1
|
Ma X, Liu S, Fan B, Jin D, Miao L, Liu L, Du S, Lin J. Enhancing mRNA translation efficiency by introducing sequence optimized AU-rich elements in 3' UTR via HuR anchorage. MOLECULAR THERAPY. NUCLEIC ACIDS 2025; 36:102485. [PMID: 40125272 PMCID: PMC11930071 DOI: 10.1016/j.omtn.2025.102485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 02/10/2025] [Indexed: 03/25/2025]
Abstract
mRNA technology holds immense promise as an innovative therapeutic approach with applications spanning infectious disease vaccines, cancer immunotherapy, protein replacement, and gene editing. However, practical use of mRNA has been hindered by challenges such as low cellular stability and transient protein expression. For addressing these, we propose a novel strategy to optimize mRNA sequences, particularly in the untranslated region, by inserting adenylate/uridylate-rich elements (AU-rich elements) to enhance stability and protein expression. Our investigation revealed that integrating AU-rich elements between the open reading frame (ORF) and the 3' untranslated region (3' UTR) significantly enhances RNA stability compared with other insertion sites. We identified cytoplasmic Human antigen R (HuR) as an essential RNA-binding protein responsible for promoting mRNA stability and translation, confirmed through HuR knockdown experiments and pull-down assays between AU-rich elements and HuR. Through rational design, we optimized the sequence of natural AU-rich elements and identified the essential "AUUUA" element, which, with certain repeats, can increase protein expression up to 5-fold. To demonstrate the universality of AU-rich element sequences in enhancing mRNA translation, we switched the coding proteins from luciferase to EGFP, mCherry, and ovalbumin (OVA), finding that both natural and engineered AU-rich element sequences amplify the expression of these proteins. In conclusion, leveraging the functionalities of RNA-binding proteins and the natural regulation of RNA stability in the untranslated region represents a novel strategy to enhance mRNA pharmacokinetics in the cytoplasm, expanding the potential applications of mRNA in therapeutic drugs.
Collapse
Affiliation(s)
- Xinghuan Ma
- MOE Key Laboratory of Bio-Intelligent Manufacturing, School of Bioengineering, Dalian University of Technology, Dalian 116024, China
| | - Sujia Liu
- MOE Key Laboratory of Bio-Intelligent Manufacturing, School of Bioengineering, Dalian University of Technology, Dalian 116024, China
| | - Bangda Fan
- MOE Key Laboratory of Bio-Intelligent Manufacturing, School of Bioengineering, Dalian University of Technology, Dalian 116024, China
| | - Danni Jin
- MOE Key Laboratory of Bio-Intelligent Manufacturing, School of Bioengineering, Dalian University of Technology, Dalian 116024, China
| | - Lei Miao
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- Beijing Key Laboratory of Molecular Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Lin Liu
- MOE Key Laboratory of Bio-Intelligent Manufacturing, School of Bioengineering, Dalian University of Technology, Dalian 116024, China
| | - Shubo Du
- MOE Key Laboratory of Bio-Intelligent Manufacturing, School of Bioengineering, Dalian University of Technology, Dalian 116024, China
| | - Jiaqi Lin
- MOE Key Laboratory of Bio-Intelligent Manufacturing, School of Bioengineering, Dalian University of Technology, Dalian 116024, China
| |
Collapse
|
2
|
Krawczyk PS, Mazur M, Orzeł W, Gewartowska O, Jeleń S, Antczak W, Kasztelan K, Brouze A, Matylla-Kulińska K, Gumińska N, Tarkowski B, Owczarek EP, Affek K, Turowski P, Tudek A, Sroka M, Śpiewla T, Kusio-Kobiałka M, Wesołowska A, Nowis D, Golab J, Kowalska J, Jemielity J, Dziembowski A, Mroczek S. Re-adenylation by TENT5A enhances efficacy of SARS-CoV-2 mRNA vaccines. Nature 2025:10.1038/s41586-025-08842-1. [PMID: 40240603 DOI: 10.1038/s41586-025-08842-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 02/26/2025] [Indexed: 04/18/2025]
Abstract
Despite the widespread use of mRNA vaccines against COVID-19, little is known about the metabolism of therapeutic RNAs. Here we use nanopore sequencing1-3 to analyse individual therapeutic mRNA molecules, focusing on their poly(A) tails. We show that the Moderna mRNA-1273 vaccine4 has a poly(A) tail of around 100 nucleotides, followed by an mΨCmΨAG sequence. In cell lines, mRNA-1273 undergoes rapid degradation initiated by mΨCmΨAG removal, followed by CCR4-NOT-mediated deadenylation. However, in medically relevant preclinical models, particularly in macrophages, mRNA-1273 poly(A) tails are extended to up to 200 nucleotides by the TENT5A poly(A) polymerase5-7, which is induced by the vaccine. Re-adenylation, which stabilizes target mRNAs, is consistently observed in synthetic mRNAs that encode proteins targeted to the endoplasmic reticulum, such as ovalbumin or antigens from Zika virus8 or the malaria parasite9. The extent of re-adenylation varies: the BioNTech-Pfizer BNT162b2 vaccine10 shows less potent re-adenylation than mRNA-1273, which correlates with a smaller proportion of membrane-associated BNT162b2. This highlights the crucial role of spatial accessibility to ER-resident TENT5A in determining re-adenylation efficiency. In vivo, TENT5A is expressed in immune cells that take up mRNA vaccine, and TENT5A deficiency reduces specific immunoglobulin production for mRNA vaccines after immunization in mice. Overall, our findings reveal a principle for enhancing the efficacy of therapeutic mRNAs, paving the way for improvement.
Collapse
Affiliation(s)
- Paweł S Krawczyk
- Laboratory of RNA Biology, International Institute of Molecular and Cell Biology, Warsaw, Poland
| | - Michał Mazur
- Laboratory of RNA Biology, International Institute of Molecular and Cell Biology, Warsaw, Poland
| | - Wiktoria Orzeł
- Laboratory of RNA Biology, International Institute of Molecular and Cell Biology, Warsaw, Poland
- Institute of Genetics and Biotechnology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Olga Gewartowska
- Genome Engineering Facility, International Institute of Molecular and Cell Biology, Warsaw, Poland
| | - Sebastian Jeleń
- Laboratory of RNA Biology, International Institute of Molecular and Cell Biology, Warsaw, Poland
- Institute of Genetics and Biotechnology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Wiktor Antczak
- Laboratory of RNA Biology, International Institute of Molecular and Cell Biology, Warsaw, Poland
- Institute of Genetics and Biotechnology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Karolina Kasztelan
- Laboratory of RNA Biology, International Institute of Molecular and Cell Biology, Warsaw, Poland
| | - Aleksandra Brouze
- Laboratory of RNA Biology, International Institute of Molecular and Cell Biology, Warsaw, Poland
- Institute of Genetics and Biotechnology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Katarzyna Matylla-Kulińska
- Laboratory of RNA Biology, International Institute of Molecular and Cell Biology, Warsaw, Poland
- Institute of Genetics and Biotechnology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Natalia Gumińska
- Laboratory of RNA Biology, International Institute of Molecular and Cell Biology, Warsaw, Poland
| | - Bartosz Tarkowski
- Laboratory of RNA Biology, International Institute of Molecular and Cell Biology, Warsaw, Poland
| | - Ewelina P Owczarek
- Laboratory of RNA Biology, International Institute of Molecular and Cell Biology, Warsaw, Poland
| | - Kamila Affek
- Laboratory of RNA Biology, International Institute of Molecular and Cell Biology, Warsaw, Poland
| | | | | | - Małgorzata Sroka
- Laboratory of Protein Structure, International Institute of Molecular and Cell Biology, Warsaw, Poland
| | - Tomasz Śpiewla
- Faculty of Physics, University of Warsaw, Warsaw, Poland
| | - Monika Kusio-Kobiałka
- Laboratory of RNA Biology, International Institute of Molecular and Cell Biology, Warsaw, Poland
| | | | - Dominika Nowis
- Laboratory of Experimental Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Jakub Golab
- Department of Immunology, Medical University of Warsaw, Warsaw, Poland
| | | | - Jacek Jemielity
- Centre of New Technologies, University of Warsaw, Warsaw, Poland
| | - Andrzej Dziembowski
- Laboratory of RNA Biology, International Institute of Molecular and Cell Biology, Warsaw, Poland.
- Department of Embryology, Faculty of Biology, University of Warsaw, Warsaw, Poland.
| | - Seweryn Mroczek
- Laboratory of RNA Biology, International Institute of Molecular and Cell Biology, Warsaw, Poland.
- Institute of Genetics and Biotechnology, Faculty of Biology, University of Warsaw, Warsaw, Poland.
| |
Collapse
|
3
|
Demissie EA, Park SY, Moon JH, Lee DY. Comparative Analysis of Codon Optimization Tools: Advancing toward a Multi-Criteria Framework for Synthetic Gene Design. J Microbiol Biotechnol 2025; 35:e2411066. [PMID: 40223268 PMCID: PMC12010093 DOI: 10.4014/jmb.2411.11066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 02/13/2025] [Accepted: 02/24/2025] [Indexed: 04/15/2025]
Abstract
Codon optimization is an essential technique in synthetic biology and biopharmaceutical production, enhancing recombinant protein expression by fine-tuning genetic sequences to match the translational machinery and codon usage preferences of specific host organisms. This study presents a comprehensive comparative analysis of widely used codon optimization tools, focusing on their capacity to reflect host-specific codon biases, design principles, and parameters. Industrially relevant target proteins were evaluated in Escherichia coli, Saccharomyces cerevisiae, and CHO cells, uncovering significant variability in sequence design and clustering patterns across tools. Tools such as JCat, OPTIMIZER, ATGme, and GeneOptimizer demonstrated strong alignment with genome-wide and highly expressed gene-level codon usage, achieving high codon adaptation index (CAI) values and efficient codon-pair utilization. Conversely, tools like TISIGNER and IDT employed different optimization strategies that frequently produced divergent results. Other key parameters, including GC content, mRNA secondary structure stability (ΔG), and codon-pair bias (CPB), were analyzed to elucidate their influence on translational efficiency. While increased GC content enhanced mRNA stability in E. coli, A/T-rich codons in S. cerevisiae minimized secondary structure formation, and moderate GC content in CHO cells balanced mRNA stability and translation efficiency. Our findings highlight the limitations of single-metric approaches and advocate for a multi-criteria framework that integrates CAI, GC content, mRNA folding energy, and codon-pair considerations. This integrative strategy enables the design of tailored genetic sequences that meet host-specific requirements, advancing synthetic gene design for biotechnological innovation and precision biopharmaceutical applications.
Collapse
Affiliation(s)
- Eden A. Demissie
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Seo-Young Park
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
- School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Je Hun Moon
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Dong-Yup Lee
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| |
Collapse
|
4
|
Luan N, Huang L, Hu J, Zhang H, Gao D, Lei Z, Zhang X, Cao H, Liu C. Immunogenicity Evaluation of Combination Respiratory Syncytial Virus and Varicella-Zoster Virus mRNA Vaccines in C57BL/6J Mice. Vaccines (Basel) 2025; 13:361. [PMID: 40333259 PMCID: PMC12030808 DOI: 10.3390/vaccines13040361] [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: 02/27/2025] [Revised: 03/19/2025] [Accepted: 03/26/2025] [Indexed: 05/09/2025] Open
Abstract
BACKGROUND Respiratory syncytial virus (RSV) and varicella-zoster virus (VZV) pose significant risks to the elderly and individuals with compromised immune systems. In this study, we investigated whether combining RSV and VZV vaccines could reduce the number of vaccination injections, thereby minimizing discomfort for elderly individuals and reducing manufacturing costs. METHODS In this study, we developed two types of combined RSV and VZV mRNA vaccines. Using RSV and VZV mRNA vaccines administered alone as controls, we evaluated the immune response elicited by the combined mRNA vaccines in C57BL/6J mice. RESULTS The results demonstrated that RSV mRNA, VZV mRNA, and a mixture of both could be effectively encapsulated in lipid nanoparticles (LNPs) with uniform particle sizes. Compared to the administration of either the RSV or VZV mRNA vaccine alone, the delivery of two kinds of mRNA LNP combination formulation-whether directly mixed or encapsulated two mRNAs in the same LNP formulation-elicited comparable IgG titers, neutralization titers, cell-mediated immunity (CMI), and CD4+ T-cell responses. CONCLUSIONS In conclusion, this study establishes the feasibility of combining RSV and VZV mRNA-LNP vaccines, laying a solid foundation for clinical trials of combined RSV and VZV vaccines.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Han Cao
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China; (N.L.); (L.H.); (J.H.); (H.Z.); (D.G.); (Z.L.); (X.Z.)
| | - Cunbao Liu
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China; (N.L.); (L.H.); (J.H.); (H.Z.); (D.G.); (Z.L.); (X.Z.)
| |
Collapse
|
5
|
Munoz-Moreno R, Allaj V, Gadee E, Button JM, Diaz F, Maddur MS, Chen W, Hu CH, Martinez L, Giannakou A, Campbell AL, Miteva Y, Guo P, Huang B, Shi S, Lotvin J, Tompkins K, Allen PS, Solórzano A. A highly stable lyophilized mRNA vaccine for Herpes Zoster provides potent cellular and humoral responses. NPJ Vaccines 2025; 10:49. [PMID: 40087280 PMCID: PMC11909110 DOI: 10.1038/s41541-025-01093-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 02/21/2025] [Indexed: 03/17/2025] Open
Abstract
Herpes zoster (HZ) is a painful vesicular rash that occurs upon varicella-zoster virus (VZV) reactivation in older adults and immunocompromised individuals. Although there is currently an approved vaccine for the prevention of shingles, its administration is commonly associated with high reactogenicity. This highlights the need to develop new vaccine alternatives with long lasting immunity and improved tolerability upon administration. In the present study, 10 different vaccine candidate designs using two different codon optimizations targeting the VZV glycoprotein E (gE) were generated. A subset of mRNA constructs were formulated into lipid nanoparticles and assessed for their ability to induce specific cellular and humoral immune responses following vaccination in mice. Notably, the selected mRNA vaccine candidates induced high levels of antibodies and robust CD4+ but also CD8+ immune responses. Moreover, we showed that our alternate lyophilized vaccine provides comparable immunogenicity to current liquid frozen formulations and is stable under long-term storage conditions.
Collapse
Affiliation(s)
| | - Viola Allaj
- Vaccine Research and Development, Pfizer, Pfizer, Pearl River, NY, USA
| | - Eddie Gadee
- Vaccine Research and Development, Pfizer, Pfizer, Pearl River, NY, USA
| | - Julie M Button
- Vaccine Research and Development, Pfizer, Pfizer, Pearl River, NY, USA
| | - Fernando Diaz
- Vaccine Research and Development, Pfizer, Pfizer, Pearl River, NY, USA
| | - Mohan S Maddur
- Vaccine Research and Development, Pfizer, Pfizer, Pearl River, NY, USA
| | - Wei Chen
- Vaccine Research and Development, Pfizer, Pfizer, Pearl River, NY, USA
| | - Cheng Hui Hu
- Vaccine Research and Development, Pfizer, Pfizer, Pearl River, NY, USA
| | - Lyndsey Martinez
- Vaccine Research and Development, Pfizer, Pfizer, Pearl River, NY, USA
| | - Andreas Giannakou
- Vaccine Research and Development, Pfizer, Pfizer, Pearl River, NY, USA
| | - Adam Lee Campbell
- Vaccine Research and Development, Pfizer, Pfizer, Pearl River, NY, USA
| | - Yana Miteva
- Vaccine Research and Development, Pfizer, Pfizer, Pearl River, NY, USA
| | - Pengbo Guo
- Vaccine Research and Development, Pfizer, Pfizer, Pearl River, NY, USA
| | - Bridget Huang
- Vaccine Research and Development, Pfizer, Pfizer, Pearl River, NY, USA
| | - Shuai Shi
- Vaccine Research and Development, Pfizer, Pfizer, Pearl River, NY, USA
| | - Jason Lotvin
- Vaccine Research and Development, Pfizer, Pfizer, Pearl River, NY, USA
| | - Kristin Tompkins
- Vaccine Research and Development, Pfizer, Pfizer, Pearl River, NY, USA
| | | | - Alicia Solórzano
- Vaccine Research and Development, Pfizer, Pfizer, Pearl River, NY, USA.
| |
Collapse
|
6
|
Ngalle Loth A, Maroquenne M, Medjmedj A, Coste F, Bizien T, Pichon C, Logeart-Avramoglou D, Perche F. Structural and functional characterization of a histidylated liposome for mRNA delivery. J Control Release 2025; 379:164-176. [PMID: 39788374 DOI: 10.1016/j.jconrel.2025.01.010] [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: 11/08/2024] [Revised: 12/23/2024] [Accepted: 01/06/2025] [Indexed: 01/12/2025]
Abstract
The development of lipid-based mRNA delivery systems has significantly facilitated recent advances in mRNA-based therapeutics. Liposomes, as the pioneering class of mRNA vectors, continue to lead in clinical trials. We previously developed a histidylated liposome that demonstrated efficient nucleic acid delivery. In this study, the liposome preparation process was optimized by freeze-drying followed by extrusion to homogenize size distribution and improve storage stability. A comprehensive characterization of these LYX liposomes was performed, including evaluation in cellular and murine animal models. LYX liposomes can be stored for up to one year at 4 °C, maintaining a stable size (150 ± 10 nm) and polydispersity index (0.10 ± 0.02), while preserving their transfection efficacy. They exhibit high encapsulation efficacy (∼95 %) and protect mRNA from RNase degradation. Lamellar organization was confirmed by Small Angle X-ray Scattering and CryoTEM, and intracellular trafficking was examined using confocal microscopy. LYX-mRNA lipoplexes can transfect both cell lines and primary cells, albeit with a lower transfection efficacy compared to the commercial Lipofectamine MessengerMAX™ vector. Our data suggest that this could be attributed to slower cell uptake and reduced endosomal escape of LYX. LYX liposomes effectively delivered mRNA encoding therapeutic BMP2 and BMP9 molecules, producing significant amounts of functional proteins that successfully induced BMP signaling. In addition, in vivo studies demonstrated the potential of LYX lipoplexes when incorporated into hydrogels and implanted subcutaneously in mice. These findings provided evidence that LYX liposomes are a promising platform for mRNA delivery, offering versatility for multiple applications.
