1
|
Guo J, Rohokale R, Kundu S, Guo Z. Monophosphoryl Lipid A-2,4-Dinitrophenylamine Conjugates Are Potent Adjuvants for Carbohydrate and Protein Vaccines. JACS AU 2025; 5:2210-2222. [PMID: 40443885 PMCID: PMC12117460 DOI: 10.1021/jacsau.5c00187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 04/23/2025] [Accepted: 04/24/2025] [Indexed: 06/02/2025]
Abstract
Adjuvants are essential for effective vaccine formulation, but currently only a few adjuvants with limited efficacies and application scopes are available. To address this issue, we explored covalent conjugates of monophosphoryl lipid A (MPLA) and 2,4-dinitrophenylamine (DNPA) as a new type of adjuvant. Immunological studies in mice prove that MPLA-DNPA conjugates can help a model vaccine induce robust IgG antibody and adaptive immune responses against carbohydrate and protein antigens and are much more potent adjuvants than alumthe positive controland the MPLA + DNPA mixture. Detailed profiling and comparison of the cytokines/chemokines elicited by various adjuvants suggest that the MPLA-DNPA conjugates can activate macrophages, monocytes, dendritic, T, T helper, and other immune cells to promote cellular immunity against vaccines. The results suggest a synergistic effect of covalently linked MPLA and DNPA, which act via interacting with the Toll-like receptor and recruiting endogenous anti-DNPA antibodies, respectively. Moreover, the linker between MPLA and DNPA shows a major impact on this synergistic effect, especially for the carbohydrate antigen. Eventually, the MPLA-DNPA conjugate with a longer linker containing a triazole moiety is identified as a promising adjuvant for both carbohydrate and protein vaccines worthy of further research and development.
Collapse
Affiliation(s)
- Jiatong Guo
- Department
of Chemistry, University of Florida, Gainesville, Florida32611, United States
| | - Rajendra Rohokale
- Department
of Chemistry, University of Florida, Gainesville, Florida32611, United States
| | - Sayan Kundu
- Department
of Chemistry, University of Florida, Gainesville, Florida32611, United States
| | - Zhongwu Guo
- Department
of Chemistry, University of Florida, Gainesville, Florida32611, United States
- UF
Health Cancer Center, University of Florida, Gainesville, Florida32611, United States
| |
Collapse
|
2
|
Alshabrmi FM. Multi-Omics Analysis of the virulence factors and designing of next-generation multi-epitopes Vaccines against Rickettsia prowazekii: a computer-aided vaccine designing approach. J Comput Aided Mol Des 2025; 39:25. [PMID: 40418389 DOI: 10.1007/s10822-025-00603-6] [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: 02/24/2025] [Accepted: 04/27/2025] [Indexed: 05/27/2025]
Abstract
Rickettsia is a genus of bacteria that are obligate intracellular parasites and are responsible for the febrile diseases known collectively as Rickettsioses. The emergence of antibiotic resistance is an escalating concern and thus developing a vaccine against Rickettsia is of paramount importance due to the significant public health threat posed by these bacteria. Thus, we employed structural vaccinology guided by machine learning algorithms to explore the virulence landscape of Rickettsia prowazekii to design a multi-epitopes-based vaccine (MEVC) that is immunogenic and safe. From a pool of virulence factors, we shortlisted five targets including sca0, sca1, sca4, sca5 and tlyA that were classified as non-allergenic as well as antigenic. The immune epitopes mapping results shortlisted five CTL epitopes, five HTL (IFN+) epitopes and five B cell epitopes as the best choice to design a vaccine construct of 475 amino acids. Various parameters were used to validate the designed MEVC which involved prediction of physiochemical properties, modeling and validation of the 3D structure, interaction with the immune receptors such as TLR2 (Toll-like receptor) and TLR4. Moreover, all-atoms simulation and binding free energy (BFE) results revealed a stable and favorable dynamic properties determined by these complexes. Jcat revealed that the improved sequence exhibits a GC content of 48.14% and a CAI (Codon Adaptation Index) value of 1.0. We used a multi-dose criterion at different time intervals i.e., 1st, 84th and 170th day to understand the immune potential of our constructed vaccine. The results provide a comprehensive overview of immune factors that ensure effective antigen memory cells generation after each injection, as predicted by the in silico pipeline. However, limitations in current algorithms particularly their inability to fully account for HLA polymorphism and the lack of experimental and clinical validation remain major shortcomings of the study. These issues should be addressed in future research to support the development of a robust immune response against Rickettsia infections.
Collapse
Affiliation(s)
- Fahad M Alshabrmi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, 51452, Saudi Arabia.
| |
Collapse
|
3
|
Kwon KW, Choi E, Kim H, Kim HW, Choi S, Lee S, Ha SJ, Shin SJ. Adjunctive beneficial effect of c-di-GMP, a STING agonist, in enhancing protective efficacy of TLR4-adjuvanted tuberculosis subunit vaccine formulations. J Biomed Sci 2025; 32:52. [PMID: 40414893 DOI: 10.1186/s12929-025-01144-8] [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: 10/29/2024] [Accepted: 05/06/2025] [Indexed: 05/27/2025] Open
Abstract
BACKGROUND Effective subunit vaccine development requires selecting appropriate adjuvant formulations to trigger desired adaptive immune responses. This study explores the immunogenicity and tuberculosis (TB) vaccine potential of antigens (Ags) combined with Toll-like receptor 4 (TLR4) adjuvants and a stimulator of interferon genes (STING) agonist. METHODS In this work, we investigated the combination of Ags with TLR4 adjuvants (monophosphoryl lipid A / dimethyldioctadecylammonium bromide; MPL/DDA or glucopyranosyl lipid adjuvant-stable emulsion; GLA-SE) and a STING agonist, c-di-GMP (CDG). Mice were immunized three times by intramuscular injections at 3-week intervals. The effects of integrating Ags in these adjuvant formulations on the immune response were evaluated, focusing on the generation of Th1-biased, polyfunctional Ag-specific CD4+ T cells and their localization in the lung and spleen. To assess protection, immunized mice were aerogenically challenged with either conventional or ultra-low doses of Mycobacterium tuberculosis (Mtb) 4 weeks after the last immunization. Subsequently, bacterial load and pulmonary inflammation were assessed. RESULTS Integrating ESAT6 Ag in TLR4 and CDG adjuvant formulations remarkably boosted Th1-biased, polyfunctional ESAT6-specific CD4+ T cells in the lungs and spleen, providing durable protection against Mtb infection. The inclusion of CDG promoted mucosal localization of ESAT6-specific CD4+ T cells resembling resident memory phenotypes in the lung parenchyma and increased Ag-specific CD4+ T cells in lung vasculature. Immunization with another vaccine Ag candidate, Ag85B, in GLA-SE plus CDG similarly increased Ag85B-specific CD4+ T cells in the spleen and both lung compartments. Following ultra-low dose Mtb challenge, ESAT6 or Ag85B/GLA-SE/CDG immunizations significantly reduced bacterial loads compared to non-, Bacillus Calmette-Guérin (BCG)-, and ESAT6 or Ag85B/GLA-SE-immunized groups. Importantly, the inclusion of CDG decreased killer cell lectin-like receptor subfamily G member 1 (KLRG1) expression among Ag-specific CD4+ T cells in the lung, correlating with enhanced lung-homing evidenced by expanded lung parenchyma Ag-specific CD4+ T cells, including less-differentiated Th1 cells. CONCLUSIONS This study highlights that CDG, when used in combination with TLR4 adjuvants, enhances long-term protective immunity, offering a promising strategy for subunit TB vaccine development.
Collapse
Affiliation(s)
- Kee Woong Kwon
- Department of Microbiology and Convergence of Medical Science, College of Medicine, Gyeongsang National University, Jinju, 52727, Republic of Korea
- Department of Microbiology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Eunsol Choi
- Department of Microbiology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Hagyu Kim
- Department of Microbiology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Hyeong Woo Kim
- Department of Microbiology and Convergence of Medical Science, College of Medicine, Gyeongsang National University, Jinju, 52727, Republic of Korea
| | - Sangwon Choi
- Department of Microbiology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Seunghyun Lee
- Department of Microbiology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Sang-Jun Ha
- Department of Biochemistry, College of Life Science & Biotechnology, Yonsei University, Seoul, 03722, South Korea
- Brain Korea 21 (BK21) FOUR Program, Yonsei Education & Research Center for Biosystems, Yonsei University, Seoul, 03722, Republic of Korea
| | - Sung Jae Shin
- Department of Microbiology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, 03722, South Korea.
- Institute for Immunology and Immunological Disease, Yonsei University College of Medicine, Seoul, 03722, South Korea.
| |
Collapse
|
4
|
Esmaeili H, Joghataei SM, Hamidiya Z, Arani EB, Shakeri AP, Rad ZN, Safari MM. Optimal immunization strategies for Saanen goats against goatpox. BMC Vet Res 2025; 21:328. [PMID: 40336081 PMCID: PMC12060465 DOI: 10.1186/s12917-025-04783-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Accepted: 04/23/2025] [Indexed: 05/09/2025] Open
Abstract
BACKGROUND Goatpox is a highly contagious disease caused by the Capripoxvirus, primarily affecting sheep and goats. Breeds like the Saanen are particularly vulnerable, especially in enzootic areas, and face risks not only from the disease itself but also from adverse reactions to live attenuated vaccines. This study compares inactivated and attenuated vaccines to find the safest and most effective vaccination strategy for Saanen goats against the Goatpox Virus (GTPV). In this study, 375 pure-breed Saanen goats were strategically divided into four groups to explore the most effective vaccination protocols for combating combinations of vaccines. In contrast, one group remained unvaccinated as a control. After vaccination, the goats were challenged by exposure to naturally infected animals to assess the vaccines' protective efficacy. PCR assays and the Virus Neutralization method were also conducted. RESULTS Group G1 exhibited no adverse reactions following two inactivated vaccines, with only mild and brief signs observed in a small number of goats (2%) after the live attenuated vaccine. Group G2, which received an inactivated vaccine followed by a live attenuated vaccine, had mild lesions in 18.66% of the goats after vaccination. In contrast, Group G3, which only received the live attenuated vaccine, showed a high morbidity rate of 82% and a mortality rate of 22%, with severe clinical signs and Pox lesions. Following the challenge, no signs of GTPV infection were observed in Groups G1, G2, and G3, whereas the control group exhibited 100% morbidity and 72% mortality, confirming the vaccine's protective efficacy. CONCLUSION This study found that a protocol using two inactivated vaccine doses at one-month intervals, followed by a live attenuated vaccine and annual boosters, effectively immunizes vulnerable breeds against GTPV without causing adverse reactions. This approach prevents complications and supports breeding in GTPV-enzootic regions.
Collapse
Affiliation(s)
- Hossein Esmaeili
- Department of Microbiology and Immunology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran.
- Faculty of Veterinary Medicine, University of Tehran, Azadi Street, Postal code: 1419963114, Tehran, Iran.
| | - Seyed Mehdi Joghataei
- Department of Microbiology and Immunology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Zeinab Hamidiya
- Department of Basic Sciences, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Ehsan Baghal Arani
- Department of Microbiology and Immunology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Amir Pasha Shakeri
- Department of Microbiology and Immunology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Zia Nosrat Rad
- Department of Microbiology and Immunology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Mohammad Mahdi Safari
- Department of Microbiology and Immunology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| |
Collapse
|
5
|
Movsisyan M, Harutyunyan H, Movsisyan K, Kasparova I, Hakobyan A, Yenkoyan K. Age-related peculiarities of antibody-mediated humoral immune response following SARS-CoV-2 infection. Exp Gerontol 2025; 203:112735. [PMID: 40120835 DOI: 10.1016/j.exger.2025.112735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 03/13/2025] [Accepted: 03/19/2025] [Indexed: 03/25/2025]
Abstract
Thousands of articles were published about the COVID-19 disease and hundreds about the immune response. But still little is known about the features of SARS-CoV-2-specific immunity in elderly. The aim of current research was to evaluate the age-related peculiarities of antibody mediated humoral immune response following SARS-CoV-2 infection. Our study presents an intriguing divergence from the classical concept of immunosenescence, where aging has been assumed to cause poor antibody responses, reduced or inefficient vaccination, and overall blunted immune responses in elderly people. Our findings were opposite to some of these expectations; participants aged over 60 expressed elevated titers of anti-SARS-CoV-2 antibodies in comparison to younger adults. Analyzing the data of relative neutralization and avidity of anti-SARS-Cov-2 (S) antibodies we propose that although older adults produce a higher quantity of antibodies, their functional efficiency appears relatively reduced exhibiting lower neutralizing capacity and binding strength per antibody compared to younger adults. We can assume that the immune system of the elderly may require a higher level of antibody production to obtain a comparable level of protection. Our findings highlight the intricate nature of immune responses in convalescent older adults. This has particular relevance to understanding immunity and vaccine responses in different age groups.
Collapse
Affiliation(s)
- M Movsisyan
- Neuroscience Laboratory, Cobrain Center, Yerevan State Medical University named after Mkhitar Heratsi, 0025 Yerevan, Armenia; Department of Allergology and Clinical Immunology, Yerevan State Medical University named after Mkhitar Heratsi, 0025 Yerevan, Armenia
| | - H Harutyunyan
- Neuroscience Laboratory, Cobrain Center, Yerevan State Medical University named after Mkhitar Heratsi, 0025 Yerevan, Armenia
| | - Kh Movsisyan
- Neuroscience Laboratory, Cobrain Center, Yerevan State Medical University named after Mkhitar Heratsi, 0025 Yerevan, Armenia
| | - I Kasparova
- Department of Histology, Yerevan State Medical University named after Mkhitar Heratsi, 0025 Yerevan, Armenia
| | - A Hakobyan
- Department of Allergology and Clinical Immunology, Yerevan State Medical University named after Mkhitar Heratsi, 0025 Yerevan, Armenia
| | - K Yenkoyan
- Neuroscience Laboratory, Cobrain Center, Yerevan State Medical University named after Mkhitar Heratsi, 0025 Yerevan, Armenia; Department of Biochemistry, Yerevan State Medical University named after Mkhitar Heratsi, 0025 Yerevan, Armenia.
| |
Collapse
|
6
|
Pang S, Wang L, Liu M, Shao M, Zhu G, Duan Q. Truncated flagellin lacking the hypervariable region: A structural basis for improved immune responses and adjuvanticity. Int J Biol Macromol 2025; 308:142742. [PMID: 40180103 DOI: 10.1016/j.ijbiomac.2025.142742] [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: 12/15/2024] [Revised: 03/02/2025] [Accepted: 03/31/2025] [Indexed: 04/05/2025]
Abstract
Bacterial flagellins are recognized for their potent immunomodulatory properties and potential as vaccine adjuvants. They activate innate and adaptive immune responses by interacting with Toll-like receptor 5 (TLR5) and the cytosolic NOD-like receptor protein 4 (NLRC4) inflammasome, thereby enhancing immune responses. This study investigates the impact of various truncated flagellin derivatives, derived from Escherichia coli (EHEC EDL933) and lacking specific domains, on TLR5 activation and their adjuvant properties. We generated several truncated flagellin mutants and assessed their ability to activate TLR5 in vitro and their immunoadjuvant effects in vivo. Our data show that only the FliCH7, FliCNC, FliCH7-FaeG, and FliCNC-FaeG proteins, which lack the hypervariable region (HVP) but retain both the amino- and carboxy-terminal regions, significantly enhanced TNF-α and IL-8 production compared to other flagellin derivatives. These findings underscore the essential roles of both conserved terminal regions in TLR5 activation. Notably, the FliCNC truncated mutant exhibited TLR5 activation comparable to that of native flagellin and induced higher antibody titers when co-administered with a model antigen or used as a fusion protein. Our results suggest that the HVR is not essential for flagellin's immunoadjuvant activity and that its removal enhances flagellin's ability to activate the innate immune system. This study provides valuable insights into optimizing flagellin derivatives for vaccine development, offering a more potent platform for enhancing immune responses against a range of pathogens.
Collapse
Affiliation(s)
- Shengmei Pang
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Joint Laboratory for International Cooperation in Agriculture and Agricultural Product Safety, Ministry of Education, Yangzhou 225009, China; Joint Laboratory of International Cooperation on Prevention and Control Technology of Important Animal Diseases and Zoonoses of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou, Jiangsu, China
| | - Longlong Wang
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Joint Laboratory for International Cooperation in Agriculture and Agricultural Product Safety, Ministry of Education, Yangzhou 225009, China; Joint Laboratory of International Cooperation on Prevention and Control Technology of Important Animal Diseases and Zoonoses of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou, Jiangsu, China
| | - Mei Liu
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Joint Laboratory for International Cooperation in Agriculture and Agricultural Product Safety, Ministry of Education, Yangzhou 225009, China; Joint Laboratory of International Cooperation on Prevention and Control Technology of Important Animal Diseases and Zoonoses of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou, Jiangsu, China
| | - Mingqing Shao
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Joint Laboratory for International Cooperation in Agriculture and Agricultural Product Safety, Ministry of Education, Yangzhou 225009, China; Joint Laboratory of International Cooperation on Prevention and Control Technology of Important Animal Diseases and Zoonoses of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou, Jiangsu, China
| | - Guoqiang Zhu
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Joint Laboratory for International Cooperation in Agriculture and Agricultural Product Safety, Ministry of Education, Yangzhou 225009, China; Joint Laboratory of International Cooperation on Prevention and Control Technology of Important Animal Diseases and Zoonoses of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou, Jiangsu, China
| | - Qiangde Duan
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Joint Laboratory for International Cooperation in Agriculture and Agricultural Product Safety, Ministry of Education, Yangzhou 225009, China; Joint Laboratory of International Cooperation on Prevention and Control Technology of Important Animal Diseases and Zoonoses of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou, Jiangsu, China.
| |
Collapse
|
7
|
Ghazizadeh Y, Salehi Shadkami H, Madani F, Niknam S, Adabi M. Advances in cancer nanovaccines: a focus on colorectal cancer. Nanomedicine (Lond) 2025; 20:1029-1041. [PMID: 40186876 PMCID: PMC12051617 DOI: 10.1080/17435889.2025.2486930] [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: 02/04/2025] [Accepted: 03/27/2025] [Indexed: 04/07/2025] Open
Abstract
Nanotechnology has revolutionized cancer treatment by providing innovative solutions through nanocancer therapies, nanovaccines, and nanoparticles. This review focuses on the application of these technologies in colorectal cancer (CRC), highlighting their progression from preclinical studies to clinical trials. Nanoparticles, including liposomes, silica, gold, and lipid nanoparticles, possess unique properties that enhance drug delivery, improve therapeutic efficacy, and minimize systemic toxicity. Additionally, nanovaccines are being developed to elicit robust immune responses against CRC cells. This paper offers a comprehensive overview of the current state of nanotechnology-based treatments for CRC, emphasizing key preclinical studies and clinical trials that demonstrate their potential. Furthermore, the review discusses the challenges faced in this field. It outlines future directions for research, underscoring the need for ongoing efforts to translate these promising technologies into practical clinical applications.
