1
|
Zhou CK, Liu ZZ, Peng ZR, Luo XY, Zhang XM, Zhang JG, Zhang L, Chen W, Yang YJ. M28 family peptidase derived from Peribacillus frigoritolerans initiates trained immunity to prevent MRSA via the complosome-phosphatidylcholine axis. Gut Microbes 2025; 17:2484386. [PMID: 40159598 PMCID: PMC11959922 DOI: 10.1080/19490976.2025.2484386] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 03/11/2025] [Accepted: 03/18/2025] [Indexed: 04/02/2025] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) represents a major global health threat due to its resistance to conventional antibiotics. The commensal microbiota maintains a symbiotic relationship with the host, playing essential roles in metabolism, energy regulation, immune modulation, and pathogen control. Mammals harbor a wide range of commensal bacteria capable of producing unique metabolites with potential therapeutic properties. This study demonstrated that M28 family peptidase (M28), derived from commensal bacteria Peribacillus frigoritolerans (P. f), provided protective effects against MRSA-induced pneumonia. M28 enhanced the phagocytosis and bactericidal activity of macrophages by inducing trained immunity. RNA sequencing and metabolomic analyses identified the CFB-C3a-C3aR-HIF-1α axis-mediated phosphatidylcholine accumulation as the key mechanism for M28-induced trained immunity. Phosphatidylcholine, like M28, also induced trained immunity. To enhance M28-mediated therapeutic potential, it was encapsulated in liposomes (M28-LNPs), which exhibited superior immune-stimulating properties compared to M28 alone. In vivo experiments revealed that M28-LNPs significantly reduced bacterial loads and lung damage following MRSA infection, which also provided enhanced protection against Klebsiella pneumoniae and Candida albicans. We first confirmed a link between complement activation and trained immunity, offering valuable insights into the treatment and prevention of complement-related autoimmune diseases.
Collapse
Affiliation(s)
- Cheng-Kai Zhou
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun Jilin, P. R China
| | - Zhen-Zhen Liu
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun Jilin, P. R China
| | - Zi-Ran Peng
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun Jilin, P. R China
| | - Xue-Yue Luo
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun Jilin, P. R China
| | - Xiao-Mei Zhang
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun Jilin, P. R China
| | - Jian-Gang Zhang
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun Jilin, P. R China
| | - Liang Zhang
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun Jilin, P. R China
| | - Wei Chen
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun Jilin, P. R China
| | - Yong-Jun Yang
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun Jilin, P. R China
| |
Collapse
|
2
|
Suzuki Y, Yakuwa M, Sato M, Samaridou E, Beck-Broichsitter M, Maeki M, Tokeshi M, Yamada Y, Harashima H, Sato Y. Splenic B cell-targeting lipid nanoparticles for safe and effective mRNA vaccine delivery. J Control Release 2025; 382:113687. [PMID: 40187650 DOI: 10.1016/j.jconrel.2025.113687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 04/01/2025] [Accepted: 04/03/2025] [Indexed: 04/07/2025]
Abstract
mRNA-loaded lipid nanoparticles (LNPs) have emerged as a potent and versatile platform that underpins the success of mRNA vaccines, but guidelines for designing safe and effective formulations with minimal off-target effects remain unclear. In this study, we focused on a rational design for a novel ionizable lipid library that is based on ionizable tri-oleoyl-tris (iTOT) compounds with a high yield via a simple 2-step synthesis. To further enhance the efficacy and safety of this potent library for vaccine applications, we identified the optimal composition for a vaccine by focusing on the molar ratio of specific lipid excipients in the formulation. This composition brought about a shift in delivery to the spleen, and the LNP formulation, which contained 15 mol% DSPC (15%DSPC-LNPs), was thoroughly taken up by both B cells and other splenic immune cells. This formulation requires neither additional lipid components nor targeting ligand modifications, and it is accompanied by antigen-specific cytotoxic T lymphocyte responses. The rigid, hydrophobic, and charge-neutral surface of 15%DSPC-LNPs minimizes apolipoprotein E-dependent hepatic uptake and maximizes complement receptor-mediated B-cell targeting. Furthermore, as an intramuscularly administered vaccine, 15%DSPC-LNPs induce antigen-specific immune responses and, importantly, results in significantly lower levels of hepatotoxicity compared with that of the mRNA vaccine formulations currently being marketed. In summary, this study demonstrated how the passive targeting of mRNA-LNPs to organs and cells could be regulated by designing novel ionizable lipids combined with adjusting the relative proportions of lipid components. The results of this study also emphasize how selective mRNA delivery to the spleen could avoid the liver, which highlights a promising strategy for the development of safe and effective vaccines.
Collapse
Affiliation(s)
- Yuichi Suzuki
- Laboratory for Molecular Design of Pharmaceutics, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12 Nishi-6, Kita-ku, Sapporo 060-0812, Japan
| | - Mai Yakuwa
- Laboratory for Molecular Design of Pharmaceutics, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12 Nishi-6, Kita-ku, Sapporo 060-0812, Japan
| | - Mina Sato
- Laboratory of Innovative Nanomedicine, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12 Nishi-6, Kita-ku, Sapporo 060-0812, Japan; Hokkaido University, Institute for Vaccine Research and Development (HU-IVReD)
| | | | | | - Masatoshi Maeki
- Division of Applied Chemistry, Faculty of Engineering, Hokkaido University, Kita-13 Nishi-8, Kita-ku, Sapporo 060-8628, Japan
| | - Manabu Tokeshi
- Division of Applied Chemistry, Faculty of Engineering, Hokkaido University, Kita-13 Nishi-8, Kita-ku, Sapporo 060-8628, Japan
| | - Yuma Yamada
- Laboratory for Molecular Design of Pharmaceutics, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12 Nishi-6, Kita-ku, Sapporo 060-0812, Japan; Hokkaido University, Institute for Vaccine Research and Development (HU-IVReD)
| | - Hideyoshi Harashima
- Laboratory of Innovative Nanomedicine, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12 Nishi-6, Kita-ku, Sapporo 060-0812, Japan; Hokkaido University, Institute for Vaccine Research and Development (HU-IVReD)
| | - Yusuke Sato
- Laboratory for Molecular Design of Pharmaceutics, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12 Nishi-6, Kita-ku, Sapporo 060-0812, Japan; Laboratory of Innovative Nanomedicine, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12 Nishi-6, Kita-ku, Sapporo 060-0812, Japan; Hokkaido University, Institute for Vaccine Research and Development (HU-IVReD).
| |
Collapse
|
3
|
Chakraborty C, Lo YH, Bhattacharya M, Das A, Wen ZH. Looking beyond the origin of SARS-CoV-2: Significant strategic aspects during the five-year journey of COVID-19 vaccine development. MOLECULAR THERAPY. NUCLEIC ACIDS 2025; 36:102527. [PMID: 40291378 PMCID: PMC12032352 DOI: 10.1016/j.omtn.2025.102527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
It has been five years since the emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), and we are also approaching the five-year mark of the COVID-19 pandemic. The vaccine is a significant weapon in combating infectious diseases like SARS-CoV-2. Several vaccines were developed against SARS-CoV-2, and they demonstrated efficacy and safety during these five years. The rapid development of multiple next-generation vaccine candidates in different platforms with very little time is the success story of the vaccine development endeavor. This remarkable success of rapid vaccine development is a new paradigm for fast vaccine development that might help develop infectious diseases and fight against the pandemic. With the completion of five years since the beginning of SARS-CoV-2 origin, we are looking back on the five years and reviewing the milestones, vaccine platforms, animal models, clinical trials, successful collaborations, vaccine safety, real-world effectiveness, and challenges. Lessons learned during these five years will help us respond to public health emergencies and to fight the battle against future pandemics.
Collapse
Affiliation(s)
- Chiranjib Chakraborty
- Department of Biotechnology, School of Life Science and Biotechnology, Adamas University, Kolkata, West Bengal 700126, India
| | - Yi-Hao Lo
- Department of Family Medicine, Zuoying Armed Forces General Hospital, Kaohsiung 81342, Taiwan
- Department of Nursing, Meiho University, Neipu Township, Pingtung County 91200, Taiwan
| | - Manojit Bhattacharya
- Department of Zoology, Fakir Mohan University, Vyasa Vihar, Balasore, Odisha 756020, India
| | - Arpita Das
- Department of Biotechnology, School of Life Science and Biotechnology, Adamas University, Kolkata, West Bengal 700126, India
| | - Zhi-Hong Wen
- Department of Marine Biotechnology and Resources, National Sun Yat-Sen University, #70 Lien-Hai Road, Kaohsiung 804201, Taiwan
- National Museum of Marine Biology & Aquarium, # 2 Houwan Road, Checheng, Pingtung 94450, Taiwan
| |
Collapse
|
4
|
Xie Y, Guo J, Hu J, Li Y, Zhang Z, Zhu Y, Deng F, Qi J, Zhou Y, Chen W. A factorial design-optimized microfluidic LNP vaccine elicits potent magnesium-adjuvating cancer immunotherapy. Mater Today Bio 2025; 32:101703. [PMID: 40230646 PMCID: PMC11994397 DOI: 10.1016/j.mtbio.2025.101703] [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: 11/13/2024] [Revised: 03/19/2025] [Accepted: 03/23/2025] [Indexed: 04/16/2025] Open
Abstract
Human papillomavirus (HPV)-associated cancers remain a critical health challenge, prompting the development of effective therapeutic vaccines. This study presents a lipid nanoparticle (LNP)-based vaccine co-loading E7 antigen peptide and magnesium ions as the adjuvant. Microfluidic technology was employed to optimize LNP preparation and formulation, ensuring efficient co-delivery of antigen and adjuvant. Magnesium ions were chosen over conventional aluminum-based adjuvants, which often suffer from limited efficacy and adverse effects, particularly for cancer immunotherapy. Compared to aluminum, magnesium ions exhibited superior capabilities in enhancing T-cell activation and promoting cellular immune response. Mechanistic insights suggest that magnesium ions facilitate dendritic cell maturation and antigen presentation via a collagen-CD36 axis, contributing to the adjuvant activity of magnesium. Through design of experiments (DoE) optimization, the LNP formulation was tailored for enhanced encapsulation and stability, positioning it as a targeted system for immune activation. These findings support the promise of magnesium ions as effective and safer adjuvants in LNP-based vaccines, marking a potential advancement for therapeutic cancer vaccination.
Collapse
Affiliation(s)
- Yongyi Xie
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Jiaxin Guo
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Jialin Hu
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Yuan Li
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Zhongqian Zhang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Yongcheng Zhu
- Department of Emergency, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, PR China
| | - Fei Deng
- Graduate School of Biomedical Engineering, ARC Centre of Excellence in Nanoscale Biophotonics, Faculty of Engineering, UNSW Sydney, NSW, 2052, Australia
| | - Jialong Qi
- Yunnan Digestive Endoscopy Clinical Medical Center, Department of Gastroenterology, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650032, PR China
| | - You Zhou
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Wenjie Chen
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, PR China
| |
Collapse
|
5
|
Amor NP, Guo K, Zhang S, Xia J, Yang Y, Lin A. Lipid Nanoparticle: Beyond Delivery Vehicle-Unveiling Its Immunological Adjuvant Potential. FASEB J 2025; 39:e70641. [PMID: 40372384 DOI: 10.1096/fj.202500622r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 04/17/2025] [Accepted: 05/07/2025] [Indexed: 05/16/2025]
Abstract
Lipid nanoparticles (LNPs) have extensively been used in drug delivery over the years, and the perspective of their significance has been well established. Latest findings have demonstrated the inherent adjuvant capacity of some specific lipid components, especially in stimulating immune compartments, which prompted the rational use of lipid-based delivery vehicles in drug R&D. In this concise review, we summarize current knowledge on the adjuvant properties of LNP, with a particular focus on the key components that mediate such effects. Specifically, we describe the vital role of ionizable lipids in triggering innate immune activation and inflammation and highlight the importance of these lipids in determining vaccine effectiveness or safety. Furthermore, the mechanisms by which LNP enhance the immune response are discussed in detail, shedding light on their potential applications in next-generation vaccine design and development. We also present compelling pre-clinical studies that serve as strong evidence for the adjuvant properties of LNP components in enhancing vaccine immunogenicity.
Collapse
Affiliation(s)
- Narh Philip Amor
- Innovation Center for Nucleic Acid Medicine, Institute for Innovative Drug Development and Life Sciences, Wuxi, China Pharmaceutical University, Nanjing, China
- Institute of Translational Medicine, China Pharmaceutical University, Nanjing, China
- Center for Infectious Medicine and Vaccine Research, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Kun Guo
- Innovation Center for Nucleic Acid Medicine, Institute for Innovative Drug Development and Life Sciences, Wuxi, China Pharmaceutical University, Nanjing, China
- Institute of Translational Medicine, China Pharmaceutical University, Nanjing, China
- Center for Infectious Medicine and Vaccine Research, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Shun Zhang
- Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, China
| | - Jun Xia
- Institute of Veterinary Medicine, Xinjiang Academy of Animal Sciences, Urumqi, China
| | - Yong Yang
- Institute of Translational Medicine, China Pharmaceutical University, Nanjing, China
- Center for Infectious Medicine and Vaccine Research, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Ang Lin
- Innovation Center for Nucleic Acid Medicine, Institute for Innovative Drug Development and Life Sciences, Wuxi, China Pharmaceutical University, Nanjing, China
- Institute of Translational Medicine, China Pharmaceutical University, Nanjing, China
- Center for Infectious Medicine and Vaccine Research, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, Jinan, China
| |
Collapse
|
6
|
Le CTT, Kim KH, Raha JR, Bhatnagar N, Pal SS, Grovenstein P, Yeasmin M, Liu R, Wang BZ, Kang SM. Dual roles of influenza B virus neuraminidase mRNA vaccine in enhancing cross-lineage protection by supplementing inactivated split vaccination. J Virol 2025; 99:e0229424. [PMID: 40265888 PMCID: PMC12090766 DOI: 10.1128/jvi.02294-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Accepted: 03/27/2025] [Indexed: 04/24/2025] Open
Abstract
The current influenza vaccine is based on immunity to hemagglutinin (HA) and provides poor cross-protection. Here, we generated mRNA vaccine encoding influenza B virus (IBV) neuraminidase (NA) conjugated to influenza A virus M2 ectodomain (M2e), encapsulated in lipid nanoparticles (LNP), capable of inducing cross-lineage IBV protection in a dose-dependent pattern. The combination of low-dose NA mRNA and inactivated split IBV vaccines was found to induce significantly higher levels of cross-reactive IgG responses, NA and HA inhibition titers, effector and memory cellular immune responses as well as cross-lineage protection than either NA mRNA or split vaccine alone. This study suggests that the NA mRNA vaccine not only provides cross-lineage protection with a high dose but also enhances the cross-protective efficacy of the combined low-dose NA mRNA and split vaccines. Our findings support a new strategy of using mRNA LNP-supplemented conventional vaccination to enhance cross-protection.IMPORTANCEThis study highlights a significant advancement in influenza vaccination strategies. To test a new vaccination strategy, we developed an influenza B virus (IBV) neuraminidase (NA) mRNA vaccine which could provide cross-lineage protection at a high dose. More importantly, the co-administration of NA mRNA and split IBV vaccine at low doses was found to significantly enhance the hemagglutinin and NA immunity as well as cross-lineage protection of seasonal IBV vaccines. This proof-of-concept study provides evidence for a novel strategy to enhance the immunogenicity and cross-protective efficacy of conventional vaccines by supplementing with new targets of mRNA vaccines.
Collapse
MESH Headings
- Neuraminidase/immunology
- Neuraminidase/genetics
- Influenza Vaccines/immunology
- Influenza Vaccines/administration & dosage
- Influenza Vaccines/genetics
- Influenza B virus/immunology
- Influenza B virus/genetics
- Influenza B virus/enzymology
- Cross Protection/immunology
- Animals
- Antibodies, Viral/immunology
- Antibodies, Viral/blood
- Vaccines, Inactivated/immunology
- Vaccines, Inactivated/administration & dosage
- Mice
- Mice, Inbred BALB C
- Female
- Orthomyxoviridae Infections/prevention & control
- Orthomyxoviridae Infections/immunology
- Orthomyxoviridae Infections/virology
- Viral Proteins/immunology
- Viral Proteins/genetics
- Vaccination
- RNA, Messenger/immunology
- RNA, Messenger/genetics
- Vaccines, Synthetic/immunology
- Vaccines, Synthetic/administration & dosage
- Humans
- Viral Matrix Proteins/immunology
- Viral Matrix Proteins/genetics
- Influenza, Human/prevention & control
- Influenza, Human/immunology
- mRNA Vaccines/immunology
- Cross Reactions
Collapse
Affiliation(s)
- Chau Thuy Tien Le
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia, USA
| | - Ki-Hye Kim
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia, USA
| | - Jannatul Ruhan Raha
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia, USA
| | - Noopur Bhatnagar
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia, USA
| | - Surya Sekhar Pal
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia, USA
| | - Phillip Grovenstein
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia, USA
| | - Mahmuda Yeasmin
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia, USA
| | - Rong Liu
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia, USA
| | - Bao-Zhong Wang
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia, USA
| | - Sang-Moo Kang
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia, USA
| |
Collapse
|
7
|
Kim J, Yang J, Heo S, Poo H. Evaluation of mRNA Transfection Reagents for mRNA Delivery and Vaccine Efficacy via Intramuscular Injection in Mice. ACS APPLIED BIO MATERIALS 2025; 8:4315-4324. [PMID: 40263125 DOI: 10.1021/acsabm.5c00424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/24/2025]
Abstract
The selection of an effective delivery carrier is crucial to assessing mRNA-based vaccines and therapeutics in vivo. Although lipid nanoparticles (LNPs) are commonly used for mRNA delivery, the LNP-mRNA formulation process is laborious and time-consuming and requires a high-cost microfluidic device. Instead, mixing with commercial reagents may simplify mRNA transfection into cells. However, their potential as in vivo carriers in intramuscular vaccination in mouse models remains unclear. In this study, we used three types of commercial RNA transfection reagents, MessengerMAX (MAX; liposome), TransIT-mRNA (IT; cationic polymer), and Invivofectamine (IVF; LNP), to produce nanoparticles directly by pipetting. The particle characteristics and mRNA delivery efficacy of the mRNA-transfection reagent mixtures were analyzed. Additionally, immune responses to vaccine efficacy and protective immunity of the mRNA mixtures as vaccine antigens were evaluated in a mouse model. Although MAX and IT showed high in vitro transfection efficiencies, their in vivo performances were limited. In contrast, IVF exhibited notable particle stability and homogeneity, making it a promising delivery carrier. Intramuscular IVF injection significantly enhanced both innate and adaptive immune responses with a robust systemic protein expression. Notably, when using SARS-CoV-2 Spike mRNA, IVF showed robust humoral immune responses, including production of IgG and neutralizing antibodies, thereby resulting in complete protection against SARS-CoV-2 infection. Therefore, these findings position IVF as an accessible and efficient mRNA carrier for evaluating mRNA vaccines and therapeutic efficacy in basic research.