Collapse
Affiliation(s)
| | - Manon Maroquenne
- Université Paris Cité, CNRS, INSERM, ENVA, B3OA, 75010, Paris, France
| | - Ayoub Medjmedj
- Centre de Biophysique Moléculaire, CBM, CNRS UPR4301, Orléans, France
| | - Franck Coste
- Centre de Biophysique Moléculaire, CBM, CNRS UPR4301, Orléans, France
| | - Thomas Bizien
- Université Paris-Saclay, Synchrotron Soleil, 91190 Saint-Aubin, France
| | - Chantal Pichon
- Inserm UMS 55 ART ARNm and LI2RSO, Université d'Orléans, F-45100 Orléans, France; Institut Universitaire de France, 1 rue Descartes, F-75035 Paris, France
| | | | - Federico Perche
- Centre de Biophysique Moléculaire, CBM, CNRS UPR4301, Orléans, France.
| |
Collapse
|
7
|
Wei Z, Zhang S, Wang X, Xue Y, Dang S, Zhai J. Technological breakthroughs and advancements in the application of mRNA vaccines: a comprehensive exploration and future prospects. Front Immunol 2025; 16:1524317. [PMID: 40103818 PMCID: PMC11913674 DOI: 10.3389/fimmu.2025.1524317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 02/17/2025] [Indexed: 03/20/2025] Open
Abstract
mRNA vaccines utilize single-stranded linear DNA as a template for in vitro transcription. The mRNA is introduced into the cytoplasm via the corresponding delivery system to express the target protein, which then performs its relevant biological function. mRNA vaccines are beneficial in various fields, including cancer vaccines, infectious disease vaccines, protein replacement therapy, and treatment of rare diseases. They offer advantages such as a simple manufacturing process, a quick development cycle, and ease of industrialization. Additionally, mRNA vaccines afford flexibility in adjusting antigen designs and combining sequences of multiple variants, thereby addressing the issue of frequent mutations in pathogenic microorganisms. This paper aims to provide an extensive review of the global development and current research status of mRNA vaccines, with a focus on immunogenicity, classification, design, delivery vector development, stability, and biomedical application. Moreover, the study highlights current challenges and offers insights into future directions for development.
Collapse
Affiliation(s)
- Zhimeng Wei
- School of Basic Medical Sciences, Inner Mongolia Minzu University, Tongliao, China
- Keerqin District First People's Hospital, Tongliao, China
| | - Shuai Zhang
- School of Basic Medical Sciences, Inner Mongolia Minzu University, Tongliao, China
| | - Xingya Wang
- School of Basic Medical Sciences, Inner Mongolia Minzu University, Tongliao, China
| | - Ying Xue
- Keerqin District First People's Hospital, Tongliao, China
| | - Sheng Dang
- Keerqin District First People's Hospital, Tongliao, China
| | - Jingbo Zhai
- School of Basic Medical Sciences, Inner Mongolia Minzu University, Tongliao, China
- Brucellosis Prevention and Treatment Engineering Research Center of Inner Mongolia Autonomous Region, Tongliao, China
- Key Laboratory of Zoonose Prevention and Control at Universities of Inner Mongolia Autonomous Region, Tongliao, China
| |
Collapse
|
8
|
Lin M, Yin Y, Zhao X, Wang C, Zhu X, Zhan L, Chen L, Wang S, Lin X, Zhang J, Xia N, Zheng Z. A truncated pre-F protein mRNA vaccine elicits an enhanced immune response and protection against respiratory syncytial virus. Nat Commun 2025; 16:1386. [PMID: 39910047 PMCID: PMC11799228 DOI: 10.1038/s41467-025-56302-1] [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: 03/26/2024] [Accepted: 01/15/2025] [Indexed: 02/07/2025] Open
Abstract
The Food and Drug Administration (FDA) has approved vaccines designed by GSK, Pfizer and Moderna to protect high-risk populations against respiratory syncytial virus (RSV). These vaccines employ the pre-fusion F (pre-F) protein as the immunogen. In this study, we explored an mRNA vaccine based on a modified pre-F protein called LC2DM-lipid nanoparticle (LC2DM-LNP). This vaccine features a truncated version of the pre-F protein that is anchored to the cell membrane. Our experiments in young and old female mice revealed that the LC2DM-LNP vaccine elicited robust neutralizing antibody titers. Moreover, LC2DM-LNP prompted a Th1-skewed T-cell immune response in female rodent models. Female cotton rats immunized with LC2DM-LNP demonstrated strong immunity to RSV, without signs of vaccine-enhanced respiratory disease (VERD), even in cases of breakthrough infection. Importantly, when administered to pregnant female cotton rats, LC2DM-LNP ensured the transfer of pre-F-specific antibodies to the offspring and provided protection against RSV without increasing lung inflammation. Our findings suggest that LC2DM-LNP could serve as an alternative RSV vaccine candidate for high-risk groups.
Collapse
Affiliation(s)
- Min Lin
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, Fujian Province, PR China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, Fujian Province, PR China
| | - Yifan Yin
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, Fujian Province, PR China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, Fujian Province, PR China
| | - Xiaomeng Zhao
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, Fujian Province, PR China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, Fujian Province, PR China
| | - Chen Wang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, Fujian Province, PR China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, Fujian Province, PR China
| | - Xueqing Zhu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, Fujian Province, PR China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, Fujian Province, PR China
| | - Letao Zhan
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, Fujian Province, PR China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, Fujian Province, PR China
| | - Li Chen
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, Fujian Province, PR China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, Fujian Province, PR China
| | - Siling Wang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, Fujian Province, PR China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, Fujian Province, PR China
| | - Xue Lin
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, Fujian Province, PR China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, Fujian Province, PR China
| | - Jun Zhang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, Fujian Province, PR China.
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, Fujian Province, PR China.
| | - Ningshao Xia
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, Fujian Province, PR China.
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, Fujian Province, PR China.
| | - Zizheng Zheng
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, Fujian Province, PR China.
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, Fujian Province, PR China.
| |
Collapse
|
9
|
Khorshid Sokhangouy S, Behzadi M, Rezaei S, Farjami M, Haghshenas M, Sefidbakht Y, Mozaffari-Jovin S. mRNA Vaccines: Design Principles, Mechanisms, and Manufacturing-Insights From COVID-19 as a Model for Combating Infectious Diseases. Biotechnol J 2025; 20:e202400596. [PMID: 39989260 DOI: 10.1002/biot.202400596] [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: 10/11/2024] [Revised: 01/24/2025] [Accepted: 02/09/2025] [Indexed: 02/25/2025]
Abstract
The full approval of two SARS-CoV-2 mRNA vaccines, Comirnaty and Spikevax, has greatly accelerated the development of numerous mRNA vaccine candidates targeting infectious diseases and cancer. mRNA vaccines provide a rapid, safe, and versatile manufacturing process while eliciting strong humoral and cellular immune responses, making them particularly beneficial for addressing emerging pandemics. Recent advancements in modified nucleotides and lipid nanoparticle delivery systems have further emphasized the potential of this vaccine platform. Despite these transformative opportunities, significant improvements are needed to enhance vaccine efficacy, stability, and immunogenicity. This review outlines the fundamentals of mRNA vaccine design, the manufacturing process, and administration strategies, along with various optimization approaches. It also offers a comprehensive overview of the mRNA vaccine candidates developed since the onset of the COVID-19 pandemic, the challenges posed by emerging SARS-CoV-2 variants, and current strategies to address these variants. Finally, we discuss the potential of broad-spectrum and combined mRNA vaccines and examine the challenges and future prospects of the mRNA vaccine platform.
Collapse
Affiliation(s)
- Saeideh Khorshid Sokhangouy
- Department of Medical Genetics and Molecular Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Medical Biotechnology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Matine Behzadi
- Department of Medical Genetics and Molecular Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Shokuh Rezaei
- Protein Research Center, Shahid Beheshti University, Tehran, Iran
| | - Mahsa Farjami
- Department of Medical Genetics and Molecular Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maryam Haghshenas
- Department of Medical Genetics and Molecular Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Yahya Sefidbakht
- Protein Research Center, Shahid Beheshti University, Tehran, Iran
| | - Sina Mozaffari-Jovin
- Department of Medical Genetics and Molecular Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
10
|
Kitte R, Serfling R, Blache U, Seitz C, Schrader S, Köhl U, Fricke S, Bär C, Tretbar US. Optimal Chimeric Antigen Receptor (CAR)-mRNA for Transient CAR T Cell Generation. Int J Mol Sci 2025; 26:965. [PMID: 39940734 PMCID: PMC11818003 DOI: 10.3390/ijms26030965] [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: 12/19/2024] [Revised: 01/10/2025] [Accepted: 01/13/2025] [Indexed: 02/16/2025] Open
Abstract
Genetically modified T lymphocytes expressing chimeric antigen receptors (CARs) are becoming increasingly important in the treatment of hematologic malignancies and are also intensively being investigated for other diseases such as autoimmune disorders and HIV. Current CAR T cell therapies predominantly use viral transduction methods which, despite their efficacy, raise safety concerns related to genomic integration and potentially associated malignancies as well as labor- and cost-intensive manufacturing. Therefore, non-viral gene transfer methods, especially mRNA-based approaches, have attracted research interest due to their transient modification and enhanced safety profile. In this study, the optimization of CAR-mRNA for T cell applications is investigated, focusing on the impact of mRNA modifications, in vitro transcription protocols, and purification techniques on the translation efficiency and immunogenicity of mRNA. Furthermore, the refined CAR-mRNA was used to generate transient CAR T cells from acute myeloid leukemia patient samples, demonstrating efficacy in vitro and proof-of-concept for clinically relevant settings. These results highlight the potential of optimized mRNA to produce transient and safe CAR T cells.
Collapse
MESH Headings
- Humans
- Receptors, Chimeric Antigen/genetics
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/metabolism
- RNA, Messenger/genetics
- RNA, Messenger/immunology
- Immunotherapy, Adoptive/methods
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Leukemia, Myeloid, Acute/therapy
- Leukemia, Myeloid, Acute/immunology
- Receptors, Antigen, T-Cell/genetics
Collapse
Affiliation(s)
- Reni Kitte
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Perlickstr. 1, 04103 Leipzig, Germany; (R.K.); (R.S.); (U.B.); (U.K.); (S.F.)
| | - Robert Serfling
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Perlickstr. 1, 04103 Leipzig, Germany; (R.K.); (R.S.); (U.B.); (U.K.); (S.F.)
| | - Ulrich Blache
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Perlickstr. 1, 04103 Leipzig, Germany; (R.K.); (R.S.); (U.B.); (U.K.); (S.F.)
- Fraunhofer Cluster of Excellence Immune-Mediated Diseases (CIMD), Perlickstr. 1, 04103 Leipzig, Germany
| | - Claudius Seitz
- Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Inhoffenstraße 7, 38124 Braunschweig, Germany; (C.S.); (S.S.)
| | - Selina Schrader
- Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Inhoffenstraße 7, 38124 Braunschweig, Germany; (C.S.); (S.S.)
| | - Ulrike Köhl
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Perlickstr. 1, 04103 Leipzig, Germany; (R.K.); (R.S.); (U.B.); (U.K.); (S.F.)
- Fraunhofer Cluster of Excellence Immune-Mediated Diseases (CIMD), Perlickstr. 1, 04103 Leipzig, Germany
- Institute of Clinical Immunology, Medical Faculty, University of Leipzig, Johannisallee 30, 04103 Leipzig, Germany
| | - Stephan Fricke
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Perlickstr. 1, 04103 Leipzig, Germany; (R.K.); (R.S.); (U.B.); (U.K.); (S.F.)
- Fraunhofer Cluster of Excellence Immune-Mediated Diseases (CIMD), Perlickstr. 1, 04103 Leipzig, Germany
- Medicine Campus MEDiC, Technical University of Dresden, Klinikum Chemnitz gGmbH, 09116 Chemnitz, Germany
| | - Christian Bär
- Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Nikolai-Fuchs-Straße 1, 30625 Hannover, Germany;
- Fraunhofer Cluster of Excellence Immune-Mediated Diseases (CIMD), Nikolai-Fuchs-Straße 1, 30625 Hannover, Germany
- Institute of Molecular and Translational Therapeutic Strategies, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - U. Sandy Tretbar
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Perlickstr. 1, 04103 Leipzig, Germany; (R.K.); (R.S.); (U.B.); (U.K.); (S.F.)
- Fraunhofer Cluster of Excellence Immune-Mediated Diseases (CIMD), Perlickstr. 1, 04103 Leipzig, Germany
| |
Collapse
|
11
|
Narasipura EA, Ma Y, Tiwade PB, VanKeulen-Miller R, Fung V, Fenton OS. A Chemoinformatic-Guided Synthesis of a Spleen-Expressing mRNA Lipid Nanoparticle Platform. Bioconjug Chem 2025; 36:54-65. [PMID: 39704424 DOI: 10.1021/acs.bioconjchem.4c00419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2024]
Abstract
mRNA lipid nanoparticles (LNPs) are a powerful technology that are actively being investigated for their ability to prevent, treat, and study disease. However, a major limitation remains: achieving extrahepatic mRNA expression. The development of new carriers could enable the expression of mRNA in non-liver targets, thus expanding the utility of mRNA-based medicines. In this study, we use a combination of chemoinformatic-guided material synthesis and design of experiment optimization for the development of a spleen-expressing lipid nanoparticle (SE-LNP). We begin with the synthesis of a novel cholesterol derivative followed by SE-LNP formulation and design of experiment-guided optimization to identify three lead SE-LNPs. We then evaluate their in vitro delivery mechanism, in vivo biodistribution, and protein expression in mice, ultimately achieving spleen-preferential expression. The goal of this paper is thus to create LNPs that preferentially express mRNA in the spleen upon intravenous delivery, demonstrating the potential of LNPs to modulate gene expression in extrahepatic tissues for disease treatment.