Collapse
Affiliation(s)
- Yalda Ghazizadeh
- Student Research Committee, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Nanomedicine Student Association (NMA), Student’s Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Hossein Salehi Shadkami
- Nanomedicine Student Association (NMA), Student’s Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Medical Science, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Madani
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Sedigheh Niknam
- Institute of Nano Science and Nano Technology, University of Kashan, Kashan, Iran
| | - Mahdi Adabi
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Food Microbiology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
8
|
Zhang W, Wang C, Meng Y, He L, Dong M. EBV Vaccines in the Prevention and Treatment of Nasopharyngeal Carcinoma. Vaccines (Basel) 2025; 13:478. [PMID: 40432090 PMCID: PMC12115577 DOI: 10.3390/vaccines13050478] [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: 03/16/2025] [Revised: 04/21/2025] [Accepted: 04/22/2025] [Indexed: 05/29/2025] Open
Abstract
Epstein-Barr virus (EBV), a ubiquitous human herpesvirus, has been robustly linked to the pathogenesis of nasopharyngeal carcinoma (NPC). The mechanism of EBV-induced NPC involves complex interactions between viral proteins and host cell pathways. This review aims to comprehensively outline the mechanism of EBV-induced NPC and the latest advances in targeted EBV vaccines for prophylaxis and treatment. This review explores the intricate molecular mechanisms by which EBV contributes to NPC pathogenesis, highlighting viral latency, genetic and epigenetic alterations, and immune evasion strategies. It emphasizes the pivotal role of key viral proteins, including EBNA1, LMP1, and LMP2A, in carcinogenesis. Subsequently, the discussion shifts towards the development of targeted EBV vaccines, including preventive vaccines aimed at preventing primary EBV infection and therapeutic vaccines aimed at treating diagnosed EBV-related NPC. The review underscores the challenges and future directions in the field, stressing the importance of developing innovative vaccine strategies and combination therapies to improve efficacy. This review synthesizes current insights into the molecular mechanisms of EBV-induced NPC and the development of EBV-targeted vaccines, highlighting the potential use of mRNA vaccines for NPC treatment.
Collapse
Affiliation(s)
- Weiwei Zhang
- Department of Oncology, Cancer Prevention and Treatment Institute of Chengdu, Chengdu Fifth People’s Hospital/The Second Clinical Medical College, Affiliated Fifth People’s Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 611130, China; (W.Z.)
| | - Chuang Wang
- Chengdu Yunce Medical Biotechnology Co., Ltd., Chengdu 611135, China;
| | - Yousheng Meng
- Department of Oncology, Cancer Prevention and Treatment Institute of Chengdu, Chengdu Fifth People’s Hospital/The Second Clinical Medical College, Affiliated Fifth People’s Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 611130, China; (W.Z.)
| | - Lang He
- Department of Oncology, Cancer Prevention and Treatment Institute of Chengdu, Chengdu Fifth People’s Hospital/The Second Clinical Medical College, Affiliated Fifth People’s Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 611130, China; (W.Z.)
| | - Mingqing Dong
- Division of Pulmonary Medicine, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou Key Laboratory of Interdiscipline and Translational Medicine, Wenzhou Key Laboratory of Heart and Lung, Wenzhou 325000, China
| |
Collapse
|
9
|
Rahman MM, Talukder A, Rahi MS, Das PK, Grice ID, Ulett GC, Wei MQ. Evaluation of Immunostimulatory Effects of Bacterial Lysate Proteins on THP-1 Macrophages: Pro-inflammatory Cytokine Response and Proteomic Profiling. J Immunol Res 2025; 2025:2289241. [PMID: 40322557 PMCID: PMC12048194 DOI: 10.1155/jimr/2289241] [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: 11/18/2024] [Accepted: 04/02/2025] [Indexed: 05/08/2025] Open
Abstract
Bacterial lysate proteins (BLPs) serve as potential immunostimulants, recognized by pattern recognition receptors (PRRs) on immune cells, eliciting a robust immune response. In this study, THP-1 macrophages were treated with varying doses of BLPs derived from Streptococcus pyogenes (SP), Streptococcus agalactiae (SA), and Serratia marcescens (SM). The results showed significant increases (p < 0.05) in pro-inflammatory cytokines such as TNF-α, IL-1β, IL-6, IL-12, granulocyte macrophage-colony stimulating factor (GM-CSF), eotaxin, and macrophage inflammatory protein (MIP)-1α, except for 5 µg of all BLPs for TNF-α and eotaxin, and 5 µg of SP for IL-12 production. No significant differences were found between the corresponding doses of SP and SA or SP and SM, except for GM-CSF in all doses, while SA and SM only showed a difference at the 5 µg dose for GM-CSF. Furthermore, there were no significant differences between the 10 and 20 µg doses of all BLPs, indicating that doses higher than 10 µg do not significantly enhance the pro-inflammatory response. Combination doses of SP + SM and SA + SM did not show significant differences, except for IL-1β, suggesting no synergistic effect. Cytotoxicity was observed to increase with higher BLP concentrations in a dose-dependent manner, with combinations of SP + SM and SA + SM exhibiting greater cytotoxicity than the individual BLPs. Proteomic analysis of BLPs identified immunostimulatory proteins, including heat shock proteins (HSPs; ClpB, DnaK, and GroEL), metabolic enzymes (glyceraldehyde 3-phosphate dehydrogenase (GAPDH), enolase, and arginine deiminase (ADI)), and surface and secreted proteins (ESAT-6-like protein, CRISPR-associated endonuclease Cas9, OmpA, porin OmpC, and serralysin), which are involved in immune modulation, bacterial clearance, and immune evasion. This study underscores the potential of bacterial proteins as vaccine adjuvants or supplementary therapies; however, further research is essential to find a balance between immune activation and inflammation reduction to develop safer and more effective immunostimulants.
Collapse
Affiliation(s)
- Md. Mijanur Rahman
- School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, Queensland, Australia
- Department of Microbiology, Noakhali Science and Technology University, Noakhali, Chittagong, Bangladesh
| | - Asma Talukder
- School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, Queensland, Australia
| | - Md. Sifat Rahi
- School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, Queensland, Australia
| | - Plabon Kumar Das
- Institute for Biomedicine and Glycomics, Griffith University, Gold Coast, Queensland, Australia
| | - I. Darren Grice
- School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, Queensland, Australia
- Institute for Biomedicine and Glycomics, Griffith University, Gold Coast, Queensland, Australia
| | - Glen C. Ulett
- School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, Queensland, Australia
- Institute for Biomedicine and Glycomics, Griffith University, Gold Coast, Queensland, Australia
| | - Ming Q. Wei
- School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, Queensland, Australia
| |
Collapse
|
10
|
Bonduelle O, Delory T, Franco-Moscardini I, Ghidi M, Bennacer S, Wokam M, Lenormand M, Petrier M, Rogeaux O, de Bernard S, Alves K, Nourikyan J, Lina B, INFLUOMICS Study group, Combadiere B, Janssen C. Boosting effect of high-dose influenza vaccination on innate immunity among elderly. JCI Insight 2025; 10:e184128. [PMID: 40036077 PMCID: PMC12016920 DOI: 10.1172/jci.insight.184128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 02/25/2025] [Indexed: 03/06/2025] Open
Abstract
BACKGROUNDThe high-dose quadrivalent influenza vaccine (QIV-HD) showed superior efficacy against laboratory-confirmed illness compared with the standard-dose quadrivalent influenza vaccine (QIV-SD) in randomized controlled trials with the elderly. However, specific underlying mechanism remains unclear.METHODSThis phase IV randomized controlled trial compared early innate responses induced by QIV-HD and QIV-SD in 59 individuals aged > 65 years. Systemic innate cells and gene signatures at day 0 (D0) and D1 as well as hemagglutinin inhibition antibody (HIA) titers at D0 and D21 after vaccination were assessed.RESULTSQIV-HD elicited robust humoral response with significantly higher antibody titers and seroconversion rates than QIV-SD. At D1 after vaccination, QIV-HD recipients showed significant reduction in innate cells, including conventional DCs and NK cells, compared with QIV-SD, correlating with significantly increased HIA titers at D21. Blood transcriptomic analysis revealed greater amplitude of gene expression in the QIV-HD arm, encompassing genes related to innate immune response, IFNs, and antigen processing and presentation, and correlated with humoral responses. Interestingly, comparative analysis with a literature dataset from young adults vaccinated with influenza standard-dose vaccine highlighted strong similarities in gene expression patterns and biological pathways with the elderly vaccinated with QIV-HD.CONCLUSIONQIV-HD induces higher HIA titers than QIV-SD, a youthful boost of the innate gene expression significantly associated with high HIA titers.TRIAL REGISTRATIONEudraCT no. 2021-004573-32.
Collapse
Affiliation(s)
- Olivia Bonduelle
- Sorbonne Université, Institut National de Santé et de Recherche Médicale, Inserm U1135, Centre d’Immunologie et des Maladies Infectieuses, Cimi-Paris, Paris, France
| | - Tristan Delory
- Centre Hospitalier Annecy Genevois, Epagny Metz-Tessy, France
| | - Isabelle Franco-Moscardini
- Sorbonne Université, Institut National de Santé et de Recherche Médicale, Inserm U1135, Centre d’Immunologie et des Maladies Infectieuses, Cimi-Paris, Paris, France
| | - Marion Ghidi
- Centre Hospitalier Annecy Genevois, Epagny Metz-Tessy, France
| | - Selma Bennacer
- Sorbonne Université, Institut National de Santé et de Recherche Médicale, Inserm U1135, Centre d’Immunologie et des Maladies Infectieuses, Cimi-Paris, Paris, France
| | - Michele Wokam
- Sorbonne Université, Institut National de Santé et de Recherche Médicale, Inserm U1135, Centre d’Immunologie et des Maladies Infectieuses, Cimi-Paris, Paris, France
| | | | - Melissa Petrier
- Centre Hospitalier Annecy Genevois, Epagny Metz-Tessy, France
| | - Olivier Rogeaux
- Centre Hospitalier Centre Hospitalier Métropole Savoie, Chambéry, France
| | | | | | | | - Bruno Lina
- Centre International de Recherche en Infectiologie, Université Claude Bernard Lyon-1, INSERM U1111, CNRS, UMR5308, ENS Lyon, Université Jean Monnet de Saint-Etienne, Lyon, France
- Laboratoire de Virologie, Institut des Agents Infectieux, Centre National de Référence des Virus des Infections Respiratoires, Hospices Civils de Lyon, Lyon, France
| | | | - Behazine Combadiere
- Sorbonne Université, Institut National de Santé et de Recherche Médicale, Inserm U1135, Centre d’Immunologie et des Maladies Infectieuses, Cimi-Paris, Paris, France
| | - Cécile Janssen
- Centre Hospitalier Annecy Genevois, Epagny Metz-Tessy, France
| |
Collapse
|
11
|
Saha A, Ghosh Roy S, Dwivedi R, Tripathi P, Kumar K, Nambiar SM, Pathak R. Beyond the Pandemic Era: Recent Advances and Efficacy of SARS-CoV-2 Vaccines Against Emerging Variants of Concern. Vaccines (Basel) 2025; 13:424. [PMID: 40333293 PMCID: PMC12031379 DOI: 10.3390/vaccines13040424] [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: 02/17/2025] [Revised: 04/10/2025] [Accepted: 04/14/2025] [Indexed: 05/09/2025] Open
Abstract
Vaccination has been instrumental in curbing the transmission of SARS-CoV-2 and mitigating the severity of clinical manifestations associated with COVID-19. Numerous COVID-19 vaccines have been developed to this effect, including BioNTech-Pfizer and Moderna's mRNA vaccines, as well as adenovirus vector-based vaccines such as Oxford-AstraZeneca. However, the emergence of new variants and subvariants of SARS-CoV-2, characterized by enhanced transmissibility and immune evasion, poses significant challenges to the efficacy of current vaccination strategies. In this review, we aim to comprehensively outline the landscape of emerging SARS-CoV-2 variants of concern (VOCs) and sub-lineages that have recently surfaced in the post-pandemic years. We assess the effectiveness of existing vaccines, including their booster doses, against these emerging variants and subvariants, such as BA.2-derived sub-lineages, XBB sub-lineages, and BA.2.86 (Pirola). Furthermore, we discuss the latest advancements in vaccine technology, including multivalent and pan-coronavirus approaches, along with the development of several next-generation coronavirus vaccines, such as exosome-based, virus-like particle (VLP), mucosal, and nanomaterial-based vaccines. Finally, we highlight the key challenges and critical areas for future research to address the evolving threat of SARS-CoV-2 subvariants and to develop strategies for combating the emergence of new viral threats, thereby improving preparedness for future pandemics.
Collapse
Affiliation(s)
- Ankita Saha
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA;
| | - Sounak Ghosh Roy
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Naval Medical Research Command, Silver Spring, MD 20910, USA;
| | - Richa Dwivedi
- Department of Microbiology, Immunology, and Physiology, Meharry Medical College, Nashville, TN 37208, USA;
| | - Prajna Tripathi
- Department of Microbiology and Immunology, Weill Cornell Medical College, New York, NY 10021, USA;
| | - Kamal Kumar
- Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA 92093, USA;
| | - Shashank Manohar Nambiar
- Division of Hepatology, Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA;
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA
| | - Rajiv Pathak
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA
| |
Collapse
|
12
|
Guerrero GG, Madrid-Marina V, Martínez-Romero A, Torres-Poveda K, Favela-Hernández JM. Host-Pathogen Interaction Interface: Promising Candidate Targets for Vaccine-Induced Protective and Memory Immune Responses. Vaccines (Basel) 2025; 13:418. [PMID: 40333316 PMCID: PMC12031405 DOI: 10.3390/vaccines13040418] [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: 02/21/2025] [Revised: 04/09/2025] [Accepted: 04/11/2025] [Indexed: 05/09/2025] Open
Abstract
Vaccine formulations are a successful strategy against pathogen transmission because vaccine candidates induce effective and long-lasting memory immune responses (B and CD4+ T cells) at systemic and mucosal sites. Extracellular vesicles of lipoproteins, bioactive compounds from plants and invertebrates (sponges) encapsulated in liposomes, and glycoproteins can target these sites. The vaccine candidates developed can mimic microbial pathogens in a way that successfully links the innate and adaptive immune responses. In addition, vaccines plus adjuvants promote and maintain an inflammatory response. In this review, we aimed to identify the host-pathogen interface as a rich source of candidate targets for vaccine-induced protective and long-lasting memory immune responses.
Collapse
Affiliation(s)
- Gloria G. Guerrero
- Unidad Académica de Ciencias Biológicas, Universidad Autónoma de Zacatecas, Zacatecas 98600, Zac., Mexico
| | - Vicente Madrid-Marina
- Centro de Investigación en Enfermedades infecciosas (CISEI), Instituto Nacional de Salud Pública (INSP), Cuernavaca 62100, Mor., Mexico
| | - Aurora Martínez-Romero
- Facultad de Química, Universidad Juárez del Estado de Durango, Gómez Palacio 34100, Dgo., Mexico
| | - Kirvis Torres-Poveda
- Centro de Investigación en Enfermedades infecciosas (CISEI), Instituto Nacional de Salud Pública (INSP), Cuernavaca 62100, Mor., Mexico
- Secretaria de Ciencia, Humanidades y Tecnologías (SECIHTI), Instituto Nacional de Salud Pública, Cuernavaca 62100, Mor., Mexico
| | - Juan Manuel Favela-Hernández
- Facultad de Química, Universidad Juárez del Estado de Durango, Gómez Palacio 34100, Dgo., Mexico
- Instituto Multidisciplinario de Ciencias “Avicena”, Torreón 27250, Coah., Mexico
| |
Collapse
|
13
|
Montgomery L, Larbi A. Monitoring Immune Responses to Vaccination: A Focus on Single-Cell Analysis and Associated Challenges. Vaccines (Basel) 2025; 13:420. [PMID: 40333304 PMCID: PMC12030821 DOI: 10.3390/vaccines13040420] [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: 03/17/2025] [Revised: 04/13/2025] [Accepted: 04/15/2025] [Indexed: 05/09/2025] Open
Abstract
Monitoring the immune response to vaccination encompasses both significant challenges and promising opportunities for scientific advancement. The primary challenge lies in the inherent complexity and interindividual variability of immune responses, influenced by factors including age, genetic background, and prior immunological history. This variability necessitates the development of sophisticated, highly sensitive assays capable of accurately quantifying immune parameters such as antibody titers, T-cell responses, and cytokine profiles. Furthermore, the temporal dynamics of the immune response require comprehensive longitudinal studies to elucidate the durability and quality of vaccine-induced immunity. Challenges of this magnitude pave the way for immunological research advancements and diagnostic methodologies. Cutting-edge monitoring techniques, such as high-throughput sequencing and advanced flow cytometry, enable deeper insights into the mechanistic underpinnings of vaccine efficacy and contribute to the iterative design of more effective vaccines. Additionally, the integration of analytical tools holds the potential to predict immune responses and tailor personalized vaccination strategies. This will be addressed in this review to provide insight for enhancing public health outcomes and fortifying preparedness against future infectious disease threats.
