1
|
Li J, Liu D, Li X, Wei J, Du W, Zhao A, Xu M. RNA vaccines: The dawn of a new age for tuberculosis? Hum Vaccin Immunother 2025; 21:2469333. [PMID: 40013818 PMCID: PMC11869779 DOI: 10.1080/21645515.2025.2469333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 02/03/2025] [Accepted: 02/14/2025] [Indexed: 02/28/2025] Open
Abstract
Since 2019, there has been a growing focus on mRNA vaccines for infectious disease prevention, particularly following the emergence of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). mRNA vaccines offer advantages such as rapid production and the ability to induce robust cellular and antibody responses, which are essential for combating infections that require cell-mediated immunity, including Tuberculosis (TB). This review explores recent progress in TB mRNA vaccines and addresses several key areas: (1) the urgent need for new TB vaccines; (2) current advancements in TB vaccine development, and the advantages and challenges of mRNA technology; (3) the design and characteristics of TB mRNA vaccines; (4) the immunological mechanisms of TB mRNA vaccines; (5) manufacturing processes for TB mRNA vaccines; and (6) safety and regulatory considerations. This interdisciplinary review aims to provide insights for researchers working to address critical questions in TB mRNA vaccine development.
Collapse
Affiliation(s)
- Junli Li
- Division of Tuberculosis Vaccine and Allergen Products, Institute of Biological Product Control, National Institutes for Food and Drug Control, Beijing, China
- State Key Laboratory of Drug Regulatory Science, National Institutes for Food and Drug Control, Beijing, China
- Key Laboratory for Quality Research and Evaluation of Biological Products, National Medical Products Administration (NMPA), Beijing, China
- Key Laboratory of Research on Quality and Standardization of Biotech Products, National Health Commission (NHC), Beijing, China
| | - Dong Liu
- Graduate School of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
- Guangzhou Laboratory, Guangzhou, China
| | - Xiaochi Li
- Division of Tuberculosis Vaccine and Allergen Products, Institute of Biological Product Control, National Institutes for Food and Drug Control, Beijing, China
- State Key Laboratory of Drug Regulatory Science, National Institutes for Food and Drug Control, Beijing, China
- Key Laboratory for Quality Research and Evaluation of Biological Products, National Medical Products Administration (NMPA), Beijing, China
- Key Laboratory of Research on Quality and Standardization of Biotech Products, National Health Commission (NHC), Beijing, China
| | - Jiazheng Wei
- College of Life Sciences and Biopharmaceuticals, Shenyang Pharmaceutical University, Shenyang, China
| | - Weixin Du
- Division of Tuberculosis Vaccine and Allergen Products, Institute of Biological Product Control, National Institutes for Food and Drug Control, Beijing, China
- State Key Laboratory of Drug Regulatory Science, National Institutes for Food and Drug Control, Beijing, China
- Key Laboratory for Quality Research and Evaluation of Biological Products, National Medical Products Administration (NMPA), Beijing, China
- Key Laboratory of Research on Quality and Standardization of Biotech Products, National Health Commission (NHC), Beijing, China
| | - Aihua Zhao
- Division of Tuberculosis Vaccine and Allergen Products, Institute of Biological Product Control, National Institutes for Food and Drug Control, Beijing, China
- State Key Laboratory of Drug Regulatory Science, National Institutes for Food and Drug Control, Beijing, China
- Key Laboratory for Quality Research and Evaluation of Biological Products, National Medical Products Administration (NMPA), Beijing, China
- Key Laboratory of Research on Quality and Standardization of Biotech Products, National Health Commission (NHC), Beijing, China
| | - Miao Xu
- Division of Tuberculosis Vaccine and Allergen Products, Institute of Biological Product Control, National Institutes for Food and Drug Control, Beijing, China
- State Key Laboratory of Drug Regulatory Science, National Institutes for Food and Drug Control, Beijing, China
- Key Laboratory for Quality Research and Evaluation of Biological Products, National Medical Products Administration (NMPA), Beijing, China
- Key Laboratory of Research on Quality and Standardization of Biotech Products, National Health Commission (NHC), Beijing, China
| |
Collapse
|
2
|
