xiamen, 361015, China
- School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Lingling Liu
- Zhongshan Hospital, Fudan University (Xiamen Branch), Xiamen, 361015, China
- Xiamen Clinical Research Center for Cancer Therapy, Xiamen, 361015, China
| | - Jing Wang
- Zhongshan Hospital, Fudan University (Xiamen Branch), Xiamen, 361015, China
- Xiamen Clinical Research Center for Cancer Therapy, Xiamen, 361015, China
| | - Hua Huang
- Zhongshan Hospital, Fudan University (Xiamen Branch), Xiamen, 361015, China
- Xiamen Clinical Research Center for Cancer Therapy, Xiamen, 361015, China
| | - Meixia Wang
- Zhongshan Hospital, Fudan University (Xiamen Branch), Xiamen, 361015, China
- Xiamen Clinical Research Center for Cancer Therapy, Xiamen, 361015, China
| | - Zhinan Guo
- Xiamen Center for Disease Control and Prevention, Xiamen 361021, China
| | - Chenghao Su
- Zhongshan Hospital, Fudan University (Xiamen Branch), Xiamen, 361015, China
- Xiamen Clinical Research Center for Cancer Therapy, Xiamen, 361015, China
- School of Public Health, Xiamen University, Xiamen, 361102, China
| |
Collapse
|
39
|
Zhang X, Zhang J, Chen S, He Q, Bai Y, Liu J, Wang Z, Liang Z, Chen L, Mao Q, Xu M. Progress and challenges in the clinical evaluation of immune responses to respiratory mucosal vaccines. Expert Rev Vaccines 2024; 23:362-370. [PMID: 38444382 DOI: 10.1080/14760584.2024.2326094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 02/28/2024] [Indexed: 03/07/2024]
Abstract
INTRODUCTION Following the coronavirus disease pandemic, respiratory mucosal vaccines that elicit both mucosal and systemic immune responses have garnered increasing attention. However, human physiological characteristics pose significant challenges in the evaluation of mucosal immunity, which directly impedes the development and application of respiratory mucosal vaccines. AREAS COVERED This study summarizes the characteristics of immune responses in the respiratory mucosa and reviews the current status and challenges in evaluating immune response to respiratory mucosal vaccines. EXPERT OPINION Secretory Immunoglobulin A (S-IgA) is a major effector molecule at mucosal sites and a commonly used indicator for evaluating respiratory mucosal vaccines. However, the unique physiological structure of the respiratory tract pose significant challenges for the clinical collection and detection of S-IgA. Therefore, it is imperative to develop a sampling method with high collection efficiency and acceptance, a sensitive detection method, reference materials for mucosal antibodies, and to establish a threshold for S-IgA that correlates with clinical protection. Sample collection is even more challenging when evaluating mucosal cell immunity. Therefore, a mucosal cell sampling method with high operability and high tolerance should be established. Targets of the circulatory system capable of reflecting mucosal cellular immunity should also be explored.
Collapse
Affiliation(s)
- Xuanxuan Zhang
- State Key Laboratory of Drug Regulatory Science, Institute of Biological Products, National Institutes for Food and Drug Control, Beijing, China
| | - Jialu Zhang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Si Chen
- Drug and Vaccine Research Center, Guangzhou National Laboratory, Guangzhou, China
| | - Qian He
- State Key Laboratory of Drug Regulatory Science, Institute of Biological Products, National Institutes for Food and Drug Control, Beijing, China
| | - Yu Bai
- State Key Laboratory of Drug Regulatory Science, Institute of Biological Products, National Institutes for Food and Drug Control, Beijing, China
| | - Jianyang Liu
- State Key Laboratory of Drug Regulatory Science, Institute of Biological Products, National Institutes for Food and Drug Control, Beijing, China
| | - Zhongfang Wang
- Drug and Vaccine Research Center, Guangzhou National Laboratory, Guangzhou, China
| | - Zhenglun Liang
- State Key Laboratory of Drug Regulatory Science, Institute of Biological Products, National Institutes for Food and Drug Control, Beijing, China
| | - Ling Chen
- Drug and Vaccine Research Center, Guangzhou National Laboratory, Guangzhou, China
- State Key Laboratory of Respiratory Disease, Guangdong Laboratory of Computational Biomedicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Qunying Mao
- State Key Laboratory of Drug Regulatory Science, Institute of Biological Products, National Institutes for Food and Drug Control, Beijing, China
| | - Miao Xu
- State Key Laboratory of Drug Regulatory Science, Institute of Biological Products, National Institutes for Food and Drug Control, Beijing, China
| |
Collapse
|
40
|