Collapse
Affiliation(s)
- LaToya Montgomery
- Medical and Scientific Affairs, Beckman Coulter Life Sciences, Brea, CA 92821, USA;
| | - Anis Larbi
- Medical and Scientific Affairs, Beckman Coulter Life Sciences, Brea, CA 92821, USA;
- Department of Medicine, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, QC J1K 2R1, Canada
| |
Collapse
|
14
|
Welch M, Krueger K, Zhang J, Piñeyro P, Mogler M, Strait E, Gauger P. Experimental Efficacy of an Alphavirus Vectored RNA Particle Vaccine Against Porcine Parainfluenza Virus-1 in Conventional Weaned Pigs. Viruses 2025; 17:565. [PMID: 40285006 PMCID: PMC12031517 DOI: 10.3390/v17040565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2025] [Revised: 04/08/2025] [Accepted: 04/08/2025] [Indexed: 04/29/2025] Open
Abstract
Parainfluenza viruses are a common cause of respiratory illness in many species. In this study, experimental, alphavirus-derived RNA particle vaccines either with or without adjuvant were evaluated against porcine parainfluenza virus 1 (PPIV1) challenge and compared to live virus exposure. Groups of ten, three-week-old pigs were vaccinated intramuscularly with an adjuvanted RNA particle (RPAdj/C) or non-adjuvanted RP (RP/C) or administered an intranasal live exposure (LE/C) dose of PPIV1 at 0- and 21-days post vaccination (DPV) followed by challenge with PPIV1 at 40 DPV. In addition, two groups were included as non-vaccinated, non-challenged (NV/NC) and non-vaccinated, challenged (NV/C) controls. Intranasal virus exposure and RP vaccination, regardless of adjuvant, reduced PPIV1 shedding in nasal swabs by 5 days post inoculation (DPI). All vaccinated or exposed pigs seroconverted as shown by enzyme-linked immunosorbent assay and serum virus neutralization. The antibody isotype detected in bronchoalveolar lavage fluid (BALF) LE/C was predominantly IgA while RP vaccination induced an IgG response. Reduced PPIV1 antigen was observed in the LE/C, RP/C and RPAdj/C groups in lung, trachea, or nasal turbinate epithelium. Additionally, the RPAdj vaccine significantly reduced nasal shedding compared to NV/C pigs although not as much as LE/C pigs. These results suggest vaccination could mitigate PPIV1 infection in commercial systems.
Collapse
Affiliation(s)
- Michael Welch
- Department of Veterinary Diagnostic and Production Animal Medicine, College of Veterinary Medicine, Iowa State University, 1800 Christensen Drive, Ames, IA 50011, USA; (M.W.); (K.K.); (J.Z.); (P.P.)
| | - Karen Krueger
- Department of Veterinary Diagnostic and Production Animal Medicine, College of Veterinary Medicine, Iowa State University, 1800 Christensen Drive, Ames, IA 50011, USA; (M.W.); (K.K.); (J.Z.); (P.P.)
| | - Jianqiang Zhang
- Department of Veterinary Diagnostic and Production Animal Medicine, College of Veterinary Medicine, Iowa State University, 1800 Christensen Drive, Ames, IA 50011, USA; (M.W.); (K.K.); (J.Z.); (P.P.)
| | - Pablo Piñeyro
- Department of Veterinary Diagnostic and Production Animal Medicine, College of Veterinary Medicine, Iowa State University, 1800 Christensen Drive, Ames, IA 50011, USA; (M.W.); (K.K.); (J.Z.); (P.P.)
| | - Mark Mogler
- Merck Animal Health, 1102 Southern Hills Dr #101, Ames, IA 50010, USA; (M.M.); (E.S.)
| | - Erin Strait
- Merck Animal Health, 1102 Southern Hills Dr #101, Ames, IA 50010, USA; (M.M.); (E.S.)
| | - Phillip Gauger
- Department of Veterinary Diagnostic and Production Animal Medicine, College of Veterinary Medicine, Iowa State University, 1800 Christensen Drive, Ames, IA 50011, USA; (M.W.); (K.K.); (J.Z.); (P.P.)
| |
Collapse
|
15
|
Liu R, Yan S, Zhang Z, Xu J, Meng Y, Zhang Q, Wei L, Zou Q, Cui F. PLM-IL4: Enhancing IL-4-inducing peptide prediction with protein language model. Comput Biol Chem 2025; 118:108448. [PMID: 40203796 DOI: 10.1016/j.compbiolchem.2025.108448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 03/12/2025] [Accepted: 03/27/2025] [Indexed: 04/11/2025]
Abstract
Despite progress in developing antiviral drugs and vaccines, infections continue to be a significant challenge. Interleukin-4 (IL-4) is crucial for regulating immune responses and mediating allergic reactions. This research aims to improve the predictive accuracy of IL-4-inducing peptides by tackling data imbalance and enhancing feature extraction. Specifically, we introduce a new approach that utilizes SMOTE and ENN for balancing the dataset and applies a 30-layer ESM-2 model for extracting deep features. The extracted features are subsequently processed through a Gated Recurrent Unit (GRU) model, which is optimized through hyperparameter tuning. Our method achieves notable improvements, with an AUC of 0.98 and an accuracy of 93.1 %, highlighting its potential to support future immunotherapy and vaccine development efforts. The PLM-IL4 web server is freely accessible at http://www.bioai-lab.com/PLM-IL4, and the datasets used in this research are also available for download from the website.
Collapse
Affiliation(s)
- Ruiqi Liu
- School of Computer Science and Technology, Hainan University, Haikou 570228, China
| | - Shankai Yan
- School of Computer Science and Technology, Hainan University, Haikou 570228, China
| | - Zilong Zhang
- School of Computer Science and Technology, Hainan University, Haikou 570228, China
| | - Junlin Xu
- School of Computer Science and Technology, Wuhan University of Science and Technology, Wuhan, Hubei 430081, China
| | - Yajie Meng
- School of Computer Science and Artificial Intelligence, Wuhan Textile University, Wuhan, Hubei 430200, China
| | - Qingchen Zhang
- School of Computer Science and Technology, Hainan University, Haikou 570228, China
| | - Leyi Wei
- Centre for Artificial Intelligence driven Drug Discovery, Faculty of Applied Science, Macao Polytechnic University, Macao; School of Informatics, Xiamen University, Xiamen, China
| | - Quan Zou
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Feifei Cui
- School of Computer Science and Technology, Hainan University, Haikou 570228, China.
| |
Collapse
|
16
|
Wagoner ZW, Yates TB, Hernandez-Davies JE, Sureshchandra S, Joloya EM, Jain A, de Assis R, Kastenschmidt JM, Sorn AM, Mitul MT, Tamburini I, Ahuja G, Zhong Q, Trask D, Seldin M, Davies DH, Wagar LE. Systems immunology analysis of human immune organoids identifies host-specific correlates of protection to different influenza vaccines. Cell Stem Cell 2025; 32:529-546.e6. [PMID: 39986275 PMCID: PMC11974613 DOI: 10.1016/j.stem.2025.01.014] [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: 04/24/2024] [Revised: 11/27/2024] [Accepted: 01/27/2025] [Indexed: 02/24/2025]
Abstract
Vaccines are an essential tool to significantly reduce pathogen-related morbidity and mortality. However, our ability to rationally design vaccines and identify correlates of protection remains limited. Here, we employed an immune organoid approach to capture human adaptive immune response diversity to influenza vaccines and systematically identify host and antigen features linked to vaccine response variability. Our investigation identified established and unique immune signatures correlated with neutralizing antibody responses across seven different influenza vaccines and antigens. Unexpectedly, heightened ex vivo tissue frequencies of T helper (Th)1 cells emerged as both a predictor and a correlate of neutralizing antibody responses to inactivated influenza vaccines (IIVs). Secondary analysis of human public data confirmed that elevated Th1 signatures are associated with antibody responses following in vivo vaccination. These findings demonstrate the utility of human in vitro models for identifying in vivo correlates of protection and establish a role for Th1 functions in influenza vaccination.
Collapse
Affiliation(s)
- Zachary W Wagoner
- Department of Physiology & Biophysics, University of California, Irvine, Irvine, CA, USA; Institute for Immunology, University of California, Irvine, Irvine, CA, USA; Center for Virus Research, University of California, Irvine, Irvine, CA, USA; Vaccine R&D Center, University of California, Irvine, Irvine, CA, USA
| | - Timothy B Yates
- Department of Physiology & Biophysics, University of California, Irvine, Irvine, CA, USA; Institute for Immunology, University of California, Irvine, Irvine, CA, USA; Center for Virus Research, University of California, Irvine, Irvine, CA, USA; Vaccine R&D Center, University of California, Irvine, Irvine, CA, USA
| | - Jenny E Hernandez-Davies
- Department of Physiology & Biophysics, University of California, Irvine, Irvine, CA, USA; Institute for Immunology, University of California, Irvine, Irvine, CA, USA; Center for Virus Research, University of California, Irvine, Irvine, CA, USA; Vaccine R&D Center, University of California, Irvine, Irvine, CA, USA
| | - Suhas Sureshchandra
- Department of Physiology & Biophysics, University of California, Irvine, Irvine, CA, USA; Institute for Immunology, University of California, Irvine, Irvine, CA, USA; Center for Virus Research, University of California, Irvine, Irvine, CA, USA; Vaccine R&D Center, University of California, Irvine, Irvine, CA, USA
| | - Erika M Joloya
- Department of Physiology & Biophysics, University of California, Irvine, Irvine, CA, USA; Institute for Immunology, University of California, Irvine, Irvine, CA, USA; Center for Virus Research, University of California, Irvine, Irvine, CA, USA; Vaccine R&D Center, University of California, Irvine, Irvine, CA, USA
| | - Aarti Jain
- Department of Physiology & Biophysics, University of California, Irvine, Irvine, CA, USA; Institute for Immunology, University of California, Irvine, Irvine, CA, USA; Vaccine R&D Center, University of California, Irvine, Irvine, CA, USA
| | - Rafael de Assis
- Department of Physiology & Biophysics, University of California, Irvine, Irvine, CA, USA; Institute for Immunology, University of California, Irvine, Irvine, CA, USA; Vaccine R&D Center, University of California, Irvine, Irvine, CA, USA
| | - Jenna M Kastenschmidt
- Department of Physiology & Biophysics, University of California, Irvine, Irvine, CA, USA; Institute for Immunology, University of California, Irvine, Irvine, CA, USA; Center for Virus Research, University of California, Irvine, Irvine, CA, USA; Vaccine R&D Center, University of California, Irvine, Irvine, CA, USA
| | - Andrew M Sorn
- Department of Physiology & Biophysics, University of California, Irvine, Irvine, CA, USA; Institute for Immunology, University of California, Irvine, Irvine, CA, USA; Center for Virus Research, University of California, Irvine, Irvine, CA, USA; Vaccine R&D Center, University of California, Irvine, Irvine, CA, USA
| | - Mahina Tabassum Mitul
- Department of Physiology & Biophysics, University of California, Irvine, Irvine, CA, USA; Institute for Immunology, University of California, Irvine, Irvine, CA, USA; Center for Virus Research, University of California, Irvine, Irvine, CA, USA; Vaccine R&D Center, University of California, Irvine, Irvine, CA, USA
| | - Ian Tamburini
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA, USA; Center for Epigenetics and Metabolism, University of California, Irvine, Irvine, CA, USA
| | - Gurpreet Ahuja
- Department of Pediatric Otolaryngology, Children's Hospital of Orange County, Orange, CA, USA; Department of Otolaryngology-Head and Neck Surgery, University of California, Irvine, Orange, CA, USA
| | - Qiu Zhong
- Department of Pediatric Otolaryngology, Children's Hospital of Orange County, Orange, CA, USA; Department of Otolaryngology-Head and Neck Surgery, University of California, Irvine, Orange, CA, USA
| | - Douglas Trask
- Department of Otolaryngology-Head and Neck Surgery, University of California, Irvine, Orange, CA, USA
| | - Marcus Seldin
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA, USA; Center for Epigenetics and Metabolism, University of California, Irvine, Irvine, CA, USA
| | - D Huw Davies
- Department of Physiology & Biophysics, University of California, Irvine, Irvine, CA, USA; Institute for Immunology, University of California, Irvine, Irvine, CA, USA; Center for Virus Research, University of California, Irvine, Irvine, CA, USA; Vaccine R&D Center, University of California, Irvine, Irvine, CA, USA
| | - Lisa E Wagar
- Department of Physiology & Biophysics, University of California, Irvine, Irvine, CA, USA; Institute for Immunology, University of California, Irvine, Irvine, CA, USA; Center for Virus Research, University of California, Irvine, Irvine, CA, USA; Vaccine R&D Center, University of California, Irvine, Irvine, CA, USA.
| |
Collapse
|
17
|
Yaraghi P, Kheyri A, Mikaeili N, Boroumand A, Abbasifard M, Farhangnia P, Rezagholizadeh F, Khorramdelazad H. Nanoparticle-mediated enhancement of DNA Vaccines: Revolutionizing immunization strategies. Int J Biol Macromol 2025; 302:140558. [PMID: 39900152 DOI: 10.1016/j.ijbiomac.2025.140558] [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: 12/10/2024] [Revised: 01/25/2025] [Accepted: 01/30/2025] [Indexed: 02/05/2025]
Abstract
DNA vaccines are a novel form of vaccination that aims to harness genetic material to produce targeted immune responses. Nevertheless, their therapeutic application is hampered by low transfection efficacy, immunogenicity, and instability. Nanoparticle (NP) - based delivery systems are beneficial in enhancing DNA stability, increasing DNA uptake by antigen-presenting cells (APCs), and controlling antigen release. Some key progress includes the polymeric, lipid-based, and hybrid NPs and biocompatible carriers with inherent adjuvant effects. These systems have helped to enhance the antigen cross-presentation and T-cell activation significantly. In addition, biocompatible hybrid nanocarriers, antigen cross-presentation strategies, and next-generation sequencing (NGS) technologies are speeding up the identification of new antigens, while AI and machine learning are facilitating the development of efficient delivery systems. This review aims to assess how NPs have contributed to improving the effectiveness of DNA vaccines for treating diseases, cancer, and emerging diseases, as well as advancing the next generation of DNA vaccines.
Collapse
Affiliation(s)
- Pegah Yaraghi
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Abbas Kheyri
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Narges Mikaeili
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Armin Boroumand
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Mitra Abbasifard
- Department of Internal Medicine, School of Medicine, Ali-Ibn Abi-Talib Hospital, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.
| | - Pooya Farhangnia
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Fereshteh Rezagholizadeh
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Hossein Khorramdelazad
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.
| |
Collapse
|
18
|
Bhadouria N, Yadav S, Bukke SPN, Narapureddy BR. Advancements in vaccine delivery: harnessing 3D printing for microneedle patch technology. Ann Med Surg (Lond) 2025; 87:2059-2067. [PMID: 40212146 PMCID: PMC11981410 DOI: 10.1097/ms9.0000000000003060] [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: 09/17/2024] [Accepted: 01/31/2025] [Indexed: 04/13/2025] Open
Abstract
The development of 3D-printed microneedle (MN) technology is a significant step in vaccine delivery, providing a painless, effective, and adaptable substitute for conventional injection-based techniques. Direct transdermal vaccination distribution without the need for needles is made possible by microneedle patches, which employ a variety of tiny needles that dissolve when they penetrate the skin. By using 3D printing to precisely customise microneedles' size, shape, and density to meet particular vaccine requirements, administration control can be improved and vaccine efficiency may even be increased. Furthermore, rapid prototyping made possible by 3D printing speeds up the development process, enabling quicker testing and improvement of vaccines. Additionally, this scalable technology can greatly increase vaccine accessibility, particularly in environments with limited resources. Research indicates that by directly interacting with the skin's immune-rich layers, microneedle patches enhance antigen delivery and elicit a strong immune response. Because MN technology offers a useful, self-administrable vaccination approach with little waste, it has significant potential for use in public health applications, notably during pandemics. This study emphasises how 3D-printed microneedle patches have the potential to revolutionise vaccination procedures and increase vaccine accessibility globally.