Collapse
Affiliation(s)
- Jungho Kim
- Department of Biomedical Science and Engineering, Konkuk University, Seoul 05029, Republic of Korea
| | - Jihyun Yang
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea
| | - Suhyeon Heo
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea
| | - Haryoung Poo
- Department of Biomedical Science and Engineering, Konkuk University, Seoul 05029, Republic of Korea
| |
Collapse
|
8
|
Gao Z, Yang H, He Z, Zhou Y, Ge X, Liu H, Yan Z, Wang H, Wei L, Qiao D, Liu Z, Zhu T, Liu L, Chen Y. Cost-effective yet high-performance ionizable lipids for mRNA-lipid nanoparticle vaccines. Biomaterials 2025; 323:123421. [PMID: 40411984 DOI: 10.1016/j.biomaterials.2025.123421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 05/03/2025] [Accepted: 05/19/2025] [Indexed: 05/27/2025]
Abstract
Ionizable lipids (ILs) are critical components in mRNA vaccines, which have been instrumental in the global response to SARS-CoV-2. However, current commercialized ILs in mRNA vaccines are typically synthesized through multiple-step organic reactions, complicating quality control and driving up production costs. To address this, we have developed novel ILs by a one-pot Ugi four-component reaction (Ugi-4CR), significantly simplifying synthesis while maintaining high yields and reducing costs. Here, from a library of 161 ILs, we chose six ILs with high expressing luciferase and investigated their performance in delivering the mRNA vaccine of SARS-CoV-2. These ILs feature distinct ionizable heads, N,N-dimethylethyl (R1), N,N-dimethylpropyl (R2), and N,N-diethylpropyl (R3), paired with hydrophobic tails of varying unsaturation, cis-9-octadecenoic (U1) and (9Z,12Z)-9,12-octadecadienoic (U2), respectively. In murine models, R2-and R3-based mSpike-LNPs induce higher antibody titers and stronger cellular immune responses compared to the R1-based counterparts, suggesting their superior mRNA delivery and expression efficiency. Notably, R2U2- and R3U2-based mSpike-LNPs further enhance IFN-γ+ splenocyte responses and activation of TNF-α+CD4+/CD8+ T cells, coupled with improved dendritic cell activation and retention in lymph nodes. We confirm that the R2U2-based LNPs on different mRNA antigens exhibit immune responses and safety profiles comparable to the commercial ALC-0315-based LNPs. Moreover, intranasal and intratracheal administration of R2U2-based mSpike-LNPs enhances mucosal immunity, as evidenced by elevated sIgA levels in mice. Further evaluation in cynomolgus macaques proves the efficacy of this LNP system, highlighting its potential for developing cost-effective mRNA vaccines.
Collapse
Affiliation(s)
- Zhan Gao
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Guangdong Engineering Technology Research Centre for Functional Biomaterials, Sun Yat-sen University, Guangzhou, 510275, China
| | - Haihong Yang
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Guangdong Engineering Technology Research Centre for Functional Biomaterials, Sun Yat-sen University, Guangzhou, 510275, China
| | - Zepeng He
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Guangdong Engineering Technology Research Centre for Functional Biomaterials, Sun Yat-sen University, Guangzhou, 510275, China
| | - Yizi Zhou
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Guangdong Engineering Technology Research Centre for Functional Biomaterials, Sun Yat-sen University, Guangzhou, 510275, China
| | - Xiaofei Ge
- Department of Ophthalmology, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, China
| | - Hong Liu
- Translational Medical Center of Huaihe Hospital, Henan University, Kaifeng, 475004, China
| | - Zhihong Yan
- CanSino Biologics Inc., Biomedical Park, 185 South Avenue, TEDA West District, Tianjin, China
| | - Haomeng Wang
- CanSino Biologics Inc., Biomedical Park, 185 South Avenue, TEDA West District, Tianjin, China
| | - Lai Wei
- Department of Ophthalmology, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, China
| | - Dongdong Qiao
- Department of Basic Medicine, Chaoshan Branch of State Key Laboratory for Esophageal Cancer Prevention and Treatment, Shantou University Medical College, Shantou, 515063, China.
| | - Zhijia Liu
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Guangdong Engineering Technology Research Centre for Functional Biomaterials, Sun Yat-sen University, Guangzhou, 510275, China.
| | - Tao Zhu
- CanSino Biologics Inc., Biomedical Park, 185 South Avenue, TEDA West District, Tianjin, China.
| | - Lixin Liu
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Guangdong Engineering Technology Research Centre for Functional Biomaterials, Sun Yat-sen University, Guangzhou, 510275, China.
| | - Yongming Chen
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Guangdong Engineering Technology Research Centre for Functional Biomaterials, Sun Yat-sen University, Guangzhou, 510275, China; College of Chemistry and Molecular Sciences, Henan University, Kaifeng, 475004, China; State Key Laboratory of Antiviral Drugs, Henan University, Zhengzhou, 450046, China.
| |
Collapse
|
9
|
Kawaguchi K, Nguyen LBT, Kinoshita M, Abe N, Oba M, Abe H, Sudo K, Inoue K, Uchida S, Sawa T. Highly pure mRNA vaccine provides robust immunization against P. aeruginosa by minimizing type I interferon responses. J Control Release 2025; 383:113860. [PMID: 40383159 DOI: 10.1016/j.jconrel.2025.113860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Revised: 05/03/2025] [Accepted: 05/16/2025] [Indexed: 05/20/2025]
Abstract
Developing effective vaccines against bacteria is critical given the growing threat of antimicrobial resistance (AMR). In this study, we developed mRNA vaccines targeting Pseudomonas aeruginosa (P. aeruginosa), a key AMR pathogen, using PureCap mRNA encapsulated in lipid nanoparticles (LNPs). The PureCap technology offers a facile method for removing immunostimulatory impurities from in vitro transcribed mRNA, such as uncapped RNA and double-stranded RNA (dsRNA). Following intramuscular vaccination of mice with mRNA encoding a model antigen, PureCap mRNA elicited antibody titers that were 26-fold higher than those induced by conventional ARCA-capped mRNA. Mechanistic analyses revealed that both uncapped RNA and dsRNA impurities in ARCA-capped mRNA were responsible for the reduced humoral immune responses. While PureCap mRNA enhanced protein expression efficiency and reduced pro-inflammatory responses compared to ARCA-capped mRNA, minimizing pro-inflammatory responses was particularly critical. When anti-interferon-α/β receptor antibodies were administered, antibody responses to ARCA-capped mRNA vaccination were restored to levels comparable to those achieved with PureCap mRNA vaccination, highlighting the negative impact of type I interferon responses on antibody responses following vaccination with ARCA-capped mRNA. In a vaccination targeting the PcrV protein of P. aeruginosa, PureCap mRNA, but not ARCA-capped mRNA, significantly prolonged the survival of mice following bacterial challenges, presumably due to enhanced antibody production. Furthermore, PureCap mRNA vaccination significantly reduced bacterial loads in the lungs and mitigated tissue damage, edema, and inflammatory responses. These findings underscore the potential of PureCap mRNA as a promising platform for bacterial vaccination, offering a valuable strategy to combat AMR.
Collapse
Affiliation(s)
- Ken Kawaguchi
- Department of Anesthesiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajiicho, Kawaramachi Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Le Bui Thao Nguyen
- Department of Advanced Nanomedical Engineering, Medical Research Laboratory, Institute of Integrated Research, Institute of Science Tokyo, 1-5-45 Yushima, Bunkyo-ku Tokyo 113-8510, Japan; Innovation Center of NanoMedicine (iCONM), Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
| | - Mao Kinoshita
- Department of Anesthesiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajiicho, Kawaramachi Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Naoko Abe
- Department of Chemistry, Graduate School of Science, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, Aichi 464-8602, Japan
| | - Makoto Oba
- Medical Chemistry, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 1-5 Shimogamohangi-cho, Sakyo-ku, Kyoto 606-0823, Japan
| | - Hiroshi Abe
- Department of Chemistry, Graduate School of Science, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, Aichi 464-8602, Japan; Institute for Glyco-core Research (iGCORE), Nagoya University, Furo-cho, Chikusa-ku, Nagoya, Aichi 464-8601, Japan
| | - Kazuki Sudo
- Department of Anesthesiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajiicho, Kawaramachi Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Keita Inoue
- Department of Anesthesiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajiicho, Kawaramachi Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Satoshi Uchida
- Department of Advanced Nanomedical Engineering, Medical Research Laboratory, Institute of Integrated Research, Institute of Science Tokyo, 1-5-45 Yushima, Bunkyo-ku Tokyo 113-8510, Japan; Innovation Center of NanoMedicine (iCONM), Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan; Medical Chemistry, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 1-5 Shimogamohangi-cho, Sakyo-ku, Kyoto 606-0823, Japan.
| | - Teiji Sawa
- Department of Anesthesiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajiicho, Kawaramachi Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan.
| |
Collapse
|
10
|
Berber E, Pantouli F, Hanley HB, Ross TM. COVID-19 Vaccination Enhances the Immunogenicity of Seasonal Influenza Vaccination in the Elderly. Vaccines (Basel) 2025; 13:531. [PMID: 40432140 PMCID: PMC12116172 DOI: 10.3390/vaccines13050531] [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: 04/20/2025] [Revised: 05/09/2025] [Accepted: 05/14/2025] [Indexed: 05/29/2025] Open
Abstract
BACKGROUND/OBJECTIVES The co-circulation of both influenza viruses and SARS-CoV-2 poses a significant health risk, especially for the elderly. While vaccination against both diseases remains an effective strategy to reduce the burden of symptomatic infections, the effect of administering COVID-19 mRNA and seasonal influenza vaccines (COV-Flu) on elicited antibody responses has not been explored. METHODS Participants between 18 and 90 years old were vaccinated with COVID-19 mRNA vaccines (n = 67), seasonal influenza vaccines (n = 130), or both (n = 201) within a three-month period between 2021 and 2024. Serum hemagglutination-inhibition (HAI) titers against influenza A (H1N1, H3N2) and B (Yamagata, Victoria) strains were measured from the COV-Flu participants or the participants vaccinated with influenza vaccines only (mono-Flu). SARS-CoV-2 neutralization assays were performed on sera collected from the COV-Flu participants and the participants receiving the mRNA vaccine only (mono-COVID-19). RESULTS The administration of influenza virus vaccines and COVID-19 mRNA vaccines within a three-month period significantly enhanced the post-vaccination HAI titers against both influenza A and B vaccine components, particularly in the elderly (65-90) participants. There were no significant differences in SARS-CoV-2 neutralization titers in COV-Flu participants compared to mono-COVID-19 participants. CONCLUSIONS Vaccination with both the COVID-19 mRNA and influenza vaccines enhances influenza-specific HAI titers without compromising the neutralization titers elicited by COVID-19 mRNA vaccination against SARS-CoV-2, especially in the elderly. These findings indicate the potential benefits of this approach, particularly for older adults, by boosting influenza virus vaccine-induced serum HAI activity while maintaining COVID-19 protective immunity.
Collapse
Affiliation(s)
- Engin Berber
- Department of Infection Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA;
| | - Fani Pantouli
- Florida Research and Innovation Center, Cleveland Clinic, Port Saint Lucie, FL 34987, USA;
| | - Hannah B. Hanley
- Center for Vaccines and Immunology, University of Georgia, Athens, GA 30602, USA;
| | - Ted M. Ross
- Department of Infection Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA;
- Florida Research and Innovation Center, Cleveland Clinic, Port Saint Lucie, FL 34987, USA;
- Center for Vaccines and Immunology, University of Georgia, Athens, GA 30602, USA;
| |
Collapse
|
11
|
Konopka EN, Edgerton AO, Kutzler MA. Nucleic acid vaccines: innovations, efficacy, and applications in at-risk populations. Front Immunol 2025; 16:1584876. [PMID: 40438110 PMCID: PMC12116436 DOI: 10.3389/fimmu.2025.1584876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Accepted: 04/09/2025] [Indexed: 06/01/2025] Open
Abstract
For more than two centuries, the field of vaccine development has progressed through the adaptation of novel platforms in parallel with technological developments. Building off the advantages and shortcomings of first and second-generation vaccine platforms, the advent of third-generation nucleic acid vaccines has enabled new approaches to tackle emerging infectious diseases, cancers, and pathogens where vaccines remain unavailable. Unlike traditional vaccine platforms, nucleic acid vaccines offer several new advantages, including their lower cost and rapid production, which was widely demonstrated during the COVID-19 pandemic. Beyond production, DNA and mRNA vaccines can elicit unique and targeted responses through specialized design and delivery approaches. Considering the growth of nucleic acid vaccine research over the past two decades, the evaluation of their efficacy in at-risk populations is paramount for refining and improving vaccine design. Importantly, the aging population represents a significant portion of individuals highly susceptible to infection and disease. This review seeks to outline the major impairments in vaccine-induced responses due to aging that may be targeted for improvement with design and delivery components encompassing mRNA and DNA vaccine formulations. Results of pre-clinical and clinical applications of these vaccines in aged animal models and humans will also be evaluated to outline current successes and limitations observed in these platforms.
Collapse
Affiliation(s)
- Emily N. Konopka
- Drexel University College of Medicine, Department of Microbiology and Immunology, Philadelphia, PA, United States
- Drexel University College of Medicine, Department of Medicine, Division of Infectious Diseases and HIV Medicine, Philadelphia, PA, United States
| | - Arden O. Edgerton
- Drexel University College of Medicine, Department of Microbiology and Immunology, Philadelphia, PA, United States
- Drexel University College of Medicine, Department of Medicine, Division of Infectious Diseases and HIV Medicine, Philadelphia, PA, United States
| | - Michele A. Kutzler
- Drexel University College of Medicine, Department of Microbiology and Immunology, Philadelphia, PA, United States
- Drexel University College of Medicine, Department of Medicine, Division of Infectious Diseases and HIV Medicine, Philadelphia, PA, United States
| |
Collapse
|
12
|
Kwon DI, Mao T, Israelow B, Santos Guedes de Sá K, Dong H, Iwasaki A. Mucosal unadjuvanted booster vaccines elicit local IgA responses by conversion of pre-existing immunity in mice. Nat Immunol 2025:10.1038/s41590-025-02156-0. [PMID: 40360777 DOI: 10.1038/s41590-025-02156-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Accepted: 04/08/2025] [Indexed: 05/15/2025]
Abstract
Mucosal delivery of vaccine boosters induces robust local protective immune responses even without any adjuvants. Yet, the mechanisms by which antigen alone induces mucosal immunity in the respiratory tract remain unclear. Here we show that an intranasal booster with an unadjuvanted recombinant SARS-CoV-2 spike protein, after intramuscular immunization with 1 μg of mRNA-LNP vaccine encoding the full-length SARS-CoV-2 spike protein (Pfizer/BioNTech BNT162b2), elicits protective mucosal immunity by retooling the lymph node-resident immune cells. On intranasal boosting, peripheral lymph node-primed B cells rapidly migrated to the lung through CXCR3-CXCL9 and CXCR3-CXCL10 signaling and differentiated into antigen-specific IgA-secreting plasma cells. Memory CD4+ T cells in the lung served as a natural adjuvant for developing mucosal IgA by inducing the expression of chemokines CXCL9 and CXCL10 for memory B cell recruitment. Furthermore, CD40 and TGFβ signaling had important roles in mucosal IgA development. Repeated mucosal boosting with an unadjuvanted protein amplified anamnestic IgA responses in both the upper and the lower respiratory tracts. These findings help explain why nasal boosters do not require an adjuvant to induce robust mucosal immunity at the respiratory mucosa and can be used to design safe and effective vaccines against respiratory pathogens.
Collapse
Affiliation(s)
- Dong-Il Kwon
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
- Center for Infection and Immunity, Yale School of Medicine, New Haven, CT, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Tianyang Mao
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Benjamin Israelow
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
- Center for Infection and Immunity, Yale School of Medicine, New Haven, CT, USA
- Section of Infectious Diseases, Department of Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Keyla Santos Guedes de Sá
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
- Center for Infection and Immunity, Yale School of Medicine, New Haven, CT, USA
| | - Huiping Dong
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
- Center for Infection and Immunity, Yale School of Medicine, New Haven, CT, USA
| | - Akiko Iwasaki
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA.
- Center for Infection and Immunity, Yale School of Medicine, New Haven, CT, USA.
- Howard Hughes Medical Institute, Chevy Chase, MD, USA.
| |
Collapse
|
13
|
Ling J, Chen H, Huang M, Wang J, Du X. An mRNA vaccine encoding proteasome-targeted antigen enhances CD8 + T cell immunity. J Control Release 2025; 381:113578. [PMID: 40015339 DOI: 10.1016/j.jconrel.2025.02.074] [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: 08/26/2024] [Revised: 02/22/2025] [Accepted: 02/24/2025] [Indexed: 03/01/2025]
Abstract
The efficient induction of antigen-specific CD8+ T cell activation is crucial in the development of mRNA tumor vaccines. Endogenous antigens are primarily degraded through the ubiquitin-proteasome system, followed by antigen presentation via major histocompatibility complex class I (MHC-I) molecules, leading to the activation of CD8+ T cells. Therefore, in this study, a novel mRNA vaccine was developed by fusing the mRNA sequence encoding the antigen with a proteasome-targeting peptide (PTP), aiming to enhance proteasomal targeting of the antigen and facilitate its degradation through the ubiquitin-proteasome system, thereby inducing a stronger CD8+ T cell immune response. This study confirmed a significant increase in antigen expression of the antigen-PTP fused mRNA vaccine upon treatment with a VHL inhibitor, as well as notable upregulation of genes associated with the MHC-I antigen-presenting pathway following treatment with the antigen-PTP fused mRNA vaccine. The intramuscular administration of the antigen-PTP fused mRNA vaccine significantly promoted the activation of dendritic cells, macrophages, and T cells in draining lymph nodes and spleens. Additionally, in TC-1 tumor-bearing mice, it markedly suppressed tumor growth, facilitated infiltration of intratumoral antigen-specific CD8+ T cells, and induced immune memory.
