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 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] [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
|
Zhang T, Jian Z, Chen J, Xu D, Yang X, Lu Y, Yan S, Pan L, Wu Q, Ouyang Z. Efficiency enhancement in main path extraction in mRNA vaccine field: A novel approach leveraging intermediate patents, with shielding origin and terminus patent edges. Hum Vaccin Immunother 2025; 21:2454078. [PMID: 39838876 DOI: 10.1080/21645515.2025.2454078] [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: 10/16/2024] [Revised: 12/24/2024] [Accepted: 01/13/2025] [Indexed: 01/23/2025] Open
Abstract
mRNA vaccines offer groundbreaking technological advantages and broad application potential. Their rapid advancement, particularly during the COVID-19 pandemic, is the result of decades of research and numerous technological breakthroughs. These discoveries build upon each other, forming dense, interconnected networks of progress. Studying the technological development paths of mRNA vaccines is therefore essential. Main path analysis (MPA) is particularly effective for mapping out development trajectories within complex and interconnected networks, which serves as a powerful tool for identifying key nodes and innovations. This study introduces a novel approach to extracting main paths from a patent citation network in the mRNA vaccine field. Initially, we shielded edges connecting the origin and terminus patents. Subsequently, we extracted the main paths from intermediate patents, and then, we reintegrated the edges connecting the origin and terminus patents based on the citation relationships, resulting in a comprehensive extraction of the main paths. The research findings indicate a consistency among the global main paths, global key-route main paths, local forward main paths, and local key-route main paths within the mRNA vaccine field. The patents on the main paths predominantly focus on nucleic acid modifications and delivery systems. The local backward main paths identify a greater number of patents, especially those related to litigation, offering a richer and more diverse set of technological insights. This study significantly advances the methodology of MPA, with the innovative shielding technique offering a fresh perspective for navigating complex networks and providing a deeper understanding of technological development in the mRNA vaccine domain.
Collapse
Affiliation(s)
- Ting Zhang
- Institute of Medical Information & Library, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Zhongquan Jian
- Institute of Artificial Intelligence, Xiamen University, Xiamen, Fujian, China
| | - Juan Chen
- Institute of Medical Information & Library, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Dongzi Xu
- Institute of Medical Information & Library, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Xiaoyi Yang
- Institute of Medical Information & Library, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Yan Lu
- Institute of Medical Information & Library, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Shu Yan
- Institute of Medical Information & Library, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Lizi Pan
- Institute of Medical Information & Library, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | | | - Zhaolian Ouyang
- Institute of Medical Information & Library, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| |
Collapse
|
3
|
Gambari R, Papi C, Gasparello J, Agostinelli E, Finotti A. Preliminary results and a theoretical perspective of co‑treatment using a miR‑93‑5p mimic and aged garlic extract to inhibit the expression of the pro‑inflammatory interleukin‑8 gene. Exp Ther Med 2025; 29:85. [PMID: 40084194 PMCID: PMC11904878 DOI: 10.3892/etm.2025.12835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 01/07/2025] [Indexed: 03/16/2025] Open
Abstract
The coronavirus disease-19 (COVID-19) pandemic has been a very significant health issue in the period between 2020 and 2023, forcing research to characterize severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) sequences and to develop novel therapeutic approaches. Interleukin-6 (IL-6) and IL-8 are considered significant therapeutic targets for COVID-19 and emerging evidence has suggested that microRNAs (miRNAs/miRs) serve a key role in regulating these genes. MiRNAs are short, 19-25 nucleotides in length, non-coding RNAs that regulate gene expression at the post-transcriptional level through the sequence-selective recognition of the 3'-untranslated region (3'-UTR) of the regulated mRNAs, eventually repressing translation, commonly, via mRNA degradation. For example, among several miRNAs involved in the regulation of the COVID-19 'cytokine storm', miR-93-5p can inhibit IL-8 gene expression by directly targeting the 3'-UTR of IL-8 mRNA. In addition, miR-93-5p can regulate Toll-like receptor-4 (TLR4) and interleukin-1 receptor-associated kinase 4 (IRAK4) expression, thus affecting the nuclear factor-κB (NF-κB) pathway and the expression of NF-κB-regulated genes, such as IL-6, IL-1β and other hyper-expressed genes during the COVID-19 'cytokine storm'. In the present study, the results provided preliminary evidence suggesting that the miR-93-5p-based miRNA therapeutics could be combined with the anti-inflammatory aged garlic extract (AGE) to more effectively inhibit IL-8 gene expression. The human bronchial epithelial IB3-1 cell line was employed as experimental model system. IB3-1 cells were stimulated with the BNT162b2 COVID-19 vaccine and transfected with pre-miR-93-5p in the absence or in the presence of AGE, to verify the inhibitory effects on the BNT162b2-induced expression of the IL-8 gene. The accumulation of IL-8 mRNA was assessed by RT-qPCR; the release of IL-8 protein was determined by Bio-Plex assay. In addition, the possible applications of TLR4/NF-κB inhibitory agents (such as miR-93-5p and AGE) for treating human pathologies at a hyperinflammatory state, such as COVID-19, cystic fibrosis and other respiratory diseases, were summarized.
Collapse
Affiliation(s)
- Roberto Gambari
- Department of Life Sciences and Biotechnology, University of Ferrara, I-44121 Ferrara, Italy
| | - Chiara Papi
- Department of Life Sciences and Biotechnology, University of Ferrara, I-44121 Ferrara, Italy
| | - Jessica Gasparello
- Department of Life Sciences and Biotechnology, University of Ferrara, I-44121 Ferrara, Italy
| | - Enzo Agostinelli
- Department of Sensory Organs, Sapienza University of Rome, Policlinico Umberto I, I-00161 Rome, Italy
- International Polyamines Foundation ‘ETS-ONLUS’, I-00159 Rome, Italy
| | - Alessia Finotti
- Department of Life Sciences and Biotechnology, University of Ferrara, I-44121 Ferrara, Italy
| |
Collapse
|
4
|
Ding YH, Song XH, Chen JS. CircRNAs in Colorectal Cancer: Unveiling Their Roles and Exploring Therapeutic Potential. Biochem Genet 2025; 63:1219-1240. [PMID: 40029586 DOI: 10.1007/s10528-025-11068-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Accepted: 02/18/2025] [Indexed: 03/05/2025]
Abstract
Colorectal cancer (CRC) is the most common malignancy of the digestive system. Although research into the causes of CRC's origin and progression has advanced over the past few decades, many details are still not fully understood. Circular RNAs (circRNAs), as a novel regulatory molecule, have been found to be closely involved in various key biological processes in CRC. CircRNAs also have been shown to encode proteins, which could offer new possibilities for therapeutic applications. This ability to produce tumor-specific proteins makes circRNA-based vaccines a potentially valuable approach for targeted cancer treatment. In this review, we summarize recent findings on the various roles of circRNAs in CRC and explore their potential in the development of protein-encoding circRNA vaccines for CRC therapy.
Collapse
Affiliation(s)
- Yi-Han Ding
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, People's Republic of China
| | - Xiao-Hang Song
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, People's Republic of China
| | - Jing-Song Chen
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, People's Republic of China.
| |
Collapse
|
5
|
Hildt E. [Platform technologies in vaccine development]. Bundesgesundheitsblatt Gesundheitsforschung Gesundheitsschutz 2025; 68:368-377. [PMID: 40035793 PMCID: PMC11950034 DOI: 10.1007/s00103-025-04024-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 02/03/2025] [Indexed: 03/06/2025]
Abstract
Platform technologies in the narrower sense refer to approaches to vaccine development in which the vaccine is always based on a consistently identical framework and differs only in terms of the antigen. One advantage of platform technologies is their rapid adaptability for the development of a vaccine against novel pathogens or variants. Currently approved vaccines in the EU use viral vectors and mRNA as platforms. Recombinant adenoviruses (Ad), vesicular stomatitis virus (VSV), and modified vaccinia virus Ankara (MVA) serve as viral vectors. The application of mRNA-based vaccines is carried out in the form of lipid nanoparticles (LNPs). The function of the LNPs is to protect the mRNA from degradation, promote the uptake of the mRNA into the cells, and provide an adjuvant effect.
Collapse
Affiliation(s)
- Eberhard Hildt
- Paul-Ehrlich-Institut, Paul-Ehrlich-Str. 51-59, 63225, Langen (Hessen), Deutschland.
| |
Collapse
|
6
|
Liu Y, Liu Q, Zhang B, Chen S, Shen Y, Li Z, Zhang J, Yang Y, Li M, Wang Y. Generation of tolerogenic antigen-presenting cells in vivo via the delivery of mRNA encoding PDL1 within lipid nanoparticles. Nat Biomed Eng 2025:10.1038/s41551-025-01373-0. [PMID: 40155762 DOI: 10.1038/s41551-025-01373-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 02/27/2025] [Indexed: 04/01/2025]
Abstract
Tolerogenic antigen-presenting cells (APCs) are promising as therapeutics for suppressing T cell activation in autoimmune diseases. However, the isolation and ex vivo manipulation of autologous APCs is costly, and the process is customized for each patient. Here we show that tolerogenic APCs can be generated in vivo by delivering, via lipid nanoparticles, messenger RNA coding for the inhibitory protein programmed death ligand 1. We optimized a lipid-nanoparticle formulation to minimize its immunogenicity by reducing the molar ratio of nitrogen atoms on the ionizable lipid and the phosphate groups on the encapsulated mRNA. In mouse models of rheumatoid arthritis and ulcerative colitis, subcutaneous delivery of nanoparticles encapsulating mRNA encoding programmed death ligand 1 reduced the fraction of activated T cells, promoted the induction of regulatory T cells and effectively prevented disease progression. The method may allow for the engineering of APCs that target specific autoantigens or that integrate additional inhibitory molecules.
Collapse
Affiliation(s)
- Yang Liu
- 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
- National Key Laboratory of Immune Response and Immunotherapy, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Qian Liu
- 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
- National Key Laboratory of Immune Response and Immunotherapy, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Baowen Zhang
- National Key Laboratory of Immune Response and Immunotherapy, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Shanshan Chen
- 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
| | - Yanqiong Shen
- National Key Laboratory of Immune Response and Immunotherapy, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, China
- RNAlfa Biotech, Hefei, China
| | - Zhibin Li
- National Key Laboratory of Immune Response and Immunotherapy, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Jiachen Zhang
- National Key Laboratory of Immune Response and Immunotherapy, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yi Yang
- RNAlfa Biotech, Hefei, China
| | - Min Li
- 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.
- National Key Laboratory of Immune Response and Immunotherapy, School of Basic Medical Sciences, Division of Life Sciences and 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
- 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.
- National Key Laboratory of Immune Response and Immunotherapy, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
- Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, China.
- RNAlfa Biotech, Hefei, China.
- Key Laboratory of Anhui Province for Emerging and Reemerging Infectious Diseases, Hefei, China.
| |
Collapse
|
7
|
Simonis A, Theobald SJ, Koch AE, Mummadavarapu R, Mudler JM, Pouikli A, Göbel U, Acton R, Winter S, Albus A, Holzmann D, Albert MC, Hallek M, Walczak H, Ulas T, Koch M, Tessarz P, Hänsel-Hertsch R, Rybniker J. Persistent epigenetic memory of SARS-CoV-2 mRNA vaccination in monocyte-derived macrophages. Mol Syst Biol 2025:10.1038/s44320-025-00093-6. [PMID: 40133533 DOI: 10.1038/s44320-025-00093-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 02/10/2025] [Accepted: 02/24/2025] [Indexed: 03/27/2025] Open
Abstract
Immune memory plays a critical role in the development of durable antimicrobial immune responses. How precisely mRNA vaccines train innate immune cells to shape protective host defense mechanisms remains unknown. Here we show that SARS-CoV-2 mRNA vaccination significantly establishes histone H3 lysine 27 acetylation (H3K27ac) at promoters of human monocyte-derived macrophages, suggesting epigenetic memory. However, we found that two consecutive vaccinations were required for the persistence of H3K27ac, which matched with pro-inflammatory innate immune-associated transcriptional changes and antigen-mediated cytokine secretion. H3K27ac at promoter regions were preserved for six months and a single mRNA booster vaccine potently restored their levels and release of macrophage-derived cytokines. Interestingly, we found that H3K27ac at promoters is enriched for G-quadruplex DNA secondary structure-forming sequences in macrophage-derived nucleosome-depleted regions, linking epigenetic memory to nucleic acid structure. Collectively, these findings reveal that mRNA vaccines induce a highly dynamic and persistent training of innate immune cells enabling a sustained pro-inflammatory immune response.
Collapse
Affiliation(s)
- Alexander Simonis
- Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, 50937, Germany
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, 50931, Germany
- German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, Cologne, Germany
| | - Sebastian J Theobald
- Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, 50937, Germany
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, 50931, Germany
- German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, Cologne, Germany
| | - Anna E Koch
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, 50931, Germany
| | - Ram Mummadavarapu
- Max Planck Research Group "Chromatin and Ageing", Max Planck Institute for Biology of Ageing, Joseph-Stelzmann-Str. 9b, Cologne, 50931, Germany
| | - Julie M Mudler
- Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, 50937, Germany
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, 50931, Germany
| | - Andromachi Pouikli
- Max Planck Research Group "Chromatin and Ageing", Max Planck Institute for Biology of Ageing, Joseph-Stelzmann-Str. 9b, Cologne, 50931, Germany
| | - Ulrike Göbel
- Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Richard Acton
- Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
- Babraham Institute, Cambridge, UK
| | - Sandra Winter
- Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, 50937, Germany
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, 50931, Germany
| | - Alexandra Albus
- Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, 50937, Germany
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, 50931, Germany
| | - Dmitriy Holzmann
- Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, 50937, Germany
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, 50931, Germany
| | - Marie-Christine Albert
- Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
- Institute of Biochemistry I, Faculty of Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Michael Hallek
- Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, 50937, Germany
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, 50931, Germany
| | - Henning Walczak
- Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
- Institute of Biochemistry I, Faculty of Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany
- Center for Cell Death, Cancer and Inflammation, UCL Cancer Institute, University College London, London, United Kingdom
| | - Thomas Ulas
- Systems Medicine, German Center for Neurodegenerative Diseases (DZNE), University of Bonn, Bonn, Germany
- PRECISE Plattform for Single Cell Genomics and Epigenomics, DZNE, University of Bonn, Bonn and West German Genome Center, Bonn, Germany
- Genomics and Immunoregulation, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Manuel Koch
- Institute of Biochemistry I, Faculty of Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany
- Institute for Dental Research and Oral Musculoskeletal Biology, Center for Dental, Oral and Maxillofacial Medicine (central facilities), Medical Faculty and University of Cologne, Cologne, Germany
| | - Peter Tessarz
- Max Planck Research Group "Chromatin and Ageing", Max Planck Institute for Biology of Ageing, Joseph-Stelzmann-Str. 9b, Cologne, 50931, Germany
- Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
- Department of Human Biology, Radboud Institute for Molecular Life Sciences, Faculty of Science, Radboud University, Nijmegen, The Netherlands
| | - Robert Hänsel-Hertsch
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, 50931, Germany.
- Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany.
- Department of Translational Genomics, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany.
- Institute of Human Genetics, University Hospital Cologne, Cologne, Germany.
| | - Jan Rybniker
- Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, 50937, Germany.
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, 50931, Germany.
- German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, Cologne, Germany.
| |
Collapse
|
8
|
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] [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
|
9
|
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
|
10
|
Svensson M, Limeres MJ, Zeyn Y, Gambaro RC, Islan GA, Berti IR, Fraude-El Ghazi S, Pretsch L, Hilbert K, Schneider P, Kaps L, Bros M, Gehring S, Cacicedo ML. mRNA-LNP vaccine strategies: Effects of adjuvants on non-parenchymal liver cells and tolerance. Mol Ther Methods Clin Dev 2025; 33:101427. [PMID: 40027262 PMCID: PMC11872076 DOI: 10.1016/j.omtm.2025.101427] [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: 08/15/2024] [Accepted: 01/31/2025] [Indexed: 03/05/2025]
Abstract
The liver, which plays pivotal roles in metabolism and immunity, often confers tolerance, suppressing immune responses to pathogens. Adjuvanted, lipid nanoparticle-encapsulated mRNA vaccines (mRNA-LNPs) offer a promising approach to overcome immune tolerance. In this study, the immunostimulatory activity of well-documented adjuvants, i.e., 2'3'-cyclic guanosine monophosphate-adenosine monophosphate (cGAMP), resiquimod (R848), and polyinosinic:polycytidylic acid (Poly I:C), on non-parenchymal liver cells was determined. When co-applied with mRNA-loaded LNPs, these adjuvants enhanced immune responses at variable extents. Moreover, the efficiency of mRNA translation in the presence of cGAMP was comparable with the non-adjuvanted control. Repetitive co-application of adjuvants with mRNA-LNPs showed improvement in cellular responses when R848 or R848/cGAMP treatments were used. These findings emphasize the need to delineate the delicate balance between immunomodulatory properties and the efficiency of mRNA translation when selecting adjuvants for mRNA-LNP vaccines and offer insights on how to enhance immunity to infectious diseases and cancers that affect the liver.
Collapse
Affiliation(s)
- Malin Svensson
- Children’s Hospital, University Medical Center Mainz of the Johannes Gutenberg University, Langenbeckstr. 1, 55131 Mainz, Germany
| | - María José Limeres
- Children’s Hospital, University Medical Center Mainz of the Johannes Gutenberg University, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Yanira Zeyn
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Rocio C. Gambaro
- Children’s Hospital, University Medical Center Mainz of the Johannes Gutenberg University, Langenbeckstr. 1, 55131 Mainz, Germany
| | - German A. Islan
- Children’s Hospital, University Medical Center Mainz of the Johannes Gutenberg University, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Ignacio Rivero Berti
- Children’s Hospital, University Medical Center Mainz of the Johannes Gutenberg University, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Silvia Fraude-El Ghazi
- Children’s Hospital, University Medical Center Mainz of the Johannes Gutenberg University, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Leah Pretsch
- Children’s Hospital, University Medical Center Mainz of the Johannes Gutenberg University, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Katja Hilbert
- Children’s Hospital, University Medical Center Mainz of the Johannes Gutenberg University, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Paul Schneider
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Leonard Kaps
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
- Department of Medicine II Saarland University Medical Center Saarland University 66421 Homburg, Germany
| | - Matthias Bros
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Stephan Gehring
- Children’s Hospital, University Medical Center Mainz of the Johannes Gutenberg University, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Maximiliano L. Cacicedo
- Children’s Hospital, University Medical Center Mainz of the Johannes Gutenberg University, Langenbeckstr. 1, 55131 Mainz, Germany
| |
Collapse
|
11
|
Haghmorad D, Eslami M, Orooji N, Halabitska I, Kamyshna I, Kamyshnyi O, Oksenych V. mRNA vaccine platforms: linking infectious disease prevention and cancer immunotherapy. Front Bioeng Biotechnol 2025; 13:1547025. [PMID: 40144393 PMCID: PMC11937095 DOI: 10.3389/fbioe.2025.1547025] [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: 12/17/2024] [Accepted: 02/24/2025] [Indexed: 03/28/2025] Open
Abstract
The advent of mRNA vaccines, accelerated by the global response to the COVID-19 pandemic, marks a transformative shift in vaccine technology. In this article, we discuss the development, current applications, and prospects of mRNA vaccines for both the prevention and treatment of infectious diseases and oncology. By leveraging the capacity to encode antigens within host cells directly, mRNA vaccines provide a versatile and scalable platform suitable for addressing a broad spectrum of pathogens and tumor-specific antigens. We highlight recent advancements in mRNA vaccine design, innovative delivery mechanisms, and ongoing clinical trials, with particular emphasis on their efficacy in combating infectious diseases, such as COVID-19, Zika, and influenza, as well as their emerging potential in cancer immunotherapy. We also address critical challenges, including vaccine stability, optimization of immune responses, and the broader issue of global accessibility. Finally, we review potential strategies for advancing next-generation mRNA vaccines, with the aim of overcoming current limitations in vaccine technology and enhancing both preventive and therapeutic approaches for infectious and oncological diseases.
Collapse
Affiliation(s)
- Dariush Haghmorad
- Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Majid Eslami
- Department of Bacteriology and Virology, Semnan University of Medical Sciences, Semnan, Iran
| | - Niloufar Orooji
- Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
- Student Research Committee, Semnan University of Medical Sciences, Semnan, Iran
| | - Iryna Halabitska
- Department of Therapy and Family Medicine, I. Horbachevsky Ternopil National Medical University, Ternopil, Ukraine
| | - Iryna Kamyshna
- Department of Medical Rehabilitation, I. Horbachevsky Ternopil National Medical University, Ternopil, Ukraine
| | - Oleksandr Kamyshnyi
- Department of Microbiology, Virology, and Immunology, I. Horbachevsky Ternopil National Medical University, Ternopil, Ukraine
| | - Valentyn Oksenych
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| |
Collapse
|
12
|
Buckley M, Araínga M, Maiorino L, Pires IS, Kim BJ, Michaels KK, Dye J, Qureshi K, Zhang YJ, Mak H, Steichen JM, Schief WR, Villinger F, Irvine DJ. Visualizing lipid nanoparticle trafficking for mRNA vaccine delivery in non-human primates. Mol Ther 2025; 33:1105-1117. [PMID: 39797396 PMCID: PMC11897755 DOI: 10.1016/j.ymthe.2025.01.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 11/23/2024] [Accepted: 01/07/2025] [Indexed: 01/13/2025] Open
Abstract
mRNA delivered using lipid nanoparticles (LNPs) has become an important subunit vaccine modality, but mechanisms of action for mRNA vaccines remain incompletely understood. Here, we synthesized a metal chelator-lipid conjugate enabling positron emission tomography (PET) tracer labeling of LNP/mRNA vaccines for quantitative visualization of vaccine trafficking in live mice and non-human primates (NHPs). Following intramuscular injection, we observed LNPs distributing through injected muscle tissue, simultaneous with rapid trafficking to draining lymph nodes (dLNs). Deltoid injection of LNPs mimicking human vaccine administration led to stochastic LNP delivery to three different sets of dLNs. LNP uptake in dLNs was confirmed by histology, and cellular analysis of tissues via flow cytometry identified antigen-presenting cells as the primary immune cell type responsible for early LNP uptake and mRNA translation. These results provide insights into the biodistribution of mRNA vaccines administered at clinically relevant doses, injection volumes, and injection sites in an important large animal model for vaccine development.
Collapse
Affiliation(s)
- Maureen Buckley
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Mariluz Araínga
- New Iberia Research Center, University of Louisiana at Lafayette, Lafayette, LA 70560, USA
| | - Laura Maiorino
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| | - Ivan S Pires
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - B J Kim
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | | - Jonathan Dye
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Kashif Qureshi
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Yiming J Zhang
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Howard Mak
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Jon M Steichen
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA 92037, USA; Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA; IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - William R Schief
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA 92037, USA; Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA; IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Francois Villinger
- New Iberia Research Center, University of Louisiana at Lafayette, Lafayette, LA 70560, USA
| | - Darrell J Irvine
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, Harvard University, Cambridge, MA 02139, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA; Department of Materials Science of Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
13
|
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 2025:10915818251320631. [PMID: 40040255 DOI: 10.1177/10915818251320631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2025]
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
|
14
|
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
|
15
|
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
|
16
|
Maniyamgama N, Bae KH, Chang ZW, Lee J, Ang MJY, Tan YJ, Ng LFP, Renia L, White KP, Yang YY. Muco-Penetrating Lipid Nanoparticles Having a Liquid Core for Enhanced Intranasal mRNA Delivery. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2407383. [PMID: 39888252 PMCID: PMC11923898 DOI: 10.1002/advs.202407383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 08/15/2024] [Indexed: 02/01/2025]
Abstract
Intranasal delivery of mRNA vaccines offers promising opportunities to combat airborne viruses like SARS-CoV-2 by provoking mucosal immunity, which not only defends against respiratory infection but also prevents contagious transmission. However, the development of nasal mRNA vaccines has been hampered by the lack of effective means to overcome the mucus barrier. Herein, ionizable lipid-incorporated liquid lipid nanoparticles (iLLNs) capable of delivering mRNA cargo across airway mucosa are designed. Adjusting the ratios of ionizable and cationic lipids allows fine-tuning of the pKa of iLLNs to the range of nasal mucosal pH (5.5-6.5), thus facilitating mucus penetration via the formation of near-neutral, PEGylated muco-inert surfaces. When nasally administered to mice, the top candidate iLLN-2/mRNA complexes enable about 60-fold greater reporter gene expression in the nasal cavity, compared to the benchmark mRNA-lipid nanoparticles (ALC-LNP) having the same lipid composition as that of BNT162b2 vaccine. Moreover, a prime-boost intranasal immunization of iLLN-2/mRNA complexes elicits a greater magnitude of SARS-CoV-2 spike-specific mucosal IgA and IgG response than ALC-LNP, without triggering any noticeable inflammatory reactions. Taken together, these results provide useful insights for the design of nasally deliverable mRNA formulations for prophylactic applications.
Collapse
Affiliation(s)
- Nipuni Maniyamgama
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, Centros #06-01, Singapore, 138668, Republic of Singapore
| | - Ki Hyun Bae
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, Centros #06-01, Singapore, 138668, Republic of Singapore
| | - Zi Wei Chang
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, Immunos #05-13, Singapore, 138648, Republic of Singapore
| | - Jialing Lee
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, Centros #06-01, Singapore, 138668, Republic of Singapore
| | - Melgious J Y Ang
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, Centros #06-01, Singapore, 138668, Republic of Singapore
| | - Yong Jie Tan
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, Immunos #05-13, Singapore, 138648, Republic of Singapore
| | - Lisa F P Ng
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, Immunos #05-13, Singapore, 138648, Republic of Singapore
| | - Laurent Renia
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, Immunos #05-13, Singapore, 138648, Republic of Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 138648, Republic of Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore, 138648, Republic of Singapore
| | - Kevin P White
- Department of Biochemistry and Precision Medicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Republic of Singapore
| | - Yi Yan Yang
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, Centros #06-01, Singapore, 138668, Republic of Singapore
| |
Collapse
|
17
|
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
|
18
|
Herman L, Guagliardo R, Zamborlin A, Liu Q, Pérez-Gil J, De Smedt SC, Raemdonck K. Surfactant protein B-derived peptides as endosomal escape enhancers for pulmonary delivery of siRNA. J Control Release 2025:S0168-3659(25)00180-4. [PMID: 40010411 DOI: 10.1016/j.jconrel.2025.02.067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 02/06/2025] [Accepted: 02/22/2025] [Indexed: 02/28/2025]
Abstract
Respiratory diseases still cause significant mortality and morbidity worldwide, highlighting the need for new inhalable drugs. RNA therapeutics, which have the potential to modulate the expression of virtually any gene, could address this unmet medical need. Nevertheless, clinical translation requires the design of RNA formulations able to overcome the extra- and intracellular barriers in the lung. We previously discovered that the endogenous cationic amphiphilic surfactant protein B (SP-B) promotes cytosolic delivery of small interfering RNA (siRNA) in lung-related cell types via endosomal membrane fusion. However, to bypass drawbacks related to the use of animal-derived SP-B, there is a keen interest in developing synthetic SP-B analogues with comparable activity. Here, we show that native SP-B can successfully be replaced by smaller peptides, with the N-terminal heptapeptide and amphipathic helix being minimally required to promote siRNA-induced gene silencing. Peptidolipid-coated nanogels were designed and demonstrated equivalent siRNA delivery efficacy compared to state-of-the-art lipid nanoparticles (LNPs). Moreover, they exhibit enhanced resistance to vibrating mesh nebulization and reduced inflammatory activation of bronchial epithelial cells. Collectively, the discovery of SP-B peptides as RNA delivery enhancers holds promise for developing potent inhalable RNA formulations with favorable safety profiles, of value for the treatment of chronic inflammatory pathologies.
Collapse
Affiliation(s)
- Lore Herman
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium.
| | - Roberta Guagliardo
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium.
| | - Agata Zamborlin
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium.
| | - Qiaoyu Liu
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium.
| | - Jesús Pérez-Gil
- Departamento de Bioquimica y Biologia Molecular, Facultad de Biologia, Research Institute Hospital 12 Octubre, Universidad Complutense, José Antonio Novais 2, 28040 Madrid, Spain.
| | - Stefaan C De Smedt
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium.
| | - Koen Raemdonck
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium.
| |
Collapse
|
19
|
Enya T, Ross SR. Innate Sensing of Viral Nucleic Acids and Their Use in Antiviral Vaccine Development. Vaccines (Basel) 2025; 13:193. [PMID: 40006739 PMCID: PMC11860339 DOI: 10.3390/vaccines13020193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 01/23/2025] [Accepted: 02/11/2025] [Indexed: 02/27/2025] Open
Abstract
Viruses pose a significant threat to humans by causing numerous infectious and potentially fatal diseases. Understanding how the host's innate immune system recognizes viruses is essential to understanding pathogenesis and ways to control viral infection. Innate immunity also plays a critical role in shaping adaptive immune responses induced by vaccines. Recently developed adjuvants often include nucleic acids that stimulate pattern recognition receptors which are essential components of innate immunity necessary for activating antigen-presentation cells and thereby bridging innate and adaptive immunity. Therefore, understanding viral nucleic acid sensing by cytosolic sensors is essential, as it provides the potential means for developing new vaccine strategies, including effective adjuvants.