Collapse
Affiliation(s)
- Namrata Bhadouria
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, Uttar Pradesh, India
| | - Shikha Yadav
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, Uttar Pradesh, India
| | - Sarad Pawar Naik Bukke
- Department of Pharmaceutics and Pharmaceutical Technology, Kampala International University, Western Campus, Ishaka-Bushenyi, Uganda
| | - Bayapa Reddy Narapureddy
- Department of Public Health, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| |
Collapse
|
19
|
Buus S, Vittrup DM, Schmidt JD, Jensen A, Stryhn A, Stensballe LG. Measles-mumps-rubella-vaccination at 6 months of age induces measles-specific T cell responses: a randomized controlled trial. Front Immunol 2025; 16:1546253. [PMID: 40165977 PMCID: PMC11955646 DOI: 10.3389/fimmu.2025.1546253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 02/11/2025] [Indexed: 04/02/2025] Open
Abstract
Background Measles is a highly contagious viral disease, particularly severe in infants. Protection in early life is provided by maternally transferred antibodies, but this period is shorter in infants of previously vaccinated mothers (PVMs) compared to infants of previously measles-infected mothers (PIMs). Earlier measles-mumps-rubella (MMR) vaccination may compensate for this. To evaluate immune responses, 6-month-old infants were randomized to receive early MMR or placebo. This study reports the cellular immune outcomes and summarizes serological and T-cell responses. Methods A double-blind, randomized trial involved 6540 Danish infants aged 5-7 months, eligible if birth weight exceeded 1000 grams and gestational age was ≥32 weeks. Participants were randomized 1:1 to receive M-M-RVaxPro or placebo. Blood samples were collected before intervention, four weeks after intervention, and four weeks after routine MMR at 15 months. Peripheral blood mononuclear cells (PBMCs) were prepared, and an IFN-γ specific ELISpot assay measured measles-specific T cells. Results Among 750 infants (341 MMR, 409 placebo) in the cellular immunogenicity trial, a significant cellular immune response was observed one-month post-intervention in the MMR group compared to placebo (geometric mean ratio [GMR]: 12.3; 95% CI: 6.9-21.9). The cellular conversion rate (CCR) in the MMR group was 45%, comparable to the previously reported seroconversion rate. However, following routine MMR at 15 months, a reduced cellular response was observed in the early MMR group (GMR: 0.6; 95% CI: 0.3-0.9). Post-routine MMR, CCRs were 66% (MMR) and 74% (placebo). The immune conversion rate (ICR, defined as seroconversion and/or T-cell response) reached 99% in both groups post-routine MMR. Conclusion Early MMR at 6 months elicited significant measles-specific cellular responses, though the CCR was lower than after routine MMR at 15 months. However, when combining serological and cellular responses, 99% of infants achieved immune conversion by 15 months. Early MMR could help reduce measles burden in infants in endemic settings without compromising subsequent immunizations. Clinical trial registration ClinicalTrials.gov, identifier NCT03780179, EudraCT 2016-001901-18.
Collapse
Affiliation(s)
- Søren Buus
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Dorthe Maria Vittrup
- The Child and Adolescent Department, The Juliane Marie Center, Copenhagen University Hospital, and Mary Elizabeth Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Jonas Damgård Schmidt
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Andreas Jensen
- The Child and Adolescent Department, The Juliane Marie Center, Copenhagen University Hospital, and Mary Elizabeth Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Anette Stryhn
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Lone Graff Stensballe
- The Child and Adolescent Department, The Juliane Marie Center, Copenhagen University Hospital, and Mary Elizabeth Hospital, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
20
|
Patel P, Patel B, Vyas SD, Patel MS, Hirani T, Haque M, Kumar S. A Narrative Review of Periodontal Vaccines: Hope or Hype? Cureus 2025; 17:e80636. [PMID: 40091902 PMCID: PMC11910667 DOI: 10.7759/cureus.80636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Accepted: 03/15/2025] [Indexed: 03/19/2025] Open
Abstract
Globally, periodontal diseases, mainly driven by polymicrobial biofilms, are a widespread concern of social medicine due to their considerable incidence and tie-up to systemic disorders like diabetes, cardiovascular diseases, and complications during pregnancy. Traditional treatments focus on mechanical debridement and antimicrobial therapies, but these approaches have limitations, including recurrence and antibiotic resistance. Periodontal vaccines offer a promising alternative by targeting the immunological mechanisms underlying periodontal disease. This review explores the current state of periodontal vaccine development, highlighting key antigens, vaccine delivery systems, and preclinical and clinical advancements. Special emphasis is placed on antigen selection, host variability, immune tolerance, and future directions to overcome these barriers. This article highlights the advancements and challenges in periodontal vaccine research, offering insights into the capability of immunoprophylaxis as a groundbreaking way to manage periodontal diseases.
Collapse
Affiliation(s)
- Pratiksha Patel
- Department of Periodontology and Implantology, Karnavati School of Dentistry, Karnavati University, Gandhinagar, IND
| | - Bhavin Patel
- Department of Periodontology and Implantology, Karnavati School of Dentistry, Karnavati University, Gandhinagar, IND
| | - Shruti D Vyas
- Department of Periodontology and Implantology, Karnavati School of Dentistry, Karnavati University, Gandhinagar, IND
| | - Maitri S Patel
- Department of Periodontology and Implantology, Karnavati School of Dentistry, Karnavati University, Gandhinagar, IND
| | - Tanvi Hirani
- Department of Periodontology and Implantology, Karnavati School of Dentistry, Karnavati University, Gandhinagar, IND
| | - Mainul Haque
- Department of Pharmacology and Therapeutics, National Defence University of Malaysia, Kuala Lumpur, MYS
- Department of Research, Karnavati School of Dentistry, Karnavati University, Gandhinagar, IND
| | - Santosh Kumar
- Department of Periodontology and Implantology, Karnavati School of Dentistry, Karnavati University, Gandhinagar, IND
| |
Collapse
|
21
|
Shinde P, Willemsen L, Anderson M, Aoki M, Basu S, Burel JG, Cheng P, Ghosh Dastidar S, Dunleavy A, Einav T, Forschmiedt J, Fourati S, Garcia J, Gibson W, Greenbaum JA, Guan L, Guan W, Gygi JP, Ha B, Hou J, Hsiao J, Huang Y, Jansen R, Kakoty B, Kang Z, Kobie JJ, Kojima M, Konstorum A, Lee J, Lewis SA, Li A, Lock EF, Mahita J, Mendes M, Meng H, Neher A, Nili S, Olsen LR, Orfield S, Overton JA, Pai N, Parker C, Qian B, Rasmussen M, Reyna J, Richardson E, Safo S, Sorenson J, Srinivasan A, Thrupp N, Tippalagama R, Trevizani R, Ventz S, Wang J, Wu CC, Ay F, Grant B, Kleinstein SH, Peters B. Putting computational models of immunity to the test-An invited challenge to predict B.pertussis vaccination responses. PLoS Comput Biol 2025; 21:e1012927. [PMID: 40163550 PMCID: PMC11978014 DOI: 10.1371/journal.pcbi.1012927] [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: 09/23/2024] [Revised: 04/08/2025] [Accepted: 03/04/2025] [Indexed: 04/02/2025] Open
Abstract
Systems vaccinology studies have been used to build computational models that predict individual vaccine responses and identify the factors contributing to differences in outcome. Comparing such models is challenging due to variability in study designs. To address this, we established a community resource to compare models predicting B. pertussis booster responses and generate experimental data for the explicit purpose of model evaluation. We here describe our second computational prediction challenge using this resource, where we benchmarked 49 algorithms from 53 scientists. We found that the most successful models stood out in their handling of nonlinearities, reducing large feature sets to representative subsets, and advanced data preprocessing. In contrast, we found that models adopted from literature that were developed to predict vaccine antibody responses in other settings performed poorly, reinforcing the need for purpose-built models. Overall, this demonstrates the value of purpose-generated datasets for rigorous and open model evaluations to identify features that improve the reliability and applicability of computational models in vaccine response prediction.
Collapse
Affiliation(s)
- Pramod Shinde
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, United States of America
| | - Lisa Willemsen
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, United States of America
| | - Michael Anderson
- Division of Biostatistics and Health Data Science, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Minori Aoki
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, United States of America
| | - Saonli Basu
- Division of Biostatistics and Health Data Science, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Julie G. Burel
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, United States of America
| | - Peng Cheng
- Department of Molecular Biology, School of Biological Sciences, University of California, San Diego, La Jolla, California, United States of America
| | - Souradipto Ghosh Dastidar
- Division of Biostatistics and Health Data Science, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Aidan Dunleavy
- School of Statistics, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Tal Einav
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, United States of America
- Department of Medicine, University of California San Diego, San Diego, California, United States of America
| | - Jamie Forschmiedt
- Division of Biostatistics and Health Data Science, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Slim Fourati
- Department of Medicine, Division of Allergy and Immunology, Feinberg School of Medicine and Center for Human Immunobiology, Northwestern University, Chicago, Illinois, United States of America
| | - Javier Garcia
- Department of Molecular Biology, School of Biological Sciences, University of California, San Diego, La Jolla, California, United States of America
| | - William Gibson
- Vaccine Research Center, National Institute of Allergy and Infectious Disease, National Institute of Health, Bethesda, Maryland, United States of America
| | - Jason A. Greenbaum
- LJI Bioinformatics Core, La Jolla Institute for Immunology, La Jolla, California, United States of America
| | - Leying Guan
- Department of Biostatistics, Yale School of Public Health, New Haven, Connecticut, United States of America
| | - Weikang Guan
- Department of Molecular Biology, School of Biological Sciences, University of California, San Diego, La Jolla, California, United States of America
| | - Jeremy P. Gygi
- Program in Computational Biology & Bioinformatics, Yale University, New Haven, Connecticut, United States of America
| | - Brendan Ha
- LJI Bioinformatics Core, La Jolla Institute for Immunology, La Jolla, California, United States of America
| | - Joe Hou
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Jason Hsiao
- Department of Molecular Biology, School of Biological Sciences, University of California, San Diego, La Jolla, California, United States of America
| | - Yunda Huang
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- Department of Global Health, University of Washington, Seattle, Washington, United States of America
| | - Rick Jansen
- Biostatistics Core, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Bhargob Kakoty
- Division of Biostatistics and Health Data Science, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Zhiyu Kang
- Division of Biostatistics and Health Data Science, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - James J. Kobie
- Department of Medicine, Division of Infectious Diseases, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Mari Kojima
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, United States of America
| | - Anna Konstorum
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut, United States of America
- Laboratory for Systems Biology, University of Florida, Gainesville, Florida, United States of America
| | - Jiyeun Lee
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, United States of America
| | - Sloan A. Lewis
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, United States of America
| | - Aixin Li
- Division of Epidemiology and Community Health, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Eric F. Lock
- Division of Biostatistics and Health Data Science, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Jarjapu Mahita
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, United States of America
| | - Marcus Mendes
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, United States of America
| | - Hailong Meng
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Aidan Neher
- Division of Biostatistics and Health Data Science, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Somayeh Nili
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, United States of America
| | - Lars Rønn Olsen
- Department of Immunology and Microbiology, LEO Foundation Skin Immunology Research Center, University of Copenhagen, Copenhagen, Denmark
| | - Shelby Orfield
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, United States of America
| | | | - Nidhi Pai
- Division of Biostatistics and Health Data Science, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Cokie Parker
- National Institute of Allergy and Infectious Diseases, National Institute of Health, Bethesda, Maryland, United States of America
| | - Brian Qian
- Department of Molecular Biology, School of Biological Sciences, University of California, San Diego, La Jolla, California, United States of America
| | - Mikkel Rasmussen
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, United States of America
| | - Joaquin Reyna
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, California, United States of America
- Bioinformatics and Systems Biology Graduate Program, University of California, San Diego, California, United States of America
| | - Eve Richardson
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, United States of America
| | - Sandra Safo
- Division of Biostatistics and Health Data Science, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Josey Sorenson
- Division of Biostatistics and Health Data Science, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Aparna Srinivasan
- Division of Biostatistics and Health Data Science, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Nicola Thrupp
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, United States of America
| | - Rashmi Tippalagama
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, United States of America
| | - Raphael Trevizani
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, United States of America
- Fundação Oswaldo Cruz, Fiocruz - Ceará, Brazil
| | - Steffen Ventz
- Division of Biostatistics and Health Data Science, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Jiuzhou Wang
- Division of Biostatistics and Health Data Science, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Cheng-Chang Wu
- Division of Biostatistics and Health Data Science, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Ferhat Ay
- Department of Medicine, University of California San Diego, San Diego, California, United States of America
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, California, United States of America
- Bioinformatics and Systems Biology Graduate Program, University of California, San Diego, California, United States of America
| | - Barry Grant
- Department of Molecular Biology, School of Biological Sciences, University of California, San Diego, La Jolla, California, United States of America
| | - Steven H. Kleinstein
- Program in Computational Biology & Bioinformatics, Yale University, New Haven, Connecticut, United States of America
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Bjoern Peters
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, United States of America
- Department of Medicine, University of California San Diego, San Diego, California, United States of America
| |
Collapse
|
22
|
Rubio-Casillas A, Redwan EM, Uversky VN. More antibodies are not always better: Fc effector functions play a critical role in SARS-CoV-2 infection and protection. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2025; 213:413-447. [PMID: 40246351 DOI: 10.1016/bs.pmbts.2025.02.001] [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: 04/19/2025]
Abstract
Traditional vaccinology has primarily focused on neutralizing antibody titers as the main correlate of vaccine efficacy, often overlooking the multifaceted roles of antibody Fc effector functions in orchestrating protective immune responses. Fc-mediated immune responses play a pivotal role in immune modulation and pathogen clearance. Emerging evidence from natural infections and vaccine studies highlights the critical contribution of Fc effector functions in determining the quality and durability of immunity. This work explores the limitations of current vaccine evaluation paradigms that prioritize neutralization over Fc effector mechanisms. It also describes findings from a study showing an unexpected role for SARS-CoV-2 anti-spike antibodies: both convalescent plasma and patient-derived monoclonal antibodies (mAbs) lead to maximum phagocytic capacity by monocytes at low concentrations, whereas at higher concentrations the phagocytic capacity was reduced. Given that the severity of COVID-19 disease and antibody titers are strongly positively correlated, this work challenges the paradigm that high antibodies offer better protection against severe disease. It is proposed that humoral and cellular responses elicited by vaccination should never be higher than those produced by natural infection. By integrating antibody Fc effector functions into vaccine development, a paradigm shift is proposed that emphasizes synergic antibody responses. Such an approach could transform vaccine efficacy assessment, enhance protection against dangerous pathogens, and drive innovation in vaccine design.
Collapse
Affiliation(s)
- Alberto Rubio-Casillas
- Autlan Regional Hospital, Jalisco Health Services, Autlan, Jalisco, Mexico; Biology Laboratory, Autlan Regional Preparatory School, University of Guadalajara, Autlan, Jalisco, Mexico.
| | - Elrashdy M Redwan
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia; Therapeutic and Protective Proteins Laboratory, Protein Research Department, Genetic Engineering and Biotechnology Research Institute, City for Scientific Research and Technology Applications, New Borg El-Arab, Alexandria, Egypt
| | - Vladimir N Uversky
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States; USF Health Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| |
Collapse
|
23
|
Enya T, Ross SR. Innate Sensing of Viral Nucleic Acids and Their Use in Antiviral Vaccine Development. Vaccines (Basel) 2025; 13:193. [PMID: 40006739 PMCID: PMC11860339 DOI: 10.3390/vaccines13020193] [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: 12/09/2024] [Revised: 01/23/2025] [Accepted: 02/11/2025] [Indexed: 02/27/2025] Open
Abstract
Viruses pose a significant threat to humans by causing numerous infectious and potentially fatal diseases. Understanding how the host's innate immune system recognizes viruses is essential to understanding pathogenesis and ways to control viral infection. Innate immunity also plays a critical role in shaping adaptive immune responses induced by vaccines. Recently developed adjuvants often include nucleic acids that stimulate pattern recognition receptors which are essential components of innate immunity necessary for activating antigen-presentation cells and thereby bridging innate and adaptive immunity. Therefore, understanding viral nucleic acid sensing by cytosolic sensors is essential, as it provides the potential means for developing new vaccine strategies, including effective adjuvants.
Collapse
Affiliation(s)
| | - Susan R. Ross
- Department of Microbiology and Immunology, University of Illinois at Chicago College of Medicine, Chicago, IL 60612, USA;
| |
Collapse
|
24
|
Ostroumov D, Benne N, Lozano Vigario F, Escalona-Rayo O, Dodz K, Sauer S, Suhl LL, Wedemeyer HH, Kühnel F, Slütter B, Wirth TC. Sequential STING and CD40 agonism drives massive expansion of tumor-specific T cells in liposomal peptide vaccines. Cell Mol Immunol 2025; 22:150-160. [PMID: 39741195 PMCID: PMC11782543 DOI: 10.1038/s41423-024-01249-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 11/12/2024] [Accepted: 11/27/2024] [Indexed: 01/02/2025] Open
Abstract
The clinical use of cancer vaccines is hampered by the low magnitude of induced T-cell responses and the need for repetitive antigen stimulation. Here, we demonstrate that liposomal formulations with incorporated STING agonists are optimally suited to deliver peptide antigens to dendritic cells in vivo and to activate dendritic cells in secondary lymphoid organs. One week after liposomal priming, systemic administration of peptides and a costimulatory agonistic CD40 antibody enables ultrarapid expansion of T cells, resulting in massive expansion of tumor-specific T cells in the peripheral blood two weeks after priming. In the MC-38 colon cancer model, this synthetic prime-boost regimen induces rapid regression and cure of large established subcutaneous cancers via the use of a single tumor-specific neoantigen. These experiments demonstrate the feasibility of liposome-based heterologous vaccination regimens to increase the therapeutic efficacy of peptide vaccines in the context of immunogenic adjuvants and costimulatory booster immunizations. Our results provide a rationale for the further development of modern liposomal peptide vaccines for cancer therapy.