Collapse
Affiliation(s)
- Jin Ling
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, PR China
| | - Hongwei Chen
- School of Medicine, South China University of Technology, Guangzhou 510006, PR China
| | - Mengwen Huang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, PR China
| | - Jun Wang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, PR China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, PR China; Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou 510006, PR China.
| | - Xiaojiao Du
- School of Medicine, South China University of Technology, Guangzhou 510006, PR China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, PR China.
| |
Collapse
|
14
|
Broomfield BJ, Tan CW, Qin RZ, Abberger H, Duckworth BC, Alvarado C, Dalit L, Lee CL, Shandre Mugan R, Mazrad ZA, Muramatsu H, Mackiewicz L, Williams BE, Chen J, Takanashi A, Fabb S, Pellegrini M, Rogers KL, Moon WJ, Pouton CW, Davis MJ, Nutt SL, Pardi N, Wimmer VC, Groom JR. Transient inhibition of type I interferon enhances CD8+ T cell stemness and vaccine protection. J Exp Med 2025; 222:e20241148. [PMID: 40062995 PMCID: PMC11893171 DOI: 10.1084/jem.20241148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 11/25/2024] [Accepted: 02/04/2025] [Indexed: 03/14/2025] Open
Abstract
Developing vaccines that promote CD8+ T cell memory is a challenge for infectious disease and cancer immunotherapy. TCF-1+ stem cell-like memory CD8+ T (TSCM) cells are important determinants of long-lived memory. Yet, the developmental requirements for TSCM cell formation are unclear. Here, we identify the temporal window for type I interferon receptor (IFNAR) blockade to drive TSCM cell generation following viral infection and mRNA-lipid nanoparticle vaccination. We reveal a reversible developmental trajectory where transcriptionally distinct TSCM cells emerged from a transitional precursor of exhausted T cellular state concomitant with viral clearance. TSCM cell differentiation correlated with T cell retention within the lymph node paracortex due to disrupted CXCR3 chemokine gradient formation. These effects were linked to increased antigen load and a counterintuitive increase in IFNγ, which controlled cell location. Vaccination with the IFNAR blockade promoted TSCM cell differentiation and enhanced protection against chronic infection. These findings propose an approach to vaccine design whereby modulation of inflammation promotes memory formation and function.
Collapse
Affiliation(s)
- Benjamin J. Broomfield
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Australia
| | - Chin Wee Tan
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Australia
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, Australia
| | - Raymond Z. Qin
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Australia
| | - Hanna Abberger
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Australia
| | - Brigette C. Duckworth
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Australia
| | - Carolina Alvarado
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
| | - Lennard Dalit
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Australia
| | - Chee Leng Lee
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Rekha Shandre Mugan
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Zihnil A.I. Mazrad
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Hiromi Muramatsu
- Department of Microbiology, Perelman School of Medicine, Philadelphia, PA, USA
| | - Liana Mackiewicz
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
| | - Bailey E. Williams
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
| | - Jinjin Chen
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Australia
| | - Asuka Takanashi
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Stewart Fabb
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Marc Pellegrini
- Centenary Institute of Cancer Medicine and Cell Biology, Camperdown, Australia
| | - Kelly L. Rogers
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Australia
| | | | - Colin W. Pouton
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Melissa J. Davis
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, Australia
- School of Biomedicine, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, Australia
| | - Stephen L. Nutt
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Australia
| | - Norbert Pardi
- Department of Microbiology, Perelman School of Medicine, Philadelphia, PA, USA
| | - Verena C. Wimmer
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Australia
| | - Joanna R. Groom
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Australia
| |
Collapse
|
15
|
Qu Y, Li Z, Yin J, Huang H, Ma J, Jiang Z, Zhou Q, Tang Y, Li Y, Huang M, Zeng Z, Guo A, Fang F, Shen Y, Zhao R, Wang Y, Gao D. cGAS mRNA-Based Immune Agonist Promotes Vaccine Responses and Antitumor Immunity. Cancer Immunol Res 2025; 13:680-695. [PMID: 40067177 DOI: 10.1158/2326-6066.cir-24-0804] [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: 08/19/2024] [Revised: 11/13/2024] [Accepted: 03/05/2025] [Indexed: 05/03/2025]
Abstract
mRNA vaccines are a potent tool for immunization against viral diseases and cancer. However, the lack of a vaccine adjuvant limits the efficacy of these treatments. In this study, we used cGAS mRNA, which encodes the DNA innate immune sensor, complexed with lipid nanoparticles (LNP), to boost the immune response. By introducing specific mutations in human cGAS mRNA (hcGASK187N/L195R), we significantly enhanced cGAS activity, resulting in a more potent and sustained stimulator of interferon gene (STING)-mediated IFN response. cGAS mRNA-LNPs exhibited stimulatory effects on maturation, antigen engulfment, and antigen presentation by antigen-presenting cells, both in vitro and in vivo. Moreover, the hcGASK187N/L195R mRNA-LNP combination demonstrated a robust adjuvant effect and amplified the potency of mRNA and protein vaccines, which was a result of strong humoral and cell-mediated responses. Remarkably, the hcGASK187N/L195R mRNA-LNP complex, either alone or in combination with antigens, demonstrated exceptional efficacy in eliciting antitumor immunity. In addition to its immune-boosting properties, hcGASK187N/L195R mRNA-LNP exerted antitumor effects with IFNγ directly on tumor cells, further promoting tumor restriction. In conclusion, we developed a cGAS mRNA-based immunostimulatory adjuvant compatible with various vaccine forms to boost the adaptive immune response and cancer immunotherapies.
Collapse
Affiliation(s)
- Yali Qu
- National Key Laboratory of Immune Response and Immunotherapy, Department of General Surgery, The First Affiliated Hospital of University of Science and Technology of China, Center for Advanced Interdisciplinary Science & Biomedicine IHM, Division of Life Sciences & Medicine, University of Science and Technology of China, Hefei, China
- Key Laboratory of Anhui Province for Emerging and Reemerging Infectious Diseases, Hefei, China
| | - Zhibin Li
- Key Laboratory of Anhui Province for Emerging and Reemerging Infectious Diseases, Hefei, China
- Department of Radiology, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Jiahao Yin
- National Key Laboratory of Immune Response and Immunotherapy, Department of General Surgery, The First Affiliated Hospital of University of Science and Technology of China, Center for Advanced Interdisciplinary Science & Biomedicine IHM, Division of Life Sciences & Medicine, University of Science and Technology of China, Hefei, China
- Key Laboratory of Anhui Province for Emerging and Reemerging Infectious Diseases, Hefei, China
| | - He Huang
- National Key Laboratory of Immune Response and Immunotherapy, Department of General Surgery, The First Affiliated Hospital of University of Science and Technology of China, Center for Advanced Interdisciplinary Science & Biomedicine IHM, Division of Life Sciences & Medicine, University of Science and Technology of China, Hefei, China
- Key Laboratory of Anhui Province for Emerging and Reemerging Infectious Diseases, Hefei, China
| | - Jialu Ma
- National Key Laboratory of Immune Response and Immunotherapy, Department of General Surgery, The First Affiliated Hospital of University of Science and Technology of China, Center for Advanced Interdisciplinary Science & Biomedicine IHM, Division of Life Sciences & Medicine, University of Science and Technology of China, Hefei, China
- Key Laboratory of Anhui Province for Emerging and Reemerging Infectious Diseases, Hefei, China
| | - Zhelin Jiang
- National Key Laboratory of Immune Response and Immunotherapy, Department of General Surgery, The First Affiliated Hospital of University of Science and Technology of China, Center for Advanced Interdisciplinary Science & Biomedicine IHM, Division of Life Sciences & Medicine, University of Science and Technology of China, Hefei, China
- Key Laboratory of Anhui Province for Emerging and Reemerging Infectious Diseases, Hefei, China
| | - Qian Zhou
- National Key Laboratory of Immune Response and Immunotherapy, Department of General Surgery, The First Affiliated Hospital of University of Science and Technology of China, Center for Advanced Interdisciplinary Science & Biomedicine IHM, Division of Life Sciences & Medicine, University of Science and Technology of China, Hefei, China
- Key Laboratory of Anhui Province for Emerging and Reemerging Infectious Diseases, Hefei, China
| | - Ying Tang
- Key Laboratory of Anhui Province for Emerging and Reemerging Infectious Diseases, Hefei, China
| | - Yuting Li
- National Key Laboratory of Immune Response and Immunotherapy, Department of General Surgery, The First Affiliated Hospital of University of Science and Technology of China, Center for Advanced Interdisciplinary Science & Biomedicine IHM, Division of Life Sciences & Medicine, University of Science and Technology of China, Hefei, China
- Key Laboratory of Anhui Province for Emerging and Reemerging Infectious Diseases, Hefei, China
| | - Minpeng Huang
- The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Zhutian Zeng
- The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Ao Guo
- Key Laboratory of Anhui Province for Emerging and Reemerging Infectious Diseases, Hefei, China
| | - Fang Fang
- The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yanqiong Shen
- Key Laboratory of Anhui Province for Emerging and Reemerging Infectious Diseases, Hefei, China
- Department of Radiology, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Ruibo Zhao
- National Key Laboratory of Immune Response and Immunotherapy, Department of General Surgery, The First Affiliated Hospital of University of Science and Technology of China, Center for Advanced Interdisciplinary Science & Biomedicine IHM, Division of Life Sciences & Medicine, University of Science and Technology of China, Hefei, China
- Key Laboratory of Anhui Province for Emerging and Reemerging Infectious Diseases, Hefei, China
| | - Yucai Wang
- Key Laboratory of Anhui Province for Emerging and Reemerging Infectious Diseases, Hefei, China
- Department of Radiology, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Daxing Gao
- National Key Laboratory of Immune Response and Immunotherapy, Department of General Surgery, The First Affiliated Hospital of University of Science and Technology of China, Center for Advanced Interdisciplinary Science & Biomedicine IHM, Division of Life Sciences & Medicine, University of Science and Technology of China, Hefei, China
- Key Laboratory of Anhui Province for Emerging and Reemerging Infectious Diseases, Hefei, China
| |
Collapse
|
16
|
You Y, Tian Y, Guo R, Shi J, Kwak KJ, Tong Y, Estania AP, Hsu WH, Liu Y, Hu S, Cao J, Yang L, Bai R, Huang P, Lee LJ, Jiang W, Kim BYS, Ma S, Liu X, Shen Z, Lan F, Phuong Nguyen PK, Lee AS. Extracellular vesicle-mediated VEGF-A mRNA delivery rescues ischaemic injury with low immunogenicity. Eur Heart J 2025; 46:1662-1676. [PMID: 39831819 DOI: 10.1093/eurheartj/ehae883] [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: 04/23/2024] [Revised: 09/09/2024] [Accepted: 12/05/2024] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND AND AIMS Lackluster results from recently completed gene therapy clinical trials of VEGF-A delivered by viral vectors have heightened the need to develop alternative delivery strategies. This study aims to demonstrate the pre-clinical efficacy and safety of extracellular vesicles (EVs) loaded with VEGF-A mRNA for the treatment of ischaemic vascular disease. METHODS After encapsulation of full-length VEGF-A mRNA into fibroblast-derived EVs via cellular nanoporation (CNP), collected VEGF-A EVs were delivered into mouse models of ischaemic injury. Target tissue delivery was verified by in situ analysis of protein and gene expression. Functional rescue was confirmed by in vivo imaging and histology. The safety of single and serial delivery was demonstrated using immune-based assays. RESULTS VEGF-A EVs were generated with high mRNA content using a CNP methodology. VEGF-A EV administration demonstrated expression of exogenous VEGF-A mRNA by in situ RNA hybridization and elevated protein expression by western blot, microscopy, and enzyme-linked immunosorbent assay. Mice treated with human VEGF-A EVs after femoral or coronary artery ligation exhibited heightened neovascularization in ischaemic tissues with increased arterial perfusion and improvement in left ventricular function, respectively. Serial delivery of VEGF-EVs in injured skin showed improved wound healing with repeat administration. Importantly, as compared with adeno-associated viral and lipid nanoparticle VEGF-A gene therapy modalities, murine VEGF-A EV delivery did not trigger innate or adaptive immune responses at the injection site or systemically. CONCLUSIONS This study demonstrated that VEGF-A EV therapy offers efficient, dose-dependent VEGF-A protein formation with low immunogenicity, resulting in new vessel formation in murine models of ischaemic vascular disease.
Collapse
Affiliation(s)
- Yi You
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 2199 Lishui Rd, Nanshan, Shenzhen, Guangdong Province 518055, China
- Institute for Cancer Research, Shenzhen Bay Laboratory, Guangqiao Road, Guangming District, Shenzhen 518055, China
| | - Yu Tian
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 2199 Lishui Rd, Nanshan, Shenzhen, Guangdong Province 518055, China
- Institute for Cancer Research, Shenzhen Bay Laboratory, Guangqiao Road, Guangming District, Shenzhen 518055, China
| | - Rui Guo
- Department of Cardiac Surgery, Peking University Third Hospital, 49 Huayuan N Rd, Haidian District, Beijing 100191, China
| | - Junfeng Shi
- Department of Chemical and Biomolecular Engineering, 151 W Woodruff Ave, Columbus, The Ohio State University, Columbus, OH 43210, USA
| | - Kwang Joo Kwak
- Department of Chemical and Biomolecular Engineering, 151 W Woodruff Ave, Columbus, The Ohio State University, Columbus, OH 43210, USA
| | - Yuhao Tong
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 2199 Lishui Rd, Nanshan, Shenzhen, Guangdong Province 518055, China
- Institute for Cancer Research, Shenzhen Bay Laboratory, Guangqiao Road, Guangming District, Shenzhen 518055, China
| | - Andreanne Poppy Estania
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 2199 Lishui Rd, Nanshan, Shenzhen, Guangdong Province 518055, China
- Institute for Cancer Research, Shenzhen Bay Laboratory, Guangqiao Road, Guangming District, Shenzhen 518055, China
| | - Wei-Hsiang Hsu
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 2199 Lishui Rd, Nanshan, Shenzhen, Guangdong Province 518055, China
- Institute for Cancer Research, Shenzhen Bay Laboratory, Guangqiao Road, Guangming District, Shenzhen 518055, China
| | - Yutong Liu
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 2199 Lishui Rd, Nanshan, Shenzhen, Guangdong Province 518055, China
- Institute for Cancer Research, Shenzhen Bay Laboratory, Guangqiao Road, Guangming District, Shenzhen 518055, China
| | - Shijun Hu
- Department of Cardiovascular Surgery for the First Affiliated Hospital & Institute for Cardiovascular Science, Suzhou Medical College, Soochow University, 899 Pinghai Road, Suzhou, Jiangsu 215000, China
| | - Jianhong Cao
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 2199 Lishui Rd, Nanshan, Shenzhen, Guangdong Province 518055, China
- Institute for Cancer Research, Shenzhen Bay Laboratory, Guangqiao Road, Guangming District, Shenzhen 518055, China
| | - Liqun Yang
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 2199 Lishui Rd, Nanshan, Shenzhen, Guangdong Province 518055, China
- Institute for Cancer Research, Shenzhen Bay Laboratory, Guangqiao Road, Guangming District, Shenzhen 518055, China
| | - Rui Bai
- Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen, Shenzhen Key Laboratory of Cardiovascular Disease, No. 12 Langshan Road, Nanshan District, Shenzhen 518057, China
| | - Pufeng Huang
- Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen, Shenzhen Key Laboratory of Cardiovascular Disease, No. 12 Langshan Road, Nanshan District, Shenzhen 518057, China
| | - Ly James Lee
- Spot Biosystems Ltd, 432 High Street, Apartment 201, Palo Alto, CA 94301, USA
| | - Wen Jiang
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, 1220 Holcombe Blvd, Houston, TX 77030, USA
| | - Betty Y S Kim
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, 1515 Holcombe Blvd, Houston, TX 77030, USA
| | - Shuhong Ma
- Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen, Shenzhen Key Laboratory of Cardiovascular Disease, No. 12 Langshan Road, Nanshan District, Shenzhen 518057, China
- State Key Laboratory of Cardiovascular Disease, Key Laboratory of Pluripotent Stem Cells in Cardiac Repair and Regeneration, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167A Beilishi Road, Xi Cheng District, Beijing 100037, China
| | - Xujie Liu
- Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen, Shenzhen Key Laboratory of Cardiovascular Disease, No. 12 Langshan Road, Nanshan District, Shenzhen 518057, China
- State Key Laboratory of Cardiovascular Disease, Key Laboratory of Pluripotent Stem Cells in Cardiac Repair and Regeneration, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167A Beilishi Road, Xi Cheng District, Beijing 100037, China
| | - Zhenya Shen
- Department of Cardiovascular Surgery for the First Affiliated Hospital & Institute for Cardiovascular Science, Suzhou Medical College, Soochow University, 899 Pinghai Road, Suzhou, Jiangsu 215000, China
| | - Feng Lan
- Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen, Shenzhen Key Laboratory of Cardiovascular Disease, No. 12 Langshan Road, Nanshan District, Shenzhen 518057, China
- State Key Laboratory of Cardiovascular Disease, Key Laboratory of Pluripotent Stem Cells in Cardiac Repair and Regeneration, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167A Beilishi Road, Xi Cheng District, Beijing 100037, China
- Beijing Laboratory for Cardiovascular Precision Medicine, The Key Laboratory of Biomedical Engineering for Cardiovascular Disease Research, Ministry of Education, Beijing Anzhen Hospital, Capital Medical University, Research Institute Building, Room 323, 2 Anzhen Road, Chaoyang District, Beijing 100029, China
| | - Patricia Kim Phuong Nguyen
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, 870 Quarry Road, Rm 183, Stanford, CA 94305, USA
- Stanford Cardiovascular Institute, 265 Campus Drive, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Andrew S Lee
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 2199 Lishui Rd, Nanshan, Shenzhen, Guangdong Province 518055, China
- Institute for Cancer Research, Shenzhen Bay Laboratory, Guangqiao Road, Guangming District, Shenzhen 518055, China
- Greater Bay Area International Clinical Trials Center, Shenzhen Medical Academy of Research and Translation, Shenzhen 518055, China
| |
Collapse
|
17
|
Brook B, Goetz M, Duval V, Micol R, Dowling DJ. mRNA vaccines: miRNA-based controlled biodistribution and directed adjuvantation. Trends Immunol 2025; 46:357-360. [PMID: 40268657 DOI: 10.1016/j.it.2025.03.004] [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: 10/17/2024] [Revised: 03/10/2025] [Accepted: 03/19/2025] [Indexed: 04/25/2025]
Abstract
The development of ionizable mRNA-lipid nanoparticle (mRNA-LNP) nucleic acid carriers facilitated the clinical translation of the Coronavirus 2019 (COVID-19) mRNA vaccines BNT162b2 and mRNA-1273. Here, we discuss insights into rational improvements to mRNA vaccines, focusing on LNP modifications for mRNA-LNP biodistribution control, miRNA-based biodistribution control of encoded transcripts, and precision adjuvantation strategies.