Collapse
Affiliation(s)
| | - Susan R. Ross
- Department of Microbiology and Immunology, University of Illinois at Chicago College of Medicine, Chicago, IL 60612, USA;
| |
Collapse
|
20
|
Ben-Akiva E, Chapman A, Mao T, Irvine DJ. Linking vaccine adjuvant mechanisms of action to function. Sci Immunol 2025; 10:eado5937. [PMID: 39951545 DOI: 10.1126/sciimmunol.ado5937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 01/23/2025] [Indexed: 02/16/2025]
Abstract
Vaccines deliver an immunogen in a manner designed to safely provoke an immune response, leading to the generation of memory T and B cells and long-lived antibody-producing plasma cells. Adjuvants play a critical role in vaccines by controlling how the immune system is exposed to the immunogen and providing inflammatory cues that enable productive immune priming. However, mechanisms of action underlying adjuvant function at the molecular, cell, and tissue levels are diverse and often poorly understood. Here, we review the current understanding of mechanisms of action underlying adjuvants used in subunit protein/polysaccharide vaccines and mRNA vaccines, discuss where possible how these mechanisms of action link to downstream effects on the immune response, and identify knowledge gaps that will be important to fill in order to enable the continued development of more effective adjuvants for challenging pathogens such as HIV and emerging threats.
Collapse
Affiliation(s)
- Elana Ben-Akiva
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Asheley Chapman
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Immunology and Microbiology, Scripps Research Institute, La Jolla, CA, USA
| | - Tianyang Mao
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Darrell J Irvine
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Immunology and Microbiology, Scripps Research Institute, La Jolla, CA, USA
- Broad Institute, Massachusetts Institute of Technology, Cambridge, MA, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| |
Collapse
|
21
|
Meng Y, Yao Z, Ke X, Hu M, Ren H, Gao S, Zhang H. Extracellular vesicles-based vaccines: Emerging immunotherapies against cancer. J Control Release 2025; 378:438-459. [PMID: 39667569 DOI: 10.1016/j.jconrel.2024.12.010] [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: 08/01/2024] [Revised: 12/03/2024] [Accepted: 12/05/2024] [Indexed: 12/14/2024]
Abstract
Cancer vaccines are promising therapeutic approaches to enhance specific T-cell immunity against most solid tumors. By stimulating anti-tumor immunity, clearing minimal residual disease, and minimizing adverse effects, these vaccines target tumor cells and are effective when combined with immune checkpoint blockade or other immunotherapies. However, the development of tumor cell-based vaccines faces quality issues due to poor immunogenicity, tumor heterogeneity, a suppressive tumor immune microenvironment, and ineffective delivery methods. In contrast, extracellular vesicles (EVs), naturally released by cells, are considered the ideal drug carriers and vaccine platforms. EVs offer highly organ-specific targeting, induce broader and more effective immune responses, and demonstrate superior tissue delivery ability. The development of EV vaccines is crucial for advancing cancer immunotherapy. Compared to cell-based vaccines, EV vaccines produced under Good Manufacturing Practices (GMP) offer advantages such as high safety, ease of preservation and transport, and a wide range of sources. This review summarizes the latest research findings on EV vaccine and potential applications in this field. It also highlights novel neoantigens for the development of EV vaccines against cancer.
Collapse
Affiliation(s)
- Yuhua Meng
- Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China; State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, and Institute of Precision Cancer Medicine and Pathology, School of Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Zhimeng Yao
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, and Institute of Precision Cancer Medicine and Pathology, School of Medicine, Jinan University, Guangzhou, Guangdong, China; Department of Urology Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Xiurong Ke
- Department of Surgery, Laboratory for Translational Surgical Oncology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Mengyuan Hu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, and Institute of Precision Cancer Medicine and Pathology, School of Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Hongzheng Ren
- Gongli Hospital of Shanghai Pudong New Area, Department of Pathology, Shanghai, China
| | - Shegan Gao
- College of Clinical Medicine, The First Affiliated Hospital of Henan University of Science and Technology, Henan Key Laboratory of Cancer Epigenetics, Luoyang, Henan, China.
| | - Hao Zhang
- Gongli Hospital of Shanghai Pudong New Area, Department of Pathology, Shanghai, China; Department of Pathology, and Institute of Precision Cancer Medicine and Pathology, School of Medicine, State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, Jinan University, Guangzhou, Guangdong, China; Department of Thoracic Surgery and General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China.
| |
Collapse
|
22
|
Bian X, Zhou L, Luo Z, Liu G, Hang Z, Li H, Li F, Wen Y. Emerging Delivery Systems for Enabling Precision Nucleic Acid Therapeutics. ACS NANO 2025; 19:4039-4083. [PMID: 39834294 DOI: 10.1021/acsnano.4c11858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Nucleic acid therapeutics represent a highly promising treatment approach in modern medicine, treating diseases at the genetic level. However, these therapeutics face numerous challenges in practical applications, particularly regarding their stability, effectiveness, cellular uptake efficiency, and limitations in delivering them specifically to target tissues. To overcome these obstacles, researchers have developed various innovative delivery systems, including viral vectors, lipid nanoparticles, polymer nanoparticles, inorganic nanoparticles, protein carriers, exosomes, antibody oligonucleotide conjugates, and DNA nanostructure-based delivery systems. These systems enhance the therapeutic efficacy of nucleic acid drugs by improving their stability, targeting specificity, and half-life in vivo. In this review, we systematically discuss different types of nucleic acid drugs, analyze the major barriers encountered in their delivery, and summarize the current research progress in emerging delivery systems. We also highlight the latest advancements in the application of these systems for treating genetic diseases, infectious diseases, cancer, brain diseases, and wound healing. This review aims to provide a comprehensive overview of nucleic acid drug delivery systems' current status and future directions by integrating the latest advancements in nanotechnology, biomaterials science, and gene editing technologies, emphasizing their transformative potential in precision medicine.
Collapse
Affiliation(s)
- Xiaochun Bian
- Beijing Key Laboratory for Bioengineering and Sensing Technology, Daxing Research Institute, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Liping Zhou
- Beijing Key Laboratory for Bioengineering and Sensing Technology, Daxing Research Institute, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Zhiwei Luo
- Beijing Key Laboratory for Bioengineering and Sensing Technology, Daxing Research Institute, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Guotao Liu
- Beijing Key Laboratory for Bioengineering and Sensing Technology, Daxing Research Institute, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Zhongci Hang
- Beijing Key Laboratory for Bioengineering and Sensing Technology, Daxing Research Institute, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Haohao Li
- Beijing Key Laboratory for Bioengineering and Sensing Technology, Daxing Research Institute, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Fengyong Li
- Plastic Surgery Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Yongqiang Wen
- Beijing Key Laboratory for Bioengineering and Sensing Technology, Daxing Research Institute, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China
| |
Collapse
|
23
|
Hickey TE, Mudunuri U, Hempel HA, Kemp TJ, Roche NV, Talsania K, Sellers BA, Cherry JM, Pinto LA. Proteomic and serologic assessments of responses to mRNA-1273 and BNT162b2 vaccines in human recipient sera. Front Immunol 2025; 15:1502458. [PMID: 39931577 PMCID: PMC11808009 DOI: 10.3389/fimmu.2024.1502458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 11/25/2024] [Indexed: 02/13/2025] Open
Abstract
Introduction The first vaccines approved against SARS-CoV-2, mRNA-1273 and BNT162b2, utilized mRNA platforms. However, little is known about the proteomic markers and pathways associated with host immune responses to mRNA vaccination. In this proof-of-concept study, sera from male and female vaccine recipients were evaluated for proteomic and immunologic responses 1-month and 6-months following homologous third vaccination. Methods An aptamer-based (7,289 marker) proteomic assay coupled with traditional serology was leveraged to generate a comprehensive evaluation of systemic responsiveness in 64 and 68 healthy recipients of mRNA-1273 and BNT162b2 vaccines, respectively. Results Sera from female recipients of mRNA-1273 showed upregulated indicators of inflammatory and immunological responses at 1-month post-third vaccination, and sera from female recipients of BNT162b2 demonstrated upregulated negative regulators of RNA sensors at 1-month. Sera from male recipients of mRNA-1273 showed no significant upregulation of pathways at 1-month post-third vaccination, though there were multiple significantly upregulated proteomic markers. Sera from male recipients of BNT162b2 demonstrated upregulated markers of immune response to doublestranded RNA and cell-cycle G(2)/M transition at 1-month. Random Forest analysis of proteomic data from pre-third-dose sera identified 85 markers used to develop a model predictive of robust or weaker IgG responses and antibody levels to SARS-CoV-2 spike protein at 6-months following boost; no specific markers were individually predictive of 6-month IgG response. Thirty markers that contributed most to the model were associated with complement cascade and activation; IL-17, TNFR pro-apoptotic, and PI3K signaling; and cell cycle progression. Discussion These results demonstrate the utility of proteomics to evaluate correlates or predictors of serological responses to SARS-CoV-2 vaccination.
Collapse
Affiliation(s)
- Thomas E. Hickey
- Vaccine, Immunity and Cancer Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Uma Mudunuri
- Advanced Biomedical Computational Science, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Heidi A. Hempel
- Vaccine, Immunity and Cancer Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Troy J. Kemp
- Vaccine, Immunity and Cancer Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Nancy V. Roche
- Vaccine, Immunity and Cancer Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Keyur Talsania
- Advanced Biomedical Computational Science, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Brian A. Sellers
- Center for Human Immunology, Inflammation and Autoimmunity, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - James M. Cherry
- Center for Human Immunology, Inflammation and Autoimmunity, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Ligia A. Pinto
- Vaccine, Immunity and Cancer Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| |
Collapse
|
24
|
Gao Z, Jing B, Wang Y, Wan W, Dong X, Liu Y. Exploring the impact of lipid nanoparticles on protein stability and cellular proteostasis. J Colloid Interface Sci 2025; 678:656-665. [PMID: 39216393 DOI: 10.1016/j.jcis.2024.08.146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 07/29/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024]
Abstract
Lipid nanoparticles (LNPs) have become pivotal in advancing modern medicine, from mRNA-based vaccines to gene editing with CRISPR-Cas9 systems. Though LNPs based therapeutics offer promising drug delivery with satisfactory clinical safety profiles, concerns are raised regarding their potential nanotoxicity. Here, we explore the impacts of LNPs on protein stability in buffer and cellular protein homeostasis (proteostasis) in HepG2 cells. First, we show that LNPs of different polyethylene glycol (PEG) molar ratios to total lipid ratio boost protein aggregation propensity by reducing protein stability in cell lysate and blood plasma. Second, in HepG2 liver cells, these LNPs induce global proteome aggregation, as imaged by a cellular protein aggregation fluorescent dye (AggStain). Such LNPs induced proteome aggregation is accompanied by decrease in cellular micro-environmental polarity as quantified by a solvatochromic protein aggregation sensor (AggRetina). The observed local polarity fluctuations may be caused by the hydrophobic contents of LNPs that promote cellular proteome aggregation. Finally, we exploit RNA sequencing analysis (RNA-Seq) to reveal activation of unfolded protein response (UPR) pathway and other proteostasis genes upon LNPs treatment. Together, these findings highlight that LNPs may induce subtle proteome stress by compromising protein stability and proteostasis even without obvious damage to cell viability.
Collapse
Affiliation(s)
- Zifan Gao
- Dalian Medical University, Dalian 116044, China
| | - Biao Jing
- State Key Laboratory of Medical Proteomics, National Chromatographic R. & A. Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Yuhui Wang
- The Second Hospital of Dalian Medical University, Dalian 116023, China
| | - Wang Wan
- State Key Laboratory of Medical Proteomics, National Chromatographic R. & A. Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China.
| | - Xuepeng Dong
- The Second Hospital of Dalian Medical University, Dalian 116023, China.
| | - Yu Liu
- State Key Laboratory of Medical Proteomics, National Chromatographic R. & A. Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China.
| |
Collapse
|
25
|
Zhang S, Wang X, Zhao T, Yang C, Huang L. Development and Evaluation of the Immunogenic Potential of an Unmodified Nucleoside mRNA Vaccine for Herpes Zoster. Vaccines (Basel) 2025; 13:68. [PMID: 39852847 PMCID: PMC11768781 DOI: 10.3390/vaccines13010068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 01/05/2025] [Accepted: 01/08/2025] [Indexed: 01/26/2025] Open
Abstract
BACKGROUND/OBJECTIVES Approved mRNA vaccines commonly use sequences modified with pseudouridine to enhance translation efficiency and mRNA stability. However, this modification can result in ribosomal frameshifts, reduced immunogenicity, and higher production costs. This study aimed to explore the potential of unmodified mRNA sequences for varicella-zoster virus (VZV) and evaluate whether codon optimization could overcome the limitations of pseudouridine modification. METHODS We utilized artificial intelligence (AI) to design several unmodified gE mRNA sequences for VZV, considering factors such as codon preference and secondary structure. The optimized mRNA sequences were assessed for protein expression levels in vitro and were subsequently used to develop a vaccine, named Vac07, encapsulated in a lipid nanoparticle (LNP) delivery system. The immunogenicity of Vac07 was evaluated in mice. RESULTS Codon-optimized mRNA sequences showed significantly higher protein expression levels in vitro compared to wild-type (WT) sequences. Vaccination with Vac07 demonstrated immunogenicity in mice that was comparable to, or even superior to, the licensed Shingrix vaccine, characterized by a stronger Th1-biased antibody response and a slightly more robust Th1-type cellular response. CONCLUSIONS Codon-optimized unmodified mRNA sequences may also represent a viable approach for mRNA vaccine development. These optimized sequences have the potential to lower production costs while possibly enhancing the immunogenicity of mRNA vaccines. Vac07, developed using this method, shows promise as a potentially more efficient and cost-effective mRNA vaccine candidate for VZV.