Collapse
Affiliation(s)
- Eshan A Narasipura
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Yutian Ma
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Palas Balakdas Tiwade
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Rachel VanKeulen-Miller
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Vincent Fung
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Owen S Fenton
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| |
Collapse
|
12
|
Kisakov DN, Karpenko LI, Kisakova LA, Sharabrin SV, Borgoyakova MB, Starostina EV, Taranov OS, Ivleva EK, Pyankov OV, Zaykovskaya AV, Dmitrienko EV, Yakovlev VA, Tigeeva EV, Bauer IA, Krasnikova SI, Rudometova NB, Rudometov AP, Sergeev AA, Ilyichev AA. Jet Injection of Naked mRNA Encoding the RBD of the SARS-CoV-2 Spike Protein Induces a High Level of a Specific Immune Response in Mice. Vaccines (Basel) 2025; 13:65. [PMID: 39852844 PMCID: PMC11769039 DOI: 10.3390/vaccines13010065] [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: 12/04/2024] [Revised: 12/26/2024] [Accepted: 01/03/2025] [Indexed: 01/26/2025] Open
Abstract
Background: Although mRNA vaccines encapsulated in lipid nanoparticles (LNPs) have demonstrated a safety profile with minimal serious adverse events in clinical trials, there is opportunity to further reduce mRNA reactogenicity. The development of naked mRNA vaccines could improve vaccine tolerability. Naked nucleic acid delivery using the jet injection method may be a solution. Methods: In the first part of the study, the optimal conditions providing low traumatization and high expression of the model mRNA-GFP molecule in the tissues of laboratory animals were determined. Then, we used the selected protocol to immunize BALB/c mice with mRNA-RBD encoding the SARS-CoV-2 receptor-binding domain (RBD). It was demonstrated that mice vaccinated with naked mRNA-RBD developed a high level of specific antibodies with virus-neutralizing activity. The vaccine also induced a strong RBD-specific T-cell response and reduced the viral load in the lungs of the animals after infection with the SARS-CoV-2 virus. The level of immune response in mice immunized with mRNA-RBD using a spring-loaded jet injector was comparable to that in animals immunized with mRNA-RBD encapsulated in LNPs. Results: In this study, the efficacy of an inexpensive, simple, and safe method of mRNA delivery using a spring-loaded jet injector was evaluated and validated. Conclusions: Our findings suggest that the jet injection method may be a possible alternative to LNPs for delivering mRNA vaccines against SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Denis N. Kisakov
- State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, World-Class Genomic Research Center for Biological Safety and Technological Independence, Federal Scientific and Technical Program on the Development of Genetic Technologies, 630559 Koltsovo, Russia; (L.I.K.); (L.A.K.); (S.V.S.); (M.B.B.); (E.V.S.); (O.S.T.); (E.K.I.); (O.V.P.); (A.V.Z.); (V.A.Y.); (E.V.T.); (S.I.K.); (N.B.R.); (A.P.R.); (A.A.S.); (A.A.I.)
| | - Larisa I. Karpenko
- State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, World-Class Genomic Research Center for Biological Safety and Technological Independence, Federal Scientific and Technical Program on the Development of Genetic Technologies, 630559 Koltsovo, Russia; (L.I.K.); (L.A.K.); (S.V.S.); (M.B.B.); (E.V.S.); (O.S.T.); (E.K.I.); (O.V.P.); (A.V.Z.); (V.A.Y.); (E.V.T.); (S.I.K.); (N.B.R.); (A.P.R.); (A.A.S.); (A.A.I.)
| | - Lyubov A. Kisakova
- State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, World-Class Genomic Research Center for Biological Safety and Technological Independence, Federal Scientific and Technical Program on the Development of Genetic Technologies, 630559 Koltsovo, Russia; (L.I.K.); (L.A.K.); (S.V.S.); (M.B.B.); (E.V.S.); (O.S.T.); (E.K.I.); (O.V.P.); (A.V.Z.); (V.A.Y.); (E.V.T.); (S.I.K.); (N.B.R.); (A.P.R.); (A.A.S.); (A.A.I.)
| | - Sergey V. Sharabrin
- State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, World-Class Genomic Research Center for Biological Safety and Technological Independence, Federal Scientific and Technical Program on the Development of Genetic Technologies, 630559 Koltsovo, Russia; (L.I.K.); (L.A.K.); (S.V.S.); (M.B.B.); (E.V.S.); (O.S.T.); (E.K.I.); (O.V.P.); (A.V.Z.); (V.A.Y.); (E.V.T.); (S.I.K.); (N.B.R.); (A.P.R.); (A.A.S.); (A.A.I.)
| | - Mariya B. Borgoyakova
- State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, World-Class Genomic Research Center for Biological Safety and Technological Independence, Federal Scientific and Technical Program on the Development of Genetic Technologies, 630559 Koltsovo, Russia; (L.I.K.); (L.A.K.); (S.V.S.); (M.B.B.); (E.V.S.); (O.S.T.); (E.K.I.); (O.V.P.); (A.V.Z.); (V.A.Y.); (E.V.T.); (S.I.K.); (N.B.R.); (A.P.R.); (A.A.S.); (A.A.I.)
| | - Ekaterina V. Starostina
- State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, World-Class Genomic Research Center for Biological Safety and Technological Independence, Federal Scientific and Technical Program on the Development of Genetic Technologies, 630559 Koltsovo, Russia; (L.I.K.); (L.A.K.); (S.V.S.); (M.B.B.); (E.V.S.); (O.S.T.); (E.K.I.); (O.V.P.); (A.V.Z.); (V.A.Y.); (E.V.T.); (S.I.K.); (N.B.R.); (A.P.R.); (A.A.S.); (A.A.I.)
| | - Oleg S. Taranov
- State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, World-Class Genomic Research Center for Biological Safety and Technological Independence, Federal Scientific and Technical Program on the Development of Genetic Technologies, 630559 Koltsovo, Russia; (L.I.K.); (L.A.K.); (S.V.S.); (M.B.B.); (E.V.S.); (O.S.T.); (E.K.I.); (O.V.P.); (A.V.Z.); (V.A.Y.); (E.V.T.); (S.I.K.); (N.B.R.); (A.P.R.); (A.A.S.); (A.A.I.)
| | - Elena K. Ivleva
- State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, World-Class Genomic Research Center for Biological Safety and Technological Independence, Federal Scientific and Technical Program on the Development of Genetic Technologies, 630559 Koltsovo, Russia; (L.I.K.); (L.A.K.); (S.V.S.); (M.B.B.); (E.V.S.); (O.S.T.); (E.K.I.); (O.V.P.); (A.V.Z.); (V.A.Y.); (E.V.T.); (S.I.K.); (N.B.R.); (A.P.R.); (A.A.S.); (A.A.I.)
| | - Oleg V. Pyankov
- State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, World-Class Genomic Research Center for Biological Safety and Technological Independence, Federal Scientific and Technical Program on the Development of Genetic Technologies, 630559 Koltsovo, Russia; (L.I.K.); (L.A.K.); (S.V.S.); (M.B.B.); (E.V.S.); (O.S.T.); (E.K.I.); (O.V.P.); (A.V.Z.); (V.A.Y.); (E.V.T.); (S.I.K.); (N.B.R.); (A.P.R.); (A.A.S.); (A.A.I.)
| | - Anna V. Zaykovskaya
- State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, World-Class Genomic Research Center for Biological Safety and Technological Independence, Federal Scientific and Technical Program on the Development of Genetic Technologies, 630559 Koltsovo, Russia; (L.I.K.); (L.A.K.); (S.V.S.); (M.B.B.); (E.V.S.); (O.S.T.); (E.K.I.); (O.V.P.); (A.V.Z.); (V.A.Y.); (E.V.T.); (S.I.K.); (N.B.R.); (A.P.R.); (A.A.S.); (A.A.I.)
| | - Elena V. Dmitrienko
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch, Russian Academy of Sciences, 630090 Novosibirsk, Russia; (E.V.D.); (I.A.B.)
| | - Vladimir A. Yakovlev
- State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, World-Class Genomic Research Center for Biological Safety and Technological Independence, Federal Scientific and Technical Program on the Development of Genetic Technologies, 630559 Koltsovo, Russia; (L.I.K.); (L.A.K.); (S.V.S.); (M.B.B.); (E.V.S.); (O.S.T.); (E.K.I.); (O.V.P.); (A.V.Z.); (V.A.Y.); (E.V.T.); (S.I.K.); (N.B.R.); (A.P.R.); (A.A.S.); (A.A.I.)
| | - Elena V. Tigeeva
- State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, World-Class Genomic Research Center for Biological Safety and Technological Independence, Federal Scientific and Technical Program on the Development of Genetic Technologies, 630559 Koltsovo, Russia; (L.I.K.); (L.A.K.); (S.V.S.); (M.B.B.); (E.V.S.); (O.S.T.); (E.K.I.); (O.V.P.); (A.V.Z.); (V.A.Y.); (E.V.T.); (S.I.K.); (N.B.R.); (A.P.R.); (A.A.S.); (A.A.I.)
| | - Irina Alekseevna Bauer
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch, Russian Academy of Sciences, 630090 Novosibirsk, Russia; (E.V.D.); (I.A.B.)
| | - Svetlana I. Krasnikova
- State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, World-Class Genomic Research Center for Biological Safety and Technological Independence, Federal Scientific and Technical Program on the Development of Genetic Technologies, 630559 Koltsovo, Russia; (L.I.K.); (L.A.K.); (S.V.S.); (M.B.B.); (E.V.S.); (O.S.T.); (E.K.I.); (O.V.P.); (A.V.Z.); (V.A.Y.); (E.V.T.); (S.I.K.); (N.B.R.); (A.P.R.); (A.A.S.); (A.A.I.)
| | - Nadezhda B. Rudometova
- State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, World-Class Genomic Research Center for Biological Safety and Technological Independence, Federal Scientific and Technical Program on the Development of Genetic Technologies, 630559 Koltsovo, Russia; (L.I.K.); (L.A.K.); (S.V.S.); (M.B.B.); (E.V.S.); (O.S.T.); (E.K.I.); (O.V.P.); (A.V.Z.); (V.A.Y.); (E.V.T.); (S.I.K.); (N.B.R.); (A.P.R.); (A.A.S.); (A.A.I.)
| | - Andrey P. Rudometov
- State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, World-Class Genomic Research Center for Biological Safety and Technological Independence, Federal Scientific and Technical Program on the Development of Genetic Technologies, 630559 Koltsovo, Russia; (L.I.K.); (L.A.K.); (S.V.S.); (M.B.B.); (E.V.S.); (O.S.T.); (E.K.I.); (O.V.P.); (A.V.Z.); (V.A.Y.); (E.V.T.); (S.I.K.); (N.B.R.); (A.P.R.); (A.A.S.); (A.A.I.)
| | - Artemiy A. Sergeev
- State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, World-Class Genomic Research Center for Biological Safety and Technological Independence, Federal Scientific and Technical Program on the Development of Genetic Technologies, 630559 Koltsovo, Russia; (L.I.K.); (L.A.K.); (S.V.S.); (M.B.B.); (E.V.S.); (O.S.T.); (E.K.I.); (O.V.P.); (A.V.Z.); (V.A.Y.); (E.V.T.); (S.I.K.); (N.B.R.); (A.P.R.); (A.A.S.); (A.A.I.)
| | - Alexander A. Ilyichev
- State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, World-Class Genomic Research Center for Biological Safety and Technological Independence, Federal Scientific and Technical Program on the Development of Genetic Technologies, 630559 Koltsovo, Russia; (L.I.K.); (L.A.K.); (S.V.S.); (M.B.B.); (E.V.S.); (O.S.T.); (E.K.I.); (O.V.P.); (A.V.Z.); (V.A.Y.); (E.V.T.); (S.I.K.); (N.B.R.); (A.P.R.); (A.A.S.); (A.A.I.)
| |
Collapse
|
13
|
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
|
14
|
Al Turihi N, Allouche D, Quéré M, Scuiller M, Legastelois I. Characterization of poly(A) and poly(T) tail lengths in plasmid DNA by liquid chromatography high-resolution mass spectrometry. Anal Bioanal Chem 2025; 417:59-68. [PMID: 39547978 PMCID: PMC11695563 DOI: 10.1007/s00216-024-05654-6] [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: 07/23/2024] [Revised: 11/04/2024] [Accepted: 11/07/2024] [Indexed: 11/17/2024]
Abstract
Rapid advances in messenger RNA (mRNA) technology necessitate effective analytical methods. This study describes the development of a novel in vitro method using ion-pair reversed-phase liquid chromatography coupled to high-resolution mass spectrometry (IP-RP-LC-HRMS) for assessing the poly(A) and complementary poly(T) tail lengths directly into the DNA template used to manufacture mRNA. Briefly, after the validation of poly(A) tail length in the plasmid by Sanger sequencing, double-stranded DNA fragments containing these tails in the plasmid of interest were amplified by the polymerase chain reaction (PCR), purified on silica column, and digested with restriction enzymes ClaI and HindIII. Gel or capillary electrophoresis confirmed sample quality and enzymatic digestion efficiency. Subsequently, poly(A) and complementary poly(T) tails were extracted and analyzed by LC-MS to determine their length and heterogeneity at a single-nucleotide resolution. Three DNA templates containing poly(A) tail lengths of 60A-G, 95A, or 108A were studied. LC-MS results correlated well with Sanger sequencing, identifying major populations of 60A-G, 95A, or 108A. Surprisingly, unlike Sanger sequencing, LC-MS analysis revealed minor poly(A) populations with lengths longer or shorter than the theoretically encoded poly(A) tail length. This finding could be explained by (i) the slippage of bacterial DNA polymerase I during plasmid replication in bacterial culture, which occurs on repeat mononucleotide sequences, or (ii) the slippage of Q5® High-Fidelity DNA Polymerase during PCR amplification. In conclusion, the method is easy, rapid, and accurate and could replace Sanger sequencing to assess the poly(A) and complementary poly(T) tail lengths in plasmid DNA.
Collapse
Affiliation(s)
- Nour Al Turihi
- RNA Sciences, Sanofi, 1541 av. Marcel Mérieux, 69280, Marcy l'Étoile, France.
| | - Delphine Allouche
- RNA Sciences, Sanofi, 1541 av. Marcel Mérieux, 69280, Marcy l'Étoile, France
| | - Maëlle Quéré
- RNA Sciences, Sanofi, 1541 av. Marcel Mérieux, 69280, Marcy l'Étoile, France
| | - Mathieu Scuiller
- RNA Sciences, Sanofi, 1541 av. Marcel Mérieux, 69280, Marcy l'Étoile, France
| | | |
Collapse
|
15
|
Yao R, Xie C, Xia X. Recent progress in mRNA cancer vaccines. Hum Vaccin Immunother 2024; 20:2307187. [PMID: 38282471 PMCID: PMC10826636 DOI: 10.1080/21645515.2024.2307187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 01/16/2024] [Indexed: 01/30/2024] Open
Abstract
The research and development of messenger RNA (mRNA) cancer vaccines have gradually overcome numerous challenges through the application of personalized cancer antigens, structural optimization of mRNA, and the development of alternative RNA-based vectors and efficient targeted delivery vectors. Clinical trials are currently underway for various cancer vaccines that encode tumor-associated antigens (TAAs), tumor-specific antigens (TSAs), or immunomodulators. In this paper, we summarize the optimization of mRNA and the emergence of RNA-based expression vectors in cancer vaccines. We begin by reviewing the advancement and utilization of state-of-the-art targeted lipid nanoparticles (LNPs), followed by presenting the primary classifications and clinical applications of mRNA cancer vaccines. Collectively, mRNA vaccines are emerging as a central focus in cancer immunotherapy, offering the potential to address multiple challenges in cancer treatment, either as standalone therapies or in combination with current cancer treatments.
Collapse
Affiliation(s)
- Ruhui Yao
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Chunyuan Xie
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xiaojun Xia
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| |
Collapse
|
16
|
Du Y, Zuber PK, Xiao H, Li X, Gordiyenko Y, Ramakrishnan V. Efficient circular RNA synthesis for potent rolling circle translation. Nat Biomed Eng 2024:10.1038/s41551-024-01306-3. [PMID: 39672985 DOI: 10.1038/s41551-024-01306-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 10/31/2024] [Indexed: 12/15/2024]
Abstract
Circular RNA (circRNA) is a candidate for next-generation messenger RNA therapeutics owing to its remarkable stability. Here we describe trans-splicing-based methods for the synthesis of circRNAs over 8,000 nucleotides. The methods are independent of bacterial sequences, outperform the permuted intron-exon method and allow for the incorporation of RNA modifications. The resulting unmodified circRNAs, which incorporate sequences from human 28S ribosomal RNA, display low immunogenicity and are translated more efficiently than permuted intron-exon-derived circRNAs. Additionally, by using viral internal ribosomal entry sites for rolling circle translation, we show that ribosomes can efficiently read through highly structured internal ribosomal entry sites, enhancing the efficiency of rolling circle translation by over 7,000-fold with respect to previous constructs. The efficient and reliable production of circRNA may facilitate its therapeutic use.
Collapse
Affiliation(s)
- Yifei Du
- MRC Laboratory of Molecular Biology, Cambridge, UK.
| | | | | | - Xueyan Li
- MRC Laboratory of Molecular Biology, Cambridge, UK
| | | | | |
Collapse
|
17
|
Xia X. How Trustworthy Are the Genomic Sequences of SARS-CoV-2 in GenBank? Microorganisms 2024; 12:2187. [PMID: 39597576 PMCID: PMC11596409 DOI: 10.3390/microorganisms12112187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 10/27/2024] [Accepted: 10/27/2024] [Indexed: 11/29/2024] Open
Abstract
Well-annotated gene and genomic sequences serve as a foundation for making inferences in molecular biology and evolution and can directly impact public health. The first SARS-CoV-2 genome was submitted to the GenBank database hosted by the U.S. National Center for Biotechnology Information and used to develop the two successful vaccines. Conserved protein domains are often chosen as targets for developing antiviral medicines or vaccines. Mutation and substitution patterns provide crucial information not only on functional motifs and genome/protein interactions but also for characterizing phylogenetic relationships among viral strains. These patterns, together with the collection time of viral samples, serve as the basis for addressing the question of when and where the host-switching event occurred. Unfortunately, viral genomic sequences submitted to GenBank undergo little quality control, and critical information in the annotation is frequently changed without being recorded. Researchers often have no choice but to hold blind faith in the authenticity of the sequences. There have been reports of incorrect genome annotation but no report that casts doubt on the genomic sequences themselves because it seems theoretically impossible to identify genomic sequences that may not be authentic. This paper takes an innovative approach to show that some SARS-CoV-2 genomes submitted to GenBank cannot possibly be authentic. Specifically, some SARS-CoV-2 genomic sequences deposited in GenBank with collection times in 2023 and 2024, isolated from saliva, nasopharyngeal, sewage, and stool, are identical to the reference genome of SARS-CoV-2 (NC_045512). The probability of such occurrence is effectively 0. I also compile SARS-CoV-2 genomes with changed sample collection times. One may be led astray in bioinformatic analysis without being aware of errors in sequences and sequence annotation.