Liu Z, Chen J, Xu M, Ho S, Wei Y, Ho HP, Yong KT. Engineered multi-domain lipid nanoparticles for targeted delivery. Chem Soc Rev 2025. [PMID: 40390667 DOI: 10.1039/d4cs00891j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/21/2025]
Abstract
Engineered lipid nanoparticles (LNPs) represent a breakthrough in targeted drug delivery, enabling precise spatiotemporal control essential to treat complex diseases such as cancer and genetic disorders. However, the complexity of the delivery process-spanning diverse targeting strategies and biological barriers-poses significant challenges to optimizing their design. To address these, this review introduces a multi-domain framework that dissects LNPs into four domains: structure, surface, payload, and environment. Engineering challenges, functional mechanisms, and characterization strategies are analyzed across each domain, along with a discussion of advantages, limitations, and in vivo fate (e.g., biodistribution and clearance). The framework also facilitates comparisons with natural exosomes and exploration of alternative administration routes, such as intranasal and intraocular delivery. We highlight current characterization techniques, such as cryo-TEM and multiscale molecular dynamics simulations, as well as the recently emerging artificial intelligence (AI) applications-ranging from LNP structure screening to the prospective use of generative models for de novo design beyond traditional experimental and simulation paradigms. Finally, we examine how engineered LNPs integrate active, passive, endogenous, and stimuli-responsive targeting mechanisms to achieve programmable delivery, potentially surpassing biological sophistication in therapeutic performance.
Collapse
Affiliation(s)
- Zhaoyu Liu
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, 999077, China.
- Department of Biomedical Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, Maryland, 21218, USA
| | - Jingxun Chen
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, 999077, China.
- Department of Biomedical Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, Maryland, 21218, USA
| | - Mingkun Xu
- Guangdong Institute of Intelligence Science and Technology, Hengqin, Zhuhai, 519031, China
| | - Sherwin Ho
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, California, 90095, USA.
| | - Yuanyuan Wei
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, 999077, China.
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, California, 90095, USA.
| | - Ho-Pui Ho
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, 999077, China.
- School of Electrical and Electronic Engineering, Yonsei University, Seoul, 03722, Korea
| | - Ken-Tye Yong
- School of Biomedical Engineering, The University of Sydney, Sydney, New South Wales 2006, Australia.
- The Biophotonics and Mechano-Bioengineering Lab, The University of Sydney, Sydney, New South Wales 2006, Australia
| |
Collapse
|
3
|
Zhang H, Barz M. Investigating the stability of RNA-lipid nanoparticles in biological fluids: Unveiling its crucial role for understanding LNP performance. J Control Release 2025; 381:113559. [PMID: 40023226 DOI: 10.1016/j.jconrel.2025.02.055] [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: 09/30/2024] [Revised: 01/30/2025] [Accepted: 02/19/2025] [Indexed: 03/04/2025]
Abstract
Lipid nanoparticles (LNPs) are the most established and clinically advanced platform for RNA delivery. While significant efforts have been made to improve RNA delivery efficiency for improved protein production, the interplay between physiological stability, target specificity, and therapeutic efficacy of RNA-LNPs remains largely unexplored. This review highlights the crucial, yet often overlooked, impact of in vivo stability or instability of RNA-LNPs in contact with biological fluids on delivery performance. We discuss the various factors, including lipid composition, particle surface properties and interactions with proteins in physiological conditions, and provide an overview of the current methods for assessing RNA-LNP stability in biological fluids, such as dynamic laser light scattering, liquid chromatography, and fluorescent and radiolabeled techniques. In the final part, we propose strategies for enhancing stability, with a focus on shielding lipids. Therefore, this work highlights the importance of investigating and understanding the balance between stability and instability of LNPs in the biological context to achieve a more meaningful correlation between formulation properties and in vivo performance.