Collapse
Affiliation(s)
- Shun Zhang
- Ningbo No. 2 Hospital, Guoke Ningbo Life Science and Health Industry Research Institute, Ningbo 315099, China;
| | - Xiaojie Wang
- Department of Clinical Pharmacy, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China;
- Technology Center, Shandong Freda Pharmaceutical Group, Jinan 250101, China
| | - Tongyi Zhao
- Vaccine Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China;
| | - Chen Yang
- School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| | - Lulu Huang
- Vaccine Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China;
| |
Collapse
|
26
|
Lee WS, Selva KJ, Audsley J, Kent HE, Reynaldi A, Schlub TE, Cromer D, Khoury DS, Peck H, Aban M, Vu MN, Zheng MZ, Chung AW, Koutsakos M, Tan HX, Wheatley AK, Juno JA, Rockman S, Davenport MP, Barr I, Kent SJ. Randomized trial of same- versus opposite-arm coadministration of inactivated influenza and SARS-CoV-2 mRNA vaccines. JCI Insight 2025; 10:e187075. [PMID: 39786918 PMCID: PMC11949032 DOI: 10.1172/jci.insight.187075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 01/02/2025] [Indexed: 01/12/2025] Open
Abstract
BACKGROUNDThe immunogenicity of current influenza vaccines needs improvement. Inactivated influenza and COVID-19 mRNA vaccines can be coadministered, but randomized controlled trial data are lacking on whether the 2 vaccines are more immunogenic if given in the same arm or opposite arms. Murine studies suggest mRNA vaccines can adjuvant influenza vaccines when coformulated and codelivered.METHODSWe randomly assigned 56 adults to receive the Afluria quadrivalent inactivated influenza and Moderna monovalent SARS-CoV-2 XBB.1.5 mRNA vaccines, either in opposite arms or both in the same arm at the same site. The primary endpoint was the difference in median combined serum hemagglutination inhibition titer to the H1, H3, and B-Vic vaccine influenza strains after vaccination.RESULTSWe found no significant difference in hemagglutination inhibition antibody levels between the groups (P = 0.30), with the same-arm group having a 1.26-fold higher titer than the opposite-arm group. There were no differences in analyses of antibodies against individual influenza strains or in nasal or saliva antibody levels. While both binding and neutralizing antibody titers against SARS-CoV-2 were not significantly different between groups postvaccination, there was a higher fold-change in BA.5 and ancestral strain neutralizing antibodies in the opposite-arm group.CONCLUSIONInfluenza vaccination is equivalently immunogenic if given in the same arm or opposite arms as the SARS-CoV-2 vaccine, but it may be preferable to administer the SARS-CoV-2 vaccine at a different site from influenza vaccines.TRIAL REGISTRATIONAustralian New Zealand Clinical Trials Registry ACTRN12624000445572.FUNDINGAustralian National Health and Medical Research Council, Australian Medical Research Future Fund, and National Institutes of Health (UH2AI176172).
Collapse
MESH Headings
- Humans
- COVID-19 Vaccines/immunology
- COVID-19 Vaccines/administration & dosage
- Influenza Vaccines/immunology
- Influenza Vaccines/administration & dosage
- Female
- Male
- Adult
- Middle Aged
- SARS-CoV-2/immunology
- COVID-19/prevention & control
- COVID-19/immunology
- Antibodies, Viral/immunology
- Antibodies, Viral/blood
- Vaccines, Inactivated/immunology
- Vaccines, Inactivated/administration & dosage
- Influenza, Human/prevention & control
- Influenza, Human/immunology
- mRNA Vaccines/immunology
- Antibodies, Neutralizing/immunology
- Antibodies, Neutralizing/blood
- Aged
- Immunogenicity, Vaccine
- Hemagglutination Inhibition Tests
- Vaccines, Synthetic/immunology
- Vaccines, Synthetic/administration & dosage
- Young Adult
- Vaccination/methods
Collapse
Affiliation(s)
- Wen Shi Lee
- Department of Microbiology and Immunology and
| | | | - Jennifer Audsley
- Department of Infectious Diseases, Peter Doherty Institute for Infection and Immunity, University of Melbourne and Royal Melbourne Hospital, Melbourne, Victoria, Australia
| | | | - Arnold Reynaldi
- Kirby Institute, University of New South Wales, Kensington, New South Wales, Australia
| | - Timothy E. Schlub
- Sydney School of Public Health, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
| | - Deborah Cromer
- Kirby Institute, University of New South Wales, Kensington, New South Wales, Australia
| | - David S. Khoury
- Kirby Institute, University of New South Wales, Kensington, New South Wales, Australia
| | - Heidi Peck
- WHO Collaborating Centre for Reference and Research on Influenza, Royal Melbourne Hospital, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Malet Aban
- WHO Collaborating Centre for Reference and Research on Influenza, Royal Melbourne Hospital, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Mai Ngoc Vu
- Department of Microbiology and Immunology and
| | | | | | | | | | | | | | - Steven Rockman
- Department of Microbiology and Immunology and
- Vaccine Innovation Unit, CSL Seqirus Ltd, Parkville, Victoria, Australia
| | - Miles P. Davenport
- Kirby Institute, University of New South Wales, Kensington, New South Wales, Australia
| | - Ian Barr
- WHO Collaborating Centre for Reference and Research on Influenza, Royal Melbourne Hospital, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Stephen J. Kent
- Department of Microbiology and Immunology and
- Melbourne Sexual Health Centre and Department of Infectious Diseases, Alfred Hospital and Central Clinical School, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
27
|
Fu Q, Zhao X, Hu J, Jiao Y, Yan Y, Pan X, Wang X, Jiao F. mRNA vaccines in the context of cancer treatment: from concept to application. J Transl Med 2025; 23:12. [PMID: 39762875 PMCID: PMC11702060 DOI: 10.1186/s12967-024-06033-6] [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: 07/29/2024] [Accepted: 12/24/2024] [Indexed: 01/11/2025] Open
Abstract
Immuno-oncology has witnessed remarkable advancements in the past decade, revolutionizing the landscape of cancer therapeutics in an encouraging manner. Among the diverse immunotherapy strategies, mRNA vaccines have ushered in a new era for the therapeutic management of malignant diseases, primarily due to their impressive impact on the COVID-19 pandemic. In this comprehensive review, we offer a systematic overview of mRNA vaccines, focusing on the optimization of structural design, the crucial role of delivery materials, and the administration route. Additionally, we summarize preclinical studies and clinical trials to provide valuable insights into the current status of mRNA vaccines in cancer treatment. Furthermore, we delve into a systematic discussion on the significant challenges facing the current development of mRNA tumor vaccines. These challenges encompass both intrinsic and external factors that are closely intertwined with the successful application of this innovative approach. To pave the way for a more promising future in cancer treatments, a deeper understanding of immunological mechanisms, an increasing number of high-quality clinical trials, and a well-established manufacturing platform are crucial. Collaborative efforts between scientists, clinicians, and industry engineers are essential to achieving these goals.
Collapse
Affiliation(s)
- Qiang Fu
- School of Pharmacology, Institute of Aging Medicine, Binzhou Medical University, Yantai, 264003, P. R. China
| | - Xiaoming Zhao
- Center of Physical Examination, Binzhou Medical University Affiliated 970 Hospital of the PLA Joint Logistic Support Force, Yantai, 264002, P. R. China
| | - Jinxia Hu
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, 346 Guanhai Road, Yantai, 264003, P. R. China
| | - Yang Jiao
- Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Hong Kong, 999077, P. R. China
| | - Yunfei Yan
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, 346 Guanhai Road, Yantai, 264003, P. R. China
| | - Xuchen Pan
- Department of Clinical Laboratory & Health Service Training, Binzhou Medical University Affiliated 970 Hospital of the PLA Joint Logistic Support Force, Yantai, 264002, P. R. China
| | - Xin Wang
- Department of Clinical Laboratory & Health Service Training, Binzhou Medical University Affiliated 970 Hospital of the PLA Joint Logistic Support Force, Yantai, 264002, P. R. China.
| | - Fei Jiao
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, 346 Guanhai Road, Yantai, 264003, P. R. China.
| |
Collapse
|
28
|
Xu L, Li C, Liao R, Xiao Q, Wang X, Zhao Z, Zhang W, Ding X, Cao Y, Cai L, Rosenecker J, Guan S, Tang J. From Sequence to System: Enhancing IVT mRNA Vaccine Effectiveness through Cutting-Edge Technologies. Mol Pharm 2025; 22:81-102. [PMID: 39601789 DOI: 10.1021/acs.molpharmaceut.4c00863] [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: 11/29/2024]
Abstract
The COVID-19 pandemic has spotlighted the potential of in vitro transcribed (IVT) mRNA vaccines with their demonstrated efficacy, safety, cost-effectiveness, and rapid manufacturing. Numerous IVT mRNA vaccines are now under clinical trials for a range of targets, including infectious diseases, cancers, and genetic disorders. Despite their promise, IVT mRNA vaccines face hurdles such as limited expression levels, nonspecific targeting beyond the liver, rapid degradation, and unintended immune activation. Overcoming these challenges is crucial to harnessing the full therapeutic potential of IVT mRNA vaccines for global health advancement. This review provides a comprehensive overview of the latest research progress and optimization strategies for IVT mRNA molecules and delivery systems, including the application of artificial intelligence (AI) models and deep learning techniques for IVT mRNA structure optimization and mRNA delivery formulation design. We also discuss recent development of the delivery platforms, such as lipid nanoparticles (LNPs), polymers, and exosomes, which aim to address challenges related to IVT mRNA protection, cellular uptake, and targeted delivery. Lastly, we offer insights into future directions for improving IVT mRNA vaccines, with the hope to spur further progress in IVT mRNA vaccine research and development.
Collapse
Affiliation(s)
- Lifeng Xu
- National Engineering Research Center of Immunological Products, Third Military Medical University, Chongqing 400038, China
| | - Chao Li
- National Engineering Research Center of Immunological Products, Third Military Medical University, Chongqing 400038, China
| | - Rui Liao
- National Engineering Research Center of Immunological Products, Third Military Medical University, Chongqing 400038, China
| | - Qin Xiao
- National Engineering Research Center of Immunological Products, Third Military Medical University, Chongqing 400038, China
| | - Xiaoran Wang
- Department of Pharmacy, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830000, China
| | - Zhuo Zhao
- National Engineering Research Center of Immunological Products, Third Military Medical University, Chongqing 400038, China
| | - Weijun Zhang
- National Engineering Research Center of Immunological Products, Third Military Medical University, Chongqing 400038, China
| | - Xiaoyan Ding
- Department of Pediatrics, Ludwig-Maximilians University of Munich, Munich 80337, Germany
| | - Yuxue Cao
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Larry Cai
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Joseph Rosenecker
- Department of Pediatrics, Ludwig-Maximilians University of Munich, Munich 80337, Germany
| | - Shan Guan
- National Engineering Research Center of Immunological Products, Third Military Medical University, Chongqing 400038, China
| | - Jie Tang
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| |
Collapse
|
29
|
Zhang M, Wang C, Pan J, Cui H, Zhao X. Advancing novel veterinary vaccines: From comprehensive antigen and adjuvant design to preparation process optimization. Int Immunopharmacol 2025; 145:113784. [PMID: 39672026 DOI: 10.1016/j.intimp.2024.113784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 12/01/2024] [Accepted: 12/02/2024] [Indexed: 12/15/2024]
Abstract
Vaccination stands as the paramount and cost-effective strategy for the prevention and management of animal infectious diseases. With the advances in biological technology, materials science and industrial optimization, substantial progress has been made in the development of innovative veterinary vaccines. A majority of the novel vaccines under current investigation tend to stimulate multiple immune pathways and to achieve long-term resistance against infectious diseases, yet it remains imperative to concentrate research efforts on the efficient utilization of vaccines, mitigating toxic side effects, and ensuring safe production processes. This article presents an overview of research progress in veterinary vaccines, encompassing comprehensive antigen design, adjuvant formulation advancements, preparation process optimization, and rigorous immune efficacy evaluation. It summarizes cutting-edge vaccines derived from in vitro synthesis and in vivo application, emphasizing immunogenic components and immune response mechanisms. It also highlights novel biological adjuvants that enhance immune efficacy, diversify raw materials, and possess targeted functions, while comprehensively exploring advancements in production methodologies and compatible vaccine products. By establishing a foundation for the integrated use of these innovative veterinary vaccines, this work facilitates future interdisciplinary cooperation in their advancement, aiming to accelerate the achievement of herd immunity through concerted efforts.
Collapse
Affiliation(s)
- Meng Zhang
- Institute of Environment and Sustainable Development in Agriculture, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Chunxin Wang
- Institute of Environment and Sustainable Development in Agriculture, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Junqian Pan
- Institute of Environment and Sustainable Development in Agriculture, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Haixin Cui
- Institute of Environment and Sustainable Development in Agriculture, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Xiang Zhao
- Institute of Environment and Sustainable Development in Agriculture, Chinese Academy of Agricultural Sciences, Beijing 100081, China.
| |
Collapse
|
30
|
Jin L, Zhou Y, Zhang S, Chen SJ. mRNA vaccine sequence and structure design and optimization: Advances and challenges. J Biol Chem 2025; 301:108015. [PMID: 39608721 PMCID: PMC11728972 DOI: 10.1016/j.jbc.2024.108015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 11/13/2024] [Accepted: 11/16/2024] [Indexed: 11/30/2024] Open
Abstract
Messenger RNA (mRNA) vaccines have emerged as a powerful tool against communicable diseases and cancers, as demonstrated by their huge success during the coronavirus disease 2019 (COVID-19) pandemic. Despite the outstanding achievements, mRNA vaccines still face challenges such as stringent storage requirements, insufficient antigen expression, and unexpected immune responses. Since the intrinsic properties of mRNA molecules significantly impact vaccine performance, optimizing mRNA design is crucial in preclinical development. In this review, we outline four key principles for optimal mRNA sequence design: enhancing ribosome loading and translation efficiency through untranslated region (UTR) optimization, improving translation efficiency via codon optimization, increasing structural stability by refining global RNA sequence and extending in-cell lifetime and expression fidelity by adjusting local RNA structures. We also explore recent advancements in computational models for designing and optimizing mRNA vaccine sequences following these principles. By integrating current mRNA knowledge, addressing challenges, and examining advanced computational methods, this review aims to promote the application of computational approaches in mRNA vaccine development and inspire novel solutions to existing obstacles.
Collapse
Affiliation(s)
- Lei Jin
- Department of Physics and Astronomy, University of Missouri, Columbia, Missouri, USA
| | - Yuanzhe Zhou
- Department of Physics and Astronomy, University of Missouri, Columbia, Missouri, USA
| | - Sicheng Zhang
- Department of Physics and Astronomy, University of Missouri, Columbia, Missouri, USA
| | - Shi-Jie Chen
- Department of Physics and Astronomy, University of Missouri, Columbia, Missouri, USA; Department of Biochemistry, MU Institute for Data Science and Informatics, University of Missouri, Columbia, Missouri, USA.
| |
Collapse
|
31
|
Bachmann MF, Krenger PS, Mohsen MO, Kramer MF, Starchenka S, Whitehead P, Vogel M, Heath MD. On the role of antibody affinity and avidity in the IgE-mediated allergic response. Allergy 2025; 80:37-46. [PMID: 39189064 PMCID: PMC11724228 DOI: 10.1111/all.16248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 07/10/2024] [Accepted: 07/11/2024] [Indexed: 08/28/2024]
Abstract
Type I hypersensitivity, also known as classical allergy, is mediated via allergen-specific IgE antibodies bound to type I FcR (FcεRI) on the surface of mast cells and basophils upon cross-linking by allergens. This IgE-mediated cellular activation may be blocked by allergen-specific IgG through multiple mechanisms, including direct neutralization of the allergen or engagement of the inhibitory receptor FcγRIIb which blocks IgE signal transduction. In addition, co-engagement of FcεRI and FcγRIIb by IgE-IgG-allergen immune complexes causes down regulation of receptor-bound IgE, resulting in desensitization of the cells. Both, activation of FcεRI by allergen-specific IgE and engagement of FcγRIIb by allergen-specific IgG are driven by allergen-binding. Here we delineate the distinct roles of antibody affinity versus avidity in driving these processes and discuss the role of IgG subclasses in inhibiting basophil and mast cell activation.