Collapse
Affiliation(s)
- Byron Brook
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA 02115, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Morgan Goetz
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA 02115, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Valerie Duval
- Combined Therapeutics Incorporated, Boston, MA 02135, USA
| | - Romain Micol
- Combined Therapeutics Incorporated, Boston, MA 02135, USA
| | - David J Dowling
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA 02115, USA; Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
18
|
Buckley LA, Sutherland JE, Borude P, Broudic K, Collin P, Hillegas A, MacLauchlin C, Saleh AF, Sharma A, Thomas J, O'Brien Laramy M. An Industry Perspective on the Use of Novel Excipients in Lipid Nanoparticles-Nonclinical Considerations. Int J Toxicol 2025; 44:196-210. [PMID: 40040255 DOI: 10.1177/10915818251320631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2025]
Abstract
Nucleic acid drug delivery with lipid nanoparticle (LNP) formulations has enabled the development of novel therapeutics and vaccines. LNP formulations are composed of both naturally occurring and synthetic lipid excipients. This perspective shares current practices in the nonclinical safety assessment of novel lipid excipients contained in LNP formulations and identifies gaps in current regulatory guidance on this topic. There is no globally harmonized regulatory guidance for the nonclinical safety assessment of novel excipients or guidance specific to safety testing of novel excipients in LNPs. Given the complexity of these LNP formulations, most nonclinical safety studies to support development are conducted with the drug product or with a LNP that contains non-active cargo. Three case studies (Onpattro®, Comirnaty®, and SpikeVax®) highlight that specific assessments may differ depending on the encapsulated modality, the intended use (e.g., therapeutic versus preventative vaccine), dose, and frequency of dosing. These case studies also suggest that regulatory agencies are open to scientific rationale to justify why certain tests should or should not be performed. As more products are approved, it will be important to understand how precedents set for approved products can be leveraged and what additional unique strategies may be applied to ensure nonclinical safety assessments are predictive, relevant, and meaningful for human safety. Proactive alignment with regulatory authorities will be critical in this context, especially as new approaches are proposed. Guidance documents may need to be revised or created as more experience is acquired to reflect the unique considerations for these novel excipients.
Collapse
Affiliation(s)
- Lorrene A Buckley
- Lilly Research Laboratories, Eli Lilly & Co., Inc., Indianapolis, IN, USA
| | | | - Prachi Borude
- Early Development, Alnylam Pharmaceuticals, Inc., Cambridge, MA, USA
| | | | - Philippe Collin
- Clinical Pharmacology and Safety Sciences, R&D, Astrazeneca, Cell and Gene Therapy Safety, Cambridge, UK
| | - Aimee Hillegas
- Immunological Toxicology & Biomarkers, Nonclinical Safety, GSK, Collegeville, PA, USA
| | - Chris MacLauchlin
- Early Development, Alnylam Pharmaceuticals, Inc., Cambridge, MA, USA
| | - Amer F Saleh
- Clinical Pharmacology and Safety Sciences, R&D, Astrazeneca, Cell and Gene Therapy Safety, Cambridge, UK
| | - Amy Sharma
- Drug Safety Research & Development, Pfizer, Inc., New York, NY, USA
| | - Justina Thomas
- Department of Pharmacology, Pharmacokinetics, and Drug Metabolism, Merck & Co., Inc., Rahway, NJ, USA
| | | |
Collapse
|
19
|
Pan M, Cao W, Zhai J, Zheng C, Xu Y, Zhang P. mRNA-based vaccines and therapies - a revolutionary approach for conquering fast-spreading infections and other clinical applications: a review. Int J Biol Macromol 2025; 309:143134. [PMID: 40233916 DOI: 10.1016/j.ijbiomac.2025.143134] [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: 01/02/2024] [Revised: 04/10/2025] [Accepted: 04/11/2025] [Indexed: 04/17/2025]
Abstract
Since the beginning of the COVID-19 pandemic, the development of messenger RNA (mRNA) vaccines has made significant progress in the pharmaceutical industry. The two COVID-19 mRNA vaccines from Moderna and Pfizer/BioNTech have been approved for marketing and have made significant contributions to preventing the spread of SARS-CoV-2. In addition, mRNA therapy has brought hope to some diseases that do not have specific treatment methods or are difficult to treat, such as the Zika virus and influenza virus infections, as well as the prevention and treatment of tumors. With the rapid development of in vitro transcription (IVT) technology, delivery systems, and adjuvants, mRNA therapy has also been applied to hereditary diseases such as Fabry's disease. This article reviews the recent development of mRNA vaccines for structural modification, treatment and prevention of different diseases; delivery carriers and adjuvants; and routes of administration to promote the clinical application of mRNA therapies.
Collapse
Affiliation(s)
- Mingyue Pan
- Department of Pharmacy, The Third Affiliated Hospital (The Affiliated Luohu Hospital) of Shenzhen University, Shenzhen 518001, China
| | - Weiling Cao
- Department of Pharmacy, The Third Affiliated Hospital (The Affiliated Luohu Hospital) of Shenzhen University, Shenzhen 518001, China
| | - Jingbo Zhai
- Key Laboratory of Zoonose Prevention and Control at Universities of Inner Mongolia Autonomous Region, Medical College, Inner Mongolia Minzu University, Tongliao 028000, China
| | - Chunfu Zheng
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, Alberta, Canada.
| | - Yingying Xu
- Department of Pharmaceutics, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China.
| | - Peng Zhang
- Department of Pharmacy, The Third Affiliated Hospital (The Affiliated Luohu Hospital) of Shenzhen University, Shenzhen 518001, China.
| |
Collapse
|
20
|
Kitaya K. B Cell Lineage in the Human Endometrium: Physiological and Pathological Implications. Cells 2025; 14:648. [PMID: 40358172 PMCID: PMC12071375 DOI: 10.3390/cells14090648] [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: 03/16/2025] [Revised: 04/12/2025] [Accepted: 04/26/2025] [Indexed: 05/15/2025] Open
Abstract
Immunocompetent cells of B lineage function in the humoral immunity system in the adaptive immune responses. B cells differentiate into plasmacytes upon antigen-induced activation and produce different subclasses of immunoglobulins/antibodies. Secreted immunoglobulins not only interact with pathogens to inactivate and neutralize them, but also involve the complement system to exert antibacterial activities and trigger opsonization. Endometrium is a mucosal tissue that lines the mammalian uterus and is indispensable for the establishment of a successful pregnancy. The lymphocytes of B cell lineage are a minority in the human cycling endometrium. Human endometrial B cells have therefore been understudied so far. However, the disorders of the female reproductive tract, including chronic endometritis and endometriosis, have highlighted the importance of further research on the endometrial B cell lineage. This review aims to revisit lymphopoiesis, maturation, commitment, and survival of B cells, shedding light on their physiological and pathological implications in the human endometrium.
Collapse
Affiliation(s)
- Kotaro Kitaya
- Infertility Center, Iryouhoujin Kouseikai Mihara Hospital, 6-8 Kamikatsura Miyanogo-cho, Nishikyo-ku, Kyoto 615-8227, Japan
| |
Collapse
|
21
|
Wojcechowskyj JA, Jong RM, Mäger I, Flach B, Munson PV, Mukherjee PP, Mertins B, Barcay KR, Folliard T. Controlling reactogenicity while preserving immunogenicity from a self-amplifying RNA vaccine by modulating nucleocytoplasmic transport. NPJ Vaccines 2025; 10:85. [PMID: 40301369 PMCID: PMC12041602 DOI: 10.1038/s41541-025-01135-8] [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: 01/12/2025] [Accepted: 04/14/2025] [Indexed: 05/01/2025] Open
Abstract
Self-amplifying RNA (saRNA)-based vaccines have emerged as a potent and durable RNA vaccine platform relative to first generation mRNA vaccines. However, RNA vaccine platforms trigger undesirable side effects at protective doses, underscoring the need for improved tolerability. To address this, we leveraged the Cardiovirus leader protein, which is well-characterized to dampen host innate signaling by modulating nucleocytoplasmic transport (NCT). Co-administration of a leader-protein-encoding mRNA (which we have named "RNAx") delivered alongside vaccine cargo saRNA reduced interferon production while enhancing Influenza hemagglutinin (HA) expression in human primary cells and murine models. RNAx potently decreased serum biomarkers of reactogenicity after immunizations with an HA-expressing saRNA-LNP vaccine while maintaining the magnitude of the antibody and cellular response. RNAx also consistently enhanced binding antibody titers after a single injection and in some conditions enhanced binding antibody and neutralization titers post-boost. These findings support RNAx as a promising platform approach for improving tolerability of saRNA-LNP vaccines while preserving or enhancing immunogenicity.
Collapse
Affiliation(s)
| | - Robyn M Jong
- ExcepGen Inc. Emeryville, San Francisco, CA, USA
| | - Imre Mäger
- ExcepGen Inc. Emeryville, San Francisco, CA, USA
| | - Britta Flach
- ExcepGen Inc. Emeryville, San Francisco, CA, USA
| | | | | | | | | | | |
Collapse
|
22
|
Ramos R, Vale N. Emerging Immunotherapies in Lung Cancer: The Latest Advances and the Future of mRNA Vaccines. Vaccines (Basel) 2025; 13:476. [PMID: 40432088 PMCID: PMC12115764 DOI: 10.3390/vaccines13050476] [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: 03/11/2025] [Revised: 04/25/2025] [Accepted: 04/28/2025] [Indexed: 05/29/2025] Open
Abstract
Lung cancer is the most lethal malignancy worldwide, having the highest incidence rate. This is a heterogeneous disease classified according to its histological and molecular characteristics. Depending on these, different therapeutic approaches have already been approved for lung cancer treatment targeting genetic alterations or even the immune system. Nonetheless, other therapies are being studied to continuously improve the care and survival of lung cancer patients. Among them, immunotherapies are one of the main targets of investigation to try and combat the ability of some malignant cells to evade anti-tumor responses mediated by the immune system. Cancer vaccine development has emerged as a promising approach to strengthen the patient's immune system and combat the disease, especially mRNA vaccines. Currently, there are several ongoing studies investigating the therapeutic efficacy of mRNA vaccines in lung cancer treatment alone or combined with other therapeutic drugs. This review aims to highlight the importance of immunotherapy in lung cancer treatment, presenting the most recent advances particularly in mRNA-based vaccines as well as the challenges and future perspectives.
Collapse
Affiliation(s)
- Raquel Ramos
- PerMed Research Group, RISE-Health, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal;
- RISE-Health, Department of Community Medicine, Health Information and Decision (MEDCIDS), Faculty of Medicine, University of Porto, Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
| | - Nuno Vale
- PerMed Research Group, RISE-Health, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal;
- RISE-Health, Department of Community Medicine, Health Information and Decision (MEDCIDS), Faculty of Medicine, University of Porto, Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
- Laboratory of Personalized Medicine, Department of Community Medicine, Health Information and Decision (MEDCIDS), Faculty of Medicine, University of Porto, Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
| |
Collapse
|
23
|
Liang S, Gao S, Fu S, Yuan S, Liu J, Liang M, Han L, Zhang Z, Liu Y, Zhang N. Screening Natural Cholesterol Analogs to Assemble Self-Adjuvant Lipid Nanoparticles for Antigens Tagging Guided Therapeutic Tumor Vaccine. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025:e2419182. [PMID: 40285566 DOI: 10.1002/adma.202419182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Revised: 03/13/2025] [Indexed: 04/29/2025]
Abstract
The clinical progress of tumor nucleotide vaccines is limited due to insufficient recognition and killing of tumor cells with low antigen expression by cytotoxic T lymphocytes (CTL). Here, natural cholesterol analogs are screened to assemble self-adjuvant lipid nanoparticles (LNPs) for antigens tagging tumor cells and dendritic cells (DC) activation. First, a library of ginsenosides are collected, and then screened according to their anti-tumor immunity. Then, ginsenoside-Rg3 based-LNPs loaded with antigens (Rg3-LNPs) are identified as the optimal formulation by investigating the physicochemical and biological properties. Finally, Rg3-LNPs and granulocyte-macrophage colony-stimulating factor (GM-CSF) are co-loaded into a macroporous hydrogel for long-term immune response. Rg3-LNPs could accumulate into both tumors and LNs. Rg3-LNPs targeted tumor cells with high glucose transporter-1 expression via the targeting ligand Rg3, and anchored antigens on the tumor cell surface, thus promoting the recognition of CTL to tumor cells; Rg3-LNPs can accumulate into the LNs to promote DC activation and antigen presentation, thus stimulating CTL activation. Besides, Rg3, as an adjuvant, cooperated with GM-CSF to remodel the tumor microenvironment, thus promoting the killing of CTL to tumor cells. Collectively, this work highlights the importance of tagging antigens to tumor cells in tumor vaccine and has great clinical value for immune-escaping tumors.
Collapse
Affiliation(s)
- Shuang Liang
- School of Pharmaceutical Sciences, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese, Qilu Hospital of Shandong University, The Second Hospital, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong, 250012, China
| | - Shuying Gao
- School of Pharmaceutical Sciences, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese, Qilu Hospital of Shandong University, The Second Hospital, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong, 250012, China
| | - Shunli Fu
- School of Pharmaceutical Sciences, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese, Qilu Hospital of Shandong University, The Second Hospital, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong, 250012, China
| | - Shijun Yuan
- School of Pharmaceutical Sciences, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese, Qilu Hospital of Shandong University, The Second Hospital, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong, 250012, China
| | - Jinhu Liu
- School of Pharmaceutical Sciences, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese, Qilu Hospital of Shandong University, The Second Hospital, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong, 250012, China
| | - Man Liang
- School of Pharmaceutical Sciences, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese, Qilu Hospital of Shandong University, The Second Hospital, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong, 250012, China
| | - Leiqiang Han
- School of Pharmaceutical Sciences, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese, Qilu Hospital of Shandong University, The Second Hospital, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong, 250012, China
| | - Zipeng Zhang
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250062, China
| | - Yongjun Liu
- School of Pharmaceutical Sciences, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese, Qilu Hospital of Shandong University, The Second Hospital, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong, 250012, China
| | - Na Zhang
- School of Pharmaceutical Sciences, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese, Qilu Hospital of Shandong University, The Second Hospital, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong, 250012, China
| |
Collapse
|
24
|
Misra B, Hughes KA, Pentz WH, Surface M, Geldenhuys WJ, Bobbala S. TLR7-Adjuvanted Ionizable Lipid Nanoparticles for mRNA Vaccine Delivery. AAPS J 2025; 27:80. [PMID: 40281311 DOI: 10.1208/s12248-025-01073-2] [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: 01/03/2025] [Accepted: 04/09/2025] [Indexed: 04/29/2025] Open
Abstract
Ionizable lipid nanoparticles (LNPs) are clinically relevant non-viral vectors that allow intracellular delivery of mRNA vaccines to immune cells. To fight against notorious pathogens and cancer, mRNA vaccines necessitate the addition of an adjuvant to induce strong and durable cell-mediated immune responses. Adjuvants that stimulate Toll-like receptor 7 (TLR7) induce the secretion of type I interferons and proinflammatory cytokines, vital for generating strong immune responses. However, the intracellular delivery of TLR7 adjuvants to precisely stimulate the endosomal TLR7 receptor remains a huge challenge. This issue can be addressed by exploiting ionizable LNP platforms, which can encapsulate and carry mRNA vaccines and small molecule hydrophobic adjuvants to immune cells. CL347 is a potent lipid-based adjuvant that selectively stimulates the TLR7 receptor. In this study, we developed ionizable LNPs incorporating SM102 and CL347 adjuvant as the ionizable lipid and TLR7 adjuvant, respectively. CL347-SM102 LNPs exhibited particle sizes of less than 150 nm with spherical morphology and mRNA encapsulation efficiency of greater than 95%. In vivo studies showed a two-fold increase in IFN-γ producing CD4 and CD8 T cells in the lymphoid organs of the mice immunized with adjuvanted LNPs compared to the non-adjuvanted LNPs. Human PBMCs treated with adjuvanted LNPs exhibited significantly higher CD40 expression and pro-inflammatory cytokine (IL-6 and IFN-γ) secretion than non-adjuvanted LNPs. Together, these results suggest the potential of ionizable LNPs as a platform for concurrent delivery of mRNA and adjuvants for prophylactic and therapeutic vaccine applications.
Collapse
Affiliation(s)
- Bishal Misra
- Department of Pharmaceutical Sciences, West Virginia University, Morgantown, West Virginia, 26506, USA
| | - Krystal A Hughes
- Department of Pharmaceutical Sciences, West Virginia University, Morgantown, West Virginia, 26506, USA
| | - William H Pentz
- Department of Pharmaceutical Sciences, West Virginia University, Morgantown, West Virginia, 26506, USA
- School of Medicine, West Virginia University, Morgantown, West Virginia, 26506, USA
| | - Morgan Surface
- Department of Pharmaceutical Sciences, West Virginia University, Morgantown, West Virginia, 26506, USA
| | - Werner J Geldenhuys
- Department of Pharmaceutical Sciences, West Virginia University, Morgantown, West Virginia, 26506, USA
| | - Sharan Bobbala
- Department of Pharmaceutical Sciences, West Virginia University, Morgantown, West Virginia, 26506, USA.
| |
Collapse
|
25
|
Li Z, Chen P, Qu A, Sun M, Xu L, Xu C, Hu S, Kuang H. Opportunities and Challenges for Nanomaterials as Vaccine Adjuvants. SMALL METHODS 2025:e2402059. [PMID: 40277301 DOI: 10.1002/smtd.202402059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 03/29/2025] [Indexed: 04/26/2025]
Abstract
Adjuvants, as a critical component of vaccines, are capable of eliciting more robust and sustained immune responses. Nanomaterials have shown unique advantages and broad application prospects in adjuvant development due to their high adjustability and distinctive physicochemical properties. This review focuses on nanoadjuvants and their immunological mechanisms. First, various types of adjuvants are introduced with an emphasis on metal and metal oxide nanoparticles, coordination polymers, liposomes, polymer nanoparticles, and other inorganic nanoparticles that can serve as vaccine adjuvants. Second, this review describes the current status of the clinical applications of nanoadjuvants. Next, the mechanisms of action for nanoadjuvants have been thoroughly elucidated, including the depot effect, NLRP3 inflammasome activation, targeting C-type lectin receptors, activation of toll-like receptors, and activation of the cGAS-STING signaling pathway. Finally, the challenges and opportunities associated with the development of nanoadjuvants have also been addressed.