Ajmera H, Lakhawat SS, Malik N, Kumar A, Bhatti JS, Kumar V, Gogoi H, Jaswal SK, Chandel S, Sharma PK. Global Emergence of SARS-CoV2 Infection and Scientific Interventions to Contain its Spread. Curr Protein Pept Sci 2024; 25:307-325. [PMID: 38265408 DOI: 10.2174/0113892037274719231212044235] [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: 08/12/2023] [Revised: 10/09/2023] [Accepted: 10/18/2023] [Indexed: 01/25/2024]
Abstract
The global pandemic caused by COVID-19 posed a significant challenge to public health, necessitating rapid scientific interventions to tackle the spread of infection. The review discusses the key areas of research on COVID-19 including viral genomics, epidemiology, pathogenesis, diagnostics, and therapeutics. The genome sequencing of the virus facilitated the tracking of its evolution, transmission dynamics, and identification of variants. Epidemiological studies have provided insights into disease spread, risk factors, and the impact of public health infrastructure and social distancing measures. Investigations of the viral pathogenesis have elucidated the mechanisms underlying immune responses and severe manifestations including the long-term effects of COVID-19. Overall, the article provides an updated overview of the diagnostic methods developed for SARS-CoV-2 and discusses their strengths, limitations, and appropriate utilization in different clinical and public health settings. Furthermore, therapeutic approaches including antiviral drugs, immunomodulatory therapies, and repurposed medications have been investigated to alleviate disease severity and improve patient outcomes. Through a comprehensive analysis of these scientific efforts, the review provides an overview of the advancements made in understanding and tackling SARS-CoV-2, while underscoring the need for continued research to address the evolving challenges posed by this global health crisis.
Collapse
Affiliation(s)
- Himanshu Ajmera
- Amity Institute of Biotechnology, Amity University Rajasthan, Jaipur, 303002, India
| | | | - Naveen Malik
- Amity Institute of Biotechnology, Amity University Rajasthan, Jaipur, 303002, India
| | - Akhilesh Kumar
- Amity Institute of Biotechnology, Amity University Rajasthan, Jaipur, 303002, India
| | - Jasvinder Singh Bhatti
- Department of Human Genetics & Molecular Medicine, Central University of Punjab, Bathinda, India
| | - Vikram Kumar
- Amity Institute of Biotechnology, Amity University Rajasthan, Jaipur, 303002, India
| | - Himanshu Gogoi
- Translational Health Science and Technology Institute, NCR Biotech Science Cluster 3rd milestone Faridabad, Haryana, India
| | - Sunil Kumar Jaswal
- Department of Biotechnology, Himachal Pradesh University Summer Hill, Shimla, India
| | - Sanjeev Chandel
- Department of Nursing, GHG College of Nursing Rajkot Road, Ludhiana, Punjab, India
| | - Pushpender Kumar Sharma
- Amity Institute of Biotechnology, Amity University Rajasthan, Jaipur, 303002, India
- Amity Centre for Nanobiotechnology and Nanomedicine, Amity University Rajasthan, Jaipur, 303002, India
| |
Collapse
|
41
|
Wang S, Cui H, Zhang C, Li W, Wang W, He W, Feng N, Zhao Y, Wang T, Tang X, Yan F, Xia X. Oral delivery of a chitosan adjuvanted COVID-19 vaccine provides long-lasting and broad-spectrum protection against SARS-CoV-2 variants of concern in golden hamsters. Antiviral Res 2023; 220:105765. [PMID: 38036065 DOI: 10.1016/j.antiviral.2023.105765] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 10/27/2023] [Accepted: 11/22/2023] [Indexed: 12/02/2023]
Abstract
Coronavirus disease 2019 (COVID-19) seriously threatens public health safety and the global economy, which warrant effective prophylactic and therapeutic approaches. Currently, vaccination and establishment of immunity have significantly reduced the severity and mortality of COVID-19. However, in regard to COVID-19 vaccines, the broad-spectrum protective efficacy against SARS-CoV-2 variants and the blocking of virus transmission need to be further improved. In this study, an optimum oral COVID-19 vaccine candidate, rVSVΔG-Sdelta, was selected from a panel of vesicular stomatitis virus (VSV)-based constructs bearing spike proteins from different SARS-CoV-2 strains. After chitosan modification, rVSVΔG-Sdelta induced both local and peripheral antibody response, particularly, broad-spectrum and long-lasting neutralizing antibodies against SARS-CoV-2 persisted for 1 year. Cross-protection against SARS-CoV-2 WT, Beta, Delta, BA.1, and BA.2 strains was achieved in golden hamsters, which presented as significantly reduced viral replication in the respiratory tract and alleviated pulmonary pathology post SARS-CoV-2 challenge. Overall, this study provides a convenient, oral-delivered, and effective oral mucosal vaccine against COVID-19, which would supplement pools and facilitate the distribution of COVID-19 vaccines.