Collapse
Affiliation(s)
- Martin F. Bachmann
- Department of Rheumatology and ImmunologyUniversity Hospital of BernBernSwitzerland
- Department for Biomedical Research Bern (DBMR)University of BernBernSwitzerland
- Nuffield Department of Medicine, The Jenner InstituteUniversity of OxfordOxfordUK
| | - Pascal S. Krenger
- Department of Rheumatology and ImmunologyUniversity Hospital of BernBernSwitzerland
- Department for Biomedical Research Bern (DBMR)University of BernBernSwitzerland
| | - Mona O. Mohsen
- Department of Rheumatology and ImmunologyUniversity Hospital of BernBernSwitzerland
- Department for Biomedical Research Bern (DBMR)University of BernBernSwitzerland
| | | | | | | | - Monique Vogel
- Department of Rheumatology and ImmunologyUniversity Hospital of BernBernSwitzerland
- Department for Biomedical Research Bern (DBMR)University of BernBernSwitzerland
| | | |
Collapse
|
32
|
Maltseva M, Keeshan A, Cooper C, Langlois MA. Immune imprinting: The persisting influence of the first antigenic encounter with rapidly evolving viruses. Hum Vaccin Immunother 2024; 20:2384192. [PMID: 39149872 PMCID: PMC11328881 DOI: 10.1080/21645515.2024.2384192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 07/08/2024] [Accepted: 07/21/2024] [Indexed: 08/17/2024] Open
Abstract
Immune imprinting is a phenomenon that stems from the fundamentals of immunological memory. Upon recurrent exposures to an evolving pathogen, the immune system must weigh the benefits of rapidly recalling established antibody repertoires with greater affinity to the initial variant or invest additional time and energy in producing de novo responses specific to the emerging variant. In this review, we delve into the mechanistic complexities of immune imprinting and its role in shaping subsequent immune responses, both de novo and recall, against rapidly evolving respiratory viruses such as influenza and coronaviruses. By exploring the duality of immune imprinting, we examine its potential to both enhance or hinder immune protection against disease, while emphasizing the role of host and viral factors. Finally, we explore how different vaccine platforms may affect immune imprinting and comment on vaccine strategies that can favor de novo variant-specific antibody responses.
Collapse
Affiliation(s)
- Mariam Maltseva
- Department of Biochemistry, Microbiology & Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Alexa Keeshan
- School of Epidemiology and Public Health, University of Ottawa Faculty of Medicine, Ottawa, ON, Canada
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Curtis Cooper
- School of Epidemiology and Public Health, University of Ottawa Faculty of Medicine, Ottawa, ON, Canada
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Department of Medicine, University of Ottawa, Ottawa, ON, Canada
- Division of Infectious Diseases, Ottawa Hospital Research Institute Clinical Epidemiology Program, Ottawa, ON, Canada
| | - Marc-André Langlois
- Department of Biochemistry, Microbiology & Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Canada
- Center for Infection, Immunity, and Inflammation (CI3), University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
33
|
Huang S, Que H, Wang M, Wei X. mRNA vaccines as cancer therapies. Chin Med J (Engl) 2024; 137:2979-2995. [PMID: 39668413 PMCID: PMC11706586 DOI: 10.1097/cm9.0000000000003455] [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: 10/14/2024] [Indexed: 12/14/2024] Open
Abstract
ABSTRACT Cancer remains a major global health challenge, with conventional treatments like chemotherapy and radiotherapy often hindered by significant side effects, lack of specificity, and limited efficacy in advanced cases. Among emerging therapeutic strategies, mRNA vaccines have shown remarkable potential due to their adaptability, rapid production, and capability for personalized cancer treatment. This review provides an in-depth analysis of messenger RNA (mRNA) vaccines as a therapeutic approach for cancer immunotherapy, focusing on their molecular biology, classification, mechanisms, and clinical studies. Derived from reported literature and data on clinicaltrials.gov, it examines studies on mRNA vaccines encoding tumor-specific antigens (TSAs), tumor-associated antigens (TAAs), immunomodulators, and chimeric antigen receptors (CARs) across various cancer types. The review highlights the ability of mRNA vaccines to encode TSAs and TAAs, enabling personalized cancer treatments, and classifies these vaccines into non-replicating and self-amplifying types. It further explores their mechanisms of action, including antigen presentation and immune activation, while emphasizing findings from clinical studies that demonstrate the potential of mRNA vaccines in cancer therapy. Despite their promise, challenges remain in enhancing delivery systems, improving immunogenicity, and addressing tumor heterogeneity. Overcoming these obstacles will require further investigation to fully harness the potential of mRNA vaccines in personalized cancer treatment.
Collapse
Affiliation(s)
- Shaoxiong Huang
- Laboratory of Aging Research and Cancer Drug Target, National/State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan 610041, China
| | - Haiying Que
- Laboratory of Aging Research and Cancer Drug Target, National/State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan 610041, China
| | - Manni Wang
- Laboratory of Aging Research and Cancer Drug Target, National/State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan 610041, China
- Department of Biotherapy, West China Hospital and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, National/State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
34
|
Dai W, Xing M, Sun L, Lv L, Wang X, Wang Y, Pang X, Guo Y, Ren J, Zhou D. Lipid nanoparticles as adjuvant of norovirus VLP vaccine augment cellular and humoral immune responses in a TLR9- and type I IFN-dependent pathway. J Virol 2024; 98:e0169924. [PMID: 39494905 DOI: 10.1128/jvi.01699-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 10/09/2024] [Indexed: 11/05/2024] Open
Abstract
Norovirus (NoV) virus-like particles (VLPs) adjuvanted with aluminum hydroxide (Alum) are common vaccine candidates in clinical studies. Alum adjuvants usually inefficiently assist recombinant proteins to induce cellular immune responses. Thus, novel adjuvants are required to develop NoV vaccines that could induce both efficient humoral and robust cellular immune responses. Lipid nanoparticles (LNPs) are well-known mRNA delivery vehicles. Increasing evidence suggests that LNPs may have intrinsic adjuvant activity and can be used as adjuvants for recombinant protein vaccines; however, the underlying mechanism remains poorly understood. In this study, we compared the adjuvant effect of LNPs and Alum for a bivalent GI.1/GII.4 NoV VLP vaccine. Compared with Alum, LNP-adjuvanted vaccines induced earlier production of binding, blocking, and neutralizing antibodies and promoted a more balanced IgG2a/IgG1 ratio. It is crucial that LNP-adjuvanted vaccines induced stronger Th1-type cytokine-producing CD4+ T cell and CD8+ T cell responses than Alum. The adjuvant activity of LNPs depended on the ionizable lipid components. Mechanistically, LNPs activated innate immune responses in a type I IFN-dependent manner and were partially dependent on Toll-like receptor (TLR) 9, thus affecting the adaptive immune responses of the vaccine. This conclusion was supported by RNA-seq analysis and in vitro cell experiments and by the deeply blunted T cell responses in IFNαR1-/- mice immunized with LNP-adjuvanted vaccines. This study not only identified LNPs as a high quality adjuvant for NoV VLP vaccines, but also clarified the underlying mechanism of LNPs as a potent immunostimulatory component for improving protein subunit vaccines.IMPORTANCEWith the rapid development of mRNA vaccines, recurrent studies show that lipid nanoparticles (LNPs) have adjuvant activity. However, the mechanism of its adjuvant effect in protein vaccines remains unknown. In this study, we found that the LNP-adjuvanted norovirus bivalent virus-like particle vaccines led to durable antibody responses as well as Th1-type cytokine-producing CD4+ T cell and CD8+ T cell responses, which exceeded the efficiency of the conventional adjuvant aluminum hydroxide. Mechanistically, LNPs activated innate immune responses in a type I IFN-dependent manner and were partially dependent on Toll-like receptor 9, thus affecting the adaptive immune responses of the vaccine. This work unveils that LNPs as a potent immunostimulatory component may be ideal for generating CD8+ T cell and B cell responses for recombinant protein vaccines.
Collapse
MESH Headings
- Animals
- Nanoparticles/administration & dosage
- Mice
- Norovirus/immunology
- Immunity, Humoral
- Vaccines, Virus-Like Particle/immunology
- Vaccines, Virus-Like Particle/administration & dosage
- Adjuvants, Immunologic/administration & dosage
- Interferon Type I/immunology
- Immunity, Cellular
- Antibodies, Viral/immunology
- Caliciviridae Infections/prevention & control
- Caliciviridae Infections/immunology
- Toll-Like Receptor 9/immunology
- Antibodies, Neutralizing/immunology
- Mice, Inbred C57BL
- Adjuvants, Vaccine
- Female
- Viral Vaccines/immunology
- Viral Vaccines/administration & dosage
- Immunoglobulin G/immunology
- Immunity, Innate
- Humans
- Mice, Knockout
- CD8-Positive T-Lymphocytes/immunology
- Lipids/immunology
- Aluminum Hydroxide/administration & dosage
- Aluminum Hydroxide/pharmacology
- Aluminum Hydroxide/immunology
- Liposomes
Collapse
Affiliation(s)
- Weiqian Dai
- Department of Pathogen Biology, Basic Medical College, Tianjin Medical University, Tianjin, China
| | - Man Xing
- Department of Pathogen Biology, Basic Medical College, Tianjin Medical University, Tianjin, China
| | - Lingjin Sun
- Department of Pathogen Biology, Basic Medical College, Tianjin Medical University, Tianjin, China
| | - Lihui Lv
- Department of Pathogen Biology, Basic Medical College, Tianjin Medical University, Tianjin, China
| | - Xiang Wang
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Yihan Wang
- Department of Pathogen Biology, Basic Medical College, Tianjin Medical University, Tianjin, China
| | - Xueyang Pang
- Department of Pathogen Biology, Basic Medical College, Tianjin Medical University, Tianjin, China
| | - Yingying Guo
- Department of Pathogen Biology, Basic Medical College, Tianjin Medical University, Tianjin, China
| | - Jiling Ren
- Department of Pathogen Biology, Basic Medical College, Tianjin Medical University, Tianjin, China
| | - Dongming Zhou
- Department of Pathogen Biology, Basic Medical College, Tianjin Medical University, Tianjin, China
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| |
Collapse
|
35
|
Gasparello J, Papi C, Marzaro G, Macone A, Zurlo M, Finotti A, Agostinelli E, Gambari R. Aged Garlic Extract (AGE) and Its Constituent S-Allyl-Cysteine (SAC) Inhibit the Expression of Pro-Inflammatory Genes Induced in Bronchial Epithelial IB3-1 Cells by Exposure to the SARS-CoV-2 Spike Protein and the BNT162b2 Vaccine. Molecules 2024; 29:5938. [PMID: 39770027 PMCID: PMC11677098 DOI: 10.3390/molecules29245938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 12/07/2024] [Accepted: 12/11/2024] [Indexed: 01/05/2025] Open
Abstract
Garlic (Allium sativum L.) is a species of the onion family (Alliaceae) widely used as a food and a folk medicine. The objective of this study was to determine the effects of AGE (aged garlic extract) on pro-inflammatory genes relevant to COVID-19. To this aim, we treated bronchial epithelial IB3-1 cells with SARS-CoV-2 spike protein (S-protein) or with the COVID-19 BNT162b2 vaccine in the absence or in the presence of AGE. The results obtained demonstrated that AGE is a potent inhibitor of the S-protein-induced expression of the IL-1β, IL-6 and IL-8 genes. Bio-Plex analysis demonstrated that AGE reduced release of IL-6 and IL-8, which were highly induced by S-protein. No inhibition of cells' growth, toxicity and pro-apoptotic effects were found in AGE-treated cells. The effects of one of the major AGE constituents (S-allyl cysteine, SAC) were studied on the same experimental model systems. SAC was able to inhibit the S-protein-induced expression of IL-1β, IL-6 and IL-8 genes and extracellular release of IL-6 and IL-8, confirming that S-allyl-cysteine is one of the constituents of AGE that is responsible for inhibiting S-protein-induced pro-inflammatory genes. Docking experiments suggest that a possible mechanism of action of SAC is an interference with the activity of Toll-like receptors (TLRs), particularly TLR4, thereby inhibiting NF-κB- and NF-κB-regulated genes, such as IL-1β, IL-6 and IL-8 genes. These results suggest that both AGE and SAC deserve further experimental efforts to verify their effects on pro-inflammatory genes in SARS-CoV-2-infected cells.