Collapse
Affiliation(s)
- Zongda Li
- International Joint Research Laboratory for Biointerface and Biodetection, State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Panpan Chen
- International Joint Research Laboratory for Biointerface and Biodetection, State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Aihua Qu
- International Joint Research Laboratory for Biointerface and Biodetection, State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Maozhong Sun
- International Joint Research Laboratory for Biointerface and Biodetection, State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Liguang Xu
- International Joint Research Laboratory for Biointerface and Biodetection, State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Chuanlai Xu
- International Joint Research Laboratory for Biointerface and Biodetection, State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Shudong Hu
- International Joint Research Laboratory for Biointerface and Biodetection, State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Hua Kuang
- International Joint Research Laboratory for Biointerface and Biodetection, State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| |
Collapse
|
26
|
Chatterjee D, Kurup D, Smeyne RJ. Environmental exposures and familial background alter the induction of neuropathology and inflammation after SARS-CoV-2 infection. NPJ Parkinsons Dis 2025; 11:86. [PMID: 40268936 PMCID: PMC12019605 DOI: 10.1038/s41531-025-00925-0] [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: 11/21/2024] [Accepted: 03/27/2025] [Indexed: 04/25/2025] Open
Abstract
Post-infection sequela of several viruses have been linked with Parkinson's disease (PD). Here, we investigated whether mice infected with SARS-CoV-2 alone or in combination with two putative Parkinsonian toxins, MPTP and paraquat, increased the susceptibility to develop Parkinsonian pathology. We also examined if G2019S LRRK2 mice had any change in sensitivity to SARS-CoV-2 as well as if vaccination against this virus altered any neuropathology. Infection with WA-1/2020 or Omicron B1.1.529 strains sensitized both WT and G2019S LRRK2 mice to the neuropathological effects of a subtoxic exposure to MPTP, but not paraquat. These neuropathologies were rescued in WT mice vaccinated with mRNA- or protein-based SARS-CoV-2 vaccines. However, G2019S LRRK2 mutant mice were only protected with the protein-based vaccine. These results highlight the role of both environmental exposures and familial background on the development of Parkinsonian pathology secondary to viral infection and the benefit of vaccines in reducing these risks.
Collapse
Affiliation(s)
- Debotri Chatterjee
- Department of Neurobiology, Thomas Jefferson University, 900 Walnut Street, Philadelphia, PA, 19107, USA
| | - Drishya Kurup
- Department of Microbiology and Immunology, Thomas Jefferson University, 233 S 10th Street, Philadelphia, PA, 19107, USA
- Jefferson Center for Vaccines and Pandemic Preparedness, 233 S 10th Street, Philadelphia, PA, 19107, USA
| | - Richard Jay Smeyne
- Department of Neurobiology, Thomas Jefferson University, 900 Walnut Street, Philadelphia, PA, 19107, USA.
| |
Collapse
|
27
|
Zelkoski AE, Lu Z, Sukumar G, Dalgard C, Said H, Alameh MG, Mitre E, Malloy AMW. Ionizable lipid nanoparticles of mRNA vaccines elicit NF-κB and IRF responses through toll-like receptor 4. NPJ Vaccines 2025; 10:73. [PMID: 40246950 PMCID: PMC12006303 DOI: 10.1038/s41541-025-01124-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 03/24/2025] [Indexed: 04/19/2025] Open
Abstract
Ionizable lipid nanoparticles (LNP) that have enabled the success of messenger RNA (mRNA) vaccines have been shown to be immunostimulatory in the absence of mRNA. However, the mechanisms through which they activate innate immune cells is incompletely understood. Using a monocyte cell line, we compared the ability of three LNP formulations to activate transcription factors Nuclear Factor-kappa B (NF-κB) and Interferon Regulatory Factor (IRF). Comparison of signaling in knockout cell lines illustrated a role for Toll-like receptor (TLR) 4 in initiation of this signaling cascade and the contribution of the ionizable lipid component. Activation induced by empty LNPs was similar to that induced by LNPs containing mRNA, indicating that LNPs may provide the majority of innate stimulation for the mRNA vaccine platform. Our findings demonstrate that ionizable lipids within LNPs signal through TLR4 to activate NF-κB and IRF, identifying a mechanism for innate activation that can be optimized for adjuvant design.
Collapse
Affiliation(s)
- Amanda E Zelkoski
- Department of Pediatrics, Uniformed Services University of Health Sciences, Bethesda, MD, USA
- Henry M Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Zhongyan Lu
- Department of Pediatrics, Uniformed Services University of Health Sciences, Bethesda, MD, USA
- Henry M Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Gauthaman Sukumar
- Henry M Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
- Department of Anatomy, Physiology & Genetics, Uniformed Services University of Health Sciences, Bethesda, MD, USA
| | - Clifton Dalgard
- Department of Anatomy, Physiology & Genetics, Uniformed Services University of Health Sciences, Bethesda, MD, USA
| | - Hooda Said
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Mohamad-Gabriel Alameh
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn Institute for RNA Innovation, University of Pennsylvania, Philadelphia, PA, USA
| | - Edward Mitre
- Department of Microbiology and Immunology, Uniformed Services University of Health Sciences, Bethesda, PA, USA
| | - Allison M W Malloy
- Department of Pediatrics, Uniformed Services University of Health Sciences, Bethesda, MD, USA.
| |
Collapse
|
28
|
Tursi NJ, Tiwari S, Bedanova N, Kannan T, Parzych E, Okba N, Liaw K, Sárközy A, Livingston C, Trullen MI, Gary EN, Vadovics M, Laenger N, Londregan J, Khan MS, Omo-Lamai S, Muramatsu H, Blatney K, Hojecki C, Machado V, Maricic I, Smith TRF, Humeau LM, Patel A, Kossenkov A, Brenner JS, Allman D, Krammer F, Pardi N, Weiner DB. Modulation of lipid nanoparticle-formulated plasmid DNA drives innate immune activation promoting adaptive immunity. Cell Rep Med 2025; 6:102035. [PMID: 40120578 PMCID: PMC12047470 DOI: 10.1016/j.xcrm.2025.102035] [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: 07/09/2024] [Revised: 11/20/2024] [Accepted: 02/25/2025] [Indexed: 03/25/2025]
Abstract
Nucleic acid vaccines have grown in importance over the past several years, with the development of new approaches remaining a focus. We describe a lipid nanoparticle-formulated DNA (DNA-LNP) formulation which induces robust innate and adaptive immunity with similar serological potency to mRNA-LNPs and adjuvanted protein. Using an influenza hemagglutinin (HA)-encoding construct, we show that priming with our HA DNA-LNP demonstrated stimulator of interferon genes (STING)-dependent upregulation and activation of migratory dendritic cell (DC) subpopulations. HA DNA-LNP induced superior antigen-specific CD8+ T cell responses relative to mRNA-LNPs or adjuvanted protein, with memory responses persisting beyond one year. In rabbits immunized with HA DNA-LNP, we observed immune responses comparable or superior to mRNA-LNPs at the same dose. In an additional model, a SARS-CoV-2 spike-encoding DNA-LNP elicited protective efficacy comparable to spike mRNA-LNPs. Our study identifies a platform-specific priming mechanism for DNA-LNPs divergent from mRNA-LNPs or adjuvanted protein, suggesting avenues for this approach in prophylactic and therapeutic vaccine development.
Collapse
Affiliation(s)
- Nicholas J Tursi
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA 19104, USA; Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sachchidanand Tiwari
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Nicole Bedanova
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Toshitha Kannan
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Elizabeth Parzych
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Nisreen Okba
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Kevin Liaw
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA 19104, USA
| | - András Sárközy
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Cory Livingston
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Maria Ibanez Trullen
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Ebony N Gary
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Máté Vadovics
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Niklas Laenger
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA 19104, USA; Biology Department, Saint Joseph's University, Philadelphia, PA 19131, USA
| | - Jennifer Londregan
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mohammad Suhail Khan
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Serena Omo-Lamai
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hiromi Muramatsu
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kerry Blatney
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Casey Hojecki
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA 19104, USA
| | | | - Igor Maricic
- Inovio Pharmaceuticals, Plymouth Meeting, PA 19462, USA
| | | | | | - Ami Patel
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Andrew Kossenkov
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Jacob S Brenner
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - David Allman
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Ignaz Semmelweis Institute, Interuniversity Institute for Infection Research, Medical University of Vienna, Vienna, Austria
| | - Norbert Pardi
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - David B Weiner
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA 19104, USA.
| |
Collapse
|
29
|
Matayoshi K, Takahashi S, Ryu S, Koide H, Yonezawa S, Ozaki N, Kurata M, Asai T. Development of a messenger RNA vaccine using pH-responsive dipeptide-conjugated lipids exhibiting reduced inflammatory properties. Int J Pharm 2025; 674:125485. [PMID: 40101873 DOI: 10.1016/j.ijpharm.2025.125485] [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: 12/25/2024] [Revised: 03/10/2025] [Accepted: 03/14/2025] [Indexed: 03/20/2025]
Abstract
Lipid nanoparticles (LNPs) are used to encapsulate messenger ribonucleic acids (mRNAs) and enhance mRNA vaccine efficacy by producing inflammatory mediators. However, the overproduction of inflammatory mediators via LNP injection causes severe side effects, presenting a potential limitation. To resolve this issue, we developed pH-responsive dipeptide-conjugated lipid (DPL)-based LNPs (DPL-LNPs) for efficient small interfering RNA delivery with excellent biocompatibility. In detail, we optimized the dipeptide sequence and lipid-tail length of DPL, the helper-lipid compositions, and the molecular weight and lipid-tail length of the polyethylene glycol (PEG)-lipid to achieve highly efficient and safe mRNA delivery. Our results revealed that the LNPs prepared using glutamic acid (E)- and arginine (R)-conjugated DPL (DPL-ER) displayed higher protein-expression efficacy than DPL-threonine-R- and DPL-aspartic acid-R-based LNPs. Additionally, the lipid-tail length of the C22-bearing DPL-ER (DPL-ER-C22)-based LNPs displayed higher protein-expression efficacies than their C18 (DPL-ER-C18)- and C24 (DPL-ER-C24)-based LNPs. Moreover, the DPL-ER-C22-based LNPs incorporating low-lipid-tail-length phospholipids and PEG-lipids exhibited efficient protein expression. Most importantly, the injection of optimized DPL-LNPs exhibited comparable antigen-specific antibody production levels, with significantly lower inflammatory-mediator production compared with those of the commercially available LNPs. These results indicate that DPL-based LNPs (DPL-LNPs) can be deployed as highly efficient, safe carriers for mRNA delivery for developing mRNA vaccine formulations.
Collapse
Affiliation(s)
- Katsuki Matayoshi
- Laboratory of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, Shizuoka 422-8526 Japan
| | - Sayaka Takahashi
- Laboratory of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, Shizuoka 422-8526 Japan
| | - Sohei Ryu
- Laboratory of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, Shizuoka 422-8526 Japan
| | - Hiroyuki Koide
- Laboratory of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, Shizuoka 422-8526 Japan
| | - Sei Yonezawa
- Laboratory of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, Shizuoka 422-8526 Japan
| | - Nahoko Ozaki
- Development & Technical Group, Sogo Pharmaceuticals Co., Ltd., 408-1 Sonegasaki, Kamisokoino, Nakama, Fukuoka 809-0003, Japan
| | - Makiko Kurata
- Development & Technical Group, Sogo Pharmaceuticals Co., Ltd., 408-1 Sonegasaki, Kamisokoino, Nakama, Fukuoka 809-0003, Japan
| | - Tomohiro Asai
- Laboratory of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, Shizuoka 422-8526 Japan.
| |
Collapse
|
30
|
Zeng A, Liu Y, Wang P, Cao Y, Guo W. Using siRNA-Based Anti-Inflammatory Lipid Nanoparticles for Gene Regulation in Psoriasis. Int J Nanomedicine 2025; 20:4519-4533. [PMID: 40248028 PMCID: PMC12003987 DOI: 10.2147/ijn.s504639] [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: 12/06/2024] [Accepted: 03/20/2025] [Indexed: 04/19/2025] Open
Abstract
Background Psoriasis is a chronic inflammatory autoimmune disease, yet it affects hundreds of millions of people. Long-term effective intervention of the disease by targeting the causative genes via RNAi (RNA interference) has become a reality. However, its further application is hindered by inflammatory side effects caused by delivery systems such as LNP (lipid nanoparticles). Purpose This study aimed to develop a novel anti-inflammatory LNP rationally tailored for topical application in psoriasis and to validate its potential to deliver Stat3 (signal transducer and activator of transcription 3) siRNA for the treatment of psoriasis. Methods To assess the transfection efficiency, anti-inflammatory capacity of LNPs. The therapeutic effect of modified anti-inflammatory LNP delivery of Stat3 siRNA on psoriasis was evaluated both in vitro and in an imiquimod-induced mice. Results LNPs exhibit both superior transfection efficiency and significant anti-inflammatory effects. In vitro functional studies showed that in an inflammatory DC model, anti-inflammatory LNP (C8B2) inhibited inflammatory mediators much better than classical LNPs by delivering Stat3 siRNA; in pathological HaCat cells, Stat3 siRNA reduced cell proliferation and promoted apoptosis. In the imiquimod-induced mouse model, the C8B2-si-Stat3 group demonstrated a clear reduction in psoriasis progression, whereas the C8B2 carrier group also exhibited a notable decrease in inflammation. Conclusion In this study, we successfully developed a novel anti-inflammatory LNP, which demonstrated notable advantages in delivery capacity, anti-inflammatory effect, and targeting therapy against STAT3, providing new ideas and strategies for nucleic acid therapy of psoriasis. This LNP platform could be broadly applicable to various inflammatory conditions, offering a versatile tool for targeted gene modulation and inflammation control.
Collapse
Affiliation(s)
- Aizhong Zeng
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 211112, People’s Republic of China
| | - Yuanyuan Liu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 211112, People’s Republic of China
| | - Ping Wang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 211112, People’s Republic of China
| | - Yufei Cao
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 211112, People’s Republic of China
| | - Wei Guo
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 211112, People’s Republic of China
| |
Collapse
|
31
|
Chiba S, Halfmann PJ, Iida S, Hirata Y, Sato Y, Kuroda M, Armbrust T, Spyra S, Suzuki T, Kawaoka Y. Correction to "Recombinant Spike protein vaccines coupled with adjuvants that have different modes of action induce protective immunity against SARS-CoV-2" [Vaccine 22 (41) (2023) 6025-6035]. Vaccine 2025; 52:126880. [PMID: 39985967 DOI: 10.1016/j.vaccine.2025.126880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2025]
Abstract
In the previously published version of the paper, the term "AS03" was used to describe the AddaS03 adjuvant used in animal experiments. This could lead to confusion among the trade and public as to a connection between the AddaS03 adjuvant and GSK's AS03. Upon request by GSK, the authors clarify that no AS03 from GSK was used in this study, and the results obtained with AddaS03 are not transposable to the GSK's AS03 adjuvant. The article has now been corrected, and the conclusions of this paper remain unchanged. Corrections highlighted in bold. The spike (S) protein of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a glycoprotein, expressed on the virion surface, that mediates infection of host cells by directly interacting with host receptors. As such, it is a reasonable target to neutralize the infectivity of the virus. Here we found that a recombinant S protein vaccine adjuvanted with Alhydrogel or the QS-21-like adjuvant Quil-A effectively induced anti-S receptor binding domain (RBD) serum IgG and neutralizing antibody titers in the Syrian hamster model, resulting in significantly low SARS-CoV-2 replication in respiratory organs and reduced body weight loss upon virus challenge. Severe lung inflammation upon virus challenge was also strongly suppressed by vaccination. We also found that the S protein vaccine adjuvanted with Alhydrogel, Quil-A, or AddaS03 elicited significantly higher neutralizing antibody titers in mice than did unadjuvanted vaccine. Although the neutralizing antibody titers against the variant viruses B.1.351 and B.1.617.2 declined markedly in mice immunized with wild-type S protein, the binding antibody levels against the variant S proteins were equivalent to those against wild-type S. When splenocytes from the immunized mice were re-stimulated with the S protein in vitro, the induced Th1 or Th2 cytokine levels were not significantly different upon re-stimulation with wild-type S or variant S, suggesting that the T-cell responses against the variants were the same as those against the wild-type virus. Upon Omicron XBB-challenge in hamsters, wild-type S-vaccination with Alhydrogel or AddaS03 reduced lung virus titers on Day 3, and the Quil-A adjuvanted group showed less body weight loss, although serum neutralizing antibody titers against XBB were barely detected in vitro. Collectively, recombinant vaccines coupled with different adjuvants may be promising modalities to combat new variant viruses by inducing various arms of the immune response.