Collapse
Affiliation(s)
- Dmitrij Ostroumov
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Naomi Benne
- Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Fernando Lozano Vigario
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden, The Netherlands
| | - Oscar Escalona-Rayo
- Department of Supramolecular and Biomaterials Chemistry, Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - Ksenia Dodz
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden, The Netherlands
| | - Sarah Sauer
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Lena Luisa Suhl
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Hans Heiner Wedemeyer
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Florian Kühnel
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Bram Slütter
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden, The Netherlands
| | - Thomas Christian Wirth
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany.
| |
Collapse
|
25
|
Wang J, Zhang Y, Jia Y, Xing H, Xu F, Xia B, Lai W, Yuan Y, Li X, Shan S, Chen J, Guo W, Zhang J, Zheng A, Li J, Gong N, Liang XJ. Targeting vaccines to dendritic cells by mimicking the processing and presentation of antigens in xenotransplant rejection. Nat Biomed Eng 2025; 9:201-214. [PMID: 39948171 DOI: 10.1038/s41551-025-01343-6] [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: 04/11/2023] [Accepted: 01/07/2025] [Indexed: 02/21/2025]
Abstract
Targeting the delivery of vaccines to dendritic cells (DCs) is challenging. Here we show that, by mimicking the fast and strong antigen processing and presentation that occurs during the rejection of xenotransplanted tissue, xenogeneic cell membrane-derived vesicles exposing tissue-specific antibodies can be leveraged to deliver peptide antigens and mRNA-encoded antigens to DCs. In mice with murine melanoma and murine thymoma, xenogeneic vesicles encapsulating a tumour-derived antigenic peptide or coated on lipid nanoparticles encapsulating an mRNA coding for a tumour antigen elicited potent tumour-specific T-cell responses that inhibited tumour growth. Mice immunized with xenogeneic vesicle-coated lipid nanoparticles encapsulating an mRNA encoding for the spike protein of severe acute respiratory syndrome coronavirus 2 elicited titres of anti-spike receptor-binding domain immunoglobulin G and of neutralizing antibodies that were approximately 32-fold and 6-fold, respectively, those elicited by a commercialized mRNA-lipid nanoparticle vaccine. The advantages of mimicking the biological recognition between immunoglobulin G on xenogeneic vesicles and fragment crystallizable receptors on DCs may justify the assessment of the safety risks of using animal-derived biological products in humans.
Collapse
Affiliation(s)
- Jinjin Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yuxuan Zhang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yaru Jia
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, China
- State Key Laboratory of New Pharmaceutical Preparations and Excipients, Hebei University, Baoding, China
| | - Haonan Xing
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, China
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Fengfei Xu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Bozhang Xia
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Wenjia Lai
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, China
| | - Yuan Yuan
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, China
| | - Xianlei Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, China
| | - Shaobo Shan
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, China
| | - Junge Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, China
| | - Weisheng Guo
- School of Biomedical Engineering, Guangzhou Medical University, Guangzhou, China
| | - Jinchao Zhang
- State Key Laboratory of New Pharmaceutical Preparations and Excipients, Hebei University, Baoding, China
| | - Aiping Zheng
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Jinghong Li
- Department of Chemistry, Center for BioAnalytical Chemistry, Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, New Cornerstone Science Institute, Tsinghua University, Beijing, China
| | - Ningqiang Gong
- Department of General Surgery, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China.
| | - Xing-Jie Liang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, China.
- University of Chinese Academy of Sciences, Beijing, China.
- State Key Laboratory of New Pharmaceutical Preparations and Excipients, Hebei University, Baoding, China.
- School of Biomedical Engineering, Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
26
|
Nakayama T, Hayashi T, Makino K, Oe K. The efficacy and safety of a quadrivalent live attenuated influenza nasal vaccine in Japanese children: A phase 3, randomized, placebo-controlled study. J Infect Chemother 2025; 31:102460. [PMID: 38959995 DOI: 10.1016/j.jiac.2024.06.023] [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: 04/26/2024] [Revised: 06/12/2024] [Accepted: 06/30/2024] [Indexed: 07/05/2024]
Abstract
BACKGROUND Vaccination is the primary method of preventing influenza infection and complications in young children. We evaluated the efficacy and safety of a single dose of MEDI3250 (intranasal, quadrivalent, live attenuated influenza vaccine) in healthy Japanese children during the 2016/17 influenza season. METHODS In this multicenter, randomized, double-blind, phase 3 study (jRCT2080223345), participants aged 2-18 years received MEDI3250 or placebo (2:1), stratified by age (2-6 years, 7-18 years). The primary and secondary endpoints were the incidence of confirmed symptomatic onset of influenza caused by a circulating wild-type strain or by a vaccine-matched strain, respectively. Safety outcomes included the incidence of adverse events (AEs) and vaccine-related AEs. RESULTS Overall, 910 participants received MEDI3250 (n = 608) or placebo (n = 302). For the primary endpoint (regardless of the influenza subtype), the incidence of influenza onset was 25.5 % (MEDI3250) and 35.9 % (placebo); relative risk reduction, 28.8 % (95 % confidence interval, 12.5 %, 42.0 %). For the secondary endpoint (vaccine-matched strain), the incidence was 10.9 % (MEDI3250) and 17.2 % (placebo); relative risk reduction, 36.6 % (95 % confidence interval, 6.5 %, 56.8 %). Solicited AEs occurred in 67.6 % (MEDI3250) and 63.6 % (placebo). Most events were mild; nasal discharge was most common (59.2 % [MEDI3250] and 52.6 % [placebo]). Unsolicited AEs occurred in 36.0 % (MEDI3250) and 33.1 % (placebo). The most common unsolicited vaccine-related AE was diarrhea (2.3 %, both groups). CONCLUSIONS MEDI3250 had a greater preventive effect against influenza onset in Japanese children than placebo; no new safety signals were observed relative to previous clinical and post-marketing studies of MEDI3250.
Collapse
Affiliation(s)
- Tetsuo Nakayama
- Laboratory of Viral Infection, Ōmura Satoshi Memorial Institute, Kitasato University, Tokyo, Japan.
| | | | | | - Keiji Oe
- Daiichi Sankyo Co., Ltd., Tokyo, Japan.
| |
Collapse
|
27
|
Farooq MA, Johnston APR, Trevaskis NL. Impact of nanoparticle properties on immune cell interactions in the lymph node. Acta Biomater 2025; 193:65-82. [PMID: 39701340 DOI: 10.1016/j.actbio.2024.12.039] [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: 08/07/2024] [Revised: 11/21/2024] [Accepted: 12/16/2024] [Indexed: 12/21/2024]
Abstract
The lymphatic system plays an important role in health and many diseases, such as cancer, autoimmune, cardiovascular, metabolic, hepatic, viral, and other infectious diseases. The lymphatic system is, therefore, an important treatment target site for a range of diseases. Lymph nodes (LNs), rich in T cells, B cells, dendritic cells, and macrophages, are also primary sites of action for vaccines and immunotherapies. Promoting the delivery of therapeutics and vaccines to LNs can, therefore, enhance treatment efficacy and facilitate avoidance of off-target side effects by enabling a reduction in therapeutic dose. Several nanoparticle (NP) based delivery systems, such as polymeric NPs, lipid NPs, liposomes, micelles, and dendrimers, have been reported to enhance the delivery of therapeutics and/or vaccines to LNs. Specific uptake into the lymph following injection into tissues is highly dependent on particle properties, particularly particle size, as small molecules are more likely to be taken up by blood capillaries due to higher blood flow rates, whereas larger molecules and NPs can be specifically transported via the lymphatic vessels to LNs as the initial lymphatic capillaries are more permeable than blood capillaries. Once NPs enter LNs, particle properties also have an important influence on their disposition within the node and association with immune cells, which has significant implications for the design of vaccines and immunotherapies. This review article focuses on the impact of NP properties, such as size, surface charge and modification, and route of administration, on lymphatic uptake, retention, and interactions with immune cells in LNs. We suggest that optimizing all these factors can enhance the efficacy of vaccines or therapeutics with targets in the lymphatics and also be helpful for the rational design of vaccines. STATEMENT OF SIGNIFICANCE: The lymphatic system plays an essential role in health and is an important treatment target site for a range of diseases. Promoting the delivery of immunotherapies and vaccines to immune cells in lymph nodes can enhance efficacy and facilitate avoidance of off-target side effects by enabling a reduction in therapeutic dose. One of the major approaches used to deliver therapeutics and vaccines to lymph nodes is via injection in nanoparticle delivery systems. This review aims to provide an overview of the impact of nanoparticle properties, such as size, surface charge, modification, and route of administration, on lymphatic uptake, lymph node retention, and interactions with immune cells in lymph nodes. This will inform the design of future improved nanoparticle systems for vaccines and immunotherapies.
Collapse
Affiliation(s)
- Muhammad Asim Farooq
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 399 Royal Parade, Parkville, VIC 3052, Australia
| | - Angus P R Johnston
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 399 Royal Parade, Parkville, VIC 3052, Australia
| | - Natalie L Trevaskis
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 399 Royal Parade, Parkville, VIC 3052, Australia.
| |
Collapse
|
28
|
Pereira DR, Pérez-Betancourt Y, Távora BCLF, Magalhães GS, Carmona-Ribeiro AM, Faquim-Mauro EL. The Role of Dendritic Cells in Adaptive Immune Response Induced by OVA/PDDA Nanoparticles. Vaccines (Basel) 2025; 13:76. [PMID: 39852855 PMCID: PMC11769024 DOI: 10.3390/vaccines13010076] [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: 10/31/2024] [Revised: 12/08/2024] [Accepted: 12/17/2024] [Indexed: 01/26/2025] Open
Abstract
Background/Objective: Cationic polymers were shown to assemble with negatively charged proteins yielding nanoparticles (NPs). Poly-diallyl-dimethyl-ammonium chloride (PDDA) combined with ovalbumin (OVA) yielded a stable colloidal dispersion (OVA/PDDA-NPs) eliciting significant anti-OVA immune response. Dendritic cells (DCs), as sentinels of foreign antigens, exert a crucial role in the antigen-specific immune response. Here, we aimed to evaluate the involvement of DCs in the immune response induced by OVA/PDDA. Methods: In vivo experiments were used to assess the ability of OVA/PDDA-NPs to induce anti-OVA antibodies by ELISA, as well as plasma cells and memory B cells using flow cytometry. Additionally, DC migration to draining lymph nodes following OVA/PDDA-NP immunization was evaluated by flow cytometry. In vitro experiments using bone marrow-derived DCs (BM-DCs) were used to analyze the binding and uptake of OVA/PDDA-NPs, DC maturation status, and their antigen-presenting capacity. Results: Our data confirmed the potent effect of OVA/PDDA-NPs inducing anti-OVA IgG1 and IgG2a antibodies with increased CD19+CD138+ plasma cells and CD19+CD38+CD27+ memory cells in immunized mice. OVA/PDDA-NPs induced DC maturation and migration to draining lymph nodes. The in vitro results showed higher binding and the uptake of OVA/PDDA-NPs by BM-DCs. In addition, the NPs were able to induce the upregulation of costimulatory and MHC-II molecules on DCs, as well as TNF-α and IL-12 production. Higher OVA-specific T cell proliferation was promoted by BM-DCs incubated with OVA/PDDA-NPs. Conclusions: The data showed the central role of DCs in the induction of antigen-specific immune response by OVA-PDDA-NPs, thus proving that these NPs are a potent adjuvant for subunit vaccine design.
Collapse
Affiliation(s)
- Daniele R. Pereira
- Laboratory of Immunopathology, Butantan Institute, São Paulo 05585-000, Brazil; (D.R.P.); (B.C.L.F.T.); (G.S.M.)
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 04021-001, Brazil
| | - Yunys Pérez-Betancourt
- Department of Biochemistry, Chemistry Institute, University of São Paulo, São Paulo 04021-001, Brazil; (Y.P.-B.); (A.M.C.-R.)
| | - Bianca C. L. F. Távora
- Laboratory of Immunopathology, Butantan Institute, São Paulo 05585-000, Brazil; (D.R.P.); (B.C.L.F.T.); (G.S.M.)
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 04021-001, Brazil
| | - Geraldo S. Magalhães
- Laboratory of Immunopathology, Butantan Institute, São Paulo 05585-000, Brazil; (D.R.P.); (B.C.L.F.T.); (G.S.M.)
| | - Ana Maria Carmona-Ribeiro
- Department of Biochemistry, Chemistry Institute, University of São Paulo, São Paulo 04021-001, Brazil; (Y.P.-B.); (A.M.C.-R.)
| | - Eliana L. Faquim-Mauro
- Laboratory of Immunopathology, Butantan Institute, São Paulo 05585-000, Brazil; (D.R.P.); (B.C.L.F.T.); (G.S.M.)
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 04021-001, Brazil
| |
Collapse
|
29
|
Veronesi G, Gianfagna F, Karachaliou M, Guasti L, Kogevinas M, Ferrario MM. Association between long-term exposure to air pollutants with breakthrough SARS-CoV-2 infections and antibody responses among COVID-19 vaccinated older adults in Northern Italy. ENVIRONMENTAL RESEARCH 2025; 265:120450. [PMID: 39603588 DOI: 10.1016/j.envres.2024.120450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 11/04/2024] [Accepted: 11/23/2024] [Indexed: 11/29/2024]
Abstract
AIMS To investigate the association between long-term exposure to PM2.5, PM10, NO2 and O3 with SARS-CoV-2 breakthrough infections and COVID-19 vaccine-induced antibody responses in a northern Italian population-based sample of older adults. METHODS Within an ongoing prospective population-based study, we followed-up 1326 vaccinated individuals aged 65-83 years, with no prior SARS-CoV-2 infection, for their first positive SARS-CoV-2 swab until December 31st, 2022. We assessed spike IgG antibody levels in most participants (n = 1206). The 2019 annual mean levels of air pollutants derived from combined use of chemical-transport and random-Forest models (spatial resolution: 1Kmq) were individually assigned based on the latest residence address. We estimated multivariable-adjusted associations (per 1 interquartile range increase, IQR) of air pollutants with breakthrough infections using Cox models with time-dependent vaccine exposure; and with percent change in the IgG geometric mean using generalized additive models. RESULTS The mean (SD) age was 74.9 ± 4.1 years, and 50% were women. An IQR (1.2 μg/m3) increase in long-term PM2.5 exposure was associated with a 52% increase in breakthrough infection risk following a second vaccine and a 26% increase following a third vaccine. The effect vanished with the further increment of vaccination doses. Associations for NO2 were inconsistent. Ozone was negatively associated with breakthrough infection risk, but this association reversed in bi-pollutant models adjusting for PM2.5. PM2.5 was associated with a -7.3% (-13.9% to -0.2%) reduction in vaccine-induced IgG levels. The reduction became more pronounced as the time delay from vaccination increased, and with adjustment for NO2 co-exposure. CONCLUSION In our population of vaccinated older adults, fine particulate matter exposure was independently associated with a higher risk of SARS-CoV-2 breakthrough infection and a lower antibody response, both effects being influenced by timely and repeated vaccination schedule.