Collapse
Affiliation(s)
- Jessica Gasparello
- Department of Life Sciences and Biotechnology, Ferrara University, 44121 Ferrara, Italy; (J.G.); (C.P.); (M.Z.); (A.F.)
| | - Chiara Papi
- Department of Life Sciences and Biotechnology, Ferrara University, 44121 Ferrara, Italy; (J.G.); (C.P.); (M.Z.); (A.F.)
| | - Giovanni Marzaro
- Department of Diagnostics and Public Health, University of Verona, 37134 Verona, Italy;
| | - Alberto Macone
- Department of Biochemical Sciences ‘A. Rossi Fanelli’, Sapienza University of Rome, 00185 Rome, Italy;
| | - Matteo Zurlo
- Department of Life Sciences and Biotechnology, Ferrara University, 44121 Ferrara, Italy; (J.G.); (C.P.); (M.Z.); (A.F.)
| | - Alessia Finotti
- Department of Life Sciences and Biotechnology, Ferrara University, 44121 Ferrara, Italy; (J.G.); (C.P.); (M.Z.); (A.F.)
| | - Enzo Agostinelli
- Department of Sensory Organs, Sapienza University of Rome, Policlinico Umberto I, Viale del Policlinico 155, 00161 Rome, Italy
- International Polyamines Foundation ‘ETS-ONLUS’, Via del Forte Tiburtino 98, 00159 Rome, Italy
| | - Roberto Gambari
- Department of Life Sciences and Biotechnology, Ferrara University, 44121 Ferrara, Italy; (J.G.); (C.P.); (M.Z.); (A.F.)
| |
Collapse
|
36
|
Abt ER, Lam AK, Noguchi M, Rashid K, McLaughlin J, Teng PL, Tran W, Cheng D, Nesterenko PA, Mao Z, Creech AL, Burton Sojo G, Jeyachandran AV, Tam YK, Henley JE, Comai L, Pardi N, Arumugaswami V, Witte ON, Radu CG, Wu TT. Staggered immunization with mRNA vaccines encoding SARS-CoV-2 polymerase or spike antigens broadens the T cell epitope repertoire. Proc Natl Acad Sci U S A 2024; 121:e2406332121. [PMID: 39589869 PMCID: PMC11626164 DOI: 10.1073/pnas.2406332121] [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: 03/27/2024] [Accepted: 10/16/2024] [Indexed: 11/28/2024] Open
Abstract
Combining a T cell-targeting mRNA vaccine encoding the conserved SARS-CoV-2 RNA-dependent RNA polymerase, RdRp, with a Spike-encoding mRNA vaccine may offer an additional pathway toward COVID-19 protection. Here, we show that a nucleoside-modified RdRp mRNA vaccine raises robust and durable CD8+ T cell responses in mice. Immunization drives a CD8+ T cell response enriched toward a specific RdRp epitope. Unexpectedly, coadministration of mRNA vaccines encoding RdRp or the Spike Receptor Binding Domain (RBD) dampens RBD-specific immune responses. Contralateral administration reduces the suppression of RBD-specific T cell responses while type I interferon signaling blockade restores RBD-specific antibodies. A staggered immunization strategy maintains both RBD vaccine-mediated antibody and T cell responses as well as protection against lethal SARS-CoV-2 challenge in human ACE2 transgenic mice. In HLA-A2.1 transgenic mice, the RdRp vaccine elicits CD8+ T cell responses against HLA-A*02:01-restricted epitopes recognized by human donor T cells. These results highlight RdRp as a candidate antigen for COVID-19 vaccines. The findings also offer insights into crafting effective multivalent mRNA vaccines to broaden CD8+ T cell responses against SARS-CoV-2 and potentially other viruses with pandemic potential.
Collapse
Affiliation(s)
- Evan R. Abt
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA90095
| | - Alex K. Lam
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA90095
| | - Miyako Noguchi
- Department of Microbiology, Immunology, and Molecular Genetics, University of California Los Angeles, Los Angeles, CA90095
| | - Khalid Rashid
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA90095
| | - Jami McLaughlin
- Department of Microbiology, Immunology, and Molecular Genetics, University of California Los Angeles, Los Angeles, CA90095
| | - Pu-Lin Teng
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA90095
| | - Wendy Tran
- Department of Microbiology, Immunology, and Molecular Genetics, University of California Los Angeles, Los Angeles, CA90095
| | - Donghui Cheng
- Department of Microbiology, Immunology, and Molecular Genetics, University of California Los Angeles, Los Angeles, CA90095
| | - Pavlo A. Nesterenko
- Department of Microbiology, Immunology, and Molecular Genetics, University of California Los Angeles, Los Angeles, CA90095
| | - Zhiyuan Mao
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA90095
| | - Amanda L. Creech
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA90095
| | - Giselle Burton Sojo
- Department of Microbiology, Immunology, and Molecular Genetics, University of California Los Angeles, Los Angeles, CA90095
| | - Arjit Vijey Jeyachandran
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA90095
| | - Ying K. Tam
- Acuitas Therapeutics, Vancouver, BCV6T 1Z3, Canada
| | - Jill E. Henley
- Department of Molecular Microbiology and Immunology, The Hastings and Wright Laboratories, Keck School of Medicine, University of Southern California, Los Angeles, CA90089
| | - Lucio Comai
- Department of Molecular Microbiology and Immunology, The Hastings and Wright Laboratories, Keck School of Medicine, University of Southern California, Los Angeles, CA90089
| | - Norbert Pardi
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | | | - Owen N. Witte
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA90095
- Department of Microbiology, Immunology, and Molecular Genetics, University of California Los Angeles, Los Angeles, CA90095
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA90095
- Molecular Biology Institute, University of California Los Angeles, Los Angeles, CA90095
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California Los Angeles, Los Angeles, CA90095
- Parker Institute for Cancer Immunotherapy, University of California Los Angeles, Los Angeles, CA90095
| | - Caius G. Radu
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA90095
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA90095
| | - Ting-Ting Wu
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA90095
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA90095
- AIDS Institute, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA90095
| |
Collapse
|
37
|
Huang L, Zhao T, Zhao W, Shao A, Zhao H, Ma W, Gong Y, Zeng X, Weng C, Bu L, Di Z, Sun S, Dai Q, Sun M, Wang L, Liu Z, Shi L, Hu J, Fang S, Zhang C, Zhang J, Wang G, Loré K, Yang Y, Lin A. Herpes zoster mRNA vaccine induces superior vaccine immunity over licensed vaccine in mice and rhesus macaques. Emerg Microbes Infect 2024; 13:2309985. [PMID: 38258878 PMCID: PMC10860463 DOI: 10.1080/22221751.2024.2309985] [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: 08/18/2023] [Accepted: 01/19/2024] [Indexed: 01/24/2024]
Abstract
Herpes zoster remains an important global health issue and mainly occurs in aged and immunocompromised individuals with an early exposure history to Varicella Zoster Virus (VZV). Although the licensed vaccine Shingrix has remarkably high efficacy, undesired reactogenicity and increasing global demand causing vaccine shortage urged the development of improved or novel VZV vaccines. In this study, we developed a novel VZV mRNA vaccine candidate (named as ZOSAL) containing sequence-optimized mRNAs encoding full-length glycoprotein E encapsulated in an ionizable lipid nanoparticle. In mice and rhesus macaques, ZOSAL demonstrated superior immunogenicity and safety in multiple aspects over Shingrix, especially in the induction of strong T-cell immunity. Transcriptomic analysis revealed that both ZOSAL and Shingrix could robustly activate innate immune compartments, especially Type-I IFN signalling and antigen processing/presentation. Multivariate correlation analysis further identified several early factors of innate compartments that can predict the magnitude of T-cell responses, which further increased our understanding of the mode of action of two different VZV vaccine modalities. Collectively, our data demonstrated the superiority of VZV mRNA vaccine over licensed subunit vaccine. The mRNA platform therefore holds prospects for further investigations in next-generation VZV vaccine development.
Collapse
Affiliation(s)
- Lulu Huang
- Vaccine Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, People’s Republic of China
- Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing, People’s Republic of China
| | - Tongyi Zhao
- Vaccine Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, People’s Republic of China
- Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing, People’s Republic of China
| | - Weijun Zhao
- Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing, People’s Republic of China
| | - Andong Shao
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, People’s Republic of China
| | - Huajun Zhao
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, Jinan, People’s Republic of China
| | - Wenxuan Ma
- Vaccine Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, People’s Republic of China
- Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing, People’s Republic of China
| | - Yingfei Gong
- Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing, People’s Republic of China
| | - Xianhuan Zeng
- Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing, People’s Republic of China
| | - Changzhen Weng
- Vaccine Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, People’s Republic of China
- Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing, People’s Republic of China
| | - Lingling Bu
- Vaccine Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, People’s Republic of China
- Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing, People’s Republic of China
| | - Zhenhua Di
- Vaccine Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, People’s Republic of China
- Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing, People’s Republic of China
| | - Shiyu Sun
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Qinsheng Dai
- Targeted Discovery Center, China Pharmaceutical University, Nanjing, People’s Republic of China
| | - Minhui Sun
- Targeted Discovery Center, China Pharmaceutical University, Nanjing, People’s Republic of China
| | - Limei Wang
- Advanced Medical Research Institute, Shandong University, Jinan, People’s Republic of China
| | - Zhenguang Liu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, People’s Republic of China
| | - Leilei Shi
- Precision Research Center for Refractory Diseases in Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Jiesen Hu
- Firestone Biotechnologies, Shanghai, People’s Republic of China
| | - Shentong Fang
- School of Biopharmacy, China Pharmaceutical University, Nanjing, People’s Republic of China
| | - Cheng Zhang
- Department of Immunology, College of Basic Medical Science, Dalian Medical University, Dalian, People’s Republic of China
| | - Jian Zhang
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, People’s Republic of China
| | - Guan Wang
- Department of Immunology, College of Basic Medical Science, Dalian Medical University, Dalian, People’s Republic of China
| | - Karin Loré
- Department of Medicine, Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Yong Yang
- Vaccine Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, People’s Republic of China
- Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing, People’s Republic of China
- School of Pharmacy, Xuzhou Medical University, Xuzhou, People’s Republic of China
| | - Ang Lin
- Vaccine Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, People’s Republic of China
- Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing, People’s Republic of China
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, Jinan, People’s Republic of China
| |
Collapse
|
38
|
Smith PP, Chicca IJ, Heaney JLJ, Muchova M, Khanim FL, Shields AM, Drayson MT, Chapple ILC, Hirschfeld J. Paracetamol suppresses neutrophilic oxygen radicals through competitive inhibition and scavenging. Chem Biol Interact 2024; 404:111283. [PMID: 39428054 DOI: 10.1016/j.cbi.2024.111283] [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: 08/15/2024] [Revised: 10/07/2024] [Accepted: 10/18/2024] [Indexed: 10/22/2024]
Abstract
Neutrophils, pivotal cells of innate and adaptive immune responses, employ reactive oxygen species (ROS) to combat pathogens and control gene expression. Paracetamol (acetaminophen) is widely used as an analgesic and antipyretic medication, yet its precise mechanisms of action are not yet fully understood. Here, we investigate the impact of both ingested and in-vitro paracetamol on neutrophil ROS activity, using flow cytometry and antioxidant assays. Our studies reveal that paracetamol significantly suppresses ROS activity ex-vivo in the short term. Additionally, both paracetamol and its metabolite N-acetyl-p-benzoquinone imine exhibited direct in vitro antioxidant effects, and paracetamol suppressed neutrophil extracellular trap formation ex vivo. These findings suggest a connection between paracetamol use and altered neutrophil responses, with potential implications for use in some patient groups, such as immunocompromised individuals. Further investigation into paracetamol's effects on neutrophil antimicrobial functions is warranted to elucidate possible risks, particularly when taken frequently or in conjunction with other treatments such as vaccinations.
Collapse
Affiliation(s)
- Peter P Smith
- College of Medicine and Health, School of Health Sciences, Dentistry, Periodontal Research Group, University of Birmingham, Birmingham, UK
| | - Ilaria J Chicca
- College of Medical and Health, School of Infection, Inflammation and Immunology, Clinical Immunology Service, University of Birmingham, Birmingham, UK
| | - Jennifer L J Heaney
- College of Medical and Health, School of Infection, Inflammation and Immunology, Clinical Immunology Service, University of Birmingham, Birmingham, UK
| | - Maria Muchova
- College of Medicine and Health, School of Health Sciences, Dentistry, Periodontal Research Group, University of Birmingham, Birmingham, UK
| | - Farhat L Khanim
- College of Medical and Health, School of Infection, Inflammation and Immunology, Clinical Immunology Service, University of Birmingham, Birmingham, UK
| | - Adrian M Shields
- College of Medical and Health, School of Infection, Inflammation and Immunology, Clinical Immunology Service, University of Birmingham, Birmingham, UK
| | - Mark T Drayson
- College of Medical and Health, School of Infection, Inflammation and Immunology, Clinical Immunology Service, University of Birmingham, Birmingham, UK
| | - Iain L C Chapple
- College of Medicine and Health, School of Health Sciences, Dentistry, Periodontal Research Group, University of Birmingham, Birmingham, UK; Birmingham NIHR Biomedical Research Centre in Inflammation, University of Birmingham, Birmingham, UK.
| | - Josefine Hirschfeld
- College of Medicine and Health, School of Health Sciences, Dentistry, Periodontal Research Group, University of Birmingham, Birmingham, UK; Birmingham NIHR Biomedical Research Centre in Inflammation, University of Birmingham, Birmingham, UK.
| |
Collapse
|
39
|
Kroemer G, Montégut L, Kepp O, Zitvogel L. The danger theory of immunity revisited. Nat Rev Immunol 2024; 24:912-928. [PMID: 39511426 DOI: 10.1038/s41577-024-01102-9] [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: 10/09/2024] [Indexed: 11/15/2024]
Abstract
The danger theory of immunity, introduced by Polly Matzinger in 1994, posits that tissue stress, damage or infection has a decisive role in determining immune responses. Since then, a growing body of evidence has supported the idea that the capacity to elicit cognate immune responses (immunogenicity) relies on the combination of antigenicity (the ability to be recognized by T cell receptors or antibodies) and adjuvanticity (additional signals arising owing to tissue damage). Here, we discuss the molecular foundations of the danger theory while focusing on immunologically relevant damage-associated molecular patterns, microorganism-associated molecular patterns, and neuroendocrine stress-associated immunomodulatory molecules, as well as on their receptors. We critically evaluate patient-relevant evidence, examining how cancer cells and pathogenic viruses suppress damage-associated molecular patterns to evade immune recognition, how intestinal dysbiosis can reduce immunostimulatory microorganism-associated molecular patterns and compromise immune responses, and which hereditary immune defects support the validity of the danger theory. Furthermore, we incorporate the danger hypothesis into a close-to-fail-safe hierarchy of immunological tolerance mechanisms that also involve the clonal deletion and inactivation of immune cells.
Collapse
Affiliation(s)
- Guido Kroemer
- Centre de Recherche des Cordeliers, INSERM U1138, Équipe Labellisée - Ligue Nationale contre le Cancer, Université Paris Cité, Sorbonne Université, Paris, France.
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France.
- Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, Paris, France.
| | - Léa Montégut
- Centre de Recherche des Cordeliers, INSERM U1138, Équipe Labellisée - Ligue Nationale contre le Cancer, Université Paris Cité, Sorbonne Université, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Oliver Kepp
- Centre de Recherche des Cordeliers, INSERM U1138, Équipe Labellisée - Ligue Nationale contre le Cancer, Université Paris Cité, Sorbonne Université, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Laurence Zitvogel
- Gustave Roussy Cancer Campus, Clinicobiome, Villejuif, France.
- INSERM UMR 1015, ClinicObiome, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France.
- Université Paris-Saclay, Ile-de-France, Paris, France.