Collapse
Affiliation(s)
- Shiho Chiba
- Influenza Research Institute, Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-, Madison, WI 53711, USA
| | - Peter J Halfmann
- Influenza Research Institute, Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-, Madison, WI 53711, USA
| | - Shun Iida
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Yuichiro Hirata
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Yuko Sato
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Makoto Kuroda
- Influenza Research Institute, Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-, Madison, WI 53711, USA
| | - Tammy Armbrust
- Influenza Research Institute, Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-, Madison, WI 53711, USA
| | - Samuel Spyra
- Influenza Research Institute, Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-, Madison, WI 53711, USA
| | - Tadaki Suzuki
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Yoshihiro Kawaoka
- Influenza Research Institute, Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-, Madison, WI 53711, USA; Division of Virology, Department of Microbiology and Immunology, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan; The Research Center for Global Viral Diseases, National Center for Global Health and Medicine Research Institute, Tokyo 162-8655, Japan.; The University of Tokyo, Pandemic Preparedness, Infection and Advanced Research Center, Tokyo 162-8655, Japan.
| |
Collapse
|
32
|
Lei H, Alu A, Yang J, He C, Shi J, Hong W, Peng D, Zhang Y, Liu J, Qin F, Huang X, Ye C, Pei L, He X, Yan H, Lu G, Song X, Wei X, Wei Y. Intranasal Inoculation of Cationic Crosslinked Carbon Dots-Adjuvanted Respiratory Syncytial Virus F Subunit Vaccine Elicits Mucosal and Systemic Humoral and Cellular Immunity. MedComm (Beijing) 2025; 6:e70146. [PMID: 40135196 PMCID: PMC11933438 DOI: 10.1002/mco2.70146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 02/19/2025] [Accepted: 02/22/2025] [Indexed: 03/27/2025] Open
Abstract
Respiratory syncytial virus (RSV) causes severe acute lower respiratory tract infections, especially in infants and the elderly. Developing an RSV vaccine that promotes a robust mucosal immune response is necessary to successfully prevent viral transmission and the development of severe disease. We previously reported that crosslinked carbon dots (CCD) may be an excellent adjuvant candidate for intranasal (IN) protein subunit vaccines. Considering the strong immunogenicity of RSV prefused F protein (preF), we prepared an IN RSV vaccine composed of the CCD adjuvant and the preF protein as antigen (CCD/preF) and evaluated the induced antigen-specific humoral and cellular immunity. We found that IN immunization with the CCD/preF vaccine elicited strong serum IgG responses and mucosal immunity, including secreted IgA antibodies, tissue-resident memory T (TRM) cells, and antigen-specific B cells, which lasted for at least 1 year. In addition, a combination of intramuscular and IN immunization with CCD/preF vaccine induced stronger systemic and mucosal immunity. Together, this study proved the high immunogenicity of the CCD/preF vaccines and supported the university of the mucosal CCD adjuvant, supporting further development of the CCD/preF vaccine in larger animal models and clinical studies.
Collapse
Affiliation(s)
- Hong Lei
- Laboratory of Aging Research and Cancer Drug TargetState Key Laboratory of Biotherapy and Cancer CenterNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengduSichuanPeople's Republic of China
| | - Aqu Alu
- Laboratory of Aging Research and Cancer Drug TargetState Key Laboratory of Biotherapy and Cancer CenterNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengduSichuanPeople's Republic of China
| | - Jingyun Yang
- Laboratory of Aging Research and Cancer Drug TargetState Key Laboratory of Biotherapy and Cancer CenterNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengduSichuanPeople's Republic of China
| | - Cai He
- Laboratory of Aging Research and Cancer Drug TargetState Key Laboratory of Biotherapy and Cancer CenterNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengduSichuanPeople's Republic of China
| | - Jie Shi
- Laboratory of Aging Research and Cancer Drug TargetState Key Laboratory of Biotherapy and Cancer CenterNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengduSichuanPeople's Republic of China
| | - Weiqi Hong
- Laboratory of Aging Research and Cancer Drug TargetState Key Laboratory of Biotherapy and Cancer CenterNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengduSichuanPeople's Republic of China
| | - Dandan Peng
- Laboratory of Aging Research and Cancer Drug TargetState Key Laboratory of Biotherapy and Cancer CenterNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengduSichuanPeople's Republic of China
| | - Yu Zhang
- Laboratory of Aging Research and Cancer Drug TargetState Key Laboratory of Biotherapy and Cancer CenterNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengduSichuanPeople's Republic of China
| | - Jian Liu
- Laboratory of Aging Research and Cancer Drug TargetState Key Laboratory of Biotherapy and Cancer CenterNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengduSichuanPeople's Republic of China
| | - Furong Qin
- Laboratory of Aging Research and Cancer Drug TargetState Key Laboratory of Biotherapy and Cancer CenterNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengduSichuanPeople's Republic of China
| | - Xiya Huang
- Laboratory of Aging Research and Cancer Drug TargetState Key Laboratory of Biotherapy and Cancer CenterNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengduSichuanPeople's Republic of China
| | - Chunjun Ye
- Laboratory of Aging Research and Cancer Drug TargetState Key Laboratory of Biotherapy and Cancer CenterNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengduSichuanPeople's Republic of China
| | - Lijiao Pei
- Laboratory of Aging Research and Cancer Drug TargetState Key Laboratory of Biotherapy and Cancer CenterNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengduSichuanPeople's Republic of China
| | - Xuemei He
- Laboratory of Aging Research and Cancer Drug TargetState Key Laboratory of Biotherapy and Cancer CenterNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengduSichuanPeople's Republic of China
| | - Hong Yan
- Laboratory of Aging Research and Cancer Drug TargetState Key Laboratory of Biotherapy and Cancer CenterNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengduSichuanPeople's Republic of China
| | - Guangwen Lu
- Laboratory of Aging Research and Cancer Drug TargetState Key Laboratory of Biotherapy and Cancer CenterNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengduSichuanPeople's Republic of China
| | - Xiangrong Song
- Laboratory of Aging Research and Cancer Drug TargetState Key Laboratory of Biotherapy and Cancer CenterNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengduSichuanPeople's Republic of China
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug TargetState Key Laboratory of Biotherapy and Cancer CenterNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengduSichuanPeople's Republic of China
| | - Yuquan Wei
- Laboratory of Aging Research and Cancer Drug TargetState Key Laboratory of Biotherapy and Cancer CenterNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengduSichuanPeople's Republic of China
| |
Collapse
|
33
|
Du X, Nakanishi H, Yamada T, Sin Y, Minegishi K, Motohashi N, Aoki Y, Itaka K. Polyplex Nanomicelle-Mediated Pgc-1α4 mRNA Delivery Via Hydrodynamic Limb Vein Injection Enhances Damage Resistance in Duchenne Muscular Dystrophy Mice. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2409065. [PMID: 40051178 PMCID: PMC12021044 DOI: 10.1002/advs.202409065] [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: 08/02/2024] [Revised: 11/30/2024] [Indexed: 04/26/2025]
Abstract
Duchenne muscular dystrophy (DMD) is caused by mutations in the DMD gene, leading to the absence of dystrophin and progressive muscle degeneration. Current therapeutic strategies, such as exon-skipping and gene therapy, face limitations including truncated dystrophin production and safety concerns. To address these issues, a novel mRNA-based therapy is explored using polyplex nanomicelles to deliver mRNA encoding peroxisome proliferator-activated receptor gamma coactivator 1 alpha isoform 4 (PGC-1α4) via hydrodynamic limb vein (HLV) administration. Using an in vivo muscle torque measurement technique, it is observed that nanomicelle-delivered Pgc-1α4 mRNA significantly improved muscle damage resistance and mitochondrial activity in mdx mice. Specifically, HLV administration of Pgc-1α4 mRNA in dystrophic muscles significantly relieved the torque reduction and myofiber injury induced by eccentric contraction (ECC), boosted metabolic gene expression, and enhanced muscle oxidative capacity. In comparison, lipid nanoparticles (LNPs), a widely used mRNA delivery system, does not achieve similar protective effects, likely due to their intrinsic immunogenicity. This foundational proof-of-concept study highlights the potential of mRNA-based therapeutics for the treatment of neuromuscular diseases such as DMD and demonstrates the capability of polyplex nanomicelles as a safe and efficient mRNA delivery system for therapeutic applications.
Collapse
Affiliation(s)
- Xuan Du
- Department of Biofunction ResearchLaboratory for Biomaterials and Bioengineering, Institute of Integrated ResearchInstitute of Science TokyoTokyo101‐0062Japan
| | - Hideyuki Nakanishi
- Department of Biofunction ResearchLaboratory for Biomaterials and Bioengineering, Institute of Integrated ResearchInstitute of Science TokyoTokyo101‐0062Japan
- Clinical Biotechnology TeamCenter for Infectious Disease Education and Research (CiDER)Osaka UniversityOsaka565‐0871Japan
| | - Takashi Yamada
- Department of Physical TherapySapporo Medical UniversitySapporo060‐8556Japan
| | - Yooksil Sin
- Department of Biofunction ResearchLaboratory for Biomaterials and Bioengineering, Institute of Integrated ResearchInstitute of Science TokyoTokyo101‐0062Japan
- Clinical Biotechnology TeamCenter for Infectious Disease Education and Research (CiDER)Osaka UniversityOsaka565‐0871Japan
| | - Katsura Minegishi
- Department of Molecular TherapyNational Institute of NeuroscienceNational Center of Neurology and Psychiatry (NCNP)Tokyo187‐8502Japan
| | - Norio Motohashi
- Department of Molecular TherapyNational Institute of NeuroscienceNational Center of Neurology and Psychiatry (NCNP)Tokyo187‐8502Japan
| | - Yoshitsugu Aoki
- Department of Molecular TherapyNational Institute of NeuroscienceNational Center of Neurology and Psychiatry (NCNP)Tokyo187‐8502Japan
| | - Keiji Itaka
- Department of Biofunction ResearchLaboratory for Biomaterials and Bioengineering, Institute of Integrated ResearchInstitute of Science TokyoTokyo101‐0062Japan
- Clinical Biotechnology TeamCenter for Infectious Disease Education and Research (CiDER)Osaka UniversityOsaka565‐0871Japan
| |
Collapse
|
34
|
Yoshida T, Takashima K, Mtali YS, Miyashita Y, Iwamoto A, Fukushima Y, Nakamura K, Oshiumi H. Regulation of IL-17A-mediated hypersensitivity by extracellular vesicles and lipid nanoparticles carrying miR-451a. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025; 214:651-665. [PMID: 40073105 DOI: 10.1093/jimmun/vkae049] [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: 03/29/2024] [Accepted: 12/04/2024] [Indexed: 03/14/2025]
Abstract
Extracellular vesicles (EVs), including exosomes, mediate intercellular communication by transporting functional molecules between donor cells and recipient cells, thereby regulating biological processes, such as immune responses. miR-451a, an immune regulatory microRNA, is highly abundant in circulating EVs; however, its precise physiological significance remains to be fully elucidated. Here, we demonstrate that miR-451a deficiency exacerbates delayed-type hypersensitivity (DTH) in mice. Notably, miR-451a knockout resulted in a significant increase in the number of interleukin (IL)-17A-expressing T helper 17 and γδ T cells infiltrating DTH-induced ear lesions. miR-451a deficiency also increased the number of γδ T cells in the secondary lymphoid tissues. Comprehensive analyses revealed that miR-451 deficiency promoted the expression of Rorc and γδ T cell-related genes following sensitization with allergens. Moreover, intravenous administration of wild-type EVs to miR-451a knockout mice increased cellular miR-451a levels in tissues and significantly attenuated the severity of DTH. Furthermore, synthetic lipid nanoparticles encapsulating miR-451a effectively mitigated DTH. Our findings indicate the importance of circulating miR-451a in the proliferation of γδ T cells and highlight the therapeutic potential of lipid nanoparticle-based microRNA delivery platforms for interventions in immune-related diseases.
Collapse
Affiliation(s)
- Takanobu Yoshida
- Department of Immunology, Faculty of Life Sciences, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
- Department of Pediatrics, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Ken Takashima
- Department of Immunology, Faculty of Life Sciences, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yohana S Mtali
- Department of Immunology, Faculty of Life Sciences, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yusuke Miyashita
- Department of Immunology, Faculty of Life Sciences, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
- Department of Pediatrics, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Asuka Iwamoto
- Department of Immunology, Faculty of Life Sciences, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yoshimi Fukushima
- Department of Immunology, Faculty of Life Sciences, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Kimitoshi Nakamura
- Department of Pediatrics, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Hiroyuki Oshiumi
- Department of Immunology, Faculty of Life Sciences, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
35
|
Ahmadivand S, Fux R, Palić D. Role of T Follicular Helper Cells in Viral Infections and Vaccine Design. Cells 2025; 14:508. [PMID: 40214462 PMCID: PMC11987902 DOI: 10.3390/cells14070508] [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: 01/25/2025] [Revised: 03/20/2025] [Accepted: 03/26/2025] [Indexed: 04/14/2025] Open
Abstract
T follicular helper (Tfh) cells are a specialized subset of CD4+ T lymphocytes that are essential for the development of long-lasting humoral immunity. Tfh cells facilitate B lymphocyte maturation, promote germinal center formation, and drive high-affinity antibody production. Our current knowledge of Tfh interactions with the humoral immune system effectors suggests that they have a critical role in supporting the immune response against viral infections. This review discusses the mechanisms through which Tfh cells influence anti-viral immunity, highlighting their interactions with B cells and their impact on antibody quality and quantity. We explore the role of Tfh cells in viral infections and examine how vaccine design can be improved to enhance Tfh cell responses. Innovative vaccine platforms, such as mRNA vaccines and self-assembling protein nanoplatforms (SAPNs), are promising strategies to enhance Tfh cell activation. Their integration and synergistic combination could further enhance immunity and Tfh responses (SAPN-RNA vaccines). In summary, we provide a comprehensive overview of the current insights into Tfh cells' role during viral infections, emphasizing their potential as strategic targets for innovative vaccine development.
Collapse
Affiliation(s)
- Sohrab Ahmadivand
- Faculty of Veterinary Medicine, Ludwig-Maximilians-University Munich, 80539 Munich, Germany
| | - Robert Fux
- Institute for Infectious Diseases and Zoonoses, Ludwig-Maximilians-University Munich, 80539 Munich, Germany;
| | - Dušan Palić
- Faculty of Veterinary Medicine, Ludwig-Maximilians-University Munich, 80539 Munich, Germany
| |
Collapse
|
36
|
Meng S, Hara T, Miura Y, Arao Y, Saito Y, Inoue K, Hirotsu T, Vecchione A, Satoh T, Ishii H. In Vivo Engineered CAR-T Cell Therapy: Lessons Built from COVID-19 mRNA Vaccines. Int J Mol Sci 2025; 26:3119. [PMID: 40243757 PMCID: PMC11988490 DOI: 10.3390/ijms26073119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 03/13/2025] [Accepted: 03/26/2025] [Indexed: 04/18/2025] Open
Abstract
Chimeric antigen receptor T cell (CAR-T) therapy has revolutionized cancer immunotherapy but continues to face significant challenges that limit its broader application, such as antigen targeting, the tumor microenvironment, and cell persistence, especially in solid tumors. Meanwhile, the global implementation of mRNA vaccines during the COVID-19 pandemic has highlighted the transformative potential of mRNA and lipid nanoparticle (LNP) technologies. These innovations, characterized by their swift development timelines, precise antigen design, and efficient delivery mechanisms, provide a promising framework to address some limitations of CAR-T therapy. Recent advancements, including mRNA-based CAR engineering and optimized LNP delivery, have demonstrated the capacity to enhance CAR-T efficacy, particularly in the context of solid tumors. This review explores how mRNA-LNP technology can drive the development of in vivo engineered CAR-T therapies to address current limitations and discusses future directions, including advancements in mRNA design, LNP optimization, and strategies for improving in vivo CAR-T functionality and safety. By bridging these technological insights, CAR-T therapy may evolve into a versatile and accessible treatment paradigm across diverse oncological landscapes.
Collapse
Grants
- grant nos. 19K22658, 20H00541, 21K19526, 22H03146, 22K19559, 23K19505, 23K18313, 23KK0153, 24K22144, and 16H06279 (PAGS) Ministry of Education, Culture, Sports, Science and Technology
- grant nos. JP23ym0126809 and JP24ym0126809 Japan Agency for Medical Research and Development
- 23-255001 Princess Takamatsu Cancer Research Fund
- G-2024-3-00 IFO Research Communications
- 2024 Oceanic Wellness Foundation
- 2024 Suzuken Memorial Foundation
Collapse
Affiliation(s)
- Sikun Meng
- Department of Medical Data Science, Center of Medical Innovation and Translational Research, Osaka University Graduate School of Medicine, Yamadaoka 2-2, Suita, Osaka 565-0871, Japan
| | - Tomoaki Hara
- Department of Medical Data Science, Center of Medical Innovation and Translational Research, Osaka University Graduate School of Medicine, Yamadaoka 2-2, Suita, Osaka 565-0871, Japan
| | - Yutaka Miura
- Laboratory for Chemistry and Life Science, Institute of Integrated Research, Institute of Science Tokyo, 4259 Nagatsutacho, Midori-ku, Yokohama 226-8501, Japan
| | - Yasuko Arao
- Department of Medical Data Science, Center of Medical Innovation and Translational Research, Osaka University Graduate School of Medicine, Yamadaoka 2-2, Suita, Osaka 565-0871, Japan
| | - Yoshiko Saito
- Department of Medical Data Science, Center of Medical Innovation and Translational Research, Osaka University Graduate School of Medicine, Yamadaoka 2-2, Suita, Osaka 565-0871, Japan
| | - Kana Inoue
- Department of Medical Data Science, Center of Medical Innovation and Translational Research, Osaka University Graduate School of Medicine, Yamadaoka 2-2, Suita, Osaka 565-0871, Japan
| | | | - Andrea Vecchione
- Department of Clinical and Molecular Medicine, University of Rome “Sapienza”, Santo Andrea Hospital, Via di Grottarossa, 1035, 00189 Rome, Italy
| | - Taroh Satoh
- Center for Cancer Genomics and Precision Medicine, Osaka University Hospital, Yamadaoka 2-2, Suita, Osaka 565-0871, Japan
| | - Hideshi Ishii
- Department of Medical Data Science, Center of Medical Innovation and Translational Research, Osaka University Graduate School of Medicine, Yamadaoka 2-2, Suita, Osaka 565-0871, Japan
| |
Collapse
|
37
|
Cheung TH, Shoichet MS. The Interplay of Endosomal Escape and RNA Release from Polymeric Nanoparticles. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2025; 41:7174-7190. [PMID: 40080875 DOI: 10.1021/acs.langmuir.4c05176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/15/2025]
Abstract
Ribonucleic acid (RNA) nanocarriers, specifically lipid nanoparticles and polymeric nanoparticles, enable RNA transfection both in vitro and in vivo; however, only a small percentage of RNA endocytosed by a cell is delivered to the cytosolic machinery, minimizing its effect. RNA nanocarriers face two major obstacles after endocytosis: endosomal escape and RNA release. Overcoming both obstacles simultaneously is challenging because endosomal escape is usually achieved by using high positive charge to disrupt the endosomal membrane. However, this high positive charge typically also inhibits RNA release because anionic RNA is strongly bound to the nanocarrier by electrostatic interactions. Many nanocarriers address one over the other despite a growing body of evidence demonstrating that both are crucial for RNA transfection. In this review, we survey the various strategies that have been employed to accomplish both endosomal escape and RNA release with a focus on polymeric nanomaterials. We first consider the various requirements a nanocarrier must achieve for RNA delivery including protection from degradation, cellular internalization, endosomal escape, and RNA release. We then discuss current polymers used for RNA delivery and examine the strategies for achieving both endosomal escape and RNA release. Finally, we review various stimuli-responsive strategies for RNA release. While RNA release continues to be a challenge in achieving efficient RNA transfection, many new innovations in polymeric materials have elucidated promising strategies.