Collapse
Affiliation(s)
- Xuhua Xia
- Department of Biology, University of Ottawa, Marie-Curie Private, Ottawa, ON K1N 6N5, Canada;
- Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| |
Collapse
|
18
|
Wan J, Wang C, Wang Z, Wang L, Wang H, Zhou M, Fu ZF, Zhao L. CXCL13 promotes broad immune responses induced by circular RNA vaccines. Proc Natl Acad Sci U S A 2024; 121:e2406434121. [PMID: 39436660 PMCID: PMC11536096 DOI: 10.1073/pnas.2406434121] [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: 03/29/2024] [Accepted: 08/22/2024] [Indexed: 10/23/2024] Open
Abstract
Antibody responses induced by current vaccines for influenza and SARS-CoV-2 often lack robust cross-reactivity. As hubs where diverse immune cells converge and interact, the alterations in the immune microenvironment within lymph nodes (LNs) are intricately linked to immune responses. Herein, we designed a lipid nanoparticle (LNP) loaded with circular RNA (circRNA) and targeted to LNs, in which CXCL13 was directly integrated into antigen-encoding circRNA strands. We demonstrated that CXCL13 alters the transcriptomic profiles of LNs, especially the upregulation of IL-21 and IL-4. Meanwhile, CXCL13 promotes the formation of germinal center and elicits robust antigen-specific T cell responses. With the codelivery of CXCL13 and the antigen, CXCL13 enhances cross-reactive antibodies against influenza virus and SARS-CoV-2, achieving protection against both homologous and heterologous influenza virus challenges in a mouse model. Notably, the targeted modification of LNP surfaces with antibodies helps address some of the challenges associated with lyophilized LNP vaccines, which is crucial for the long-term storage of LNP-circRNA vaccines. Overall, the circRNA-based antigen-CXCL13 coexpression system developed herein provides a simple and robust platform that enhances the magnitude and breadth of antibody responses against multiple viral glycoproteins, highlighting the potential utility of CXCL13 in inducing broad immune responses.
Collapse
Affiliation(s)
- Jiawu Wan
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan430070, China
- Hubei Hongshan Laboratory, Wuhan430070, China
- Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan430070, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan430070, China
| | - Caiqian Wang
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan430070, China
- Hubei Hongshan Laboratory, Wuhan430070, China
- Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan430070, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan430070, China
| | - Zongmei Wang
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan430070, China
- Hubei Hongshan Laboratory, Wuhan430070, China
- Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan430070, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan430070, China
| | - Lingli Wang
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan430070, China
- Hubei Hongshan Laboratory, Wuhan430070, China
- Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan430070, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan430070, China
| | - Haoran Wang
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan430070, China
- Hubei Hongshan Laboratory, Wuhan430070, China
- Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan430070, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan430070, China
| | - Ming Zhou
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan430070, China
- Hubei Hongshan Laboratory, Wuhan430070, China
- Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan430070, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan430070, China
| | - Zhen F. Fu
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan430070, China
- Hubei Hongshan Laboratory, Wuhan430070, China
- Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan430070, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan430070, China
| | - Ling Zhao
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan430070, China
- Hubei Hongshan Laboratory, Wuhan430070, China
- Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan430070, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan430070, China
| |
Collapse
|
19
|
Liu X, Li Z, Li X, Wu W, Jiang H, Zheng Y, Zhou J, Ye X, Lu J, Wang W, Yu L, Li Y, Qu L, Wang J, Li F, Chen L, Wu L, Feng L. A single-dose circular RNA vaccine prevents Zika virus infection without enhancing dengue severity in mice. Nat Commun 2024; 15:8932. [PMID: 39414822 PMCID: PMC11484855 DOI: 10.1038/s41467-024-53242-0] [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: 03/07/2024] [Accepted: 10/07/2024] [Indexed: 10/18/2024] Open
Abstract
Antibody-dependent enhancement (ADE) is a potential concern for the development of Zika virus (ZIKV) vaccines. Cross-reactive but poorly neutralizing antibodies, usually targeting viral pre-membrane or envelope (E) proteins, can potentially enhance dengue virus (DENV) infection. Although E domain III (EDIII) contains ZIKV-specific epitopes, its immunogenicity is poor. Here, we show that dimeric EDIII, fused to human IgG1 Fc fragment (EDIII-Fc) and encoded by circular RNA (circRNA), induces better germinal center reactions and higher neutralizing antibodies compared to circRNAs encoding monomeric or trimeric EDIII. Two doses of circRNAs encoding EDIII-Fc and ZIKV nonstructural protein NS1, another protective antigen, prevent lethal ZIKV infection in neonates born to immunized C57BL/6 mice and in interferon-α/β receptor knockout adult C57BL/6 mice. Importantly, a single-dose optimized circRNA vaccine with improved antigen expression confers potent and durable protection without inducing obvious DENV ADE in mice, laying the groundwork for developing flavivirus vaccines based on circRNAs encoding EDIII-Fc and NS1.
Collapse
Affiliation(s)
- Xinglong Liu
- State Key Laboratory of Respiratory Disease, CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhengfeng Li
- State Key Laboratory of Respiratory Disease, CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Xiaoxia Li
- State Key Laboratory of Respiratory Disease, CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Weixuan Wu
- State Key Laboratory of Respiratory Disease, CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Huadong Jiang
- State Key Laboratory of Respiratory Disease, CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
- School of Life Science, University of Science and Technology of China, Hefei, 230026, China
| | - Yufen Zheng
- State Key Laboratory of Respiratory Disease, CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Junjie Zhou
- State Key Laboratory of Respiratory Disease, CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Xianmiao Ye
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China
| | - Junnan Lu
- State Key Laboratory of Respiratory Disease, CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Wei Wang
- Bioland Laboratory, Guangzhou, 510005, China
| | - Lei Yu
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, 510440, China
| | - Yiping Li
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 501180, China
| | - Linbing Qu
- State Key Laboratory of Respiratory Disease, CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Jianhua Wang
- State Key Laboratory of Respiratory Disease, CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Feng Li
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, 510440, China
| | - Ling Chen
- State Key Laboratory of Respiratory Disease, CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China.
- Guangzhou National Laboratory, Guangzhou, 510005, China.
| | - Linping Wu
- State Key Laboratory of Respiratory Disease, CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Liqiang Feng
- State Key Laboratory of Respiratory Disease, CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
20
|
He W, Geng Q, Ji G, Li J, Wang D, He Y, Jin Q, Ye J. Effective Synthesis of mRNA during In Vitro Transcription with Fewer Impurities Produced. Molecules 2024; 29:4713. [PMID: 39407643 PMCID: PMC11477551 DOI: 10.3390/molecules29194713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 09/28/2024] [Accepted: 09/30/2024] [Indexed: 10/20/2024] Open
Abstract
The remarkable efficacy of COVID-19 vaccines has established mRNA as a highly promising biomedical technology. However, the adequate application of mRNA therapeutics necessitates additional measures to mitigate the inherent immunogenicity, which is predominantly caused by dsRNA. As a byproduct of the in vitro transcription of mRNA, dsRNA was reported to be originated through several distinct mechanisms, including the extension of 3' loop-back hairpins, the extension of hybridized abortive transcripts, and promoter-independent transcription. The intricate mechanisms involved pose a dilemma as the reduction in dsRNA results in a concomitant decrease in other critical quality attributes of mRNA. Here, we demonstrate that the promoter binding motifs of T7 RNA polymerase directly impact the production of promoter-independent transcription-based dsRNA. Specifically, the G753A mutation significantly reduces the formation of dsRNA byproducts, which can further combine with modified nucleotides to enhance the effectiveness of dsRNA mitigation and with previously reported high-integrity mutation K389A to minimize side effects. Accordingly, the present study reports a cost-effective approach to synthesize high-purity, less immunostimulatory mRNA by using an engineered T7 RNA polymerase mutant.
Collapse
Affiliation(s)
- Wei He
- College of Forestry, Nanjing Forestry University, Nanjing 210037, China;
- Vazyme Biotech Co., Ltd., Nanjing 210037, China; (Q.G.); (G.J.); (J.L.); (D.W.)
| | - Qi Geng
- Vazyme Biotech Co., Ltd., Nanjing 210037, China; (Q.G.); (G.J.); (J.L.); (D.W.)
| | - Guiying Ji
- Vazyme Biotech Co., Ltd., Nanjing 210037, China; (Q.G.); (G.J.); (J.L.); (D.W.)
| | - Ji Li
- Vazyme Biotech Co., Ltd., Nanjing 210037, China; (Q.G.); (G.J.); (J.L.); (D.W.)
| | - Dan Wang
- Vazyme Biotech Co., Ltd., Nanjing 210037, China; (Q.G.); (G.J.); (J.L.); (D.W.)
| | - Yucai He
- School of Pharmacy, Changzhou University, Changzhou 213164, China
| | - Qiuheng Jin
- Vazyme Biotech Co., Ltd., Nanjing 210037, China; (Q.G.); (G.J.); (J.L.); (D.W.)
| | - Jianren Ye
- College of Forestry, Nanjing Forestry University, Nanjing 210037, China;
| |
Collapse
|
21
|
Zhang X, Li M, Zhang N, Li Y, Teng F, Li Y, Zhang X, Xu X, Li H, Zhu Y, Wang Y, Jia Y, Qin C, Wang B, Guo S, Wang Y, Yu X. SARS-CoV-2 Evolution: Immune Dynamics, Omicron Specificity, and Predictive Modeling in Vaccinated Populations. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2402639. [PMID: 39206813 PMCID: PMC11516136 DOI: 10.1002/advs.202402639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 07/25/2024] [Indexed: 09/04/2024]
Abstract
Host immunity is central to the virus's spread dynamics, which is significantly influenced by vaccination and prior infection experiences. In this work, we analyzed the co-evolution of SARS-CoV-2 mutation, angiotensin-converting enzyme 2 (ACE2) receptor binding, and neutralizing antibody (NAb) responses across various variants in 822 human and mice vaccinated with different non-Omicron and Omicron vaccines is analyzed. The link between vaccine efficacy and vaccine type, dosing, and post-vaccination duration is revealed. The classification of immune protection against non-Omicron and Omicron variants is co-evolved with genetic mutations and vaccination. Additionally, a model, the Prevalence Score (P-Score) is introduced, which surpasses previous algorithm-based models in predicting the potential prevalence of new variants in vaccinated populations. The hybrid vaccination combining the wild-type (WT) inactivated vaccine with the Omicron BA.4/5 mRNA vaccine may provide broad protection against both non-Omicron variants and Omicron variants, albeit with EG.5.1 still posing a risk. In conclusion, these findings enhance understanding of population immunity variations and provide valuable insights for future vaccine development and public health strategies.
Collapse
Affiliation(s)
- Xiaohan Zhang
- State Key Laboratory of Medical ProteomicsBeijing Proteome Research CenterNational Center for Protein Sciences‐Beijing (PHOENIX Center)Beijing Institute of LifeomicsBeijing102206China
- School of MedicineNanjing University of Chinese MedicineNanjing210023China
| | - Mansheng Li
- State Key Laboratory of Medical ProteomicsBeijing Proteome Research CenterNational Center for Protein Sciences‐Beijing (PHOENIX Center)Beijing Institute of LifeomicsBeijing102206China
| | - Nana Zhang
- Department of VirologyState Key Laboratory of Pathogen and BiosecurityBeijing Institute of Microbiology and EpidemiologyAcademy of Military Medical SciencesBeijing100071China
| | - Yunhui Li
- Department of Clinical LaboratoryBeijing Ditan HospitalCapital Medical UniversityBeijing100015China
| | - Fei Teng
- Emergency Medicine Clinical Research CenterBeijing Chao‐Yang HospitalCapital Medical UniversityBeijing Key Laboratory of Cardiopulmonary Cerebral ResuscitationBeijing100020China
| | - Yongzhe Li
- Department of Clinical LaboratoryPeking Union Medical College HospitalChinese Academy of Medical Science & Peking Union Medical CollegeBeijing100730China
| | - Xiaomei Zhang
- State Key Laboratory of Medical ProteomicsBeijing Proteome Research CenterNational Center for Protein Sciences‐Beijing (PHOENIX Center)Beijing Institute of LifeomicsBeijing102206China
| | - Xingming Xu
- State Key Laboratory of Medical ProteomicsBeijing Proteome Research CenterNational Center for Protein Sciences‐Beijing (PHOENIX Center)Beijing Institute of LifeomicsBeijing102206China
| | - Haolong Li
- Department of Clinical LaboratoryPeking Union Medical College HospitalChinese Academy of Medical Science & Peking Union Medical CollegeBeijing100730China
| | - Yunping Zhu
- State Key Laboratory of Medical ProteomicsBeijing Proteome Research CenterNational Center for Protein Sciences‐Beijing (PHOENIX Center)Beijing Institute of LifeomicsBeijing102206China
| | - Yumin Wang
- The First Affiliated Hospital of Wenzhou Medical UniversityWenzhou325000China
| | - Yan Jia
- ProteomicsEra Medical Co. Ltd.Beijing102206China
| | - Chengfeng Qin
- Department of VirologyState Key Laboratory of Pathogen and BiosecurityBeijing Institute of Microbiology and EpidemiologyAcademy of Military Medical SciencesBeijing100071China
| | - Bingwei Wang
- School of MedicineNanjing University of Chinese MedicineNanjing210023China
| | - Shubin Guo
- Emergency Medicine Clinical Research CenterBeijing Chao‐Yang HospitalCapital Medical UniversityBeijing Key Laboratory of Cardiopulmonary Cerebral ResuscitationBeijing100020China
| | - Yajie Wang
- Department of Clinical LaboratoryBeijing Ditan HospitalCapital Medical UniversityBeijing100015China
| | - Xiaobo Yu
- State Key Laboratory of Medical ProteomicsBeijing Proteome Research CenterNational Center for Protein Sciences‐Beijing (PHOENIX Center)Beijing Institute of LifeomicsBeijing102206China
- The First Affiliated Hospital of Wenzhou Medical UniversityWenzhou325000China
| |
Collapse
|
22
|
Gao Y, Zhu S, Li H, Hao X, Chen W, Pan D, Qian Z. AntigenBoost: enhanced mRNA-based antigen expression through rational amino acid substitution. Brief Bioinform 2024; 25:bbae468. [PMID: 39400114 PMCID: PMC11472322 DOI: 10.1093/bib/bbae468] [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: 06/28/2024] [Revised: 07/29/2024] [Accepted: 09/09/2024] [Indexed: 10/15/2024] Open
Abstract
Messenger RNA (mRNA) vaccines represent a groundbreaking advancement in immunology and public health, particularly highlighted by their role in combating the COVID-19 pandemic. Optimizing mRNA-based antigen expression is a crucial focus in this emerging industry. We have developed a bioinformatics tool named AntigenBoost to address the challenge posed by destabilizing dipeptides that hinder ribosomal translation. AntigenBoost identifies these dipeptides within specific antigens and provides a range of potential amino acid substitution strategies using a two-dimensional scoring system. Through a combination of bioinformatics analysis and experimental validation, we significantly enhanced the in vitro expression of mRNA-derived Respiratory Syncytial Virus fusion glycoprotein and Influenza A Hemagglutinin antigen. Notably, a single amino acid substitution improved the immune response in mice, underscoring the effectiveness of AntigenBoost in mRNA vaccine design.
Collapse
Affiliation(s)
- Yumiao Gao
- NanoRibo (Shanghai) Biotechnology Co., Ltd., No. 1188 Lianhang Road, Minhang District, Shanghai 200003, China
| | - Siran Zhu
- NanoRibo (Shanghai) Biotechnology Co., Ltd., No. 1188 Lianhang Road, Minhang District, Shanghai 200003, China
| | - Huichun Li
- NanoRibo (Shanghai) Biotechnology Co., Ltd., No. 1188 Lianhang Road, Minhang District, Shanghai 200003, China
| | - Xueting Hao
- NanoRibo (Shanghai) Biotechnology Co., Ltd., No. 1188 Lianhang Road, Minhang District, Shanghai 200003, China
| | - Wen Chen
- NanoRibo (Shanghai) Biotechnology Co., Ltd., No. 1188 Lianhang Road, Minhang District, Shanghai 200003, China
| | - Deng Pan
- NanoRibo (Shanghai) Biotechnology Co., Ltd., No. 1188 Lianhang Road, Minhang District, Shanghai 200003, China
| | - Zhikang Qian
- NanoRibo (Shanghai) Biotechnology Co., Ltd., No. 1188 Lianhang Road, Minhang District, Shanghai 200003, China
| |
Collapse
|
23
|
Guimaraes GJ, Kim J, Bartlett MG. Characterization of mRNA therapeutics. MASS SPECTROMETRY REVIEWS 2024; 43:1066-1090. [PMID: 37401740 DOI: 10.1002/mas.21856] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 04/25/2023] [Accepted: 05/17/2023] [Indexed: 07/05/2023]
Abstract
Therapeutic messenger RNAs (mRNAs) have emerged as powerful tools in the treatment of complex diseases, especially for conditions that lack efficacious treatment. The successful application of this modality can be attributed to its ability to encode entire proteins. While the large nature of these molecules has supported their success as therapeutics, its extended size creates several analytical challenges. To further support therapeutic mRNA development and its deployment in clinical trials, appropriate methods to support their characterization must be developed. In this review, we describe current analytical methods that have been used in the characterization of RNA quality, identity, and integrity. Advantages and limitations from several analytical techniques ranging from gel electrophoresis to liquid chromatography-mass spectrometry and from shotgun sequencing to intact mass measurements are discussed. We comprehensively describe the application of analytical methods in the measurements of capping efficiency, poly A tail analysis, as well as their applicability in stability studies.