- Center of Clinical Investigations in Biotherapies of Cancer (BIOTHERIS), Villejuif, France.
| |
Collapse
|
40
|
Scholaert M, Peries M, Braun E, Martin J, Serhan N, Loste A, Bruner A, Basso L, Chaput B, Merle E, Descargues P, Pagès E, Gaudenzio N. Multimodal profiling of biostabilized human skin modules reveals a coordinated ecosystem response to injected mRNA-1273 COVID-19 vaccine. Allergy 2024; 79:3341-3359. [PMID: 39157907 PMCID: PMC11657073 DOI: 10.1111/all.16273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 06/26/2024] [Accepted: 06/28/2024] [Indexed: 08/20/2024]
Abstract
BACKGROUND The field of drug development is witnessing a remarkable surge in the development of innovative strategies. There is a need to develop technological platforms capable of generating human data prior to progressing to clinical trials. METHODS Here we introduce a new flexible solution designed for the comprehensive monitoring of the natural human skin ecosystem's response to immunogenic drugs over time. Based on unique bioengineering to preserve surgical resections in a long survival state, it allows for the first time a comprehensive analysis of resident immune cells response at both organ and single-cell levels. RESULTS Upon injection of the mRNA-1273 COVID-19 vaccine, we characterized precise sequential molecular events triggered upon detection of the exogenous substance. The vaccine consistently targets DC/macrophages and mast cells, regardless of the administration route, while promoting specific cell-cell communications in surrounding immune cell subsets. CONCLUSION Given its direct translational relevance, this approach provides a multiscale vision of genuine human tissue immunity that could pave the way toward the development of new vaccination and drug development strategies.
Collapse
Affiliation(s)
- Manon Scholaert
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity) INSERM UMR1291—CNRS UMR5051University Toulouse IIIToulouseFrance
- Genoskin SASToulouseFrance
| | | | | | - Jeremy Martin
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity) INSERM UMR1291—CNRS UMR5051University Toulouse IIIToulouseFrance
| | - Nadine Serhan
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity) INSERM UMR1291—CNRS UMR5051University Toulouse IIIToulouseFrance
| | - Alexia Loste
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity) INSERM UMR1291—CNRS UMR5051University Toulouse IIIToulouseFrance
| | - Audrey Bruner
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity) INSERM UMR1291—CNRS UMR5051University Toulouse IIIToulouseFrance
| | - Lilian Basso
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity) INSERM UMR1291—CNRS UMR5051University Toulouse IIIToulouseFrance
| | - Benoît Chaput
- Department of Plastic, Reconstructive and Aesthetic Surgery, Rangueil HospitalCHU ToulouseToulouseFrance
| | | | | | | | - Nicolas Gaudenzio
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity) INSERM UMR1291—CNRS UMR5051University Toulouse IIIToulouseFrance
- Genoskin SASToulouseFrance
| |
Collapse
|
41
|
Cahuapaza-Gutierrez NL. Aplastic Anemia in the light of the COVID-19 pandemic: infection, vaccination, and pathophysiologic mechanisms. Ann Hematol 2024; 103:4989-5005. [PMID: 39441353 DOI: 10.1007/s00277-024-06052-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 10/18/2024] [Indexed: 10/25/2024]
Abstract
Patients infected with SARS-CoV-2 and vaccinated against COVID-19 could develop aplastic anemia (AA). Comprehensive review and analysis were conducted through a selective literature search in PubMed, Scopus, EMBASE, and Web of Science. For this analysis, 26 studies were included, comprising 16 case reports, 7 case series, and 3 observational studies, totaling 53 patients. The causes of acquired or idiopathic AA are diverse; this review presents recent findings, including possible new etiologies such as SARS-CoV-2 infection and COVID-19 vaccines. This possible association is explored, addressing the existing gap, and aiming to improve daily medical practice. This article reviews the relationship between AA and SARS-CoV-2 infection, as well as COVID-19 vaccines, analyzing cases of de novo occurrence and relapses of AA. Although a definitive mechanistic link has not yet been established, possible underlying pathophysiological mechanisms are explored.
Collapse
Affiliation(s)
- Nelson Luis Cahuapaza-Gutierrez
- Facultad de Ciencias de La Salud, Carrera de Medicina Humana, Universidad Científica del Sur, Lima, Perú.
- CHANGE Research Working Group, Universidad Científica del Sur, Lima, Perú.
| |
Collapse
|
42
|
Gupta A, Rudra A, Reed K, Langer R, Anderson DG. Advanced technologies for the development of infectious disease vaccines. Nat Rev Drug Discov 2024; 23:914-938. [PMID: 39433939 DOI: 10.1038/s41573-024-01041-z] [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] [Accepted: 08/28/2024] [Indexed: 10/23/2024]
Abstract
Vaccines play a critical role in the prevention of life-threatening infectious disease. However, the development of effective vaccines against many immune-evading pathogens such as HIV has proven challenging, and existing vaccines against some diseases such as tuberculosis and malaria have limited efficacy. The historically slow rate of vaccine development and limited pan-variant immune responses also limit existing vaccine utility against rapidly emerging and mutating pathogens such as influenza and SARS-CoV-2. Additionally, reactogenic effects can contribute to vaccine hesitancy, further undermining the ability of vaccination campaigns to generate herd immunity. These limitations are fuelling the development of novel vaccine technologies to more effectively combat infectious diseases. Towards this end, advances in vaccine delivery systems, adjuvants, antigens and other technologies are paving the way for the next generation of vaccines. This Review focuses on recent advances in synthetic vaccine systems and their associated challenges, highlighting innovation in the field of nano- and nucleic acid-based vaccines.
Collapse
Affiliation(s)
- Akash Gupta
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Arnab Rudra
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Boston, MA, USA
| | - Kaelan Reed
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Robert Langer
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Harvard and MIT Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Daniel G Anderson
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Boston, MA, USA.
- Harvard and MIT Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
43
|
Tan C, xiao Y, Liu T, Chen S, Zhou J, Zhang S, Hu Y, Wu A, Li C. Development of multi-epitope mRNA vaccine against Clostridioides difficile using reverse vaccinology and immunoinformatics approaches. Synth Syst Biotechnol 2024; 9:667-683. [PMID: 38817826 PMCID: PMC11137598 DOI: 10.1016/j.synbio.2024.05.008] [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: 01/08/2024] [Revised: 04/28/2024] [Accepted: 05/15/2024] [Indexed: 06/01/2024] Open
Abstract
Clostridioides difficile (C. difficile), as the major pathogen of diarrhea in healthcare settings, has become increasingly prevalent within community populations, resulting in significant morbidity and mortality. However, the therapeutic options for Clostridioides difficile infection (CDI) remain limited, and as of now, no authorized vaccine is available to combat this disease. Therefore, the development of a novel vaccine against C. difficile is of paramount importance. In our study, the complete proteome sequences of 118 strains of C. difficile were downloaded and analyzed. We found four antigenic proteins that were highly conserved and can be used for epitope identification. We designed two vaccines, WLcd1 and WLcd2, that contain the ideal T-cell and B-cell epitopes, adjuvants, and the pan HLA DR-binding epitope (PADRE) sequences. The biophysical and chemical assessments of these vaccine candidates indicated that they were suitable for immunogenic applications. Molecular docking analyses revealed that WLcd1 bonded with higher affinity to Toll-like receptors (TLRs) than WLcd2. Furthermore, molecular dynamics (MD) simulations, performed using Gmx_MMPBSA v1.56, confirmed the binding stability of WLcd1 with TLR2 and TLR4. The preliminary findings suggested that this multi-epitope vaccine could be a promising candidate for protection against CDI; however, experimental studies are necessary to confirm these predictions.
Collapse
Affiliation(s)
- Caixia Tan
- Infection Control Center, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China
- National Clinical Research Center for Geriatric Disorders (XiangYa Hospital), Changsha, Hunan Province, 410008, China
| | - Yuanyuan xiao
- Infection Control Center, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China
- National Clinical Research Center for Geriatric Disorders (XiangYa Hospital), Changsha, Hunan Province, 410008, China
| | - Ting Liu
- Infection Control Center, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China
- National Clinical Research Center for Geriatric Disorders (XiangYa Hospital), Changsha, Hunan Province, 410008, China
| | - Siyao Chen
- Infection Control Center, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China
- National Clinical Research Center for Geriatric Disorders (XiangYa Hospital), Changsha, Hunan Province, 410008, China
| | - Juan Zhou
- Infection Control Center, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China
- National Clinical Research Center for Geriatric Disorders (XiangYa Hospital), Changsha, Hunan Province, 410008, China
| | - Sisi Zhang
- Infection Control Center, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China
- National Clinical Research Center for Geriatric Disorders (XiangYa Hospital), Changsha, Hunan Province, 410008, China
| | - Yiran Hu
- Infection Control Center, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China
- National Clinical Research Center for Geriatric Disorders (XiangYa Hospital), Changsha, Hunan Province, 410008, China
| | - Anhua Wu
- Infection Control Center, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China
- National Clinical Research Center for Geriatric Disorders (XiangYa Hospital), Changsha, Hunan Province, 410008, China
| | - Chunhui Li
- Infection Control Center, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China
- National Clinical Research Center for Geriatric Disorders (XiangYa Hospital), Changsha, Hunan Province, 410008, China
| |
Collapse
|
44
|
Catenacci L, Rossi R, Sechi F, Buonocore D, Sorrenti M, Perteghella S, Peviani M, Bonferoni MC. Effect of Lipid Nanoparticle Physico-Chemical Properties and Composition on Their Interaction with the Immune System. Pharmaceutics 2024; 16:1521. [PMID: 39771501 PMCID: PMC11728546 DOI: 10.3390/pharmaceutics16121521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 11/19/2024] [Accepted: 11/24/2024] [Indexed: 01/16/2025] Open
Abstract
Lipid nanoparticles (LNPs) have shown promise as a delivery system for nucleic acid-based therapeutics, including DNA, siRNA, and mRNA vaccines. The immune system plays a critical role in the response to these nanocarriers, with innate immune cells initiating an early response and adaptive immune cells mediating a more specific reaction, sometimes leading to potential adverse effects. Recent studies have shown that the innate immune response to LNPs is mediated by Toll-like receptors (TLRs) and other pattern recognition receptors (PRRs), which recognize the lipid components of the nanoparticles. This recognition can trigger the activation of inflammatory pathways and the production of cytokines and chemokines, leading to potential adverse effects such as fever, inflammation, and pain at the injection site. On the other hand, the adaptive immune response to LNPs appears to be primarily directed against the protein encoded by the mRNA cargo, with little evidence of an ongoing adaptive immune response to the components of the LNP itself. Understanding the relationship between LNPs and the immune system is critical for the development of safe and effective nucleic acid-based delivery systems. In fact, targeting the immune system is essential to develop effective vaccines, as well as therapies against cancer or infections. There is a lack of research in the literature that has systematically studied the factors that influence the interaction between LNPs and the immune system and further research is needed to better elucidate the mechanisms underlying the immune response to LNPs. In this review, we discuss LNPs' composition, physico-chemical properties, such as size, shape, and surface charge, and the protein corona formation which can affect the reactivity of the immune system, thus providing a guide for the research on new formulations that could gain a favorable efficacy/safety profile.
Collapse
Affiliation(s)
- Laura Catenacci
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy; (L.C.); (R.R.); (F.S.); (M.S.); (M.C.B.)
| | - Rachele Rossi
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy; (L.C.); (R.R.); (F.S.); (M.S.); (M.C.B.)
| | - Francesca Sechi
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy; (L.C.); (R.R.); (F.S.); (M.S.); (M.C.B.)
| | - Daniela Buonocore
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, 27100 Pavia, Italy;
| | - Milena Sorrenti
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy; (L.C.); (R.R.); (F.S.); (M.S.); (M.C.B.)
| | - Sara Perteghella
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy; (L.C.); (R.R.); (F.S.); (M.S.); (M.C.B.)
| | - Marco Peviani
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, 27100 Pavia, Italy;
| | - Maria Cristina Bonferoni
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy; (L.C.); (R.R.); (F.S.); (M.S.); (M.C.B.)
| |
Collapse
|
45
|
Dowell W, Dearborn J, Languon S, Miller Z, Kirch T, Paige S, Garvin O, Kjendal L, Harby E, Zuchowski AB, Clark E, Lescieur-Garcia C, Vix J, Schumer A, Mistri SK, Snoke DB, Doiron AL, Freeman K, Toth MJ, Poynter ME, Boyson JE, Majumdar D. Distinct Inflammatory Programs Underlie the Intramuscular Lipid Nanoparticle Response. ACS NANO 2024. [PMID: 39563529 DOI: 10.1021/acsnano.4c08490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2024]
Abstract
Developments in mRNA/lipid nanoparticle (LNP) technology have advanced the fields of vaccinology and gene therapy, raising questions about immunogenicity. While some mRNA/LNPs generate an adjuvant-like environment in muscle tissue, other mRNA/LNPs are distinct in their capacity for multiple rounds of therapeutic delivery. We evaluate the adjuvancy of components of mRNA/LNPs by phenotyping cellular infiltrate at injection sites, tracking uptake by immune cells, and assessing the inflammatory state. Delivery of 9 common, but chemically distinct, LNPs to muscle revealed two classes of inflammatory gene expression programs: inflammatory (Class A) and noninflammatory (Class B). We find that intramuscular injection with Class A, but not Class B, empty LNPs (eLNPs) induce robust neutrophil infiltration into muscle within 2 h and a diverse myeloid population within 24 h. Single-cell RNA sequencing revealed SM-102-mediated expression of inflammatory chemokines by myeloid infiltrates within muscle 1 day after injection. Surprisingly, we found direct transfection of muscle infiltrating myeloid cells and splenocytes 24 h after intramuscular mRNA/LNP administration. Transfected myeloid cells within the muscle exhibit an activated phenotype 24 h after injection. Similarly, directly transfected splenic lymphocytes and dendritic cells (DCs) are differentially activated by Class A or Class B containing mRNA/LNP. Within the splenic DC compartment, type II conventional DCs (cDC2s) are directly transfected and activated by Class A mRNA/LNP. Together, we show that mRNA and LNPs work synergistically to provide the necessary innate immune stimuli required for effective vaccination. Importantly, this work provides a design framework for vaccines and therapeutics alike.