Collapse
Affiliation(s)
- Heyang Zhang
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research (LACDR), Leiden University, 2333CC Leiden, Netherlands.
| | - Matthias Barz
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research (LACDR), Leiden University, 2333CC Leiden, Netherlands; Department of Dermatology, University Medical Center of the Johannes Gutenberg-University, 55128 Mainz, Germany.
| |
Collapse
|
4
|
Wang L, Li Y, Jiang P, Bai H, Wu C, Shuai Q, Yan Y. Enhanced mRNA delivery via incorporating hydrophobic amines into lipid nanoparticles. Colloids Surf B Biointerfaces 2025; 249:114528. [PMID: 39847891 DOI: 10.1016/j.colsurfb.2025.114528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 01/15/2025] [Accepted: 01/17/2025] [Indexed: 01/25/2025]
Abstract
Lipid nanoparticles (LNPs) have shown promising performance in mRNA delivery. Nevertheless, a thorough understanding of the relationship between mRNA delivery efficacy and the structure of LNPs remains imperative. In this study, we systematically investigated the effects of additional hydrophobic amines on the physicochemical properties of mRNA LNPs and their delivery efficacy. The results indicated that this influence depended on the chemical structure of the additional amines and the structure of the lipid carriers. The appropriate addition of the hydrophobic amine 2C8 to lipid carriers with structural 2C8 or 2C6 tails significantly increased their mRNA delivery efficiency. In contrast, the addition of hydrophobic amine C18 to LNPs resulted in a decrease in mRNA delivery efficiency, while the addition of hydrophobic amines 2C6 and C8, as well as alkanes C12' and C16', had relatively little effect on mRNA delivery. Further investigations demonstrated that the appropriate addition of 2C8 could reduce LNP size, moderate internal hydrophobicity and LNP stability, facilitate mRNA release, enhance cellular uptake, and improve intracellular transportation of LNPs, thereby achieving superior mRNA delivery efficiency. These findings highlight the important role of additional hydrophobic amines in mRNA delivery with LNPs and provide valuable insights for the advancement of mRNA delivery carriers.
Collapse
Affiliation(s)
- Longyu Wang
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, China
| | - Yichen Li
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, China
| | - Pingge Jiang
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, China
| | - Hao Bai
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, China
| | - Chengfan Wu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, China
| | - Qi Shuai
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, China
| | - Yunfeng Yan
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, China.
| |
Collapse
|
5
|
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
|
6
|
Madden PJ, Marina-Zárate E, Rodrigues KA, Steichen JM, Shil M, Ni K, Michaels KK, Maiorino L, Upadhyay AA, Saha S, Pradhan A, Kalyuzhiny O, Liguori A, Lopez PG, Phung I, Flynn C, Zhou A, Melo MB, Lemnios A, Phelps N, Georgeson E, Alavi N, Kubitz M, Lu D, Eskandarzadeh S, Metz A, Rodriguez OL, Shields K, Schultze S, Smith ML, Healy BS, Lim D, Lewis VR, Ben-Akiva E, Pinney W, Gregory J, Xiao S, Carnathan DG, Pai Kasturi S, Watson CT, Bosinger SE, Silvestri G, Schief WR, Irvine DJ, Crotty S. Diverse priming outcomes under conditions of very rare precursor B cells. Immunity 2025; 58:997-1014.e11. [PMID: 40168992 PMCID: PMC12060733 DOI: 10.1016/j.immuni.2025.03.003] [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: 10/11/2024] [Revised: 02/18/2025] [Accepted: 03/04/2025] [Indexed: 04/03/2025]
Abstract
Rare naive B cells have special pathogen-recognition features that enable outsized contributions to protective immunity but infrequently participate in immune responses. We investigatee how germline-targeting vaccine delivery and adjuvant selection affect priming of exceptionally rare BG18-like HIV broadly neutralizing antibody-precursor B cells (<1-in-50 million) in non-human primates. Only escalating dose (ED) priming immunization using the saponin adjuvant SMNP elicited detectable BG18-like cells in germinal centers (GCs) compared with other conditions. All groups had strong GC responses, but only ED+SMNP and bolus+SMNP induced BG18-like memory B cells in >50% of animals. One group had vaccine-specific GC responses equivalent to ED+SMNP but scarce BG18-like B cells. Following homologous boosting, BG18-like memory B cells were present in a bolus priming group but with lower somatic hypermutation and affinities than ED+SMNP. This outcome inversely associated with post-prime antibody titers, suggesting antibody feedback significantly influences rare precursor B cell responses. Thus, antigen and inflammatory stimuli extensively impact priming and affinity maturation of rare B cells.