Collapse
Affiliation(s)
- Guilherme J Guimaraes
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, Georgia, USA
| | - Jaeah Kim
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, Georgia, USA
| | - Michael G Bartlett
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, Georgia, USA
| |
Collapse
|
24
|
Hulscher N, McCullough PA, Marotta DE. Strategic deactivation of mRNA COVID-19 vaccines: New applications for siRNA therapy and RIBOTACs. J Gene Med 2024; 26:e3733. [PMID: 39183706 DOI: 10.1002/jgm.3733] [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: 06/02/2024] [Revised: 07/19/2024] [Accepted: 08/13/2024] [Indexed: 08/27/2024] Open
Abstract
The rapid development and authorization of messenger ribonucleic acid (mRNA) vaccines by Pfizer-BioNTech (BNT162b2) and Moderna (mRNA-1273) in 2020 marked a significant milestone in human mRNA product application, overcoming previous obstacles such as mRNA instability and immunogenicity. This paper reviews the strategic modifications incorporated into these vaccines to enhance mRNA stability and translation efficiency, such as the inclusion of nucleoside modifications and optimized mRNA design elements including the 5' cap and poly(A) tail. We highlight emerging concerns regarding the wide systemic biodistribution of these mRNA vaccines leading to prolonged inflammatory responses and other safety concerns. The regulatory framework guiding the biodistribution studies is pivotal in assessing the safety profiles of new mRNA formulations in use today. The stability of mRNA vaccines, their pervasive distribution, and the longevity of the encapsulated mRNA along with unlimited production of the damaging and potentially lethal spike (S) protein call for strategies to mitigate potential adverse effects. Here, we explore the potential of small interfering RNA (siRNA) and ribonuclease targeting chimeras (RIBOTACs) as promising solutions to target, inactivate, and degrade residual and persistent vaccine mRNA, thereby potentially preventing uncontrolled S protein production and reducing toxicity. The targeted nature of siRNA and RIBOTACs allows for precise intervention, offering a path to prevent and mitigate adverse events of mRNA-based therapies. This review calls for further research into siRNA and RIBOTAC applications as antidotes and detoxication products for mRNA vaccine technology.
Collapse
|
25
|
Ma Q, Zhang X, Yang J, Li H, Hao Y, Feng X. Optimization of the 5' untranslated region of mRNA vaccines. Sci Rep 2024; 14:19845. [PMID: 39191885 DOI: 10.1038/s41598-024-70792-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 08/21/2024] [Indexed: 08/29/2024] Open
Abstract
To investigate the impact of different 5' untranslated regions (UTRs) on mRNA vaccine translation efficiency, five dual-reporter gene expression plasmids with different 5'UTRs were constructed. The corresponding mRNA transcripts were transcribed and capped in vitro. By comparing the expression levels of reporter genes with different 5'UTRs, we identified the 5'UTR associated with the highest expression level. Subsequently, HIVgp145 mRNA vaccines containing various 5'UTRs were constructed and verified. The results demonstrated that mRNA 3 (β-globin 5'UTR) displayed the greatest number of green fluorescence-positive cells and the highest luciferase fluorescence intensity in the reporter gene expression system. Further, among the HIVgp145 mRNA vaccines with different 5'UTRs, mRNA 7 (β-globin 5'UTR) exhibited the highest level of expression. These findings indicate that it is feasible to use the 5'UTR of β-globin in an mRNA vaccine, laying the foundation for animal immunogenicity testing.
Collapse
Affiliation(s)
- Qi Ma
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Yingxin Street #100, Xicheng District, Beijing, 100052, China
| | - Xiaoguang Zhang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Yingxin Street #100, Xicheng District, Beijing, 100052, China
| | - Jing Yang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Yingxin Street #100, Xicheng District, Beijing, 100052, China
| | - Hongxia Li
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Yingxin Street #100, Xicheng District, Beijing, 100052, China
| | - Yanzhe Hao
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Yingxin Street #100, Xicheng District, Beijing, 100052, China.
| | - Xia Feng
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Yingxin Street #100, Xicheng District, Beijing, 100052, China.
| |
Collapse
|
26
|
Mueller S. Existing and emerging mRNA vaccines and their environmental impact: a transdisciplinary assessment. ENVIRONMENTAL SCIENCES EUROPE 2024; 36:144. [DOI: 10.1186/s12302-024-00966-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 07/30/2024] [Indexed: 01/05/2025]
|
27
|
Kuzmin IV, Soto Acosta R, Pruitt L, Wasdin PT, Kedarinath K, Hernandez KR, Gonzales KA, Hill K, Weidner NG, Mire C, Engdahl TB, Moon WJ, Popov V, Crowe JE, Georgiev IS, Garcia-Blanco MA, Abbott RK, Bukreyev A. Comparison of uridine and N1-methylpseudouridine mRNA platforms in development of an Andes virus vaccine. Nat Commun 2024; 15:6421. [PMID: 39080316 PMCID: PMC11289437 DOI: 10.1038/s41467-024-50774-3] [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: 07/29/2023] [Accepted: 07/19/2024] [Indexed: 08/02/2024] Open
Abstract
The rodent-borne Andes virus (ANDV) causes a severe disease in humans. We developed an ANDV mRNA vaccine based on the M segment of the viral genome, either with regular uridine (U-mRNA) or N1-methylpseudouridine (m1Ψ-mRNA). Female mice immunized by m1Ψ-mRNA developed slightly greater germinal center (GC) responses than U-mRNA-immunized mice. Single cell RNA and BCR sequencing of the GC B cells revealed similar levels of activation, except an additional cluster of cells exhibiting interferon response in animals vaccinated with U-mRNA but not m1Ψ-mRNA. Similar immunoglobulin class-switching and somatic hypermutations were observed in response to the vaccines. Female Syrian hamsters were immunized via a prime-boost regimen with two doses of each vaccine. The titers of glycoprotein-binding antibodies were greater for U-mRNA construct than for m1Ψ-mRNA construct; however, the titers of ANDV-neutralizing antibodies were similar. Vaccinated animals were challenged with a lethal dose of ANDV, along with a naïve control group. All control animals and two animals vaccinated with a lower dose of m1Ψ-mRNA succumbed to infection whereas other vaccinated animals survived without evidence of virus replication. The data demonstrate the development of a protective vaccine against ANDV and the lack of a substantial effect of m1Ψ modification on immunogenicity and protection in rodents.
Collapse
MESH Headings
- Animals
- Female
- Mice
- Mesocricetus
- Uridine
- Viral Vaccines/immunology
- Viral Vaccines/administration & dosage
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- RNA, Messenger/immunology
- Antibodies, Viral/immunology
- Orthohantavirus/immunology
- Orthohantavirus/genetics
- Antibodies, Neutralizing/immunology
- Germinal Center/immunology
- Pseudouridine/immunology
- Cricetinae
- mRNA Vaccines
- Hemorrhagic Fever, American/prevention & control
- Hemorrhagic Fever, American/immunology
- Hemorrhagic Fever, American/virology
- RNA, Viral/genetics
- RNA, Viral/immunology
- B-Lymphocytes/immunology
- Humans
- Vaccine Development
Collapse
Affiliation(s)
- Ivan V Kuzmin
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
- Galveston National Laboratory, Galveston, TX, USA
| | - Ruben Soto Acosta
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
- Galveston National Laboratory, Galveston, TX, USA
| | - Layne Pruitt
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
| | - Perry T Wasdin
- Vanderbilt University Medical Center, Vanderbilt Vaccine Center, Nashville, TN, USA
| | - Kritika Kedarinath
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
- Galveston National Laboratory, Galveston, TX, USA
| | - Keziah R Hernandez
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
- Galveston National Laboratory, Galveston, TX, USA
| | - Kristyn A Gonzales
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
| | - Kharighan Hill
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
| | - Nicole G Weidner
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
| | - Chad Mire
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
- Galveston National Laboratory, Galveston, TX, USA
| | - Taylor B Engdahl
- Vanderbilt University Medical Center, Vanderbilt Vaccine Center, Nashville, TN, USA
| | | | - Vsevolod Popov
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
| | - James E Crowe
- Vanderbilt University Medical Center, Vanderbilt Vaccine Center, Nashville, TN, USA
| | - Ivelin S Georgiev
- Vanderbilt University Medical Center, Vanderbilt Vaccine Center, Nashville, TN, USA
| | - Mariano A Garcia-Blanco
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA, USA
| | - Robert K Abbott
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA.
| | - Alexander Bukreyev
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA.
- Galveston National Laboratory, Galveston, TX, USA.
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA.
| |
Collapse
|
28
|
Chiu CT, Tsai HH, Chen JY, Hu CMJ, Chen HW. An Immunoreceptor-Targeting Strategy with Minimalistic C3b Peptide Fusion Enhances SARS-CoV-2 RBD mRNA Vaccine Immunogenicity. Int J Nanomedicine 2024; 19:7201-7214. [PMID: 39050877 PMCID: PMC11268571 DOI: 10.2147/ijn.s463546] [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: 02/15/2024] [Accepted: 07/11/2024] [Indexed: 07/27/2024] Open
Abstract
Introduction The clinical success of mRNA vaccine during the COVID-19 pandemic has inspired emerging approaches to elevate mRNA vaccine immunogenicity. Among them, antigen fusion protein designs for improved immune cell targeting have been shown to augment humoral immunity against small antigen targets. Methods This research demonstrates that SARS-CoV-2 receptor binding domain (RBD) fusion with a minimalistic peptide segment of complement component 3b (C3b, residues 727-767) ligand can improve mRNA vaccine immunogenicity through antigen targeting to complement receptor 1 (CR1). We affirm vaccines' antigenicity and targeting ability towards specific receptors through Western blot and immunofluorescence assay. Furthermore, mice immunization studies help the investigation of the antibody responses. Results Using SARS-CoV-2 Omicron RBD antigen, we compare mRNA vaccine formulations expressing RBD fusion protein with mouse C3b peptide (RBD-mC3), RBD fusion protein with mouse Fc (RBD-Fc), and wild-type RBD. Our results confirm the proper antigenicity and normal functionality of RBD-mC3. Upon validating comparable antigen expression by the different vaccine formulations, receptor-targeting capability of the fusion antigens is further confirmed. In mouse immunization studies, we show that while both RBD-mC3 and RBD-Fc elevate vaccine immunogenicity, RBD-mC3 leads to more sustained RBD-specific titers over the RBD-Fc design, presumably due to reduced antigenic diversion by the minimalistic targeting ligand. Conclusion The study demonstrates a novel C3b-based antigen design strategy for immune cell targeting and mRNA vaccine enhancement.
Collapse
Affiliation(s)
- Chun-Ta Chiu
- Department of Veterinary Medicine, National Taiwan University, Taipei, 10617, Taiwan
| | - Hsiao-Han Tsai
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 115, Taiwan
| | - Jing-Yuan Chen
- Department of Veterinary Medicine, National Taiwan University, Taipei, 10617, Taiwan
- Animal Resource Center, National Taiwan University, Taipei, 10617, Taiwan
| | - Che-Ming Jack Hu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 115, Taiwan
| | - Hui-Wen Chen
- Department of Veterinary Medicine, National Taiwan University, Taipei, 10617, Taiwan
- Animal Resource Center, National Taiwan University, Taipei, 10617, Taiwan
| |
Collapse
|
29
|
Yang CH, Li HC, Lo SY. Enhancing recombinant antibody yield in Chinese hamster ovary cells. Tzu Chi Med J 2024; 36:240-250. [PMID: 38993821 PMCID: PMC11236083 DOI: 10.4103/tcmj.tcmj_315_23] [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: 12/17/2023] [Revised: 03/18/2024] [Accepted: 04/12/2024] [Indexed: 07/13/2024] Open
Abstract
A range of recombinant monoclonal antibodies (rMAbs) have found application in treating diverse diseases, spanning various cancers and immune system disorders. Chinese hamster ovary (CHO) cells have emerged as the predominant choice for producing these rMAbs due to their robustness, ease of transfection, and capacity for posttranslational modifications akin to those in human cells. Transient transfection and/or stable expression could be conducted to express rMAbs in CHO cells. To bolster the yield of rMAbs in CHO cells, a multitude of approaches have been developed, encompassing vector optimization, medium formulation, cultivation parameters, and cell engineering. This review succinctly outlines these methodologies when also addressing challenges encountered in the production process, such as issues with aggregation and fucosylation.
Collapse
Affiliation(s)
- Chee-Hing Yang
- Department of Microbiology and Immunology, School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Hui-Chun Li
- Department of Biochemistry, School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Shih-Yen Lo
- Department of Laboratory Medicine and Biotechnology, Tzu Chi University, Hualien, Taiwan
- Department of Laboratory Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical, Hualien, Taiwan
| |
Collapse
|
30
|
Javidanbardan A, Messerian KO, Zydney AL. Membrane technology for the purification of RNA and DNA therapeutics. Trends Biotechnol 2024; 42:714-727. [PMID: 38212210 DOI: 10.1016/j.tibtech.2023.11.016] [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: 08/29/2023] [Revised: 11/29/2023] [Accepted: 11/30/2023] [Indexed: 01/13/2024]
Abstract
Nucleic acid therapeutics have the potential to revolutionize the biopharmaceutical industry, providing highly effective vaccines and novel treatments for cancers and genetic disorders. The successful commercialization of these therapeutics will require development of manufacturing strategies specifically tailored to the purification of nucleic acids. Membrane technologies already play a critical role in the downstream processing of nucleic acid therapeutics, ranging from clarification to concentration to selective purification. This review provides an overview of how membrane systems are currently used for nucleic acid purification, while highlighting areas of future need and opportunity, including adoption of membranes in continuous bioprocessing.
Collapse
Affiliation(s)
- Amin Javidanbardan
- Department of Chemical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
| | - Kevork Oliver Messerian
- Department of Chemical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
| | - Andrew L Zydney
- Department of Chemical Engineering, The Pennsylvania State University, University Park, PA 16802, USA.
| |
Collapse
|
31
|
He W, Zhang X, Zou Y, Li J, Wang C, He Y, Jin Q, Ye J. Effective Synthesis of High-Integrity mRNA Using In Vitro Transcription. Molecules 2024; 29:2461. [PMID: 38893337 PMCID: PMC11173937 DOI: 10.3390/molecules29112461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 05/12/2024] [Accepted: 05/14/2024] [Indexed: 06/21/2024] Open
Abstract
mRNA vaccines are entering a period of rapid development. However, their synthesis is still plagued by challenges related to mRNA impurities and fragments (incomplete mRNA). Most impurities of mRNA products transcribed in vitro are mRNA fragments. Only full-length mRNA transcripts containing both a 5'-cap and a 3'-poly(A) structure are viable for in vivo expression. Therefore, RNA fragments are the primary product-related impurities that significantly hinder mRNA efficacy and must be effectively controlled; these species are believed to originate from either mRNA hydrolysis or premature transcriptional termination. In the manufacturing of commercial mRNA vaccines, T7 RNA polymerase-catalyzed in vitro transcription (IVT) synthesis is a well-established method for synthesizing long RNA transcripts. This study identified a pivotal domain on the T7 RNA polymerase that is associated with erroneous mRNA release. By leveraging the advantageous properties of a T7 RNA polymerase mutant and precisely optimized IVT process parameters, we successfully achieved an mRNA integrity exceeding 91%, thereby further unlocking the immense potential of mRNA therapeutics.
Collapse
Affiliation(s)
- Wei He
- College of Biology and the Environment, Nanjing Forestry University, Nanjing 210037, China;
- Vazyme Biotech Co., Ltd., Nanjing 210037, China; (X.Z.); (Y.Z.); (J.L.); (C.W.)
| | - Xinya Zhang
- Vazyme Biotech Co., Ltd., Nanjing 210037, China; (X.Z.); (Y.Z.); (J.L.); (C.W.)
| | - Yangxiaoyu Zou
- Vazyme Biotech Co., Ltd., Nanjing 210037, China; (X.Z.); (Y.Z.); (J.L.); (C.W.)
| | - Ji Li
- Vazyme Biotech Co., Ltd., Nanjing 210037, China; (X.Z.); (Y.Z.); (J.L.); (C.W.)
| | - Chong Wang
- Vazyme Biotech Co., Ltd., Nanjing 210037, China; (X.Z.); (Y.Z.); (J.L.); (C.W.)
| | - Yucai He
- School of Pharmacy, Changzhou University, Changzhou 213164, China
| | - Qiuheng Jin
- Vazyme Biotech Co., Ltd., Nanjing 210037, China; (X.Z.); (Y.Z.); (J.L.); (C.W.)
| | - Jianren Ye
- College of Biology and the Environment, Nanjing Forestry University, Nanjing 210037, China;
| |
Collapse
|
32
|
Park S, Kim M, Lee JW. Optimizing Nucleic Acid Delivery Systems through Barcode Technology. ACS Synth Biol 2024; 13:1006-1018. [PMID: 38526308 DOI: 10.1021/acssynbio.3c00602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/26/2024]
Abstract
Conventional biological experiments often focus on in vitro assays because of the inherent limitations when handling multiple variables in vivo, including labor-intensive and time-consuming procedures. Often only a subset of samples demonstrating significant efficacy in the in vitro assays can be evaluated in vivo. Nonetheless, because of the low correlation between the in vitro and in vivo tests, evaluation of the variables under examination in vivo and not solely in vitro is critical. An emerging approach to achieve high-throughput in vivo tests involves using a barcode system consisting of various nucleotide combinations. Unique barcodes for each variant enable the simultaneous testing of multiple entities, eliminating the need for separate individual tests. Subsequently, to identify crucial parameters, samples were collected and analyzed using barcode sequencing. This review explores the development of barcode design and its applications, including the evaluation of nucleic acid delivery systems and the optimization of gene expression in vivo.