Collapse
Affiliation(s)
- Giovanni Veronesi
- Research Center in Epidemiology and Preventive Medicine (EPIMED), Department of Medicine and Surgery, University of Insubria, Varese, Italy.
| | - Francesco Gianfagna
- Research Center in Epidemiology and Preventive Medicine (EPIMED), Department of Medicine and Surgery, University of Insubria, Varese, Italy; Mediterranea Cardiocentro, Napoli, Italy
| | | | - Luigina Guasti
- Research Center in Epidemiology and Preventive Medicine (EPIMED), Department of Medicine and Surgery, University of Insubria, Varese, Italy; Geriatric Division, Azienda Socio Sanitaria Territoriale Sette Laghi, Varese, Italy
| | - Manolis Kogevinas
- ISGlobal, Barcelona, Spain; CIBER Epidemiologia y Salud Pública (CIBERESP), Madrid, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain; IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - Marco M Ferrario
- Research Center in Epidemiology and Preventive Medicine (EPIMED), Department of Medicine and Surgery, University of Insubria, Varese, Italy
| |
Collapse
|
30
|
Hamid FA, Le NMN, Song D, Amin H, Hicks L, Bird S, Siram K, Hoppe B, Demeler B, Evans JT, Burkhart D, Pravetoni M. A cationic liposome-formulated Toll Like Receptor (TLR)7/8 agonist enhances the efficacy of a vaccine against fentanyl toxicity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.08.631964. [PMID: 39868149 PMCID: PMC11761771 DOI: 10.1101/2025.01.08.631964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
The U.S. opioid epidemic is an extraordinary public health crisis that started in 1990 and significantly accelerated in the last decade. Since 2020, over 100,000 fatal drug overdoses have been reported annually, and 75% of those involved fentanyl and its analogs (F/FA). Accelerating the translation of innovative, effective, and safe treatments is needed to augment existing measures to counteract such a crisis. Active immunization against F/FA and other opioids represents a promising therapeutic and prophylactic strategy for opioid use disorder (OUD) and opioid-induced overdose toxicity. Previously we demonstrated that the anti-fentanyl vaccine comprising a fentanyl-based hapten (F) conjugated to the diphtheria cross-reactive material (CRM), admixed with the novel lipidated toll-like receptor 7/8 (TLR7/8) agonist INI-4001 adsorbed on Alhydrogel ® (alum) induced high-affinity fentanyl-specific polyclonal antibodies that protected against fentanyl-induced pharmacological effects in mice, rats, and mini-pigs. Here, INI-4001 was formulated into liposomes with different surface charges, and their impact on F-CRM adsorption, INI-4001 adjuvanticity, and vaccine efficacy were explored. Additionally, as the role of innate immunity in mediating the efficacy of addiction vaccines is largely unknown, we tested these formulations on the activation of innate immunity in vitro . Cationic INI-4001 liposomes surpassed other liposomal and aluminum-based formulations of INI-4001 by enhancing the efficacy of fentanyl vaccines and protecting rats against bradycardia and respiratory depression by blocking the distribution of fentanyl to the brain. Fentanyl vaccines adjuvanted with either cationic INI-4001 liposomes or the aqueous INI-4001 adsorbed to alum induced significant surface expression of co-stimulatory molecules and maturation markers in a murine dendritic cell line (JAWS II), while the former was superior in enhancing the macrophages surface expression of CD40, CD86 and inducible nitric oxide synthase (iNOS), indicative of maturation and activation. These results warrant further investigation of liposome-based formulations of TLR7/8 agonists for improving the efficacy of vaccines targeting F/FA and other opioids of public health interest. Graphical abstract
Collapse
|
31
|
Liu S, Lin M, Zhou X. T4 Phage Displaying Dual Antigen Clusters Against H3N2 Influenza Virus Infection. Vaccines (Basel) 2025; 13:70. [PMID: 39852849 PMCID: PMC11769387 DOI: 10.3390/vaccines13010070] [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: 11/07/2024] [Revised: 01/09/2025] [Accepted: 01/11/2025] [Indexed: 01/26/2025] Open
Abstract
BACKGROUND The current H3N2 influenza subunit vaccine exhibits weak immunogenicity, which limits its effectiveness in preventing and controlling influenza virus infections. METHODS In this study, we aimed to develop a T4 phage-based nanovaccine designed to enhance the immunogenicity of two antigens by displaying the HA1 and M2e antigens of the H3N2 influenza virus on each phage nanoparticle. Specifically, we fused the Soc protein with the HA1 antigen and the Hoc protein with the M2e antigen, assembling them onto a T4 phage that lacks Soc and Hoc proteins (Soc-Hoc-T4), thereby constructing a nanovaccine that concurrently presents both HA1 and M2e antigens. RESULTS The analysis of the optical density of the target protein bands indicated that each particle could display approximately 179 HA1 and 68 M2e antigen molecules. Additionally, animal experiments demonstrated that this nanoparticle vaccine displaying dual antigen clusters induced a stronger specific immune response, higher antibody titers, a more balanced Th1/Th2 immune response, and enhanced CD4+ and CD8+ T cell effects compared to immunization with HA1 and M2e antigen molecules alone. Importantly, mice immunized with the T4 phage displaying dual antigen clusters achieved full protection (100% protection) against the H3N2 influenza virus, highlighting its robust protective efficacy. CONCLUSIONS In summary, our findings indicate that particles based on a T4 phage displaying antigen clusters exhibit ideal immunogenicity and protective effects, providing a promising strategy for the development of subunit vaccines against various viruses beyond influenza.
Collapse
Affiliation(s)
- Shenglong Liu
- College of Veterinary Medicine, Institute of Comparative Medicine, Yangzhou University, Yangzhou 225009, China; (S.L.); (M.L.)
| | - Mengzhou Lin
- College of Veterinary Medicine, Institute of Comparative Medicine, Yangzhou University, Yangzhou 225009, China; (S.L.); (M.L.)
| | - Xin Zhou
- College of Veterinary Medicine, Institute of Comparative Medicine, Yangzhou University, Yangzhou 225009, China; (S.L.); (M.L.)
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou 225009, China
| |
Collapse
|
32
|
Zhang M, Wang C, Pan J, Cui H, Zhao X. Advancing novel veterinary vaccines: From comprehensive antigen and adjuvant design to preparation process optimization. Int Immunopharmacol 2025; 145:113784. [PMID: 39672026 DOI: 10.1016/j.intimp.2024.113784] [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: 10/15/2024] [Revised: 12/01/2024] [Accepted: 12/02/2024] [Indexed: 12/15/2024]
Abstract
Vaccination stands as the paramount and cost-effective strategy for the prevention and management of animal infectious diseases. With the advances in biological technology, materials science and industrial optimization, substantial progress has been made in the development of innovative veterinary vaccines. A majority of the novel vaccines under current investigation tend to stimulate multiple immune pathways and to achieve long-term resistance against infectious diseases, yet it remains imperative to concentrate research efforts on the efficient utilization of vaccines, mitigating toxic side effects, and ensuring safe production processes. This article presents an overview of research progress in veterinary vaccines, encompassing comprehensive antigen design, adjuvant formulation advancements, preparation process optimization, and rigorous immune efficacy evaluation. It summarizes cutting-edge vaccines derived from in vitro synthesis and in vivo application, emphasizing immunogenic components and immune response mechanisms. It also highlights novel biological adjuvants that enhance immune efficacy, diversify raw materials, and possess targeted functions, while comprehensively exploring advancements in production methodologies and compatible vaccine products. By establishing a foundation for the integrated use of these innovative veterinary vaccines, this work facilitates future interdisciplinary cooperation in their advancement, aiming to accelerate the achievement of herd immunity through concerted efforts.
Collapse
Affiliation(s)
- Meng Zhang
- Institute of Environment and Sustainable Development in Agriculture, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Chunxin Wang
- Institute of Environment and Sustainable Development in Agriculture, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Junqian Pan
- Institute of Environment and Sustainable Development in Agriculture, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Haixin Cui
- Institute of Environment and Sustainable Development in Agriculture, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Xiang Zhao
- Institute of Environment and Sustainable Development in Agriculture, Chinese Academy of Agricultural Sciences, Beijing 100081, China.
| |
Collapse
|
33
|
Liang J, Yao L, Liu Z, Chen Y, Lin Y, Tian T. Nanoparticles in Subunit Vaccines: Immunological Foundations, Categories, and Applications. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2407649. [PMID: 39501996 DOI: 10.1002/smll.202407649] [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: 08/28/2024] [Revised: 10/12/2024] [Indexed: 01/11/2025]
Abstract
Subunit vaccines, significant in next-generation vaccine development, offer precise targeting of immune responses by focusing on specific antigens. However, this precision often comes at the cost of eliciting strong and durable immunity, posing a great challenge to vaccine design. To address this limitation, recent advancements in nanoparticles (NPs) are utilized to enhance antigen delivery efficiency and boost vaccine efficacy. This review examines how the physicochemical properties of NPs influence various stages of the immune response during vaccine delivery and analyzes how different NP types contribute to immune activation and enhance vaccine performance. It then explores the unique characteristics and immune activation mechanisms of these NPs, along with their recent advancements, and highlights their application in subunit vaccines targeting infectious diseases and cancer. Finally, it discusses the challenges in NP-based vaccine development and proposes future directions for innovation in this promising field.
Collapse
Affiliation(s)
- Jiale Liang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West ChinaHospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Lan Yao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West ChinaHospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Zhiqiang Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West ChinaHospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Ye Chen
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West ChinaHospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yunfeng Lin
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West ChinaHospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Taoran Tian
- Sichuan Provincial Engineering Research Center of Oral Biomaterials, Chengdu, Sichuan, 610041, China
| |
Collapse
|
34
|
Song Z, Zhou Y, Jiao L, Zhu T, Yu R, Wang Z, Qiu Y, Miao J, Cai T, Zhang S, Liu H, Sun H, Sun Y, Wang D, Liu Z. Lovastatin enhances humoral and cellular immune responses to H1N1 influenza vaccine. Vet Microbiol 2025; 300:110331. [PMID: 39662203 DOI: 10.1016/j.vetmic.2024.110331] [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: 08/03/2024] [Revised: 11/29/2024] [Accepted: 12/03/2024] [Indexed: 12/13/2024]
Abstract
The Swine Influenza Virus (SIV) is a major respiratory pathogen in swine, causing acute, febrile, and highly transmissible infections. This virus is widespread globally and poses significant risks to human health and social development. Traditional prevention strategies for SIV rely on the use of inactivated vaccines combined with Alum adjuvants. However, this method is limited by insufficient protection due to the lack of cellular immunity provided by Alum adjuvants. In this study, we investigated the effect of lovastatin, a specific inhibitor of the mevalonate pathway, on the immune response in mice vaccinated with the H1N1 vaccine. We focused on its impact on antibody production, as well as T-cell and B-cell development. Our findings reveal that the combination of lovastatin and H1N1 vaccine (Lov/H1N1) significantly enhances the production of H1N1-specific serum IgG and hemagglutination inhibition (HI) antibodies. Additionally, it promotes T-cell activation in both draining lymph nodes (dLNs) and the spleen. Analysis of cytokines produced after antigenic restimulation of splenic lymphocytes from immunized mice showed that the Lov/H1N1 combination induces both Th1-type (IFNγ, TNFα) and Th2-type (IL4, IL6) responses. Moreover, Lov/H1N1 facilitates the formation of germinal centers (GCs), which are crucial for the generation of memory B cells and long-lived plasma cells. These results indicate that lovastatin is a promising adjuvant candidate, capable of inducing robust cellular and humoral immune responses, thereby overcoming the limitations of Alum adjuvants. Our study provides a foundation for future research on combined vaccine strategies, highlighting Lovastatin's potential to enhance vaccine efficacy through improved immune responses.
Collapse
Affiliation(s)
- Zuchen Song
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Yantong Zhou
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Lina Jiao
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Tianyu Zhu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Ruihong Yu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Zheng Wang
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Yawei Qiu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Jinfeng Miao
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Ting Cai
- Guoke Ningbo Life Science and Health Industry Research Institute, Ningbo, Zhejiang 315032, PR China
| | - Shun Zhang
- Guoke Ningbo Life Science and Health Industry Research Institute, Ningbo, Zhejiang 315032, PR China
| | - Huina Liu
- Guoke Ningbo Life Science and Health Industry Research Institute, Ningbo, Zhejiang 315032, PR China
| | - Haifeng Sun
- Key Laboratory of Bacteriology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Yuechao Sun
- Guoke Ningbo Life Science and Health Industry Research Institute, Ningbo, Zhejiang 315032, PR China
| | - Deyun Wang
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China.
| | - Zhenguang Liu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China; Guoke Ningbo Life Science and Health Industry Research Institute, Ningbo, Zhejiang 315032, PR China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China.
| |
Collapse
|
35
|
Joshi R, Sheth D, Beladiya J, Patel C, Solanki N, Dalal M, Kyada A, Patel SB. Novel Targets for the Development of Tuberculosis Vaccine. Curr Drug Discov Technol 2025; 22:e070624230860. [PMID: 38859789 DOI: 10.2174/0115701638285518240601075811] [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: 02/08/2024] [Revised: 04/10/2024] [Accepted: 04/22/2024] [Indexed: 06/12/2024]
Abstract
In underdeveloped nations, tuberculosis (TB) continues to be a major source of morbidity and mortality. The currently available vaccine against tuberculosis in endemic areas is mainly ineffective, which triggers the need for a clinically effective vaccine against tuberculosis. In the present review, we emphasized the impact of genetic variations in the BCG strains, which influence the efficacy of BCG vaccines. We also discussed the current status of BCG vaccines and their potential mechanisms on the modulation of B cells and, thereby, humoral immunity, which trigger immune responses against various intracellular pathogens. Further, we also elaborated upon the pre-clinical and clinical studies demonstrating the efficacy and safety of the vaccines. Moreover, we also presented the putative novel targets such as polysaccharide-induced antibodies for the protection against Mtb, PGRS domain as an important target for Humoral immunity, HLA-E pathway-Target strategy for new TB vaccine, Coronin-1a - Novel player for Mycobacterial survival, IRGM, IFN-I3, an autophagy inducer with Irgm1 serving as a core part in the Tuberculosis vaccine development.
Collapse
Affiliation(s)
- Rushika Joshi
- Department of Pharmacology, L.M. College of Pharmacy, Navrangpura, Ahmedabad, 380009, Gujarat, India
| | - Devang Sheth
- Department of Pharmacology, L.M. College of Pharmacy, Navrangpura, Ahmedabad, 380009, Gujarat, India
| | - Jayesh Beladiya
- Department of Pharmacology, L.M. College of Pharmacy, Navrangpura, Ahmedabad, 380009, Gujarat, India
| | - Chirag Patel
- Department of Pharmacology, L.M. College of Pharmacy, Navrangpura, Ahmedabad, 380009, Gujarat, India
| | - Nilay Solanki
- Department of Pharmacology, Ramanbhai Patel College of Pharmacy, Charotar University of Science and Technology, CHARUSAT Campus, Changa, Petlad, Anand, Gujarat, 388421, India
| | - Mittal Dalal
- Department of Pharmacology, L.M. College of Pharmacy, Navrangpura, Ahmedabad, 380009, Gujarat, India
| | - Ashish Kyada
- Department of Pharmaceutical Sciences, Marwadi University, Rajkot, 360003, Gujarat, India
| | - Sandip B Patel
- Department of Pharmacology, L.M. College of Pharmacy, Navrangpura, Ahmedabad, 380009, Gujarat, India
| |
Collapse
|
36
|
Almansour ZH. Influence of immunological awareness on enhancing the overall complying with health instructions and necessary vaccines during epidemics and pandemics. Hum Vaccin Immunother 2024; 20:2406066. [PMID: 39314074 PMCID: PMC11423657 DOI: 10.1080/21645515.2024.2406066] [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: 06/25/2024] [Revised: 09/03/2024] [Accepted: 09/15/2024] [Indexed: 09/25/2024] Open
Abstract
Immunological awareness plays a pivotal role in promoting adherence to health instructions during public health emergencies. This study assessed the influence of specialized immunology education on compliance behaviors among higher education students. To assess the influence of specialized immunology education on compliance behaviors toward health instructions and necessary vaccines during epidemics and pandemics among higher education students. A cross-sectional survey was conducted among 532 students at King Faisal University, Saudi Arabia using stratified random sampling. A validated questionnaire examined demographics, health awareness, and specific immunological knowledge. Regression analysis was performed to assess predictors of compliance with health instructions. Considerable knowledge gaps were found around fundamental immunological concepts despite fair awareness about vaccines. Marked disparities existed across gender and academic disciplines. Regression modeling established specialized immunology training as a significant predictor of compliance with guidelines like masking and vaccination during epidemics (p < .001). The findings highlight the need for customized immunology curricula targeting students from nonscientific backgrounds. Incorporating immunology training in continuing education programs for healthcare professionals and public officials can further promote compliance. Multifaceted public health campaigns combining immunology education with initiatives to address socio-cultural barriers are warranted. Further research should explore sustainable educational interventions to enhance long-term retention and integration of immunology knowledge into health behaviors.
Collapse
Affiliation(s)
- Zainab H. Almansour
- Biological Sciences Department, College of Science, King Faisal University, Al Ahsa, Saudi Arabia
| |
Collapse
|
37
|
Lee RU, Watson NL, Glickman GL, White L, Isidean SD, Porter CK, Hollis-Perry M, Walther SR, Maiolatesi S, Sedegah M, Ganeshan H, Huang J, Boulifard DA, Ewing D, Sundaram AK, Harrison EM, DeTizio K, Belmonte M, Belmonte A, Inoue S, Easterling A, Cooper ES, Danko J. A randomized clinical trial of the impact of melatonin on influenza vaccine: Outcomes from the melatonin and vaccine response immunity and chronobiology study (MAVRICS). Hum Vaccin Immunother 2024; 20:2419742. [PMID: 39539030 PMCID: PMC11572083 DOI: 10.1080/21645515.2024.2419742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 10/03/2024] [Accepted: 10/18/2024] [Indexed: 11/16/2024] Open
Abstract
Vaccine immunogenicity is affected by a variety of factors. Melatonin has been reported to affect immune responses to vaccines and infection. This was a randomized open-label trial - in which adults scheduled to receive the influenza vaccine were randomized to 5 mg melatonin or control to evaluate the effect of post-vaccination melatonin on humoral (hemagglutination-inhibition assays, HAI) and cellular (FluoroSpot) vaccine-specific cytokine responses 14-21 days post-vaccination. A total of 108 participants (melatonin treatment group: 53; control group: 55) completed the study. The groups were similar in baseline characteristics, including sleep as measured by the Pittsburgh Sleep Quality Index. Seroconversion rates or geometric mean fold rises (GMFR) in HAI titers did not vary by treatment group. There were also no statistically significant differences between pre- and post-vaccination levels of interferon gamma (IFN-γ) or granzyme B (GzB) by treatment; however, there was a significantly higher fold rise in the double secretor (IFN-γ + GzB) peripheral blood mononuclear cells for influenza vaccine in subjects taking daily melatonin (GMFR 1.7; 95% CI 1.3, 2.3) compared to those who did not (GMFR 0.9; 95% CI 0.7, 1.1) (p < .001). Daily melatonin for 14 days post-influenza vaccination significantly increased the cellular co-expression of IFN-γ + GzB; however, there were no other differences in the cellular or humoral responses. Future studies of the potential utility of melatonin for enhancing vaccine response with larger sample sizes may help elucidate candidate mechanisms for these limited effects, including any interactions with the circadian system.