Collapse
Affiliation(s)
- Patrick J Madden
- La Jolla Institute for Immunology, La Jolla, CA, USA; Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA, USA
| | - Ester Marina-Zárate
- La Jolla Institute for Immunology, La Jolla, CA, USA; Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA, USA
| | - Kristen A Rodrigues
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jon M Steichen
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA, USA; Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Monolina Shil
- La Jolla Institute for Immunology, La Jolla, CA, USA; Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA, USA
| | - Kaiyuan Ni
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | - Laura Maiorino
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Amit A Upadhyay
- Emory National Primate Research Center and Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA; Department of Pathology and Laboratory Medicine, Emory School of Medicine, Atlanta, GA, USA
| | - Swati Saha
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY, USA
| | - Arpan Pradhan
- Emory National Primate Research Center and Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Oleksandr Kalyuzhiny
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA, USA; Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA; IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA, USA
| | - Alessia Liguori
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA, USA; Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA; IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA, USA
| | - Paul G Lopez
- La Jolla Institute for Immunology, La Jolla, CA, USA; Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA, USA
| | - Ivy Phung
- La Jolla Institute for Immunology, La Jolla, CA, USA; Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA, USA
| | - Claudia Flynn
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA, USA
| | - Amelia Zhou
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA, USA
| | - Mariane B Melo
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Ashley Lemnios
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Nicole Phelps
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA, USA; Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Erik Georgeson
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA, USA; Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Nushin Alavi
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA, USA; Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Michael Kubitz
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA, USA; Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Danny Lu
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA, USA; Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Saman Eskandarzadeh
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA, USA; Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Amanda Metz
- Emory National Primate Research Center and Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA; Department of Pathology and Laboratory Medicine, Emory School of Medicine, Atlanta, GA, USA
| | - Oscar L Rodriguez
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY, USA
| | - Kaitlyn Shields
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY, USA
| | - Steven Schultze
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY, USA
| | - Melissa L Smith
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY, USA
| | - Brandon S Healy
- Emory National Primate Research Center and Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Deuk Lim
- Emory National Primate Research Center and Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Vanessa R Lewis
- Emory National Primate Research Center and Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Elana Ben-Akiva
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - William Pinney
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Justin Gregory
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Shuhao Xiao
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Diane G Carnathan
- Emory National Primate Research Center and Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Sudhir Pai Kasturi
- Emory National Primate Research Center and Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Corey T Watson
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY, USA
| | - Steven E Bosinger
- Emory National Primate Research Center and Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA; Department of Pathology and Laboratory Medicine, Emory School of Medicine, Atlanta, GA, USA
| | - Guido Silvestri
- Emory National Primate Research Center and Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
| | - William R Schief
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA, USA; Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA; IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA, USA; Moderna, Inc., Cambridge, MA, USA
| | - Darrell J Irvine
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA, USA; Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA; Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA; Howard Hughes Medical Institute, Chevy Chase, MD, USA.
| | - Shane Crotty
- La Jolla Institute for Immunology, La Jolla, CA, USA; Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA, USA; Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
7
|
Li Y, Meagher RB, Lin X. Tailoring mRNA lipid nanoparticles for antifungal vaccines. PLoS Pathog 2025; 21:e1013091. [PMID: 40293964 PMCID: PMC12036839 DOI: 10.1371/journal.ppat.1013091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2025] Open
Abstract
Vaccination is one of the most effective public health measures for preventing and managing infectious diseases. Despite intensive efforts from the relatively small medical mycology community, developing effective vaccines against invasive fungal infections remains a scientific challenge. This is predominantly due to large antigenic repertoires, complicated life cycles, and the capacity of fungal pathogens to evade the host immune system. Additionally, antifungal vaccines often need to work for at-risk individuals who are immunodeficient. We anticipate that the success of mRNA vaccines against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and its exploration for various infectious diseases and cancers will usher a new wave of antifungal vaccine research. Herein, we discuss recent advancements and key scientific areas that need to be explored to actualize the development of effective antifungal mRNA vaccines.