Collapse
Affiliation(s)
- Soan Park
- Department of Chemical Engineering, Pohang University of Science and Technology, 77 CheongamRo, Gyeongbuk, 37673 NamGu, Pohang, Republic of Korea
| | - Mibang Kim
- Department of Chemical Engineering, Pohang University of Science and Technology, 77 CheongamRo, Gyeongbuk, 37673 NamGu, Pohang, Republic of Korea
| | - Jeong Wook Lee
- Department of Chemical Engineering, Pohang University of Science and Technology, 77 CheongamRo, Gyeongbuk, 37673 NamGu, Pohang, Republic of Korea
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology, 77 CheongamRo, Gyeongbuk, 37673 NamGu, Pohang, Republic of Korea
| |
Collapse
|
33
|
Farookhi H, Xia X. Differential Selection for Translation Efficiency Shapes Translation Machineries in Bacterial Species. Microorganisms 2024; 12:768. [PMID: 38674712 PMCID: PMC11052298 DOI: 10.3390/microorganisms12040768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 04/01/2024] [Accepted: 04/09/2024] [Indexed: 04/28/2024] Open
Abstract
Different bacterial species have dramatically different generation times, from 20-30 min in Escherichia coli to about two weeks in Mycobacterium leprae. The translation machinery in a cell needs to synthesize all proteins for a new cell in each generation. The three subprocesses of translation, i.e., initiation, elongation, and termination, are expected to be under stronger selection pressure to optimize in short-generation bacteria (SGB) such as Vibrio natriegens than in the long-generation Mycobacterium leprae. The initiation efficiency depends on the start codon decoded by the initiation tRNA, the optimal Shine-Dalgarno (SD) decoded by the anti-SD (aSD) sequence on small subunit rRNA, and the secondary structure that may embed the initiation signals and prevent them from being decoded. The elongation efficiency depends on the tRNA pool and codon usage. The termination efficiency in bacteria depends mainly on the nature of the stop codon and the nucleotide immediately downstream of the stop codon. By contrasting SGB with long-generation bacteria (LGB), we predict (1) SGB to have more ribosome RNA operons to produce ribosomes, and more tRNA genes for carrying amino acids to ribosomes, (2) SGB to have a higher percentage of genes using AUG as the start codon and UAA as the stop codon than LGB, (3) SGB to exhibit better codon and anticodon adaptation than LGB, and (4) SGB to have a weaker secondary structure near the translation initiation signals than LGB. These differences between SGB and LGB should be more pronounced in highly expressed genes than the rest of the genes. We present empirical evidence in support of these predictions.
Collapse
Affiliation(s)
- Heba Farookhi
- Department of Biology, University of Ottawa, Ottawa, ON K1N 6N5, Canada;
| | - Xuhua Xia
- Department of Biology, University of Ottawa, Ottawa, ON K1N 6N5, Canada;
- Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| |
Collapse
|
34
|
Paremskaia AI, Kogan AA, Murashkina A, Naumova DA, Satish A, Abramov IS, Feoktistova SG, Mityaeva ON, Deviatkin AA, Volchkov PY. Codon-optimization in gene therapy: promises, prospects and challenges. Front Bioeng Biotechnol 2024; 12:1371596. [PMID: 38605988 PMCID: PMC11007035 DOI: 10.3389/fbioe.2024.1371596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 03/19/2024] [Indexed: 04/13/2024] Open
Abstract
Codon optimization has evolved to enhance protein expression efficiency by exploiting the genetic code's redundancy, allowing for multiple codon options for a single amino acid. Initially observed in E. coli, optimal codon usage correlates with high gene expression, which has propelled applications expanding from basic research to biopharmaceuticals and vaccine development. The method is especially valuable for adjusting immune responses in gene therapies and has the potenial to create tissue-specific therapies. However, challenges persist, such as the risk of unintended effects on protein function and the complexity of evaluating optimization effectiveness. Despite these issues, codon optimization is crucial in advancing gene therapeutics. This study provides a comprehensive review of the current metrics for codon-optimization, and its practical usage in research and clinical applications, in the context of gene therapy.
Collapse
Affiliation(s)
- Anastasiia Iu Paremskaia
- Federal Research Center for Innovator and Emerging Biomedical and Pharmaceutical Technologies, Moscow, Russia
| | - Anna A. Kogan
- Federal Research Center for Innovator and Emerging Biomedical and Pharmaceutical Technologies, Moscow, Russia
| | - Anastasiia Murashkina
- Federal Research Center for Innovator and Emerging Biomedical and Pharmaceutical Technologies, Moscow, Russia
| | - Daria A. Naumova
- Federal Research Center for Innovator and Emerging Biomedical and Pharmaceutical Technologies, Moscow, Russia
| | - Anakha Satish
- Federal Research Center for Innovator and Emerging Biomedical and Pharmaceutical Technologies, Moscow, Russia
| | - Ivan S. Abramov
- Federal Research Center for Innovator and Emerging Biomedical and Pharmaceutical Technologies, Moscow, Russia
- The MCSC named after A. S. Loginov, Moscow, Russia
| | - Sofya G. Feoktistova
- Federal Research Center for Innovator and Emerging Biomedical and Pharmaceutical Technologies, Moscow, Russia
| | - Olga N. Mityaeva
- Federal Research Center for Innovator and Emerging Biomedical and Pharmaceutical Technologies, Moscow, Russia
| | - Andrei A. Deviatkin
- Federal Research Center for Innovator and Emerging Biomedical and Pharmaceutical Technologies, Moscow, Russia
| | - Pavel Yu Volchkov
- Federal Research Center for Innovator and Emerging Biomedical and Pharmaceutical Technologies, Moscow, Russia
- The MCSC named after A. S. Loginov, Moscow, Russia
| |
Collapse
|
35
|
Huang X, Zhu X, Yang H, Li Q, Gai L, Sui X, Lu H, Feng J. Nanomaterial Delivery Vehicles for the Development of Neoantigen Tumor Vaccines for Personalized Treatment. Molecules 2024; 29:1462. [PMID: 38611742 PMCID: PMC11012694 DOI: 10.3390/molecules29071462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 03/16/2024] [Accepted: 03/22/2024] [Indexed: 04/14/2024] Open
Abstract
Tumor vaccines have been considered a promising therapeutic approach for treating cancer in recent years. With the development of sequencing technologies, tumor vaccines based on neoantigens or genomes specifically expressed in tumor cells, mainly in the form of peptides, nucleic acids, and dendritic cells, are beginning to receive widespread attention. Therefore, in this review, we have introduced different forms of neoantigen vaccines and discussed the development of these vaccines in treating cancer. Furthermore, neoantigen vaccines are influenced by factors such as antigen stability, weak immunogenicity, and biosafety in addition to sequencing technology. Hence, the biological nanomaterials, polymeric nanomaterials, inorganic nanomaterials, etc., used as vaccine carriers are principally summarized here, which may contribute to the design of neoantigen vaccines for improved stability and better efficacy.
Collapse
Affiliation(s)
- Xiaoyu Huang
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (X.H.); (X.Z.); (H.Y.); (Q.L.); (X.S.)
| | - Xiaolong Zhu
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (X.H.); (X.Z.); (H.Y.); (Q.L.); (X.S.)
| | - Huan Yang
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (X.H.); (X.Z.); (H.Y.); (Q.L.); (X.S.)
| | - Qinyi Li
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (X.H.); (X.Z.); (H.Y.); (Q.L.); (X.S.)
| | - Lizhi Gai
- College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology of Ministry of Education, and Key Laboratory of Organosilicon Material Technology of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China;
| | - Xinbing Sui
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (X.H.); (X.Z.); (H.Y.); (Q.L.); (X.S.)
| | - Hua Lu
- College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology of Ministry of Education, and Key Laboratory of Organosilicon Material Technology of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China;
| | - Jiao Feng
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (X.H.); (X.Z.); (H.Y.); (Q.L.); (X.S.)
| |
Collapse
|
36
|
Patel DR, Minns AM, Sim DG, Field CJ, Kerr AE, Heinly TA, Luley EH, Rossi RM, Bator CM, Moustafa IM, Norton EB, Hafenstein SL, Lindner SE, Sutton TC. Intranasal SARS-CoV-2 RBD decorated nanoparticle vaccine enhances viral clearance in the Syrian hamster model. Microbiol Spectr 2024; 12:e0499822. [PMID: 38334387 PMCID: PMC10923206 DOI: 10.1128/spectrum.04998-22] [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: 12/05/2022] [Accepted: 01/17/2024] [Indexed: 02/10/2024] Open
Abstract
Multiple vaccines have been developed and licensed for severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). While these vaccines reduce disease severity, they do not prevent infection. To prevent infection and limit transmission, vaccines must be developed that induce immunity in the respiratory tract. Therefore, we performed proof-of-principle studies with an intranasal nanoparticle vaccine against SARS-CoV-2. The vaccine candidate consisted of the self-assembling 60-subunit I3-01 protein scaffold covalently decorated with the SARS-CoV-2 receptor-binding domain (RBD) using the SpyCatcher-SpyTag system. We verified the intended antigen display features by reconstructing the I3-01 scaffold to 3.4 A using cryogenicelectron microscopy. Using this RBD-grafted SpyCage scaffold (RBD + SpyCage), we performed two intranasal vaccination studies in the "gold-standard" pre-clinical Syrian hamster model. The initial study focused on assessing the immunogenicity of RBD + SpyCage combined with the LTA1 intranasal adjuvant. These studies showed RBD + SpyCage vaccination induced an antibody response that promoted viral clearance but did not prevent infection. Inclusion of the LTA1 adjuvant enhanced the magnitude of the antibody response but did not enhance protection. Thus, in an expanded study, in the absence of an intranasal adjuvant, we evaluated if covalent bonding of RBD to the scaffold was required to induce an antibody response. Covalent grafting of RBD was required for the vaccine to be immunogenic, and animals vaccinated with RBD + SpyCage more rapidly cleared SARS-CoV-2 from both the upper and lower respiratory tract. These findings demonstrate the intranasal SpyCage vaccine platform can induce protection against SARS-CoV-2 and, with additional modifications to improve immunogenicity, is a versatile platform for the development of intranasal vaccines targeting respiratory pathogens.IMPORTANCEDespite the availability of efficacious COVID vaccines that reduce disease severity, SARS-CoV-2 continues to spread. To limit SARS-CoV-2 transmission, the next generation of vaccines must induce immunity in the mucosa of the upper respiratory tract. Therefore, we performed proof-of-principle, intranasal vaccination studies with a recombinant protein nanoparticle scaffold, SpyCage, decorated with the RBD of the S protein (SpyCage + RBD). We show that SpyCage + RBD was immunogenic and enhanced SARS-CoV-2 clearance from the nose and lungs of Syrian hamsters. Moreover, covalent grafting of the RBD to the scaffold was required to induce an immune response when given via the intranasal route. These proof-of-concept findings indicate that with further enhancements to immunogenicity (e.g., adjuvant incorporation and antigen optimization), the SpyCage scaffold has potential as a versatile, intranasal vaccine platform for respiratory pathogens.
Collapse
Affiliation(s)
- Devanshi R. Patel
- Department of Veterinary and Biomedical Sciences, Pennsylvania State University, University Park, Pennsylvania, USA
- The Huck Institutes of Life Sciences, Pennsylvania State University, University Park, Pennsylvania, USA
| | - Allen M. Minns
- The Huck Institutes of Life Sciences, Pennsylvania State University, University Park, Pennsylvania, USA
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, Pennsylvania, USA
- The Huck Center for Malaria Research, University Park, Pennsylvania, USA
| | - Derek G. Sim
- The Huck Institutes of Life Sciences, Pennsylvania State University, University Park, Pennsylvania, USA
- Department of Biology, Pennsylvania State University, University Park, Pennsylvania, USA
| | - Cassandra J. Field
- Department of Veterinary and Biomedical Sciences, Pennsylvania State University, University Park, Pennsylvania, USA
- The Huck Institutes of Life Sciences, Pennsylvania State University, University Park, Pennsylvania, USA
| | - Abigail E. Kerr
- Department of Veterinary and Biomedical Sciences, Pennsylvania State University, University Park, Pennsylvania, USA
- The Huck Institutes of Life Sciences, Pennsylvania State University, University Park, Pennsylvania, USA
| | - Talia A. Heinly
- Department of Veterinary and Biomedical Sciences, Pennsylvania State University, University Park, Pennsylvania, USA
- The Huck Institutes of Life Sciences, Pennsylvania State University, University Park, Pennsylvania, USA
| | - Erin H. Luley
- Department of Veterinary and Biomedical Sciences, Pennsylvania State University, University Park, Pennsylvania, USA
- Animal Diagnostic Laboratory, Pennsylvania State University, University Park, Pennsylvania, USA
| | - Randall M. Rossi
- The Huck Institutes of Life Sciences, Pennsylvania State University, University Park, Pennsylvania, USA
| | - Carol M. Bator
- The Huck Institutes of Life Sciences, Pennsylvania State University, University Park, Pennsylvania, USA
| | - Ibrahim M. Moustafa
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, Pennsylvania, USA
| | - Elizabeth B. Norton
- Department of Microbiology and Immunology, Tulane University, New Orleans, Louisiana, USA
| | - Susan L. Hafenstein
- The Huck Institutes of Life Sciences, Pennsylvania State University, University Park, Pennsylvania, USA
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, Pennsylvania, USA
- Department of Medicine, Pennsylvania State University, University Park, Pennsylvania, USA
| | - Scott E. Lindner
- The Huck Institutes of Life Sciences, Pennsylvania State University, University Park, Pennsylvania, USA
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, Pennsylvania, USA
- The Huck Center for Malaria Research, University Park, Pennsylvania, USA
| | - Troy C. Sutton
- Department of Veterinary and Biomedical Sciences, Pennsylvania State University, University Park, Pennsylvania, USA
- The Huck Institutes of Life Sciences, Pennsylvania State University, University Park, Pennsylvania, USA
| |
Collapse
|
37
|
Reis-Claro I, Silva MI, Moutinho A, Garcia BC, Pereira-Castro I, Moreira A. Application of the iPLUS non-coding sequence in improving biopharmaceuticals production. Front Bioeng Biotechnol 2024; 12:1355957. [PMID: 38380261 PMCID: PMC10876878 DOI: 10.3389/fbioe.2024.1355957] [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: 12/14/2023] [Accepted: 01/25/2024] [Indexed: 02/22/2024] Open
Abstract
The biotechnological landscape has witnessed significant growth in biological therapeutics particularly in the field of recombinant protein production. Here we investigate the function of 3'UTR cis-regulatory elements in increasing mRNA and protein levels in different biological therapeutics and model systems, spanning from monoclonal antibodies to mRNA vaccines. We explore the regulatory function of iPLUS - a universal sequence capable of consistently augmenting recombinant protein levels. By incorporating iPLUS in a vector to express a monoclonal antibody used in immunotherapy, in a mammalian cell line used by the industry (ExpiCHO), trastuzumab production increases by 2-fold. As yeast Pichia pastoris is widely used in the manufacture of industrial enzymes and pharmaceuticals, we then used iPLUS in tandem (3x) and iPLUSv2 (a variant of iPLUS) to provide proof-of-concept data that it increases the production of a reporter protein more than 100-fold. As iPLUS functions by also increasing mRNA levels, we hypothesize that these sequences could be used as an asset in the mRNA vaccine industry. In fact, by including iPLUSv2 downstream of Spike we were able to double its production. Moreover, the same effect was observed when we introduced iPLUSv2 downstream of MAGEC2, a tumor-specific antigen tested for cancer mRNA vaccines. Taken together, our study provides data (TLR4) showing that iPLUS may be used as a valuable asset in a variety of systems used by the biotech and biopharmaceutical industry. Our results underscore the critical role of non-coding sequences in controlling gene expression, offering a promising avenue to accelerate, enhance, and cost-effectively optimize biopharmaceutical production processes.
Collapse
Affiliation(s)
- Inês Reis-Claro
- Gene Regulation, i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Maria Inês Silva
- Gene Regulation, i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Ana Moutinho
- Gene Regulation, i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Beatriz C. Garcia
- Gene Regulation, i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Isabel Pereira-Castro
- Gene Regulation, i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Alexandra Moreira
- Gene Regulation, i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| |
Collapse
|
38
|
Schreckenberg R, Woitasky N, Itani N, Czech L, Ferdinandy P, Schulz R. Cardiac side effects of RNA-based SARS-CoV-2 vaccines: Hidden cardiotoxic effects of mRNA-1273 and BNT162b2 on ventricular myocyte function and structure. Br J Pharmacol 2024; 181:345-361. [PMID: 37828636 DOI: 10.1111/bph.16262] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 07/16/2023] [Accepted: 08/03/2023] [Indexed: 10/14/2023] Open
Abstract
BACKGROUND AND PURPOSE To protect against SARS-CoV-2 infection, the first mRNA-based vaccines, Spikevax (mRNA-1273, Moderna) and Comirnaty (BNT162b2, Pfizer/Biontech), were approved in 2020. The structure and assembly of the immunogen-in both cases, the SARS-CoV-2 spike (S) glycoprotein-are determined by a messenger RNA sequence that is translated by endogenous ribosomes. Cardiac side-effects, which for the most part can be classified by their clinical symptoms as myo- and/or pericarditis, can be caused by both mRNA-1273 and BNT162b2. EXPERIMENTAL APPROACH As persuasive theories for the underlying pathomechanisms have yet to be developed, this study investigated the effect of mRNA-1273 and BNT162b2 on the function, structure, and viability of isolated adult rat cardiomyocytes over a 72 h period. KEY RESULTS In the first 24 h after application, both mRNA-1273 and BNT162b2 caused neither functional disturbances nor morphological abnormalities. After 48 h, expression of the encoded spike protein was detected in ventricular cardiomyocytes for both mRNAs. At this point in time, mRNA-1273 induced arrhythmic as well as completely irregular contractions associated with irregular as well as localized calcium transients, which provide indications of significant dysfunction of the cardiac ryanodine receptor (RyR2). In contrast, BNT162b2 increased cardiomyocyte contraction via significantly increased protein kinase A (PKA) activity at the cellular level. CONCLUSION AND IMPLICATIONS Here, we demonstrated for the first time, that in isolated cardiomyocytes, both mRNA-1273 and BNT162b2 induce specific dysfunctions that correlate pathophysiologically to cardiomyopathy. Both RyR2 impairment and sustained PKA activation may significantly increase the risk of acute cardiac events.