Collapse
Affiliation(s)
- Timothy H Cheung
- Department of Chemistry, University of Toronto, 80 St. George Street, Toronto, Ontario M5S 3H6, Canada
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 160 College Street, Toronto, Ontario M5S 3E1, Canada
| | - Molly S Shoichet
- Department of Chemistry, University of Toronto, 80 St. George Street, Toronto, Ontario M5S 3H6, Canada
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 160 College Street, Toronto, Ontario M5S 3E1, Canada
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, Ontario M5S 3E5, Canada
- Institute of Biomedical Engineering, University of Toronto, 164 College Street, Toronto, Ontario M5S 3G9, Canada
| |
Collapse
|
38
|
Lindsay S, Hussain M, Binici B, Perrie Y. Exploring the Challenges of Lipid Nanoparticle Development: The In Vitro-In Vivo Correlation Gap. Vaccines (Basel) 2025; 13:339. [PMID: 40333261 PMCID: PMC12031360 DOI: 10.3390/vaccines13040339] [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: 01/30/2025] [Revised: 03/09/2025] [Accepted: 03/11/2025] [Indexed: 05/09/2025] Open
Abstract
BACKGROUND/OBJECTIVES The development of lipid nanoparticles (LNPs) as delivery platforms for nucleic acids has revolutionised possibilities for both therapeutic and vaccine applications. However, emerging studies highlight challenges in achieving reliable in vitro-in vivo correlation (IVIVC), which delays the translation of experimental findings into clinical applications. This study investigates these potential discrepancies by evaluating the physicochemical properties, in vitro efficacy (across three commonly used cell lines), and in vivo performance (mRNA expression and vaccine efficacy) of four LNP formulations. METHODS LNPs composed of DSPC, cholesterol, a PEGylated lipid, and one of four ionizable lipids (SM-102, ALC-0315, MC3, or C12-200) were manufactured using microfluidics. RESULTS All formulations exhibited comparable physicochemical properties, as expected (size 70-100 nm, low PDI, near-neutral zeta potential, and high mRNA encapsulation). In vitro studies demonstrated variable LNP-mediated mRNA expression in both immortalised and immune cells, with SM-102 inducing significantly higher protein expression (p < 0.05) than the other formulations in immortalised and immune cells. However, in vivo results revealed that ALC-0315 and SM-102-based LNPs achieved significantly (p < 0.05) higher protein expression without a significant difference between them, while MC3- and C12-200-based LNPs exhibited lower expression levels. As vaccine formulations, all LNPs elicited strong immune responses with no significant differences among them. CONCLUSIONS These findings highlight the complexities of correlating in vitro and in vivo outcomes in LNP development and demonstrate the importance of holistic evaluation strategies to optimise their clinical translation.
Collapse
Affiliation(s)
| | | | | | - Yvonne Perrie
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK; (S.L.); (M.H.); (B.B.)
| |
Collapse
|
39
|
Fernandes RS, de Assis Burle-Caldas G, Sergio SAR, Bráz AF, da Silva Leite NP, Pereira M, de Oliveira Silva J, Hojo-Souza NS, de Oliveira B, Fernandes APSM, da Fonseca FG, Gazzinelli RT, Dos Santos Ferreira D, Teixeira SMR. The immunogenic potential of an optimized mRNA lipid nanoparticle formulation carrying sequences from virus and protozoan antigens. J Nanobiotechnology 2025; 23:221. [PMID: 40102899 PMCID: PMC11921523 DOI: 10.1186/s12951-025-03201-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Accepted: 02/04/2025] [Indexed: 03/20/2025] Open
Abstract
BACKGROUND Lipid nanoparticles (LNP) are a safe and effective messenger RNA (mRNA) delivery system for vaccine applications, as shown by the COVID-19 mRNA vaccines. One of the main challenges faced during the development of these vaccines is the production of new and versatile LNP formulations capable of efficient encapsulation and delivery to cells in vivo. This study aimed to develop a new mRNA vaccine formulation that could potentially be used against existing diseases as well as those caused by pathogens that emerge every year. RESULTS Using firefly luciferase (Luc) as a reporter mRNA, we evaluated the physical-chemical properties, stability, and biodistribution of an LNP-mRNA formulation produced using a novel lipid composition and a microfluidic organic-aqueous precipitation method. Using mRNAs encoding a dengue virus or a Leishmania infantum antigen, we evaluated the immunogenicity of LNP-mRNA formulations and compared them with the immunization with the corresponding recombinant protein or plasmid-encoded antigens. For all tested LNP-mRNAs, mRNA encapsulation efficiency was higher than 85%, their diameter was around 100 nm, and their polydispersity index was less than 0.3. Following an intramuscular injection of 10 µg of the LNP-Luc formulation in mice, we detected luciferase activity in the injection site, as well as in the liver and spleen, as early as 6 h post-administration. LNPs containing mRNA encoding virus and parasite antigens were highly immunogenic, as shown by levels of antigen-specific IgG antibody as well as IFN-γ production by splenocytes of immunized animals that were similar to the levels that resulted from immunization with the corresponding recombinant protein or plasmid DNA. CONCLUSIONS Altogether, these results indicate that these novel LNP-mRNA formulations are highly immunogenic and may be used as novel vaccine candidates for different infectious diseases.
Collapse
Affiliation(s)
- Renata S Fernandes
- Centro de Tecnologia de Vacinas da, Universidade Federal de Minas Gerais, Belo Horizonte, Belo Horizonte, MG, 31310-260, Brazil
| | - Gabriela de Assis Burle-Caldas
- Centro de Tecnologia de Vacinas da, Universidade Federal de Minas Gerais, Belo Horizonte, Belo Horizonte, MG, 31310-260, Brazil
| | | | - Ana Flávia Bráz
- Centro de Tecnologia de Vacinas da, Universidade Federal de Minas Gerais, Belo Horizonte, Belo Horizonte, MG, 31310-260, Brazil
| | - Nathália Pereira da Silva Leite
- Centro de Tecnologia de Vacinas da, Universidade Federal de Minas Gerais, Belo Horizonte, Belo Horizonte, MG, 31310-260, Brazil
| | - Milton Pereira
- Centro de Tecnologia de Vacinas da, Universidade Federal de Minas Gerais, Belo Horizonte, Belo Horizonte, MG, 31310-260, Brazil
| | - Juliana de Oliveira Silva
- Centro de Tecnologia de Vacinas da, Universidade Federal de Minas Gerais, Belo Horizonte, Belo Horizonte, MG, 31310-260, Brazil
- Department of Pharmaceuticals, School of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, MG, 31270-901, Brazil
| | - Natália Satchiko Hojo-Souza
- Centro de Tecnologia de Vacinas da, Universidade Federal de Minas Gerais, Belo Horizonte, Belo Horizonte, MG, 31310-260, Brazil
- Instituto René Rachou, Fundação Oswaldo Cruz-Minas, Belo Horizonte, MG, 30190-002, Brazil
| | - Bianca de Oliveira
- Centro de Tecnologia de Vacinas da, Universidade Federal de Minas Gerais, Belo Horizonte, Belo Horizonte, MG, 31310-260, Brazil
| | - Ana Paula S Moura Fernandes
- Centro de Tecnologia de Vacinas da, Universidade Federal de Minas Gerais, Belo Horizonte, Belo Horizonte, MG, 31310-260, Brazil
- Department of Clinical & Toxicological Analysis, School of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, MG, 31270-901, Brazil
| | - Flávio Guimarães da Fonseca
- Centro de Tecnologia de Vacinas da, Universidade Federal de Minas Gerais, Belo Horizonte, Belo Horizonte, MG, 31310-260, Brazil
- Department of Microbiology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, 31270-901, Brazil
| | - Ricardo Tostes Gazzinelli
- Centro de Tecnologia de Vacinas da, Universidade Federal de Minas Gerais, Belo Horizonte, Belo Horizonte, MG, 31310-260, Brazil
- Instituto René Rachou, Fundação Oswaldo Cruz-Minas, Belo Horizonte, MG, 30190-002, Brazil
- Department of Biochemistry & Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, 31270-901, Brazil
| | - Diego Dos Santos Ferreira
- Centro de Tecnologia de Vacinas da, Universidade Federal de Minas Gerais, Belo Horizonte, Belo Horizonte, MG, 31310-260, Brazil
- Department of Pharmaceuticals, School of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, MG, 31270-901, Brazil
| | - Santuza M Ribeiro Teixeira
- Centro de Tecnologia de Vacinas da, Universidade Federal de Minas Gerais, Belo Horizonte, Belo Horizonte, MG, 31310-260, Brazil.
- Department of Biochemistry & Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, 31270-901, Brazil.
| |
Collapse
|
40
|
Alshehry Y, Liu X, Li W, Wang Q, Cole J, Zhu G. Lipid Nanoparticles for mRNA Delivery in Cancer Immunotherapy. AAPS J 2025; 27:66. [PMID: 40102316 DOI: 10.1208/s12248-025-01051-8] [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: 01/02/2025] [Accepted: 02/23/2025] [Indexed: 03/20/2025] Open
Abstract
Cancer immunotherapy is poised to be one of the major modalities for cancer treatment. Messenger RNA (mRNA) has emerged as a versatile and promising platform for the development of effective cancer immunotherapy. Delivery systems for mRNA therapeutics are pivotal for their optimal therapeutic efficacy and minimal adverse side effects. Lipid nanoparticles (LNPs) have demonstrated a great success for mRNA delivery. Numerous LNPs have been designed and optimized to enhance mRNA stability, facilitate transfection, and ensure intracellular delivery for subsequent processing. Nevertheless, challenges remain to, for example, improve the efficiency of endosomal escape and passive targeting. This review highlights key advancements in the development of mRNA LNPs for cancer immunotherapy. We delve into the design of LNPs for mRNA delivery, encompassing the chemical structures, characterization, and structure-activity relationships (SAR) of LNP compositions. We discuss the key factors influencing the transfection efficiency, passive targeting, and tropism of mRNA-loaded LNPs. We also review the preclinical and clinical applications of mRNA LNPs in cancer immunotherapy. This review can enhance our understanding in the design and application of LNPs for mRNA delivery in cancer immunotherapy.
Collapse
Affiliation(s)
- Yasir Alshehry
- Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, 23298, United States of America
- Department of Pharmaceutics, College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, 31441, Dammam, Saudi Arabia
| | - Xiang Liu
- Department of Pharmaceutical Sciences, College of Pharmacy, Biointerfaces Institute, University of Michigan, Ann Arbor, MI, 48109, United States of America
| | - Wenhua Li
- Department of Pharmaceutical Sciences, College of Pharmacy, Biointerfaces Institute, University of Michigan, Ann Arbor, MI, 48109, United States of America
| | - Qiyan Wang
- Department of Pharmaceutical Sciences, College of Pharmacy, Biointerfaces Institute, University of Michigan, Ann Arbor, MI, 48109, United States of America
| | - Janét Cole
- Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, 23298, United States of America
| | - Guizhi Zhu
- Department of Pharmaceutical Sciences, College of Pharmacy, Biointerfaces Institute, University of Michigan, Ann Arbor, MI, 48109, United States of America.
- Bioinnovations in Brain Cancer, Biointerfaces Institute, Rogel Cancer Center, Center for RNA Biomedicine, University of Michigan, Ann Arbor, MI, 48109, United States of America.
| |
Collapse
|
41
|
Banda O, Adams SE, Omer L, Jung SK, Said H, Phoka T, Tam Y, Weissman D, Rivella S, Alameh MG, Kurre P. Restoring hematopoietic stem and progenitor cell function in Fancc -/- mice by in situ delivery of RNA lipid nanoparticles. MOLECULAR THERAPY. NUCLEIC ACIDS 2025; 36:102423. [PMID: 39811495 PMCID: PMC11730543 DOI: 10.1016/j.omtn.2024.102423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 12/10/2024] [Indexed: 01/16/2025]
Abstract
Fanconi anemia (FA) is a congenital multisystem disorder characterized by early-onset bone marrow failure (BMF) and cancer susceptibility. While ex vivo gene addition and repair therapies are being considered as treatment options, depleted hematopoietic stem cell (HSC) pools, poor HSC mobilization, compromised survival during ex vivo transduction, and increased sensitivity to conventional conditioning strategies limit eligibility for FA patients to receive gene therapies. As an alternative approach, we explored in vivo protein replacement by mRNA delivery via lipid nanoparticles (LNPs). Our study aims to address several key obstacles to current mRNA-LNP treatment: access to the HSC niche, effective expression half-life, and potential mRNA LNP immunogenicity. Results demonstrate efficient in vivo LNP transfection of murine BM via intravenous or intrafemoral injections, yielding reporter expression across hematopoietic and non-hematopoietic BM niche populations. Functionally, LNP delivery of modified Fancc mRNA restored ex vivo expansion. In a proof of principle approach, LNP-treated murine Fancc -/- HSPCs engrafted with restored alkylator resistance up to 120 h post-treatment using circularized mRNA constructs. In vitro delivery of mRNA LNPs resulted in modest differences in innate immune target gene expression in both FA and wild-type HSPCs. Our results suggest that mRNA-LNP-based protein replacement therapy holds promise for clinical translation.
Collapse
Affiliation(s)
- Omar Banda
- Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Penn Institute for RNA Innovation, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sarah E. Adams
- Comprehensive Bone Marrow Failure Center, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Linah Omer
- Comprehensive Bone Marrow Failure Center, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Seul K. Jung
- Comprehensive Bone Marrow Failure Center, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Hooda Said
- Department of Bioengineering, George Mason University, Fairfax, VA 22030, USA
| | - Theerapat Phoka
- Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ying Tam
- Acuitas Therapeutics, Vancouver, BC, Canada
| | - Drew Weissman
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Penn Institute for RNA Innovation, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Stefano Rivella
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Pediatrics, Division of Hematology, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Penn Institute for RNA Innovation, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mohamad-Gabriel Alameh
- Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Penn Institute for RNA Innovation, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Peter Kurre
- Comprehensive Bone Marrow Failure Center, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
42
|
Baimanov D, Wang J, Liu Y, Zheng P, Yu S, Liu F, Wang J, Boraschi D, Zhao Y, Chen C, Wang L. Identification of Cell Receptors Responsible for Recognition and Binding of Lipid Nanoparticles. J Am Chem Soc 2025; 147:7604-7616. [PMID: 39993835 DOI: 10.1021/jacs.4c16987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/26/2025]
Abstract
Effective delivery of lipid nanoparticles (LNPs) and their organ- or cell-type targeting are paramount for therapeutic success. Achieving this requires a comprehensive understanding of protein corona dynamics and the identification of cell receptors involved in the recognition and uptake of LNPs. We introduce a simple, fast, and in situ strategy by a biosensor-based "Fishing" method to uncover protein corona formation on LNPs and identify key receptors of human blood cells that are responsible for the recognition and binding of human plasma corona on the surface of LNPs. Unexpectedly, we observed a significant presence of immunoglobulins with high abundance, especially anti-PEG antibodies, within the LNP corona. These antibodies, along with complement opsonization, drive colony-stimulating factor 2 receptor β (CSF2RB)-mediated phagocytosis by human myeloid cells. These compositions of the human plasma corona and their interactions with neighboring proteins are critical for the recognition and binding of LNPs by cell receptors and cellular uptake. Our findings highlight the pivotal role of anti-PEG antibodies in the circulation and phagocytosis of LNPs in vivo. This approach offers profound insights into nanomaterial behavior in vivo, paving the way for the enhanced design and efficacy of LNP-based therapies.
Collapse
Affiliation(s)
- Didar Baimanov
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, New Cornerstone Science Laboratory, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100049, P. R. China
| | - Jing Wang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, P. R. China
- Peking University Ningbo Institute of Marine Medicines, Ningbo 315832, P. R. China
| | - Yuchen Liu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, P. R. China
| | - Pingping Zheng
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, New Cornerstone Science Laboratory, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100049, P. R. China
| | - Shengtao Yu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, New Cornerstone Science Laboratory, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100049, P. R. China
| | - Fen Liu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, P. R. China
| | - Jian Wang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, P. R. China
| | - Diana Boraschi
- Laboratory of Inflammation and Vaccines, China-Italy Joint Laboratory of Pharmacobiotechnology for Medical Immunomodulation, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, P. R. China
| | - Yuliang Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, New Cornerstone Science Laboratory, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100049, P. R. China
- GBA Research Innovation Institute for Nanotechnology, Guangzhou 510700, Guangdong, P. R. China
- Research Unit of Nanoscience and Technology, Chinese Academy of Medical Sciences, Beijing 100730, P. R. China
| | - Chunying Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, New Cornerstone Science Laboratory, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100049, P. R. China
- GBA Research Innovation Institute for Nanotechnology, Guangzhou 510700, Guangdong, P. R. China
- Research Unit of Nanoscience and Technology, Chinese Academy of Medical Sciences, Beijing 100730, P. R. China
| | - Liming Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, New Cornerstone Science Laboratory, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100049, P. R. China
| |
Collapse
|
43
|
Hong X, Chen T, Liu Y, Li J, Huang D, Ye K, Liao W, Wang Y, Liu M, Luan P. Design, current states, and challenges of nanomaterials in anti-neuroinflammation: A perspective on Alzheimer's disease. Ageing Res Rev 2025; 105:102669. [PMID: 39864562 DOI: 10.1016/j.arr.2025.102669] [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: 12/04/2024] [Revised: 01/08/2025] [Accepted: 01/21/2025] [Indexed: 01/28/2025]
Abstract
Alzheimer's disease (AD), an age-related neurodegenerative disease, brings huge damage to the society, to the whole family and even to the patient himself. However, until now, the etiological factor of AD is still unknown and there is no effective treatment for it. Massive deposition of amyloid-beta peptide(Aβ) and hyperphosphorylation of Tau proteins are acknowledged pathological features of AD. Recent studies have revealed that neuroinflammation plays a pivotal role in the pathology of AD. With the rise of nanomaterials in the biomedical field, researchers are exploring how the unique properties of these materials can be leveraged to develop effective treatments for AD. This article has summarized the influence of neuroinflammation in AD, the design of nanoplatforms, and the current research status and inadequacy of nanomaterials in improving neuroinflammation in AD.