Collapse
Affiliation(s)
- Shen Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China
| | - Huan Cui
- College of Veterinary Medicine, Hebei Agricultural University, 2596 Lucky South Street, Baoding, 071000, China
| | - Cheng Zhang
- College of Veterinary Medicine, Hebei Agricultural University, 2596 Lucky South Street, Baoding, 071000, China
| | - Wujian Li
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China; College of Veterinary Medicine, Jilin University, Changchun, 130062, Jilin, China
| | - Weiqi Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China; College of Veterinary Medicine, Jilin University, Changchun, 130062, Jilin, China
| | - Wenwen He
- The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 42100, China
| | - Na Feng
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China
| | - Yongkun Zhao
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China
| | - Tiecheng Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China
| | - Xiaoqing Tang
- The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 42100, China.
| | - Feihu Yan
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China.
| | - Xianzhu Xia
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China.
| |
Collapse
|
42
|
Huang CQ, Vishwanath S, Carnell GW, Chan ACY, Heeney JL. Immune imprinting and next-generation coronavirus vaccines. Nat Microbiol 2023; 8:1971-1985. [PMID: 37932355 DOI: 10.1038/s41564-023-01505-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 09/13/2023] [Indexed: 11/08/2023]
Abstract
Vaccines based on historical virus isolates provide limited protection from continuously evolving RNA viruses, such as influenza viruses or coronaviruses, which occasionally spill over between animals and humans. Despite repeated booster immunizations, population-wide declines in the neutralization of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants have occurred. This has been compared to seasonal influenza vaccinations in humans, where the breadth of immune responses induced by repeat exposures to antigenically distinct influenza viruses is confounded by pre-existing immunity-a mechanism known as imprinting. Since its emergence, SARS-CoV-2 has evolved in a population with partial immunity, acquired by infection, vaccination or both. Here we critically examine the evidence for and against immune imprinting in host humoral responses to SARS-CoV-2 and its implications for coronavirus disease 2019 (COVID-19) booster vaccine programmes.
Collapse
Affiliation(s)
- Chloe Qingzhou Huang
- Laboratory of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Sneha Vishwanath
- Laboratory of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - George William Carnell
- Laboratory of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Andrew Chun Yue Chan
- Laboratory of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Jonathan Luke Heeney
- Laboratory of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Cambridge, UK.