Collapse
Affiliation(s)
- Rolf Schreckenberg
- Institute of Physiology, Faculty of Medicine, Justus-Liebig University, Gießen, Germany
| | - Nadine Woitasky
- Institute of Physiology, Faculty of Medicine, Justus-Liebig University, Gießen, Germany
| | - Nadja Itani
- Institute of Physiology, Faculty of Medicine, Justus-Liebig University, Gießen, Germany
| | - Laureen Czech
- Institute of Physiology, Faculty of Medicine, Justus-Liebig University, Gießen, Germany
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, National Heart Laboratory, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
| | - Rainer Schulz
- Institute of Physiology, Faculty of Medicine, Justus-Liebig University, Gießen, Germany
| |
Collapse
|
39
|
Zhang H, Vandesompele J, Braeckmans K, De Smedt SC, Remaut K. Nucleic acid degradation as barrier to gene delivery: a guide to understand and overcome nuclease activity. Chem Soc Rev 2024; 53:317-360. [PMID: 38073448 DOI: 10.1039/d3cs00194f] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2024]
Abstract
Gene therapy is on its way to revolutionize the treatment of both inherited and acquired diseases, by transferring nucleic acids to correct a disease-causing gene in the target cells of patients. In the fight against infectious diseases, mRNA-based therapeutics have proven to be a viable strategy in the recent Covid-19 pandemic. Although a growing number of gene therapies have been approved, the success rate is limited when compared to the large number of preclinical and clinical trials that have been/are being performed. In this review, we highlight some of the hurdles which gene therapies encounter after administration into the human body, with a focus on nucleic acid degradation by nucleases that are extremely abundant in mammalian organs, biological fluids as well as in subcellular compartments. We overview the available strategies to reduce the biodegradation of gene therapeutics after administration, including chemical modifications of the nucleic acids, encapsulation into vectors and co-administration with nuclease inhibitors and discuss which strategies are applied for clinically approved nucleic acid therapeutics. In the final part, we discuss the currently available methods and techniques to qualify and quantify the integrity of nucleic acids, with their own strengths and limitations.
Collapse
Affiliation(s)
- Heyang Zhang
- Laboratory for General Biochemistry and Physical Pharmacy, Department of Pharmaceutical Sciences, Ghent University, 9000 Ghent, Belgium.
- Leiden Academic Centre for Drug Research, Leiden University, 2333 CC Leiden, The Netherlands
| | - Jo Vandesompele
- Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Kevin Braeckmans
- Laboratory for General Biochemistry and Physical Pharmacy, Department of Pharmaceutical Sciences, Ghent University, 9000 Ghent, Belgium.
- Centre for Nano- and Biophotonics, Ghent University, 9000 Ghent, Belgium
| | - Stefaan C De Smedt
- Laboratory for General Biochemistry and Physical Pharmacy, Department of Pharmaceutical Sciences, Ghent University, 9000 Ghent, Belgium.
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- Centre for Nano- and Biophotonics, Ghent University, 9000 Ghent, Belgium
| | - Katrien Remaut
- Laboratory for General Biochemistry and Physical Pharmacy, Department of Pharmaceutical Sciences, Ghent University, 9000 Ghent, Belgium.
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| |
Collapse
|
40
|
Valdes Angues R, Perea Bustos Y. SARS-CoV-2 Vaccination and the Multi-Hit Hypothesis of Oncogenesis. Cureus 2023; 15:e50703. [PMID: 38234925 PMCID: PMC10792266 DOI: 10.7759/cureus.50703] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/16/2023] [Indexed: 01/19/2024] Open
Abstract
Cancer is a complex and dynamic disease. The "hallmarks of cancer" were proposed by Hanahan and Weinberg (2000) as a group of biological competencies that human cells attain as they progress from normalcy to neoplastic transformation. These competencies include self-sufficiency in proliferative signaling, insensitivity to growth-suppressive signals and immune surveillance, the ability to evade cell death, enabling replicative immortality, reprogramming energy metabolism, inducing angiogenesis, and activating tissue invasion and metastasis. Underlying these competencies are genome instability, which expedites their acquisition, and inflammation, which fosters their function(s). Additionally, cancer exhibits another dimension of complexity: a heterogeneous repertoire of infiltrating and resident host cells, secreted factors, and extracellular matrix, known as the tumor microenvironment, that through a dynamic and reciprocal relationship with cancer cells supports immortality, local invasion, and metastatic dissemination. This staggering intricacy calls for caution when advising all people with cancer (or a previous history of cancer) to receive the COVID-19 primary vaccine series plus additional booster doses. Moreover, because these patients were not included in the pivotal clinical trials, considerable uncertainty remains regarding vaccine efficacy, safety, and the risk of interactions with anticancer therapies, which could reduce the value and innocuity of either medical treatment. After reviewing the available literature, we are particularly concerned that certain COVID-19 vaccines may generate a pro-tumorigenic milieu (i.e., a specific environment that could lead to neoplastic transformation) that predisposes some (stable) oncologic patients and survivors to cancer progression, recurrence, and/or metastasis. This hypothesis is based on biological plausibility and fulfillment of the multi-hit hypothesis of oncogenesis (i.e., induction of lymphopenia and inflammation, downregulation of angiotensin-converting enzyme 2 (ACE2) expression, activation of oncogenic cascades, sequestration of tumor suppressor proteins, dysregulation of the RNA-G quadruplex-protein binding system, alteration of type I interferon responses, unsilencing of retrotransposable elements, etc.) together with growing evidence and safety reports filed to Vaccine Adverse Effects Report System (VAERS) suggesting that some cancer patients experienced disease exacerbation or recurrence following COVID-19 vaccination. In light of the above and because some of these concerns (i.e., alteration of oncogenic pathways, promotion of inflammatory cascades, and dysregulation of the renin-angiotensin system) also apply to cancer patients infected with SARS-CoV-2, we encourage the scientific and medical community to urgently evaluate the impact of both COVID-19 and COVID-19 vaccination on cancer biology and tumor registries, adjusting public health recommendations accordingly.
Collapse
Affiliation(s)
- Raquel Valdes Angues
- Neurology, Oregon Health and Science University School of Medicine, Portland, USA
| | | |
Collapse
|
41
|
Kurkin DV, Morkovin EI, Bakulin DA, Gorbunova YV, Kolosov YA, Dzhavakhyan MA, Makarenko IE, Drai RV, Zaborovsky AV, Shatalova OV, Strygin AV, Petrov VI, Pleten AP, Prokopov AA, Tatarenko-Kozmina TY. Overview of drugs approved by the FDA in 2022. PHARMACY & PHARMACOLOGY 2023; 11:193-210. [DOI: 10.19163/2307-9266-2023-11-3-193-210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Abstract
The aim of the work is to conduct a review of medications approved by the FDA in 2022.Materials and methods. In searching for the materials to write this review article, bibliographic databases including PubMed, Google Scholar and e-library.ru were utilized. The search was conducted for the publications spanning the period from 2008 to 2023. Herewith, the following keywords and word combinations were used: new drug approval, NDA, drug authorization, approval package, breakthrough medicine.Results. The discovery and development of medications are among the most crucial scientific processes in healthcare. Developing a new drug is a highly intricate, expensive, and time-consuming process. Nowadays, the problem of costs reduction and the process of expedited discovering of new medications are particularly pertinent. To optimize the search for active compounds, virtual and high-throughput screenings, machine learning, artificial intelligence, cryo-electron microscopy, and drug repurposing are employed. Simultaneously, the search for original molecules to serve as the basis for innovative drugs continues. This article presents a review of medications approved by the FDA in 2022 for the treatment of various pathologies.Conclusion. A drug development is a complex and resource-intensive process, with only a small fraction of candidates advancing to clinical trials. A drug design evolves in tandem with societal needs, and this review highlights some of the medications approved by the FDA in 2022. Technological advancements are expected to expedite drug development, potentially reducing the time to the market. Biotechnology, including cell therapy, holds significant prospects, and achievements in genetic mapping and chip technologies will enhance the accessibility of personalized pharmacology.
Collapse
Affiliation(s)
- D. V. Kurkin
- 1.Yevdokimov Moscow State University of Medicine and Dentistry.
2.Volgograd State Medical University
| | | | | | | | - Yu. A. Kolosov
- Yevdokimov Moscow State University of Medicine and Dentistry
| | - M. A. Dzhavakhyan
- 1.Yevdokimov Moscow State University of Medicine and Dentistry.
2.All-Russian Scientific Research Institute of Medicinal and Aromatic Plants
| | - I. E. Makarenko
- 1.Yevdokimov Moscow State University of Medicine and Dentistry.
2.Farm-Holding
| | | | | | | | | | | | - A. P. Pleten
- Yevdokimov Moscow State University of Medicine and Dentistry
| | - A. A. Prokopov
- Yevdokimov Moscow State University of Medicine and Dentistry
| | | |
Collapse
|
42
|
Deo S, Desai K, Patare A, Wadapurkar R, Rade S, Mahudkar S, Sathe M, Srivastava S, Prasanna P, Singh A. Evaluation of self-amplifying mRNA platform for protein expression and genetic stability: Implication for mRNA therapies. Biochem Biophys Res Commun 2023; 680:108-118. [PMID: 37738900 DOI: 10.1016/j.bbrc.2023.09.016] [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: 06/20/2023] [Revised: 09/01/2023] [Accepted: 09/07/2023] [Indexed: 09/24/2023]
Abstract
The consecutive launch of mRNA vaccines like mRNA-1273, BNT 162b2, and GEMCOVAC®-19 against COVID-19 has triggered the debate of long-term expression, safety, and genomic integration of the mRNA vaccine platforms. In the present study, we examined the longevity of antigenic protein expression of mRNA-614 and mRNA-S1LC based on self-amplifying mRNA (SAM) in Expi-293F™, HEK-293 T, and ARPE-19 cells. The protein expression was checked by sandwich-ELISA, FACS, luciferase activity assay, and Western blot. The transcribed antigenic mRNA was sequenced and found to be un-mutated. Additionally, no genomic integration of the reverse transcribed mRNA was observed even up to 7 days post-transfection as verified by PCR. Furthermore, we have generated high-quality 3D structures of non-structural proteins (nsPs) in silico and the genes encoding for the nsPs were cloned and expressed using the T7 system. Findings from the current study have strengthened the fact that the alphavirus-based SAM platform has the potential to become a modality in the upcoming years.
Collapse
Affiliation(s)
- Swarda Deo
- Gennova Biopharmaceuticals Ltd. ITBT Park, Hinjawadi Phase 2 Road, Hinjawadi Rajiv Gandhi Infotech Park, Hinjawadi, Pune, Maharashtra, 411057, India
| | - Kaushik Desai
- Gennova Biopharmaceuticals Ltd. ITBT Park, Hinjawadi Phase 2 Road, Hinjawadi Rajiv Gandhi Infotech Park, Hinjawadi, Pune, Maharashtra, 411057, India
| | - Aishwarya Patare
- Gennova Biopharmaceuticals Ltd. ITBT Park, Hinjawadi Phase 2 Road, Hinjawadi Rajiv Gandhi Infotech Park, Hinjawadi, Pune, Maharashtra, 411057, India
| | - Rucha Wadapurkar
- Gennova Biopharmaceuticals Ltd. ITBT Park, Hinjawadi Phase 2 Road, Hinjawadi Rajiv Gandhi Infotech Park, Hinjawadi, Pune, Maharashtra, 411057, India
| | - Saniya Rade
- Gennova Biopharmaceuticals Ltd. ITBT Park, Hinjawadi Phase 2 Road, Hinjawadi Rajiv Gandhi Infotech Park, Hinjawadi, Pune, Maharashtra, 411057, India
| | - Siddhi Mahudkar
- Gennova Biopharmaceuticals Ltd. ITBT Park, Hinjawadi Phase 2 Road, Hinjawadi Rajiv Gandhi Infotech Park, Hinjawadi, Pune, Maharashtra, 411057, India
| | - Madhura Sathe
- Gennova Biopharmaceuticals Ltd. ITBT Park, Hinjawadi Phase 2 Road, Hinjawadi Rajiv Gandhi Infotech Park, Hinjawadi, Pune, Maharashtra, 411057, India
| | - Shalini Srivastava
- Gennova Biopharmaceuticals Ltd. ITBT Park, Hinjawadi Phase 2 Road, Hinjawadi Rajiv Gandhi Infotech Park, Hinjawadi, Pune, Maharashtra, 411057, India
| | - Pragya Prasanna
- Gennova Biopharmaceuticals Ltd. ITBT Park, Hinjawadi Phase 2 Road, Hinjawadi Rajiv Gandhi Infotech Park, Hinjawadi, Pune, Maharashtra, 411057, India
| | - Ajay Singh
- Gennova Biopharmaceuticals Ltd. ITBT Park, Hinjawadi Phase 2 Road, Hinjawadi Rajiv Gandhi Infotech Park, Hinjawadi, Pune, Maharashtra, 411057, India.
| |
Collapse
|
43
|
Perenkov AD, Sergeeva AD, Vedunova MV, Krysko DV. In Vitro Transcribed RNA-Based Platform Vaccines: Past, Present, and Future. Vaccines (Basel) 2023; 11:1600. [PMID: 37897003 PMCID: PMC10610676 DOI: 10.3390/vaccines11101600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 10/07/2023] [Accepted: 10/10/2023] [Indexed: 10/29/2023] Open
Abstract
mRNA was discovered in 1961, but it was not used as a vaccine until after three decades. Recently, the development of mRNA vaccine technology gained great impetus from the pursuit of vaccines against COVID-19. To improve the properties of RNA vaccines, and primarily their circulation time, self-amplifying mRNA and trans-amplifying mRNA were developed. A separate branch of mRNA technology is circular RNA vaccines, which were developed with the discovery of the possibility of translation on their protein matrix. Circular RNA has several advantages over mRNA vaccines and is considered a fairly promising platform, as is trans-amplifying mRNA. This review presents an overview of the mRNA platform and a critical discussion of the more modern self-amplifying mRNA, trans-amplifying mRNA, and circular RNA platforms created on its basis. Finally, the main features, advantages, and disadvantages of each of the presented mRNA platforms are discussed. This discussion will facilitate the decision-making process in selecting the most appropriate platform for creating RNA vaccines against cancer or viral diseases.
Collapse
Affiliation(s)
- Alexey D. Perenkov
- Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhny Novgorod, 603022 Nizhny Novgorod, Russia
| | - Alena D. Sergeeva
- Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhny Novgorod, 603022 Nizhny Novgorod, Russia
| | - Maria V. Vedunova
- Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhny Novgorod, 603022 Nizhny Novgorod, Russia
| | - Dmitri V. Krysko
- Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhny Novgorod, 603022 Nizhny Novgorod, Russia
- Cell Death Investigation and Therapy (CDIT) Laboratory, Anatomy and Embryology Unit, Department of Human Structure and Repair, Faculty of Medicine and Health Science, Ghent University, 9000 Ghent, Belgium
- Cancer Research Institute Ghent, 9000 Ghent, Belgium
| |
Collapse
|
44
|
Zhang L, More KR, Ojha A, Jackson CB, Quinlan BD, Li H, He W, Farzan M, Pardi N, Choe H. Effect of mRNA-LNP components of two globally-marketed COVID-19 vaccines on efficacy and stability. NPJ Vaccines 2023; 8:156. [PMID: 37821446 PMCID: PMC10567765 DOI: 10.1038/s41541-023-00751-6] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 09/25/2023] [Indexed: 10/13/2023] Open
Abstract
During the COVID-19 pandemic, Pfizer-BioNTech and Moderna successfully developed nucleoside-modified mRNA lipid nanoparticle (LNP) vaccines. SARS-CoV-2 spike protein expressed by those vaccines are identical in amino acid sequence, but several key components are distinct. Here, we compared the effect of ionizable lipids, untranslated regions (UTRs), and nucleotide composition of the two vaccines, focusing on mRNA delivery, antibody generation, and long-term stability. We found that the ionizable lipid, SM-102, in Moderna's vaccine performs better than ALC-0315 in Pfizer-BioNTech's vaccine for intramuscular delivery of mRNA and antibody production in mice and long-term stability at 4 °C. Moreover, Pfizer-BioNTech's 5' UTR and Moderna's 3' UTR outperform their counterparts in their contribution to transgene expression in mice. We further found that varying N1-methylpseudouridine content at the wobble position of mRNA has little effect on vaccine efficacy. These findings may contribute to the further improvement of nucleoside-modified mRNA-LNP vaccines and therapeutics.