Collapse
Affiliation(s)
- Xinyang Hong
- Department of Alzheimer's Disease Clinical Research Center, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, China.
| | - Tongkai Chen
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China.
| | - Yunyun Liu
- Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-sen University), Ministry of Education, Guangzhou, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Department of Neurology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Jun Li
- Department of Alzheimer's Disease Clinical Research Center, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, China.
| | - Dongqing Huang
- Department of Alzheimer's Disease Clinical Research Center, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, China.
| | - Kaiyu Ye
- Department of Alzheimer's Disease Clinical Research Center, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, China.
| | - Wanchen Liao
- Department of Alzheimer's Disease Clinical Research Center, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, China.
| | - Yulin Wang
- Department of Alzheimer's Disease Clinical Research Center, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, China.
| | - Mengling Liu
- Department of Alzheimer's Disease Clinical Research Center, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, China.
| | - Ping Luan
- Department of Alzheimer's Disease Clinical Research Center, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, China; School of Basic Medical Sciences, Shenzhen University, Shenzhen 518060, China.
| |
Collapse
|
44
|
Fatima M, An T, Hong KJ. Revolutionizing mRNA Vaccines Through Innovative Formulation and Delivery Strategies. Biomolecules 2025; 15:359. [PMID: 40149895 PMCID: PMC11940278 DOI: 10.3390/biom15030359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 02/12/2025] [Accepted: 02/19/2025] [Indexed: 03/29/2025] Open
Abstract
Modernization of existing methods for the delivery of mRNA is vital in advanced therapeutics. Traditionally, mRNA has faced obstacles of poor stability due to enzymatic degradation. This work examines cutting-edge formulation and emerging techniques for safer delivery of mRNA vaccines. Inspired by the success of lipid nanoparticles (LNP) in delivering mRNA vaccines for COVID-19, a variety of other formulations have been developed to deliver mRNA vaccines for diverse infections. The meritorious features of nanoparticle-based mRNA delivery strategies, including LNP, polymeric, dendrimers, polysaccharide-based, peptide-derived, carbon and metal-based, DNA nanostructures, hybrid, and extracellular vesicles, have been examined. The impact of these delivery platforms on mRNA vaccine delivery efficacy, protection from enzymatic degradation, cellular uptake, controlled release, and immunogenicity has been discussed in detail. Even with significant developments, there are certain limitations to overcome, including toxicity concerns, limited information about immune pathways, the need to maintain a cold chain, and the necessity of optimizing administration methods. Continuous innovation is essential for improving delivery systems for mRNA vaccines. Future research directions have been proposed to address the existing challenges in mRNA delivery and to expand their potential prophylactic and therapeutic application.
Collapse
Affiliation(s)
- Munazza Fatima
- Department of Microbiology, Gachon University College of Medicine, Incheon 21936, Republic of Korea;
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Republic of Korea
| | - Timothy An
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Republic of Korea
| | - Kee-Jong Hong
- Department of Microbiology, Gachon University College of Medicine, Incheon 21936, Republic of Korea;
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Republic of Korea
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Republic of Korea
- Korea mRNA Vaccine Initiative, Gachon University, Seongnam 13120, Republic of Korea
| |
Collapse
|
45
|
Shi Y, Mao J, Wang S, Ma S, Luo L, You J. Pharmaceutical strategies for optimized mRNA expression. Biomaterials 2025; 314:122853. [PMID: 39342919 DOI: 10.1016/j.biomaterials.2024.122853] [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: 09/19/2024] [Accepted: 09/26/2024] [Indexed: 10/01/2024]
Abstract
Messenger RNA (mRNA)-based immunotherapies and protein in situ production therapies hold great promise for addressing theoretically all the diseases characterized by aberrant protein levels. The safe, stable, and precise delivery of mRNA to target cells via appropriate pharmaceutical strategies is a prerequisite for its optimal efficacy. In this review, we summarize the structural characteristics, mode of action, development prospects, and limitations of existing mRNA delivery systems from a pharmaceutical perspective, with an emphasis on the impacts from formulation adjustments and preparation techniques of non-viral vectors on mRNA stability, target site accumulation and transfection efficiency. In addition, we introduce strategies for synergistical combination of mRNA and small molecules to augment the potency or mitigate the adverse effects of mRNA therapeutics. Lastly, we delve into the challenges impeding the development of mRNA drugs while exploring promising avenues for future advancements.
Collapse
Affiliation(s)
- Yingying Shi
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, 310058, PR China
| | - Jiapeng Mao
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, 310058, PR China
| | - Sijie Wang
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, 310058, PR China
| | - Siyao Ma
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, 166 Qiutaobei Road, Hangzhou, Zhejiang, 310017, PR China
| | - Lihua Luo
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, 310058, PR China.
| | - Jian You
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, 310058, PR China; State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, 79 Qingchun Road, Shangcheng District, Hangzhou, Zhejiang, 310006, PR China; The First Affiliated Hospital, College of Medicine, Zhejiang University, 79 QingChun Road, Hangzhou, Zhejiang, 310000, PR China; Jinhua Institute of Zhejiang University, 498 Yiwu Street, Jinhua, Zhejiang, 321299, PR China.
| |
Collapse
|
46
|
Zhang Y, Lian C, Lai W, Jiang L, Xing Y, Liang H, Li J, Zhang X, Gan J, Li Z, Yin F. Programmable Stapling Peptide Based on Sulfonium as Universal Vaccine Adjuvants for Multiple Types of Vaccines. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2409567. [PMID: 39878394 PMCID: PMC11923873 DOI: 10.1002/advs.202409567] [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: 08/13/2024] [Revised: 12/22/2024] [Indexed: 01/31/2025]
Abstract
Adjuvants are non-specific immune enhancers commonly used to improve the responsiveness and persistence of the immune system toward antigens. However, due to the undefined chemical structure, toxicity, non-biodegradability, and lack of design technology in many existing adjuvants, it remains difficult to achieve substantive breakthroughs in the adjuvant research field. Here, a novel adjuvant development strategy based on stapling peptides is reported to overcome this challenge. The nano-vaccine incorporating peptide adjuvant and recombinant HBsAg protein not only induced strong antibody titers that are equivalent to aluminum adjuvanted vaccines but also simultaneously activated T-cell immune response. Similar results are also observed in herpes zoster vaccine and more complex influenza vaccine. The mechanism analysis demonstrates that antigen is efficiently carried into antigen-presenting cells (APCs) by peptide, further promoting the secretion of cytokines and activation of APCs. In addition, by redesigning the adjuvant, it is found that the sulfonium centers, rather than the sequence of peptide played an important role in immune activation. This discovery may provide a new paradigm for the rational design of peptide-based adjuvants. In brief, this study demonstrates that stapling peptides with sulfonium centers can provide a well-defined, programmable, biocompatible, and effective adjuvant for multiple types of vaccines.
Collapse
Affiliation(s)
- Yaping Zhang
- State Key Laboratory of Chemical OncogenomicsSchool of Chemical Biology and BiotechnologyPeking University Shenzhen Graduate SchoolShenzhen518055P. R. China
- Pingshan Translational Medicine CenterShenzhen Bay LaboratoryShenzhen518118P. R. China
| | - Chenshan Lian
- State Key Laboratory of Chemical OncogenomicsSchool of Chemical Biology and BiotechnologyPeking University Shenzhen Graduate SchoolShenzhen518055P. R. China
| | - Wenlong Lai
- Shenzhen Kangtai Biological Products Co. Ltd.Shenzhen518057P. R. China
| | - Leying Jiang
- State Key Laboratory of Chemical OncogenomicsSchool of Chemical Biology and BiotechnologyPeking University Shenzhen Graduate SchoolShenzhen518055P. R. China
| | - Yun Xing
- State Key Laboratory of Chemical OncogenomicsSchool of Chemical Biology and BiotechnologyPeking University Shenzhen Graduate SchoolShenzhen518055P. R. China
| | - Huiting Liang
- Pingshan Translational Medicine CenterShenzhen Bay LaboratoryShenzhen518118P. R. China
| | - Jin Li
- Shenzhen Kangtai Biological Products Co. Ltd.Shenzhen518057P. R. China
| | - Xinming Zhang
- Beijing Minhai Biotechnology Co. Ltd.Beijing102609P. R. China
| | - Jianhui Gan
- Shenzhen Kangtai Biological Products Co. Ltd.Shenzhen518057P. R. China
| | - Zigang Li
- State Key Laboratory of Chemical OncogenomicsSchool of Chemical Biology and BiotechnologyPeking University Shenzhen Graduate SchoolShenzhen518055P. R. China
- Pingshan Translational Medicine CenterShenzhen Bay LaboratoryShenzhen518118P. R. China
| | - Feng Yin
- State Key Laboratory of Chemical OncogenomicsSchool of Chemical Biology and BiotechnologyPeking University Shenzhen Graduate SchoolShenzhen518055P. R. China
- Pingshan Translational Medicine CenterShenzhen Bay LaboratoryShenzhen518118P. R. China
| |
Collapse
|
47
|
Imani S, Lv S, Qian H, Cui Y, Li X, Babaeizad A, Wang Q. Current innovations in mRNA vaccines for targeting multidrug-resistant ESKAPE pathogens. Biotechnol Adv 2025; 79:108492. [PMID: 39637949 DOI: 10.1016/j.biotechadv.2024.108492] [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: 08/19/2024] [Revised: 10/30/2024] [Accepted: 11/28/2024] [Indexed: 12/07/2024]
Abstract
The prevalence of multidrug-resistant (MDR) ESKAPE pathogens, including Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, and Pseudomonas aeruginosa, represents a critical global public health challenge. In response, mRNA vaccines offer an adaptable and scalable platform for immunotherapy against ESKAPE pathogens by encoding specific antigens that stimulate B-cell-driven antibody production and CD8+ T-cell-mediated cytotoxicity, effectively neutralizing these pathogens and combating resistance. This review examines recent advancements and ongoing challenges in the development of mRNA vaccines targeting MDR ESKAPE pathogens. We explore antigen selection, the nuances of mRNA vaccine technology, and the complex interactions between bacterial infections and antibiotic resistance. By assessing the potential efficacy of mRNA vaccines and addressing key barriers to their paraclinical implementation, this review highlights the promising function of mRNA-based immunization in combating MDR ESKAPE pathogens.
Collapse
Affiliation(s)
- Saber Imani
- Key Laboratory of Artificial Organs and Computational Medicine of Zhejiang Province, Key Laboratory of Pollution Exposure and Health Intervention of Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, Zhejiang, China
| | - Shuojie Lv
- Key Laboratory of Artificial Organs and Computational Medicine of Zhejiang Province, Key Laboratory of Pollution Exposure and Health Intervention of Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, Zhejiang, China
| | - Hongbo Qian
- Key Laboratory of Artificial Organs and Computational Medicine of Zhejiang Province, Key Laboratory of Pollution Exposure and Health Intervention of Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, Zhejiang, China
| | - Yulan Cui
- Key Laboratory of Artificial Organs and Computational Medicine of Zhejiang Province, Key Laboratory of Pollution Exposure and Health Intervention of Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, Zhejiang, China
| | - XiaoYan Li
- Key Laboratory of Artificial Organs and Computational Medicine of Zhejiang Province, Key Laboratory of Pollution Exposure and Health Intervention of Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, Zhejiang, China
| | - Ali Babaeizad
- Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Qingjing Wang
- Key Laboratory of Artificial Organs and Computational Medicine of Zhejiang Province, Key Laboratory of Pollution Exposure and Health Intervention of Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, Zhejiang, China.
| |
Collapse
|
48
|
Baharom F, Hermans D, Delamarre L, Seder RA. Vax-Innate: improving therapeutic cancer vaccines by modulating T cells and the tumour microenvironment. Nat Rev Immunol 2025; 25:195-211. [PMID: 39433884 DOI: 10.1038/s41577-024-01091-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/02/2024] [Indexed: 10/23/2024]
Abstract
T cells have a critical role in mediating antitumour immunity. The success of immune checkpoint inhibitors (ICIs) for cancer treatment highlights how enhancing endogenous T cell responses can mediate tumour regression. However, mortality remains high for many cancers, especially in the metastatic setting. Based on advances in the genetic characterization of tumours and identification of tumour-specific antigens, individualized therapeutic cancer vaccines targeting mutated tumour antigens (neoantigens) are being developed to generate tumour-specific T cells for improved therapeutic responses. Early clinical trials using individualized neoantigen vaccines for patients with advanced disease had limited clinical efficacy despite demonstrated induction of T cell responses. Therefore, enhancing T cell activity by improving the magnitude, quality and breadth of T cell responses following vaccination is one current goal for improving outcome against metastatic tumours. Another major consideration is how T cells can be further optimized to function within the tumour microenvironment (TME). In this Perspective, we focus on neoantigen vaccines and propose a new approach, termed Vax-Innate, in which vaccination through intravenous delivery or in combination with tumour-targeting immune modulators may improve antitumour efficacy by simultaneously increasing the magnitude, quality and breadth of T cells while transforming the TME into a largely immunostimulatory environment for T cells.
Collapse
Affiliation(s)
| | - Dalton Hermans
- Vaccine Research Center, National Institutes of Health, Bethesda, MD, USA
| | | | - Robert A Seder
- Vaccine Research Center, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
49
|
Kirtane AR, Traverso G. Improving the Efficacy of Cancer mRNA Vaccines. Cancer J 2025; 31:e0764. [PMID: 40126883 DOI: 10.1097/ppo.0000000000000764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Accepted: 02/05/2025] [Indexed: 03/26/2025]
Abstract
mRNA vaccines consist of antigen-encoding mRNA, which produces the antigenic protein upon translation. Coupling antigen production with innate immune activation can generate a potent, antigen-specific T-cell response. Clinical reports have demonstrated the ability of mRNA vaccines to elicit an anticancer immune response against various tumor types. Here, we discuss strategies to enhance the potency of mRNA vaccines. We provide an overview of existing knowledge regarding the activation and trafficking mechanisms of mRNA vaccines and share optimization strategies to boost mRNA-mediated antigen production. In addition, we address methods to target mRNA vaccines to dendritic cells and lymph nodes, key initiators of the immune response. Finally, we review strategies for enhancing immune activation using adjuvants compatible with mRNA vaccines. mRNA vaccines offer unique advantages that can be utilized for oncology applications. However, significant work is needed to understand their underlying mechanisms and develop technologies to improve their effectiveness.
Collapse
Affiliation(s)
- Ameya R Kirtane
- Department of Pharmaceutics, University of Minnesota, Minneapolis, MN
| | - Giovanni Traverso
- Department of Mechanical Engineering, Massachusetts Institute of Technology
- Broad Institute, Massachusetts Institute of Technology, Cambridge, MA
- Department of Gastroenterology, Hepatology and Endoscopy, Brigham and Women's Hospital, Boston, MA
| |
Collapse
|
50
|
Wang X, Yang X, Zhang X, Yan H, Jin J, Ma Z, Duan J, Zhang G, Huang T, Li Y, Wu H, Zhang T, Zhu A, Jin C, Song X, Su B. Dynamic SARS-CoV-2-specific B-cell and T-cell responses induced in people living with HIV after a full course of inactivated SARS-CoV-2 vaccine. Front Immunol 2025; 16:1554409. [PMID: 40070834 PMCID: PMC11893571 DOI: 10.3389/fimmu.2025.1554409] [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: 01/02/2025] [Accepted: 02/04/2025] [Indexed: 03/14/2025] Open
Abstract
Objective Both B-cell- and T-cell-mediated immunity are crucial for the effective clearance of viral infection, but little is known about the dynamic characteristics of SARS-CoV-2-specific B-cell and T-cell responses in people living with HIV (PLWH) after a full course of inactivated SARS-CoV-2 vaccination. Methods In this study, fifty people living with HIV (PLWH) and thirty healthy controls (HCs) were enrolled to assess B-cell and T-cell responses at the day before the vaccination (T0), two weeks after the first dose (T1), two months after the first dose (T2), the day of the third dose (T3), one month after the third dose (T4), three months after the third dose (T5) and 12 months (T6) after the third dose. Results SARS-CoV-2-specific B-cell and T-cell responses were induced in people living with HIV (PLWH), and these responses lasted at least one year after the third vaccine dose. However, the peak frequencies of Spike-specific B-cell and T-cell responses in PLWH were lower than those in HIV-negative controls. In addition, the expansion of activated B cells, memory B cells and plasma cells after primary vaccination was observed, but the percentages of these cells were decreased at T6 and were comparable to those at T0. Additionally, the percentages of activated T cells, exhausted T cells and SARS-CoV-2-specific T cells with enhanced functional activity were increased following the administration of inactivated SARS-CoV-2 vaccine. In addition, PLWH had lower percentages of plasma cells, RBD-specific B cells, circulating Tfh (cTfh) cells and CD38+ cTfh cells, and the percentages of the latter two types of cells were positively correlated with the titer of neutralizing antibodies, indicating these differences may account for the weaker immune responses induced in PLWH. Conclusion These data suggest that specific B-cell and T-cell responses could be sustained for at least one year after receiving the third vaccination. Our findings emphasize that the weak SARS-CoV-2-specific B-cell and T-cell responses induced in PLWH have implications for clinical decision-making and public health policy for PLWH with respect to SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Xiuwen Wang
- Beijing Key Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
- Center for General Practice Medicine, Department of Rheumatology and Immunology, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Xiaodong Yang
- Department of Infectious Diseases, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Xin Zhang
- Beijing Key Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Hongxia Yan
- Beijing Key Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Junyan Jin
- Beijing Key Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Zhenglai Ma
- Beijing Key Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Junyi Duan
- Tian Yuan Studio, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Guanghui Zhang
- Tian Yuan Studio, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Tao Huang
- Tian Yuan Studio, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Yongzheng Li
- Department of Pathogeny Biology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| | - Hao Wu
- Beijing Key Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Tong Zhang
- Beijing Key Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Aiwei Zhu
- Beijing Key Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Cong Jin
- National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Xiangrong Song
- Department of Critical Care Medicine, Frontiers Science Center for Disease related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Bin Su
- Beijing Key Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
- Central Laboratory, Beijing Youan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|