Collapse
Affiliation(s)
- Rachel U. Lee
- Department of Medicine, Walter Reed National Military Medical Center, Bethesda, MD, USA
- Department of Psychiatry and Neuroscience, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Nora L. Watson
- Department of Medicine, Walter Reed National Military Medical Center, Bethesda, MD, USA
| | - Gena L. Glickman
- Department of Psychiatry and Neuroscience, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Lindsey White
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, USA
- Department of Translational and Clinical Research, Naval Medical Research Command, Silver Spring, MD, USA
| | - Sandra D. Isidean
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, USA
- Department of Translational and Clinical Research, Naval Medical Research Command, Silver Spring, MD, USA
| | - Chad K. Porter
- Department of Translational and Clinical Research, Naval Medical Research Command, Silver Spring, MD, USA
| | - Monique Hollis-Perry
- Department of Psychiatry and Neuroscience, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Samuel R. Walther
- Department of Translational and Clinical Research, Naval Medical Research Command, Silver Spring, MD, USA
| | - Santina Maiolatesi
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, USA
- Department of Translational and Clinical Research, Naval Medical Research Command, Silver Spring, MD, USA
| | - Martha Sedegah
- Department of Translational and Clinical Research, Naval Medical Research Command, Silver Spring, MD, USA
| | - Harini Ganeshan
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, USA
- Department of Translational and Clinical Research, Naval Medical Research Command, Silver Spring, MD, USA
| | - Jun Huang
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, USA
- Department of Translational and Clinical Research, Naval Medical Research Command, Silver Spring, MD, USA
| | - David A. Boulifard
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, USA
- Department of Translational and Clinical Research, Naval Medical Research Command, Silver Spring, MD, USA
| | - Daniel Ewing
- Department of Translational and Clinical Research, Naval Medical Research Command, Silver Spring, MD, USA
| | - Appavu K. Sundaram
- Department of Translational and Clinical Research, Naval Medical Research Command, Silver Spring, MD, USA
| | - Elizabeth M. Harrison
- Department of Psychiatry and Neuroscience, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, USA
| | - Katherine DeTizio
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, USA
- Department of Translational and Clinical Research, Naval Medical Research Command, Silver Spring, MD, USA
| | - Maria Belmonte
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, USA
- Department of Translational and Clinical Research, Naval Medical Research Command, Silver Spring, MD, USA
| | - Arnel Belmonte
- Department of Translational and Clinical Research, Naval Medical Research Command, Silver Spring, MD, USA
- General Dynamics Information Technology, Falls Church, Virginia, MD, USA
| | - Sandra Inoue
- Department of Translational and Clinical Research, Naval Medical Research Command, Silver Spring, MD, USA
- General Dynamics Information Technology, Falls Church, Virginia, MD, USA
| | - Alexandra Easterling
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, USA
| | - Elizabeth S. Cooper
- Department of Translational and Clinical Research, Naval Medical Research Command, Silver Spring, MD, USA
| | - Janine Danko
- Department of Translational and Clinical Research, Naval Medical Research Command, Silver Spring, MD, USA
| |
Collapse
|
38
|
Zhou J, Bowman CJ, Markiewicz VR, Manickam B, Gomme E, Sellers RS, Rohde CM. Favorable Nonclinical Safety Profile of RSVpreF Bivalent Vaccine in Rats and Rabbits. Vaccines (Basel) 2024; 13:26. [PMID: 39852805 PMCID: PMC11769190 DOI: 10.3390/vaccines13010026] [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: 11/22/2024] [Revised: 12/21/2024] [Accepted: 12/29/2024] [Indexed: 01/26/2025] Open
Abstract
Background: Respiratory syncytial virus (RSV) infections usually cause mild, cold-like symptoms in most people, but are a leading infectious disease causing infant death and hospitalization and can result in increased morbidity and mortality in older adults and at-risk individuals. Pfizer has developed Abrysvo®, an unadjuvanted bivalent recombinant protein subunit vaccine containing prefusion-stabilized fusion (F) proteins representing RSV A and RSV B subgroups (RSVpreF). It is the only RSV vaccine approved for both maternal immunization to protect infants and active immunization of older adults (≥60 years) and 18-59-year-old individuals with high-risk conditions for prevention of RSV disease. Methods: Nonclinical safety studies, including a repeat-dose toxicity (RDT) study in rats and a combined developmental and reproductive toxicity (DART) study in rabbits, were conducted to support early clinical development. Study designs and parameters evaluated in these studies were consistent with principles and practices as outlined in relevant regulatory guidelines. RSVpreF bivalent vaccine, with or without Al(OH)3, was administered intramuscularly (IM) at 2× the human dose to animals in both studies. Results: Locally tolerated, reversible, inflammatory responses at the injection sites and the draining lymph nodes were observed as typical findings following vaccination. No effect of RSVpreF, with or without Al(OH)3, was observed on female fertility or on embryo-fetal or postnatal survival, growth, or development in the DART study. In both studies, robust immune responses to both RSV A and B antigens were observed, especially with the Al(OH)3 formulation. Conclusions: RSVpreF was well-tolerated both locally and systemically without any adverse effects on reproductive and developmental endpoints.
Collapse
Affiliation(s)
- Jun Zhou
- Drug Safety Research and Development, Pfizer Research & Development, Groton, CT 06340, USA; (C.J.B.)
| | - Christopher J. Bowman
- Drug Safety Research and Development, Pfizer Research & Development, Groton, CT 06340, USA; (C.J.B.)
| | - Vicki R. Markiewicz
- Drug Safety Research and Development, Pfizer Research & Development, Groton, CT 06340, USA; (C.J.B.)
- Independent Researcher, East Lyme, CT 06333, USA
| | - Balasubramanian Manickam
- Drug Safety Research and Development, Pfizer Research & Development, Groton, CT 06340, USA; (C.J.B.)
| | - Emily Gomme
- Clinical Immunology and High-Throughput Assays, Vaccine Research and Development, Pfizer Research & Development, Pearl River, NY 10965, USA
| | - Rani S. Sellers
- Drug Safety Research and Development, Pfizer Research & Development, Pearl River, NY 10965, USA
| | - Cynthia M. Rohde
- Drug Safety Research and Development, Pfizer Research & Development, Pearl River, NY 10965, USA
| |
Collapse
|
39
|
Copenhaver WK, Goodwin BJ, Simonetti A, Shah KP, Averell NJ, Lo DF, Jermyn RT. Immunization-related complex regional pain syndrome: A systematic review of case reports. PCN REPORTS : PSYCHIATRY AND CLINICAL NEUROSCIENCES 2024; 3:e70041. [PMID: 39664886 PMCID: PMC11631839 DOI: 10.1002/pcn5.70041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 10/28/2024] [Accepted: 11/14/2024] [Indexed: 12/13/2024]
Abstract
Aim Vaccines have been shown to have the highest efficacy in preventing infectious diseases through their ability to induce immunological memory against pathogens. An adverse reaction to a vaccine is an unexpected medical occurrence following immunization. Complex regional pain syndrome (CRPS) is a disease that has undergone much controversy regarding its onset post-vaccination. This systematic review aims to evaluate cases of CRPS post-vaccination to better understand the manifestation of the disease and its potential association with vaccines. Methods A systematic review of case reports was conducted employing the PRISMA 2020 guidelines. Outcomes of interest include type of vaccination, patient age, patient sex, time to symptom onset, and medical history including but not limited to previous autoimmune diseases, psychological illness, physical tissue trauma, and neurological disease. Results Initial querying of the five databases yielded 404 articles. Following a thorough review of articles, only 14 remained, comprising 18 cases. Studies included cases of CRPS development following tetanus, hepatitis B, hepatitis A, rubella, influenza, tetanus-diphtheria, human papillomavirus, and COVID-19 vaccine administration. Conclusion The limitations of evidence used in this study highlight the need for a greater output of higher-level evidence in the form of controlled trials and retrospective studies to help further elucidate the connection between vaccine use and the development of CRPS in patients. Currently, vaccines continue to be safe for global public use.
Collapse
Affiliation(s)
| | | | | | - Kunal P. Shah
- Futures Forward Research InstituteToms RiverNew JerseyUSA
| | | | - David F. Lo
- Futures Forward Research InstituteToms RiverNew JerseyUSA
- Department of BiologyRutgers, The State University of New JerseyNew BrunswickNew JerseyUSA
| | - Richard T. Jermyn
- Department of MedicineRowan University School of Osteopathic MedicineStratfordNew JerseyUSA
| |
Collapse
|
40
|
Hasan A, Ibrahim M, Alonazi WB, Shen J. Application of immunoinformatics to develop a novel and effective multiepitope chimeric vaccine against Variovorax durovernensis. Comput Biol Chem 2024; 113:108266. [PMID: 39504600 DOI: 10.1016/j.compbiolchem.2024.108266] [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: 05/08/2024] [Revised: 08/31/2024] [Accepted: 10/20/2024] [Indexed: 11/08/2024]
Abstract
Bloodstream infections pose a significant public health challenge caused by resistant bacteria such as Variovorax durovernensis, a recently reported Gram-negative bacterium, worsening the burden on healthcare systems. The design of a vaccine using chimeric peptides derived from a representative V. durovernensis strain holds significant promise for preventing disease onset. The current study aimed to employ reverse vaccinology (RV) approaches such as the retrieval of V. durovernensis proteomics data, removal of redundant proteins by CD-HIT, filtering of non-homologous proteins to humans and essential proteins, identification of outer membrane (OM) proteins by CELLO and PSORTb. Following these steps immunoinformatic approaches were applied, such as epitope prediction by IEDB, vaccine design using linkers and adjuvant and analysis of antigenicity, allergenicity, safety and stability. Among the 4208 nonredundant proteins, an OmpA family protein (A0A940EKP4) was designated a potential candidate for the development of a multiepitope vaccine construct. Upon analysis of OM protein, six immunodominant (B cell) epitopes were found on the basis of the chimeric construct following the prediction of CTL stands cytotoxic T lymphocyte and HTL stands helper T lymphocyte epitopes. To ensure comprehensive population coverage globally, the CTL and HTL coverage rates were 58.18 % and 46.56 %, respectively, and 77.23 % overall. By utilizing EAAAK, GPGPG, and AAY linkers, Cholera toxin B subunit adjuvants, and appropriate epitopes were smoothly incorporated into a chimeric vaccine effectively triggering both adaptive and innate immune responses. For example, the administered antigen showed a peak in counts on the fifthday post injection and then gradually declined until the fifteenth day. Elevated levels of several antibodies (IgG + IgM > 700,000; IgM > 600,000; IgG1 + IgG2; IgG1 > 500,000) were observed as decreased in the antigen concentration. Molecular dynamics simulations carried out via iMODS revealed strong correlations between residue pairs, highlighting the stability of the docked complex. The designed vaccine has promising potential in eliciting specific immunogenic responses, thereby facilitating future research for vaccine development against V. durovernensis.
Collapse
Affiliation(s)
- Ahmad Hasan
- Institute of Biotechnology, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, PR China
| | - Muhammad Ibrahim
- Institute of Biotechnology, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, PR China
| | - Wadi B Alonazi
- Health Administration Department, College of Business Administration, King Saud University, Riyadh, Saudi Arabia
| | - Jian Shen
- Department of Medical Administration, Zhejiang Province People Hospital, Affiliated People Hospital, Hangzhou Medical College Hangzhou, Zhejiang, PR China.
| |
Collapse
|
41
|
Khan A, Ammar Zahid M, Farrukh F, Salah Abdelsalam S, Mohammad A, Al-Zoubi RM, Shkoor M, Ait Hssain A, Wei DQ, Agouni A. Integrated structural proteomics and machine learning-guided mapping of a highly protective precision vaccine against mycoplasma pulmonis. Int Immunopharmacol 2024; 141:112833. [PMID: 39153303 DOI: 10.1016/j.intimp.2024.112833] [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: 06/13/2024] [Revised: 07/09/2024] [Accepted: 07/27/2024] [Indexed: 08/19/2024]
Abstract
Mycoplasma pulmonis (M. pulmonis) is an emerging respiratory infection commonly linked to prostate cancer, and it is classified under the group of mycoplasmas. Improved management of mycoplasma infections is essential due to the frequent ineffectiveness of current antibiotic treatments in completely eliminating these pathogens from the host. The objective of this study is to design and construct effective and protective vaccines guided by structural proteomics and machine learning algorithms to provide protection against the M. pulmonis infection. Through a thorough examination of the entire proteome of M. pulmonis, four specific targets Membrane protein P80, Lipoprotein, Uncharacterized protein and GGDEF domain-containing protein have been identified as appropriate for designing a vaccine. The proteins underwent mapping of cytotoxic T lymphocyte (CTL), helper T lymphocyte (HTL) (IFN)-γ ±, and B-cell epitopes using artificial and recurrent neural networks. The design involved the creation of mRNA and peptide-based vaccine, which consisted of 8 CTL epitopes associated by GGS linkers, 7 HTL (IFN-positive) epitopes, and 8 B-cell epitopes joined by GPGPG linkers. The vaccine designed exhibit antigenic behavior, non-allergenic qualities, and exceptional physicochemical attributes. Structural modeling revealed that correct folding is crucial for optimal functioning. The coupling of the MEVC and Toll-like Receptors (TLR)1, TLR2, and TLR6 was examined through molecular docking experiments. This was followed by molecular simulation investigations, which included binding free energy estimations. The results indicated that the dynamics of the interaction were stable, and the binding was strong. In silico cloning and optimization analysis revealed an optimized sequence with a GC content of 49.776 % and a CAI of 0.982. The immunological simulation results showed strong immune responses, with elevated levels of active and plasma B-cells, regulatory T-cells, HTL, and CTL in both IgM+IgG and secondary immune responses. The antigen was completely cleared by the 50th day. This study lays the foundation for creating a potent and secure vaccine candidate to combat the newly identified M. pulmonis infection in people.
Collapse
Affiliation(s)
- Abbas Khan
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, P.O. Box 2713, Doha, Qatar
| | - Muhammad Ammar Zahid
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, P.O. Box 2713, Doha, Qatar.
| | - Farheen Farrukh
- Gujranwala Medical College, 5 KM Alipur Chatha Rd, Gondlanwala Rd, Gujranwala, Pakistan
| | - Shahenda Salah Abdelsalam
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, P.O. Box 2713, Doha, Qatar.
| | - Anwar Mohammad
- Department of Biochemistry and Molecular Biology, Dasman Diabetes Institute, Dasman, Kuwait
| | - Raed M Al-Zoubi
- Surgical Research Section, Department of Surgery, Hamad Medical Corporation, Doha, Qatar; Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, P.O. Box 2713, Doha, Qatar; Department of Chemistry, Jordan University of Science and Technology, P.O. Box 3030, Irbid 22110, Jordan.
| | - Mohanad Shkoor
- Department of Chemistry, College of Arts and Science, Qatar University, P.O. Box 2713, Doha, Qatar.
| | - Ali Ait Hssain
- Medical Intensive Care Unit, Hamad Medical Corporation, Doha, Qatar
| | - Dong-Qing Wei
- Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China.
| | - Abdelali Agouni
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, P.O. Box 2713, Doha, Qatar.
| |
Collapse
|
42
|
Dhall A, Patiyal S, Raghava GPS. A hybrid method for discovering interferon-gamma inducing peptides in human and mouse. Sci Rep 2024; 14:26859. [PMID: 39501025 PMCID: PMC11538504 DOI: 10.1038/s41598-024-77957-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 10/28/2024] [Indexed: 11/08/2024] Open
Abstract
Interferon-gamma (IFN-γ) is a versatile pleiotropic cytokine essential for both innate and adaptive immune responses. It exhibits both pro-inflammatory and anti-inflammatory properties, making it a promising therapeutic candidate for treating various infectious diseases and cancers. We present IFNepitope2, a host-specific technique to annotate IFN-γ inducing peptides, it is an updated version of IFNepitope introduced by Dhanda et al. In this study, dataset used for developing prediction method contain experimentally validated 25,492 and 7983 IFN-γ inducing peptides in human and mouse host, respectively. In initial phase, machine learning techniques have been exploited to develop classification model using wide range of peptide features. Further, to improve machine learning based models or alignment free models, we explore potential of similarity-based technique BLAST. Finally, a hybrid model has been developed that combine best machine learning based model with BLAST. In most of the case, models based on extra tree perform better than other machine learning techniques. In case of peptide features, compositional feature particularly dipeptide composition performs better than one-hot encoding or binary profile. Our best machine learning based models achieved AUROC 0.89 and 0.83 for human and mouse host, respectively. The hybrid model achieved the AUROC 0.90 and 0.85 for human and mouse host, respectively. All models have been evaluated on an independent/validation dataset not used for training or testing these models. Newly developed method performs better than existing method on independent dataset. The major objective of this study is to predict, design and scan IFN-γ inducing peptides, thus server/software have been developed ( https://webs.iiitd.edu.in/raghava/ifnepitope2/ ). This method is also available as standalone at https://github.com/raghavagps/ifnepitope2 and python package index at https://pypi.org/project/ifnepitope2/ .