| |
Collapse
|
43
|
Guan X, Verma AK, Wang G, Shi J, Perlman S, Du L. Glycosylated Delta-receptor-binding domain mucosal vaccine elicits broadly neutralizing antibodies with protection against SARS-CoV-2 challenge. iScience 2023; 26:108033. [PMID: 37822493 PMCID: PMC10563057 DOI: 10.1016/j.isci.2023.108033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 08/15/2023] [Accepted: 09/21/2023] [Indexed: 10/13/2023] Open
Abstract
Mucosal COVID-19 vaccines are needed to block SARS-CoV-2 infection at the mucosal site. Intranasal delivery of a glycosylated Delta variant receptor-binding domain (Delta-RBD) mucosal vaccine elicited potent and balanced systemic antibody titers comparable to those induced by the intramuscular injection of the same vaccine or Omicron-S subunit vaccine, as well as high mucosal IgA antibody responses. It elicited broadly neutralizing antibodies against the original SARS-CoV-2 strain, Delta and Omicron BA1/BA2 variants, completely protecting transgenic mice from lethal challenge with a Delta variant, including complete absence of weight loss. Of note, intramuscular priming with the Omicron-S protein followed by intranasal boosting with the Delta-RBD protein improved the vaccine's ability to generate broad-spectrum neutralizing antibodies against recent BA5 and XBB Omicron variants. Overall, this vaccine has the potential to prevent the SARS-CoV-2 infection of the respiratory mucosa, while the i.m. priming and i.n. boosting vaccination strategy may offer protection against known and emerging SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Xiaoqing Guan
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Abhishek K. Verma
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, USA
| | - Gang Wang
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Juan Shi
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Stanley Perlman
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, USA
- Department of Pediatrics, University of Iowa, Iowa City, IA, USA
| | - Lanying Du
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| |
Collapse
|
44
|
Perrotta C, Fenizia C, Carnovale C, Pozzi M, Trabattoni D, Cervia D, Clementi E. Updated Considerations for the Immunopharmacological Aspects of the "Talented mRNA Vaccines". Vaccines (Basel) 2023; 11:1481. [PMID: 37766157 PMCID: PMC10534931 DOI: 10.3390/vaccines11091481] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 09/08/2023] [Accepted: 09/09/2023] [Indexed: 09/29/2023] Open
Abstract
Messenger RNA (mRNA) vaccines belong to a new class of medications, RNA therapeutics, including both coding and non-coding RNAs. The use of mRNA as a therapy is based on the biological role of mRNA itself, namely its translation into a functional protein. The goal of mRNA vaccines is to produce a specific antigen in cells to elicit an immune response that might be prophylactic or therapeutic. The potential of mRNA as vaccine has been envisaged for years but its efficacy has been clearly demonstrated with the approval of COVID-19 vaccines in 2021. Since then, mRNA vaccines have been in the pipeline for diseases that are still untreatable. There are many advantages of mRNA vaccines over traditional vaccines, including easy and cost-effective production, high safety, and high-level antigen expression. However, the nature of mRNA itself and some technical issues pose challenges associated with the vaccines' development and use. Here we review the immunological and pharmacological features of mRNA vaccines by discussing their pharmacokinetics, mechanisms of action, and safety, with a particular attention on the advantages and challenges related to their administration. Furthermore, we present an overview of the areas of application and the clinical trials that utilize a mRNA vaccine as a treatment.
Collapse
Affiliation(s)
- Cristiana Perrotta
- Department of Biomedical and Clinical Sciences (DIBIC), Università degli Studi di Milano, 20157 Milano, Italy; (C.C.); (D.T.)
| | - Claudio Fenizia
- Department of Pathophysiology and Transplantation (DEPT), Università degli Studi di Milano, 20122 Milano, Italy;
| | - Carla Carnovale
- Department of Biomedical and Clinical Sciences (DIBIC), Università degli Studi di Milano, 20157 Milano, Italy; (C.C.); (D.T.)
| | - Marco Pozzi
- Scientific Institute IRCCS Eugenio Medea, 23842 Bosisio Parini, Italy;
| | - Daria Trabattoni
- Department of Biomedical and Clinical Sciences (DIBIC), Università degli Studi di Milano, 20157 Milano, Italy; (C.C.); (D.T.)
| | - Davide Cervia
- Department for Innovation in Biological, Agro-Food and Forest Systems (DIBAF), Università degli Studi della Tuscia, 01100 Viterbo, Italy;
| | - Emilio Clementi
- Department of Biomedical and Clinical Sciences (DIBIC), Università degli Studi di Milano, 20157 Milano, Italy; (C.C.); (D.T.)
- Scientific Institute IRCCS Eugenio Medea, 23842 Bosisio Parini, Italy;
| |
Collapse
|
45
|
Sonvico F, Colombo G, Quarta E, Guareschi F, Banella S, Buttini F, Scherließ R. Nasal delivery as a strategy for the prevention and treatment of COVID-19. Expert Opin Drug Deliv 2023; 20:1115-1130. [PMID: 37755135 DOI: 10.1080/17425247.2023.2263363] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Accepted: 09/22/2023] [Indexed: 09/28/2023]
Abstract
INTRODUCTION The upper respiratory tract is a major route of infection for COVID-19 and other respiratory diseases. Thus, it appears logical to exploit the nose as administration site to prevent, fight, or minimize infectious spread and treat the disease. Numerous nasal products addressing these aspects have been considered and developed for COVID-19. AREAS COVERED This review gives a comprehensive overview of the different approaches involving nasal delivery, i.e., nasal vaccination, barrier products, and antiviral pharmacological treatments that have led to products on the market or under clinical evaluation, highlighting the peculiarities of the nose as application and absorption site and pointing at key aspects of nasal drug delivery. EXPERT OPINION From the analysis of nasal delivery strategies to prevent or fight COVID-19, it emerges that, especially for nasal immunization, formulations appear the same as originally designed for parenteral administration, leading to suboptimal results. On the other hand, mechanical barrier and antiviral products, designed to halt or treat the infection at early stage, have been proven effective but were rarely brought to the clinics. If supported by robust and targeted product development strategies, intranasal immunization and drug delivery can represent valid and sometimes superior alternatives to more conventional parenteral and oral medications.