Collapse
Affiliation(s)
- William Dowell
- Department of Surgery; Larner College of Medicine, University of Vermont, Burlington, Vermont 05405, United States
- Cellular, Molecular, and Biomedical Sciences Program, Burlington, Vermont 05405, United States
| | - Jacob Dearborn
- Department of Surgery; Larner College of Medicine, University of Vermont, Burlington, Vermont 05405, United States
- Cellular, Molecular, and Biomedical Sciences Program, Burlington, Vermont 05405, United States
| | - Sylvester Languon
- Department of Surgery; Larner College of Medicine, University of Vermont, Burlington, Vermont 05405, United States
- Cellular, Molecular, and Biomedical Sciences Program, Burlington, Vermont 05405, United States
| | - Zachary Miller
- Department of Surgery; Larner College of Medicine, University of Vermont, Burlington, Vermont 05405, United States
- Cellular, Molecular, and Biomedical Sciences Program, Burlington, Vermont 05405, United States
| | - Tylar Kirch
- Department of Surgery; Larner College of Medicine, University of Vermont, Burlington, Vermont 05405, United States
- Cellular, Molecular, and Biomedical Sciences Program, Burlington, Vermont 05405, United States
| | - Stephen Paige
- Department of Electrical and Biomedical Engineering, University of Vermont, Burlington, Vermont 05405, United States
| | - Olivia Garvin
- Department of Surgery; Larner College of Medicine, University of Vermont, Burlington, Vermont 05405, United States
| | - Lily Kjendal
- Department of Surgery; Larner College of Medicine, University of Vermont, Burlington, Vermont 05405, United States
| | - Ethan Harby
- Department of Surgery; Larner College of Medicine, University of Vermont, Burlington, Vermont 05405, United States
| | - Adam B Zuchowski
- Department of Surgery; Larner College of Medicine, University of Vermont, Burlington, Vermont 05405, United States
| | - Emily Clark
- Department of Surgery; Larner College of Medicine, University of Vermont, Burlington, Vermont 05405, United States
| | - Carlos Lescieur-Garcia
- Department of Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont 05405, United States
| | - Jesse Vix
- Department of Surgery; Larner College of Medicine, University of Vermont, Burlington, Vermont 05405, United States
| | - Amy Schumer
- Department of Surgery; Larner College of Medicine, University of Vermont, Burlington, Vermont 05405, United States
- Department of Obstetrics, Gynecology and Reproductive Sciences, Larner College of Medicine, University of Vermont, Burlington, Vermont 05405, United States
| | - Somen K Mistri
- Department of Surgery; Larner College of Medicine, University of Vermont, Burlington, Vermont 05405, United States
| | - Deena B Snoke
- Department of Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont 05405, United States
| | - Amber L Doiron
- Department of Electrical and Biomedical Engineering, University of Vermont, Burlington, Vermont 05405, United States
| | - Kalev Freeman
- Department of Emergency Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont 05405, United States
| | - Michael J Toth
- Department of Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont 05405, United States
| | - Matthew E Poynter
- Department of Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont 05405, United States
| | - Jonathan E Boyson
- Department of Surgery; Larner College of Medicine, University of Vermont, Burlington, Vermont 05405, United States
| | - Devdoot Majumdar
- Department of Surgery; Larner College of Medicine, University of Vermont, Burlington, Vermont 05405, United States
| |
Collapse
|
46
|
Tani H. Recent Advances and Prospects in RNA Drug Development. Int J Mol Sci 2024; 25:12284. [PMID: 39596348 PMCID: PMC11594839 DOI: 10.3390/ijms252212284] [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: 10/23/2024] [Revised: 11/12/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024] Open
Abstract
RNA therapeutics have undergone remarkable evolution since their inception in the late 1970s, revolutionizing medicine by offering new possibilities for treating previously intractable diseases. The field encompasses various modalities, including antisense oligonucleotides (ASOs), small interfering RNAs (siRNAs), microRNAs (miRNAs), and messenger RNAs (mRNAs), each with unique mechanisms and applications. The foundation was laid in 1978 with the discovery that synthetic oligonucleotides could inhibit viral replication, followed by pivotal developments such as RNA interference's discovery in 1998. The COVID-19 pandemic marked a crucial turning point, demonstrating the potential of mRNA vaccines and accelerating interest in RNA-based approaches. However, significant challenges remain, including stability issues, delivery to target tissues, potential off-target effects, and immunogenicity concerns. Recent advancements in chemical modifications, delivery systems, and the integration of AI technologies are addressing these challenges. The field has seen notable successes, such as approved treatments for spinal muscular atrophy and hereditary transthyretin-mediated amyloidosis. Looking ahead, RNA therapeutics show promise for personalized medicine approaches, particularly in treating genetic disorders and cancer. The continued evolution of this field, driven by technological innovations and deeper understanding of RNA biology, suggests a transformative impact on future medical treatments. The purpose of this review is to provide a comprehensive overview of the evolution, current state, and prospects of RNA therapeutics.
Collapse
Affiliation(s)
- Hidenori Tani
- Department of Health Pharmacy, Yokohama University of Pharmacy, 601 Matano, Totsuka, Yokohama 245-0066, Japan
| |
Collapse
|
47
|
Haghighi E, Abolmaali SS, Dehshahri A, Mousavi Shaegh SA, Azarpira N, Tamaddon AM. Navigating the intricate in-vivo journey of lipid nanoparticles tailored for the targeted delivery of RNA therapeutics: a quality-by-design approach. J Nanobiotechnology 2024; 22:710. [PMID: 39543630 PMCID: PMC11566655 DOI: 10.1186/s12951-024-02972-w] [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: 08/29/2024] [Accepted: 11/03/2024] [Indexed: 11/17/2024] Open
Abstract
RNA therapeutics, such as mRNA, siRNA, and CRISPR-Cas9, present exciting avenues for treating diverse diseases. However, their potential is commonly hindered by vulnerability to degradation and poor cellular uptake, requiring effective delivery systems. Lipid nanoparticles (LNPs) have emerged as a leading choice for in vivo RNA delivery, offering protection against degradation, enhanced cellular uptake, and facilitation of endosomal escape. However, LNPs encounter numerous challenges for targeted RNA delivery in vivo, demanding advanced particle engineering, surface functionalization with targeting ligands, and a profound comprehension of the biological milieu in which they function. This review explores the structural and physicochemical characteristics of LNPs, in-vivo fate, and customization for RNA therapeutics. We highlight the quality-by-design (QbD) approach for targeted delivery beyond the liver, focusing on biodistribution, immunogenicity, and toxicity. In addition, we explored the current challenges and strategies associated with LNPs for in-vivo RNA delivery, such as ensuring repeated-dose efficacy, safety, and tissue-specific gene delivery. Furthermore, we provide insights into the current clinical applications in various classes of diseases and finally prospects of LNPs in RNA therapeutics.
Collapse
Affiliation(s)
- Elahe Haghighi
- Department of Pharmaceutical Nanotechnology, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Samira Sadat Abolmaali
- Department of Pharmaceutical Nanotechnology, Shiraz University of Medical Sciences, Shiraz, Iran.
- Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Ali Dehshahri
- Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seyed Ali Mousavi Shaegh
- Laboratory of Microfluidics and Medical Microsystems, Research Institute for Medical Sciences, Mashhad University of Medical Sciences, Mashhad, Iran
- Orthopedic Research Center, Ghaem Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
- Clinical Research Development Unit, Ghaem Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Negar Azarpira
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Mohammad Tamaddon
- Department of Pharmaceutical Nanotechnology, Shiraz University of Medical Sciences, Shiraz, Iran.
- Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz, Iran.
- Department of Pharmaceutics, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
48
|
Wu K, Xu F, Dai Y, Jin S, Zheng A, Zhang N, Xu Y. Characterization of mRNA-LNP structural features and mechanisms for enhanced mRNA vaccine immunogenicity. J Control Release 2024; 376:1288-1299. [PMID: 39522562 DOI: 10.1016/j.jconrel.2024.11.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 10/30/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
Lipid nanoparticles (LNPs) used for nonviral gene delivery have achieved significant success, particularly in COVID-19 mRNA vaccines. LNPs are routinely characterized by their particle size, polydispersity, and mRNA loading efficiency. However, the internal structure of these particles has not been specified, despite evidence showing that LNPs can be highly heterogeneous, with variations in lipid composition and preparation methods. How these structural features contributed to mRNA LNP vaccine activities is also unclear. In this study, we prepared LNPs with distinctly different internal structures. They were named the emulsion-like LNPs (eLNPs) and membrane-rich LNPs (mLNPs) respectively and compared with the classic "bleb" structure LNPs (cLNPs). The eLNPs contained higher molar percent of the ionizable lipid and lower molar percent of DSPC and cholesterol. The different lipid organization structures lead to varying mRNA delivery activities in vitro and in vivo. After intramuscular injection, eLNPs remained at the injection site and expressed antigens locally. The resulted immune responses had a very fast onset (higher titer at week 2) and lasted longer and stronger (higher titers at week 8) than other LNPs (cLNPs and mLNPs). We hypothesize that the rapid onset and local expression of antigens by muscle cells in the eLNP groups may be favored by the antigen recognition and presentation process, despite the overall mRNA expression activities was not as high especially in liver and other organ. Our data support that eLNPs are potentially the more suitable delivery system for mRNA vaccine due to their high immunogenicity and low systemic toxicity.
Collapse
Affiliation(s)
- Kangzeng Wu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, PR China; Zhejiang-California International Nanosystems Institute, Zhejiang University, Hangzhou 310058, PR China
| | - Fengwei Xu
- HighField Biopharmaceuticals Inc., Hangzhou 310000, PR China
| | - Yongchao Dai
- HighField Biopharmaceuticals Inc., Hangzhou 310000, PR China
| | - Shanshan Jin
- HighField Biopharmaceuticals Inc., Hangzhou 310000, PR China
| | - Anjie Zheng
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, PR China
| | - Ning Zhang
- Wisdom Lake Academy of Pharmacy, Xi'an Jiaotong-Liverpool University, Suzhou, PR China.
| | - Yuhong Xu
- College of Pharmacy, Dali University, Dali 671003, PR China; HighField Biopharmaceuticals Inc., Hangzhou 310000, PR China.
| |
Collapse
|
49
|
Zhang J, Xu Y, Chen M, Wang S, Lin G, Huang Y, Yang C, Yang Y, Song Y. Spatial Engineering of Heterotypic Antigens on a DNA Framework for the Preparation of Mosaic Nanoparticle Vaccines with Enhanced Immune Activation against SARS-CoV-2 Variants. Angew Chem Int Ed Engl 2024; 63:e202412294. [PMID: 39030890 DOI: 10.1002/anie.202412294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 07/11/2024] [Accepted: 07/17/2024] [Indexed: 07/22/2024]
Abstract
Mosaic nanoparticle vaccines with heterotypic antigens exhibit broad-spectrum antiviral capabilities, but the impact of antigen proportions and distribution patterns on vaccine-induced immunity remains largely unexplored. Here, we present a DNA nanotechnology-based strategy for spatially assembling heterotypic antigens to guide the rational design of mosaic nanoparticle vaccines. By utilizing two aptamers with orthogonal selectivity for the original SARS-CoV-2 spike trimer and Omicron receptor-binding domain (RBD), along with a DNA soccer-ball framework, we precisely manipulate the spacing, stoichiometry, and overall distribution of heterotypic antigens to create mosaic nanoparticles with average, bipolar, and unipolar antigen distributions. Systematic in vitro and in vivo immunological investigations demonstrate that 30 heterotypic antigens in equivalent proportions, with an average distribution, lead to higher production of broad-spectrum neutralizing antibodies compared to the bipolar and unipolar distributions. Furthermore, the precise assembly utilizing our developed methodology reveals that a mere increment of five Omicron RBD antigens on a nanoparticle (from 15 to 20) not only diminishes neutralization against the Omicron variant but also triggers excessive inflammation. This work provides a unique perspective on the rational design of mosaic vaccines by highlighting the significance of the spatial placement and proportion of heterotypic antigens in their structure-activity mechanisms.
Collapse
Affiliation(s)
- Jialu Zhang
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, the Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Xiamen University, Xiamen, Fujian, 361005, China
| | - Yunyun Xu
- Institute of Molecular Medicine and Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, State Key Laboratory of Oncogenes and Related Genes, Department of Laboratory Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Mingying Chen
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, the Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Xiamen University, Xiamen, Fujian, 361005, China
| | - Shengwen Wang
- Institute of Molecular Medicine and Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, State Key Laboratory of Oncogenes and Related Genes, Department of Laboratory Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
- College of Chemistry and Materials Science, Shanghai Normal University, Shanghai, 200234, China
| | - Guihong Lin
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, the Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Xiamen University, Xiamen, Fujian, 361005, China
| | - Yihao Huang
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, the Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Xiamen University, Xiamen, Fujian, 361005, China
| | - Chaoyong Yang
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, the Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Xiamen University, Xiamen, Fujian, 361005, China
- Institute of Molecular Medicine and Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, State Key Laboratory of Oncogenes and Related Genes, Department of Laboratory Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Yang Yang
- Institute of Molecular Medicine and Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, State Key Laboratory of Oncogenes and Related Genes, Department of Laboratory Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Yanling Song
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, the Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Xiamen University, Xiamen, Fujian, 361005, China
| |
Collapse
|
50
|
Manzo C, Isetta M, Castagna A. Infective agents and polymyalgia rheumatica: key discussion points emerging from a narrative review of published literature. Reumatologia 2024; 62:360-367. [PMID: 39677882 PMCID: PMC11635615 DOI: 10.5114/reum/194687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 10/16/2024] [Indexed: 12/17/2024] Open
Abstract
Introduction The aetiology of polymyalgia rheumatica (PMR) is unknown. Recently, reports on cases of PMR following the coronavirus disease 2019 (COVID-19) have revived the role of infection as an aetiological or triggering factor. It is estimated that patients with PMR have manifestations of giant cell arteritis (GCA) in < 20% of cases. To date, little is known on the potential role of infectious agents in facilitating this association. Given this background, we performed a review of published literature. Our first aim was to review and discuss the relationship between PMR and infective agents. Secondly, we compared data of PMR-only patients with PMR and overlapping GCA to seek any commonalities or differences regarding the type of infectious agent in these two subgroups. Material and methods We performed a non-systematic literature search on Embase and Medline (COVID interface) with the following search terms: "polymyalgia rheumatica" AND "infections" OR "infectious agents", both MESH headings and free-text (in each language they were written). Each paper's reference list was scanned for additional publications meeting this study's aim. When papers reported data partially presented in previous articles, we referred to the most recent published data. Abstracts submitted at conferences or from non-peer-reviewed sources were not included. Polymyalgia rheumatica following vaccinations was an additional exclusion criterion. Results Several infectious agents have been held responsible for PMR. However, no definite causal link has been identified so far. According to our review, the search for a specific infectious agent, however intriguing, appears to be stagnating. Genetic background and epigenetic regulation probably play a key role. However, topical studies are lacking. Polymyalgia rheumatica as an adverse event following immunization should be kept methodologically distinct from PMR following an acute infection, as the adjuvants in the vaccine can make a significant difference. Conclusions Finally, some infectious agents are able to replicate in human arteries or have an endothelium tropism. Whilst these can theoretically trigger GCA, their role in isolated PMR seems minimal.
Collapse
Affiliation(s)
- Ciro Manzo
- Department of Internal and Geriatric Medicine, Azienda Sanitaria Locale Napoli 3 sud, Rheumatologic Outpatient Clinic, Health District No. 59, Naples, Sant’Agnello, Italy
| | - Marco Isetta
- Central and North West London NHS Trust, England
| | - Alberto Castagna
- Department of Primary Care, Health District of Soverato, Azienda Sanitaria Provinciale Catanzaro, Italy
| |
Collapse
|