Collapse
Affiliation(s)
- Lizhou Zhang
- Division of Infectious Disease, Boston Children's Hospital, Boston, MA, USA.
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA.
- Department of Immunology and Microbiology, UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, USA.
| | - Kunal R More
- Department of Immunology and Microbiology, UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, USA
| | - Amrita Ojha
- Department of Immunology and Microbiology, UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, USA
| | - Cody B Jackson
- Division of Infectious Disease, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Department of Immunology and Microbiology, UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, USA
| | - Brian D Quinlan
- Department of Immunology and Microbiology, UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, USA
| | - Hao Li
- Division of Infectious Disease, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Department of Immunology and Microbiology, UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, USA
- Skaggs Graduate School, The Scripps Research Institute, La Jolla, CA, USA
| | - Wenhui He
- Division of Infectious Disease, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Department of Immunology and Microbiology, UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, USA
- Center For Integrated Solutions for Infectious Diseases, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Michael Farzan
- Division of Infectious Disease, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Department of Immunology and Microbiology, UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, USA
- Skaggs Graduate School, The Scripps Research Institute, La Jolla, CA, USA
- Center For Integrated Solutions for Infectious Diseases, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Norbert Pardi
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Hyeryun Choe
- Division of Infectious Disease, Boston Children's Hospital, Boston, MA, USA.
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA.
- Department of Immunology and Microbiology, UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, USA.
| |
Collapse
|
45
|
Youssef M, Hitti C, Puppin Chaves Fulber J, Kamen AA. Enabling mRNA Therapeutics: Current Landscape and Challenges in Manufacturing. Biomolecules 2023; 13:1497. [PMID: 37892179 PMCID: PMC10604719 DOI: 10.3390/biom13101497] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 10/05/2023] [Accepted: 10/06/2023] [Indexed: 10/29/2023] Open
Abstract
Recent advances and discoveries in the structure and role of mRNA as well as novel lipid-based delivery modalities have enabled the advancement of mRNA therapeutics into the clinical trial space. The manufacturing of these products is relatively simple and eliminates many of the challenges associated with cell culture production of viral delivery systems for gene and cell therapy applications, allowing rapid production of mRNA for personalized treatments, cancer therapies, protein replacement and gene editing. The success of mRNA vaccines during the COVID-19 pandemic highlighted the immense potential of this technology as a vaccination platform, but there are still particular challenges to establish mRNA as a widespread therapeutic tool. Immunostimulatory byproducts can pose a barrier for chronic treatments and different production scales may need to be considered for these applications. Moreover, long-term storage of mRNA products is notoriously difficult. This review provides a detailed overview of the manufacturing steps for mRNA therapeutics, including sequence design, DNA template preparation, mRNA production and formulation, while identifying the challenges remaining in the dose requirements, long-term storage and immunotolerance of the product.
Collapse
Affiliation(s)
| | | | | | - Amine A. Kamen
- Department of Bioengineering, McGill University, Montreal, QC H3A 0G4, Canada; (M.Y.); (C.H.); (J.P.C.F.)
| |
Collapse
|
46
|
Yang L, Wang T, Zhang D, Huang X, Dong Y, Gao W, Ye Y, Ren K, Zhao W, Qiao H, Jia L. Black Phosphorus Nanosheets Assist Nanoerythrosomes for Efficient mRNA Vaccine Delivery and Immune Activation. Adv Healthc Mater 2023; 12:e2300935. [PMID: 37363954 DOI: 10.1002/adhm.202300935] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 06/06/2023] [Indexed: 06/28/2023]
Abstract
Messenger RNA (mRNA)-based vaccines have enormous potential in infectious disease prevention and tumor neoantigen application. However, developing an advanced delivery system for efficient mRNA delivery and intracellular release for protein translation remains a challenge. Herein, a biocompatible biomimetic system is designed using red blood cell-derived nanoerythrosomes (NER) and black phosphorus nanosheets (BP) for mRNA delivery. BP is covalently modified with polyethyleneimine (PEI), serving as a core to efficiently condense mRNA via electrostatic interactions. To facilitate the spleen targeting of the mRNA-loaded BP (BPmRNA ), NER is co-extruded with BPmRNA to construct a stable "core-shell" nanovaccine (NER@BPmRNA ). The mRNA nanovaccine exhibits efficient protein expression and immune activation via BP-mediated adjuvant effect and enhanced lysosomal escape. In vivo evaluation demonstrates that the system delivery of mRNA encoding coronavirus receptor-binding domain (RBD) significantly increases the antibody titer and pseudovirus neutralization effect compared with that of NER without BP assistance. Furthermore, the mRNA extracted from mouse melanoma tissues is utilized to simulate tumor neoantigen delivered by NER@BPmRNA . In the vaccinated mice, BP-assisted NER for the delivery of melanoma mRNA can induce more antibodies that specifically recognize tumor antigens. Thus, BP-assisted NER can serve as a safe and effective delivery vehicle in mRNA-based therapy.
Collapse
Affiliation(s)
- Lixin Yang
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, 210009, China
| | - Tengqi Wang
- Central Lab, Bayannur Hospital, Bayannur, 015000, China
| | - Dexin Zhang
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, 210009, China
| | - Xin Huang
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, 210009, China
| | - Yuqin Dong
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, 210009, China
| | - Wen Gao
- Central Lab, Bayannur Hospital, Bayannur, 015000, China
| | - Youqing Ye
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, 210009, China
| | - Ke Ren
- School of Laboratory Medicine, Chengdu Medical College, Chengdu, 610500, China
| | - Wei Zhao
- Department of Pathology, China Pharmaceutical University Nanjing First Hospital, Nanjing, 210012, China
| | - Haishi Qiao
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, 210009, China
| | - Lizhou Jia
- Central Lab, Bayannur Hospital, Bayannur, 015000, China
| |
Collapse
|
47
|
Gunter HM, Idrisoglu S, Singh S, Han DJ, Ariens E, Peters JR, Wong T, Cheetham SW, Xu J, Rai SK, Feldman R, Herbert A, Marcellin E, Tropee R, Munro T, Mercer TR. mRNA vaccine quality analysis using RNA sequencing. Nat Commun 2023; 14:5663. [PMID: 37735471 PMCID: PMC10514319 DOI: 10.1038/s41467-023-41354-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Accepted: 08/24/2023] [Indexed: 09/23/2023] Open
Abstract
The success of mRNA vaccines has been realised, in part, by advances in manufacturing that enabled billions of doses to be produced at sufficient quality and safety. However, mRNA vaccines must be rigorously analysed to measure their integrity and detect contaminants that reduce their effectiveness and induce side-effects. Currently, mRNA vaccines and therapies are analysed using a range of time-consuming and costly methods. Here we describe a streamlined method to analyse mRNA vaccines and therapies using long-read nanopore sequencing. Compared to other industry-standard techniques, VAX-seq can comprehensively measure key mRNA vaccine quality attributes, including sequence, length, integrity, and purity. We also show how direct RNA sequencing can analyse mRNA chemistry, including the detection of nucleoside modifications. To support this approach, we provide supporting software to automatically report on mRNA and plasmid template quality and integrity. Given these advantages, we anticipate that RNA sequencing methods, such as VAX-seq, will become central to the development and manufacture of mRNA drugs.
Collapse
Affiliation(s)
- Helen M Gunter
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, QLD, Australia
- BASE facility, University of Queensland, Brisbane, QLD, Australia
| | - Senel Idrisoglu
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, QLD, Australia
- BASE facility, University of Queensland, Brisbane, QLD, Australia
| | - Swati Singh
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, QLD, Australia
- BASE facility, University of Queensland, Brisbane, QLD, Australia
| | - Dae Jong Han
- BASE facility, University of Queensland, Brisbane, QLD, Australia
| | - Emily Ariens
- BASE facility, University of Queensland, Brisbane, QLD, Australia
| | | | - Ted Wong
- Garvan Institute of Medical Research, Sydney, NSW, Australia
| | - Seth W Cheetham
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, QLD, Australia
- BASE facility, University of Queensland, Brisbane, QLD, Australia
| | - Jun Xu
- Genome Innovation Hub, University of Queensland, Brisbane, QLD, Australia
| | - Subash Kumar Rai
- Genome Innovation Hub, University of Queensland, Brisbane, QLD, Australia
| | - Robert Feldman
- COVID19 Vaccine Corporation Limited (CVC), Auckland, New Zealand
| | - Andy Herbert
- COVID19 Vaccine Corporation Limited (CVC), Auckland, New Zealand
| | - Esteban Marcellin
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, QLD, Australia
| | - Romain Tropee
- BASE facility, University of Queensland, Brisbane, QLD, Australia
| | - Trent Munro
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, QLD, Australia
- BASE facility, University of Queensland, Brisbane, QLD, Australia
| | - Tim R Mercer
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, QLD, Australia.
- BASE facility, University of Queensland, Brisbane, QLD, Australia.
| |
Collapse
|
48
|
Wang Y, Cao H, Lin K, Hu J, Luan N, Liu C. Evaluation of the Immunological Efficacy of an LNP-mRNA Vaccine Prepared from Varicella Zoster Virus Glycoprotein gE with a Double-Mutated Carboxyl Terminus in Different Untranslated Regions in Mice. Vaccines (Basel) 2023; 11:1475. [PMID: 37766151 PMCID: PMC10534744 DOI: 10.3390/vaccines11091475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 09/08/2023] [Accepted: 09/09/2023] [Indexed: 09/29/2023] Open
Abstract
Cell-mediated immunity (CMI) plays a key role in the effectiveness of varicella zoster virus (VZV) vaccines, and mRNA vaccines have an innate advantage in inducing CMI. Glycoprotein E (gE) has been used widely as an antigen for VZV vaccines, and carboxyl-terminal mutations of gE are associated with VZV titer and infectivity. In addition, the untranslated regions (UTRs) of mRNA affect the stability and half-life of mRNA in the cell and are crucial for protein expression and antigenic translational efficiency. In this study, three UTRs were designed and connected to the nucleic acid sequence of gE-M, which is double mutated in the extracellular region of gE. Then, mRNA with different nucleic acids was encapsulated in lipid nanoparticles (LNPs), forming three LNP-mRNA VZV vaccines, named gE-M-Z, gE-M-M, and gE-M-P. The immune response elicited by these vaccines in mice was evaluated at intervals of 4 weeks, and the mice were sacrificed 2 weeks after the final immunization. In the results, the gE-M-P group, which retains the nucleic acid sequence of gE-M and is connected to Pfizer/BioNTech's BNT162b2 UTRs, induced the strongest humoral immune response and CMI. Because CMI is crucial for protection against VZV and for the design of VZV vaccines, this study provides a feasible strategy for improving the effectiveness and economy of future VZV vaccines.
Collapse
Affiliation(s)
- Yunfei Wang
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China
| | - Han Cao
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China
| | - Kangyang Lin
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China
| | - Jingping Hu
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China
| | - Ning Luan
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China
| | - Cunbao Liu
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China
| |
Collapse
|
49
|
Pflumm D, Seidel A, Klein F, Groß R, Krutzke L, Kochanek S, Kroschel J, Münch J, Stifter K, Schirmbeck R. Heterologous DNA-prime/protein-boost immunization with a monomeric SARS-CoV-2 spike antigen redundantizes the trimeric receptor-binding domain structure to induce neutralizing antibodies in old mice. Front Immunol 2023; 14:1231274. [PMID: 37753087 PMCID: PMC10518615 DOI: 10.3389/fimmu.2023.1231274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 08/09/2023] [Indexed: 09/28/2023] Open
Abstract
A multitude of alterations in the old immune system impair its functional integrity. Closely related, older individuals show, for example, a reduced responsiveness to severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) vaccines. However, systematic strategies to specifically improve the efficacy of vaccines in the old are missing or limited to simple approaches like increasing the antigen concentration or injection frequencies. We here asked whether the intrinsic, trimeric structure of the SARS-CoV-2 spike (S) antigen and/or a DNA- or protein-based antigen delivery platform affects priming of functional antibody responses particularly in old mice. The used S-antigens were primarily defined by the presence/absence of the membrane-anchoring TM domain and the closely interlinked formation/non-formation of a trimeric structure of the receptor binding domain (S-RBD). Among others, we generated vectors expressing prefusion-stabilized, cell-associated (TM+) trimeric "S2-P" or secreted (TM-) monomeric "S6-PΔTM" antigens. These proteins were produced from vector-transfected HEK-293T cells under mild conditions by Strep-tag purification, revealing that cell-associated but not secreted S proteins tightly bound Hsp73 and Grp78 chaperones. We showed that both, TM-deficient S6-PΔTM and full-length S2-P antigens elicited very similar S-RBD-specific antibody titers and pseudovirus neutralization activities in young (2-3 months) mice through homologous DNA-prime/DNA-boost or protein-prime/protein-boost vaccination. The trimeric S2-P antigen induced high S-RBD-specific antibody responses in old (23-24 months) mice through DNA-prime/DNA-boost vaccination. Unexpectedly, the monomeric S6-PΔTM antigen induced very low S-RBD-specific antibody titers in old mice through homologous DNA-prime/DNA-boost or protein-prime/protein-boost vaccination. However, old mice efficiently elicited an S-RBD-specific antibody response after heterologous DNA-prime/protein-boost immunization with the S6-PΔTM antigen, and antibody titers even reached similar levels and neutralizing activities as in young mice and also cross-reacted with different S-variants of concern. The old immune system thus distinguished between trimeric and monomeric S protein conformations: it remained antigen responsive to the trimeric S2-P antigen, and a simple change in the vaccine delivery regimen was sufficient to unleash its reactivity to the monomeric S6-PΔTM antigen. This clearly shows that both the antigen structure and the delivery platform are crucial to efficiently prime humoral immune responses in old mice and might be relevant for designing "age-adapted" vaccine strategies.
Collapse
Affiliation(s)
- Dominik Pflumm
- Department of Internal Medicine I, University Hospital of Ulm, Ulm, Germany
| | - Alina Seidel
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | - Fabrice Klein
- Department of Gene Therapy, University Hospital of Ulm, Ulm, Germany
| | - Rüdiger Groß
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | - Lea Krutzke
- Department of Gene Therapy, University Hospital of Ulm, Ulm, Germany
| | - Stefan Kochanek
- Department of Gene Therapy, University Hospital of Ulm, Ulm, Germany
| | - Joris Kroschel
- Institute of Clinical Chemistry, Ulm University Medical Center, Ulm, Germany
| | - Jan Münch
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | - Katja Stifter
- Department of Internal Medicine I, University Hospital of Ulm, Ulm, Germany
| | | |
Collapse
|
50
|
Rojas M, Herrán M, Ramírez-Santana C, Leung PSC, Anaya JM, Ridgway WM, Gershwin ME. Molecular mimicry and autoimmunity in the time of COVID-19. J Autoimmun 2023; 139:103070. [PMID: 37390745 PMCID: PMC10258587 DOI: 10.1016/j.jaut.2023.103070] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 05/26/2023] [Accepted: 06/03/2023] [Indexed: 07/02/2023]
Abstract
Infectious diseases are commonly implicated as potential initiators of autoimmune diseases (ADs) and represent the most commonly known factor in the development of autoimmunity in susceptible individuals. Epidemiological data and animal studies on multiple ADs suggest that molecular mimicry is one of the likely mechanisms for the loss of peripheral tolerance and the development of clinical disease. Besides molecular mimicry, other mechanisms such as defects in central tolerance, nonspecific bystander activation, epitope-determinant spreading, and/or constant antigenic stimuli, may also contribute for breach of tolerance and to the development of ADs. Linear peptide homology is not the only mechanism by which molecular mimicry is established. Peptide modeling (i.e., 3D structure), molecular docking analyses, and affinity estimation for HLAs are emerging as critical strategies when studying the links of molecular mimicry in the development of autoimmunity. In the current pandemic, several reports have confirmed an influence of SARS-CoV-2 on subsequent autoimmunity. Bioinformatic and experimental evidence support the potential role of molecular mimicry. Peptide dimensional analysis requires more research and will be increasingly important for designing and distributing vaccines and better understanding the role of environmental factors related to autoimmunity.
Collapse
Affiliation(s)
- Manuel Rojas
- Division of Rheumatology, Allergy and Clinical Immunology, University of California, Davis, CA, 95616, USA; Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogota, Colombia.
| | - María Herrán
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogota, Colombia
| | - Carolina Ramírez-Santana
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogota, Colombia
| | - Patrick S C Leung
- Division of Rheumatology, Allergy and Clinical Immunology, University of California, Davis, CA, 95616, USA
| | - Juan-Manuel Anaya
- Health Research and Innovation Center at Coosalud, Cartagena, 130001, Colombia
| | - William M Ridgway
- Division of Rheumatology, Allergy and Clinical Immunology, University of California, Davis, CA, 95616, USA
| | - M Eric Gershwin
- Division of Rheumatology, Allergy and Clinical Immunology, University of California, Davis, CA, 95616, USA
| |
Collapse
|