Collapse
Affiliation(s)
- Fabio Sonvico
- Department of Food and Drug, University of Parma, Parma, Italy
| | - Gaia Colombo
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Eride Quarta
- Department of Food and Drug, University of Parma, Parma, Italy
| | | | - Sabrina Banella
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | | | - Regina Scherließ
- Department of Pharmaceutics and Biopharmaceutics, Kiel University, Kiel, Germany
- Priority Research Area Kiel Nano, Surface and Interface Sciences (KiNSIS), Kiel University, Kiel, Germany
| |
Collapse
|
46
|
Tsai CJY, Loh JMS, Fujihashi K, Kiyono H. Mucosal vaccination: onward and upward. Expert Rev Vaccines 2023; 22:885-899. [PMID: 37817433 DOI: 10.1080/14760584.2023.2268724] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 10/05/2023] [Indexed: 10/12/2023]
Abstract
INTRODUCTION The unique mucosal immune system allows the generation of robust protective immune responses at the front line of pathogen encounters. The needle-free delivery route and cold chain-free logistic requirements also provide additional advantages in ease and economy. However, the development of mucosal vaccines faces several challenges, and only a handful of mucosal vaccines are currently licensed. These vaccines are all in the form of live attenuated or inactivated whole organisms, whereas no subunit-based mucosal vaccine is available. AREAS COVERED The selection of antigen, delivery vehicle, route and adjuvants for mucosal vaccination are highly important. This is particularly crucial for subunit vaccines, as they often fail to elicit strong immune responses. Emerging research is providing new insights into the biological and immunological uniqueness of mucosal tissues. However, many aspects of the mucosal immunology still await to be investigated. EXPERT OPINION This article provides an overview of the current understanding of mucosal vaccination and discusses the remaining knowledge gaps. We emphasize that because of the potential benefits mucosal vaccines can bring from the biomedical, social and economic standpoints, the unmet goal to achieve mucosal vaccine success is worth the effort.
Collapse
Affiliation(s)
- Catherine J Y Tsai
- Department of Molecular Medicine & Pathology, University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, New Zealand, Auckland
- Department of Human Mucosal Vaccinology, Chiba University Hospital, Chiba, Japan
- Chiba University Synergy Institute for Futuristic Mucosal Vaccine Research and Development (cSIMVa), Chiba University, Chiba, Japan
| | - Jacelyn M S Loh
- Department of Molecular Medicine & Pathology, University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, New Zealand, Auckland
| | - Kohtaro Fujihashi
- Department of Human Mucosal Vaccinology, Chiba University Hospital, Chiba, Japan
- Chiba University Synergy Institute for Futuristic Mucosal Vaccine Research and Development (cSIMVa), Chiba University, Chiba, Japan
- Division of Infectious Disease Vaccine R&D, Research Institute of Disaster Medicine, Chiba University, Chiba, Japan
- Division of Mucosal Vaccines, International Vaccine Design Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Department of Pediatric Dentistry, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Hiroshi Kiyono
- Department of Human Mucosal Vaccinology, Chiba University Hospital, Chiba, Japan
- Chiba University Synergy Institute for Futuristic Mucosal Vaccine Research and Development (cSIMVa), Chiba University, Chiba, Japan
- Division of Infectious Disease Vaccine R&D, Research Institute of Disaster Medicine, Chiba University, Chiba, Japan
- Institute for Advanced Academic Research, Chiba University, Chiba, Japan
- CU-UCSD Center for Mucosal Immunology, Allergy and Vaccines (cMAV), Division of Gastroenterology, Department of Medicine, University of California, San Diego, CA, USA
- Future Medicine Education and Research Organization, Mucosal Immunology and Allergy Therapeutics, Institute for Global Prominent Research, Chiba University, Chiba, Japan
| |
Collapse
|