Collapse
Affiliation(s)
- Anjali Dhall
- Department of Computational Biology, Indraprastha Institute of Information Technology, Okhla Industrial Estate, Phase III, (Near Govind Puri Metro Station), New Delhi, 110020, India
| | - Sumeet Patiyal
- Department of Computational Biology, Indraprastha Institute of Information Technology, Okhla Industrial Estate, Phase III, (Near Govind Puri Metro Station), New Delhi, 110020, India
| | - Gajendra P S Raghava
- Department of Computational Biology, Indraprastha Institute of Information Technology, Okhla Industrial Estate, Phase III, (Near Govind Puri Metro Station), New Delhi, 110020, India.
| |
Collapse
|
43
|
Tang H, Liu X, Ke J, Tang Y, Luo S, Li XK, Huang M. New perspectives of exosomes in urologic malignancies - Mainly focus on biomarkers and tumor microenvironment. Pathol Res Pract 2024; 263:155645. [PMID: 39476607 DOI: 10.1016/j.prp.2024.155645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 08/09/2024] [Accepted: 10/02/2024] [Indexed: 11/10/2024]
Abstract
Bladder cancer (BCa) and renal cell carcinoma (RCC) are prevalent urologic malignancies (UM) characterized by high morbidity and frequent recurrence. Current diagnostic approaches, often invasive, often indicate an advanced disease stage. And the complex tumor microenvironment often promotes tumor progression and induces resistance to chemotherapy. Current diagnostic and therapeutic modalities often fail to achieve satisfactory outcomes for patients. Exosomes transport diverse cargoes, including cytokines, proteins, lipids, non-coding RNAs, and microRNAs, crucial for intercellular communication. Exosomes have shown potential as biomarkers for UM, participating in tumor progression, especially within the tumor microenvironment (TME), including tumor cell apoptosis, proliferation, migration, invasion, depletion of immune cell function, epithelial-mesenchymal transition (EMT), angiogenesis, and more.In this review, we summarize research advances related to exosomes in UM, focusing on the role of exosomes as biomarkers in bladder and renal cancer, highlighting their significance within the TME.
Collapse
Affiliation(s)
- Hai Tang
- Urology department, the Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Xing Liu
- Urology department, the Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Jingwei Ke
- Urology department, the Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Yiquan Tang
- Urology department, the Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Songtao Luo
- Urology department, the Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Xu Kun Li
- Urology department, the Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Mingwei Huang
- Urology department, the Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China.
| |
Collapse
|
44
|
Kim MY, Mason HS, Ma JKC, Reljic R. Recombinant immune complexes as vaccines against infectious diseases. Trends Biotechnol 2024; 42:1427-1438. [PMID: 38825437 DOI: 10.1016/j.tibtech.2024.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 05/03/2024] [Accepted: 05/10/2024] [Indexed: 06/04/2024]
Abstract
New vaccine technologies are needed to combat many existing infections and prepare better for those that may emerge in the future. The conventional technologies that rely on protein-based vaccines are still severely restricted by the sparsity and poor accessibility of available adjuvants. One possible solution to this problem is to enhance antigen immunogenicity by a more natural means by complexing it with antibodies in the form of immune complexes (ICs). However, natural ICs are impractical as vaccines, and significant research efforts have been made to generate them in recombinant form, with plant bioengineering being at the forefront of these efforts. Here, we describe the challenges and progress made to date to make recombinant IC vaccines applicable to humans.
Collapse
Affiliation(s)
- Mi-Young Kim
- St. George's University of London, London, UK; Jeonbuk National University, Jeonju, South Korea
| | - Hugh S Mason
- School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | | | | |
Collapse
|
45
|
Mohagheghi M, Abisoye-Ogunniyan A, Evans AC, Peterson AE, Bude GA, Hoang-Phou S, Vannest BD, Hall D, Rasley A, Weilhammer DR, Fischer NO, He W, Robinson BV, Pal S, Slepenkin A, de la Maza L, Coleman MA. Cell-Free Screening, Production and Animal Testing of a STI-Related Chlamydial Major Outer Membrane Protein Supported in Nanolipoproteins. Vaccines (Basel) 2024; 12:1246. [PMID: 39591149 PMCID: PMC11598365 DOI: 10.3390/vaccines12111246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Revised: 10/22/2024] [Accepted: 10/23/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Vaccine development against Chlamydia, a prevalent sexually transmitted infection (STI), is imperative due to its global public health impact. However, significant challenges arise in the production of effective subunit vaccines based on recombinant protein antigens, particularly with membrane proteins like the Major Outer Membrane Protein (MOMP). METHODS Cell-free protein synthesis (CFPS) technology is an attractive approach to address these challenges as a method of high-throughput membrane protein and protein complex production coupled with nanolipoprotein particles (NLPs). NLPs provide a supporting scaffold while allowing easy adjuvant addition during formulation. Over the last decade, we have been working toward the production and characterization of MOMP-NLP complexes for vaccine testing. RESULTS The work presented here highlights the expression and biophysical analyses, including transmission electron microscopy (TEM) and dynamic light scattering (DLS), which confirm the formation and functionality of MOMP-NLP complexes for use in animal studies. Moreover, immunization studies in preclinical models compare the past and present protective efficacy of MOMP-NLP formulations, particularly when co-adjuvanted with CpG and FSL1. CONCLUSION Ex vivo assessments further highlight the immunomodulatory effects of MOMP-NLP vaccinations, emphasizing their potential to elicit robust immune responses. However, further research is warranted to optimize vaccine formulations further, validate efficacy against Chlamydia trachomatis, and better understand the underlying mechanisms of immune response.
Collapse
Affiliation(s)
- Mariam Mohagheghi
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA 94551, USA (A.A.-O.); (S.H.-P.); (B.D.V.); (A.R.); (B.V.R.)
| | - Abisola Abisoye-Ogunniyan
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA 94551, USA (A.A.-O.); (S.H.-P.); (B.D.V.); (A.R.); (B.V.R.)
| | - Angela C. Evans
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA 94551, USA (A.A.-O.); (S.H.-P.); (B.D.V.); (A.R.); (B.V.R.)
| | - Alexander E. Peterson
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA 94551, USA (A.A.-O.); (S.H.-P.); (B.D.V.); (A.R.); (B.V.R.)
| | - Gregory A. Bude
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA 94551, USA (A.A.-O.); (S.H.-P.); (B.D.V.); (A.R.); (B.V.R.)
| | - Steven Hoang-Phou
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA 94551, USA (A.A.-O.); (S.H.-P.); (B.D.V.); (A.R.); (B.V.R.)
| | - Byron Dillon Vannest
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA 94551, USA (A.A.-O.); (S.H.-P.); (B.D.V.); (A.R.); (B.V.R.)
| | - Dominique Hall
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA 94551, USA (A.A.-O.); (S.H.-P.); (B.D.V.); (A.R.); (B.V.R.)
| | - Amy Rasley
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA 94551, USA (A.A.-O.); (S.H.-P.); (B.D.V.); (A.R.); (B.V.R.)
| | - Dina R. Weilhammer
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA 94551, USA (A.A.-O.); (S.H.-P.); (B.D.V.); (A.R.); (B.V.R.)
| | - Nicholas O. Fischer
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA 94551, USA (A.A.-O.); (S.H.-P.); (B.D.V.); (A.R.); (B.V.R.)
| | - Wei He
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA 94551, USA (A.A.-O.); (S.H.-P.); (B.D.V.); (A.R.); (B.V.R.)
| | - Beverly V. Robinson
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA 94551, USA (A.A.-O.); (S.H.-P.); (B.D.V.); (A.R.); (B.V.R.)
| | - Sukumar Pal
- Department of Pathology and Laboratory Medicine, University of California, Irvine, CA 92697, USA
| | - Anatoli Slepenkin
- Department of Pathology and Laboratory Medicine, University of California, Irvine, CA 92697, USA
| | - Luis de la Maza
- Department of Pathology and Laboratory Medicine, University of California, Irvine, CA 92697, USA
| | - Matthew A. Coleman
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA 94551, USA (A.A.-O.); (S.H.-P.); (B.D.V.); (A.R.); (B.V.R.)
| |
Collapse
|
46
|
Wang C, Macintyre AN, Oguin TH, McCarthy KR, Moody MA, Yuan F. Spatiotemporal control of immune responses with nucleic acid cocktail vaccine. ADVANCED THERAPEUTICS 2024; 7:2400263. [PMID: 40248361 PMCID: PMC12002596 DOI: 10.1002/adtp.202400263] [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: 06/18/2024] [Indexed: 04/19/2025]
Abstract
Nucleic acid vaccines play important roles in prevention and treatment of diseases. However, limited immunogenicity remains a major obstacle for DNA vaccine applications in the clinic. To address the issue, the present study investigates a cocktail approach to DNA vaccination. In this proof-of-the-concept study, the cocktail consists of two DNAs encoding viral hemagglutinin (HA) and granulocyte-macrophage colony stimulatory factor (GM-CSF), respectively. Data from the study demonstrate that recruitment and activation of antigen-presenting cells (APCs) can be substantially improved by spatiotemporal regulation of GM-CSF and HA expressions at the site of vaccination. The types of recruited APCs and their phenotypes are also controllable by adjusting the cocktail compositions. Compared to mono-ingredient vaccine, the optimized cocktail vaccine is able to enhance the anti-viral humoral and T cell immune responses. No significant systemic inflammation has been detected after either prime or boost immunization using the cocktail vaccine. Data in the study suggest that the DNA cocktail is a safe, effective, and controllable platform for improving vaccine efficacy.
Collapse
Affiliation(s)
- Chunxi Wang
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Andrew N. Macintyre
- Duke Human Vaccine Institute (DHVI), Duke University, Durham, NC 27708, USA
- Department of Medicine, Duke University School of Medicine, Durham, NC 27708, USA
| | - Thomas H. Oguin
- Duke Human Vaccine Institute (DHVI), Duke University, Durham, NC 27708, USA
| | - Kevin R. McCarthy
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine; Pittsburgh, PA 15261, USA
| | - M. Anthony Moody
- Duke Human Vaccine Institute (DHVI), Duke University, Durham, NC 27708, USA
- Department of Pediatrics, Duke University School of Medicine, Durham, NC 27708, USA
| | - Fan Yuan
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| |
Collapse
|
47
|
Qian H, Tian L, Liu W, Liu L, Li M, Zhao Z, Lei X, Zheng W, Zhao Z, Zheng X. Adenovirus type 5-expressing Gn induces better protective immunity than Gc against SFTSV infection in mice. NPJ Vaccines 2024; 9:194. [PMID: 39426985 PMCID: PMC11490641 DOI: 10.1038/s41541-024-00993-y] [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: 03/16/2024] [Accepted: 10/10/2024] [Indexed: 10/21/2024] Open
Abstract
Severe fever with thrombocytopenia syndrome (SFTS) is caused by the SFTS virus (SFTSV) with high morbidity and mortality. The major immunodominant region of SFTSV surface glycoprotein (G) remains unclear. In this study, we constructed adenovirus type 5 (Ad5) vectored vaccine candidates expressing different regions of SFTSV G (Gn, Gc and Gn-Gc) and evaluated their immunogenicity and protective efficacy in mice. In wild-type mice, compared with Ad5-Gc or Ad5-Gn-Gc, Ad5-Gn recruited/activated more dendritic cells and B cells in lymph nodes or peripheral blood, causing Th1-/Th2-mediated responses in splenocytes and triggered a greater level of SFTSV-neutralizing antibodies. In IFNAR Ab-treated mice, immunization of Ad5-Gn exhibited better protection against SFTSV challenge than Ad5-Gc or Ad5-Gn-Gc. Furthermore, passive immunization revealed complete protective immunity of Gn-specific serum rather than Gc. Collectively, our data demonstrated that Gn is the immunodominant fragment of SFTSV G and could be a potential candidate for SFTSV vaccine development.
Collapse
Affiliation(s)
- Hua Qian
- The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Li Tian
- The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Wenkai Liu
- Department of Virology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Lele Liu
- Department of Virology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Menghua Li
- Department of Virology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Zhongxin Zhao
- Department of Laboratory Medicine, Linyi People's Hospital, Linyi, Shandong, China
| | - Xiaoying Lei
- Department of Virology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Wenwen Zheng
- Department of Virology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China.
| | - Zhongpeng Zhao
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
| | - Xuexing Zheng
- The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China.
| |
Collapse
|
48
|
Gopalaswamy R, Aravindhan V, Subbian S. The Ambivalence of Post COVID-19 Vaccination Responses in Humans. Biomolecules 2024; 14:1320. [PMID: 39456253 PMCID: PMC11506738 DOI: 10.3390/biom14101320] [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: 08/20/2024] [Revised: 10/09/2024] [Accepted: 10/14/2024] [Indexed: 10/28/2024] Open
Abstract
The Coronavirus disease 2019 (COVID-19) pandemic, caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), has prompted a massive global vaccination campaign, leading to the rapid development and deployment of several vaccines. Various COVID-19 vaccines are under different phases of clinical trials and include the whole virus or its parts like DNA, mRNA, or protein subunits administered directly or through vectors. Beginning in 2020, a few mRNA (Pfizer-BioNTech BNT162b2 and Moderna mRNA-1273) and adenovirus-based (AstraZeneca ChAdOx1-S and the Janssen Ad26.COV2.S) vaccines were recommended by WHO for emergency use before the completion of the phase 3 and 4 trials. These vaccines were mostly administered in two or three doses at a defined frequency between the two doses. While these vaccines, mainly based on viral nucleic acids or protein conferred protection against the progression of SARS-CoV-2 infection into severe COVID-19, and prevented death due to the disease, their use has also been accompanied by a plethora of side effects. Common side effects include localized reactions such as pain at the injection site, as well as systemic reactions like fever, fatigue, and headache. These symptoms are generally mild to moderate and resolve within a few days. However, rare but more serious side effects have been reported, including allergic reactions such as anaphylaxis and, in some cases, myocarditis or pericarditis, particularly in younger males. Ongoing surveillance and research efforts continue to refine the understanding of these adverse effects, providing critical insights into the risk-benefit profile of COVID-19 vaccines. Nonetheless, the overall safety profile supports the continued use of these vaccines in combating the pandemic, with regulatory agencies and health organizations emphasizing the importance of vaccination in preventing COVID-19's severe outcomes. In this review, we describe different types of COVID-19 vaccines and summarize various adverse effects due to autoimmune and inflammatory response(s) manifesting predominantly as cardiac, hematological, neurological, and psychological dysfunctions. The incidence, clinical presentation, risk factors, diagnosis, and management of different adverse effects and possible mechanisms contributing to these effects are discussed. The review highlights the potential ambivalence of human response post-COVID-19 vaccination and necessitates the need to mitigate the adverse side effects.
Collapse
Affiliation(s)
- Radha Gopalaswamy
- Directorate of Distance Education, Madurai Kamaraj University, Madurai 625021, India;
| | - Vivekanandhan Aravindhan
- Department of Genetics, Dr Arcot Lakshmanasamy Mudaliyar Post Graduate Institute of Basic Medical Sciences (Dr ALM PG IBMS), University of Madras, Taramani, Chennai 600005, India;
| | - Selvakumar Subbian
- Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA
| |
Collapse
|
49
|
Shrestha KR, Kim S, Jo A, Ragothaman M, Yoo SY. In vivo safety evaluation and tracing of arginylglycylaspartic acid-engineered phage nanofiber in murine model. J Mater Chem B 2024; 12:10258-10271. [PMID: 39300937 DOI: 10.1039/d4tb00823e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/22/2024]
Abstract
The engineered phage YSY184, mimicking the extracellular matrix nanofiber, effectively promotes stem cell differentiation and angiogenesis. This study evaluated its safety in a mouse model, monitoring weight, immunogenicity, spleen immune responses, and macrophage infiltration. Rapid clearance of YSY184 was observed, with peak tissue presence within three hours, significantly reduced by 24 hours, and negligible after one month. No adverse physiological or pathological effects were detected post-administration, affirming YSY184's safety and underscore its potential for therapeutic use, warranting further clinical exploration.
Collapse
Affiliation(s)
- Kshitiz Raj Shrestha
- Institute of Nanobio Convergence, Pusan National University, Busan 46241, Republic of Korea.
| | - Sehoon Kim
- Institute of Nanobio Convergence, Pusan National University, Busan 46241, Republic of Korea.
| | - Anna Jo
- Institute of Nanobio Convergence, Pusan National University, Busan 46241, Republic of Korea.
| | - Murali Ragothaman
- Institute of Nanobio Convergence, Pusan National University, Busan 46241, Republic of Korea.
| | - So Young Yoo
- Institute of Nanobio Convergence, Pusan National University, Busan 46241, Republic of Korea.
| |
Collapse
|
50
|
Cao Q, Fang H, Tian H. mRNA vaccines contribute to innate and adaptive immunity to enhance immune response in vivo. Biomaterials 2024; 310:122628. [PMID: 38820767 DOI: 10.1016/j.biomaterials.2024.122628] [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: 01/06/2024] [Revised: 05/02/2024] [Accepted: 05/19/2024] [Indexed: 06/02/2024]
Abstract
Messenger RNA (mRNA) therapeutics have been widely employed as strategies for the treatment and prevention of diseases. Amid the global outbreak of COVID-19, mRNA vaccines have witnessed rapid development. Generally, in the case of mRNA vaccines, the initiation of the innate immune system serves as a prerequisite for triggering subsequent adaptive immune responses. Critical cells, cytokines, and chemokines within the innate immune system play crucial and beneficial roles in coordinating tailored immune reactions towards mRNA vaccines. Furthermore, immunostimulators and delivery systems play a significant role in augmenting the immune potency of mRNA vaccines. In this comprehensive review, we systematically delineate the latest advancements in mRNA vaccine research, present an in-depth exploration of strategies aimed at amplifying the immune effectiveness of mRNA vaccines, and offer some perspectives and recommendations regarding the future advancements in mRNA vaccine development.
Collapse
Affiliation(s)
- Qiannan Cao
- State Key Laboratory of Physical Chemistry of Solid Surfaces, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China
| | - Huapan Fang
- State Key Laboratory of Physical Chemistry of Solid Surfaces, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China; Innovation Laboratory for Sciences and Technologies of Energy Materials of Fujian Province (IKKEM), Xiamen, 361005, China; Institute of Functional Nano and Soft Materials, Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China.
| | - Huayu Tian
- State Key Laboratory of Physical Chemistry of Solid Surfaces, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China; Innovation Laboratory for Sciences and Technologies of Energy Materials of Fujian Province (IKKEM), Xiamen, 361005, China.
| |
Collapse
|