Collapse
Affiliation(s)
- Yeqi Li
- Department of Microbiology, University of Georgia, Athens, GeorgiaUnited States of America
| | - Richard B. Meagher
- Department of Genetics, University of Georgia, Athens, GeorgiaUnited States of America
| | - Xiaorong Lin
- Department of Microbiology, University of Georgia, Athens, GeorgiaUnited States of America
| |
Collapse
|
8
|
Fernandes RS, de Assis Burle-Caldas G, Sergio SAR, Bráz AF, da Silva Leite NP, Pereira M, de Oliveira Silva J, Hojo-Souza NS, de Oliveira B, Fernandes APSM, da Fonseca FG, Gazzinelli RT, Dos Santos Ferreira D, Teixeira SMR. The immunogenic potential of an optimized mRNA lipid nanoparticle formulation carrying sequences from virus and protozoan antigens. J Nanobiotechnology 2025; 23:221. [PMID: 40102899 PMCID: PMC11921523 DOI: 10.1186/s12951-025-03201-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Accepted: 02/04/2025] [Indexed: 03/20/2025] Open
Abstract
BACKGROUND Lipid nanoparticles (LNP) are a safe and effective messenger RNA (mRNA) delivery system for vaccine applications, as shown by the COVID-19 mRNA vaccines. One of the main challenges faced during the development of these vaccines is the production of new and versatile LNP formulations capable of efficient encapsulation and delivery to cells in vivo. This study aimed to develop a new mRNA vaccine formulation that could potentially be used against existing diseases as well as those caused by pathogens that emerge every year. RESULTS Using firefly luciferase (Luc) as a reporter mRNA, we evaluated the physical-chemical properties, stability, and biodistribution of an LNP-mRNA formulation produced using a novel lipid composition and a microfluidic organic-aqueous precipitation method. Using mRNAs encoding a dengue virus or a Leishmania infantum antigen, we evaluated the immunogenicity of LNP-mRNA formulations and compared them with the immunization with the corresponding recombinant protein or plasmid-encoded antigens. For all tested LNP-mRNAs, mRNA encapsulation efficiency was higher than 85%, their diameter was around 100 nm, and their polydispersity index was less than 0.3. Following an intramuscular injection of 10 µg of the LNP-Luc formulation in mice, we detected luciferase activity in the injection site, as well as in the liver and spleen, as early as 6 h post-administration. LNPs containing mRNA encoding virus and parasite antigens were highly immunogenic, as shown by levels of antigen-specific IgG antibody as well as IFN-γ production by splenocytes of immunized animals that were similar to the levels that resulted from immunization with the corresponding recombinant protein or plasmid DNA. CONCLUSIONS Altogether, these results indicate that these novel LNP-mRNA formulations are highly immunogenic and may be used as novel vaccine candidates for different infectious diseases.
Collapse
Affiliation(s)
- Renata S Fernandes
- Centro de Tecnologia de Vacinas da, Universidade Federal de Minas Gerais, Belo Horizonte, Belo Horizonte, MG, 31310-260, Brazil
| | - Gabriela de Assis Burle-Caldas
- Centro de Tecnologia de Vacinas da, Universidade Federal de Minas Gerais, Belo Horizonte, Belo Horizonte, MG, 31310-260, Brazil
| | | | - Ana Flávia Bráz
- Centro de Tecnologia de Vacinas da, Universidade Federal de Minas Gerais, Belo Horizonte, Belo Horizonte, MG, 31310-260, Brazil
| | - Nathália Pereira da Silva Leite
- Centro de Tecnologia de Vacinas da, Universidade Federal de Minas Gerais, Belo Horizonte, Belo Horizonte, MG, 31310-260, Brazil
| | - Milton Pereira
- Centro de Tecnologia de Vacinas da, Universidade Federal de Minas Gerais, Belo Horizonte, Belo Horizonte, MG, 31310-260, Brazil
| | - Juliana de Oliveira Silva
- Centro de Tecnologia de Vacinas da, Universidade Federal de Minas Gerais, Belo Horizonte, Belo Horizonte, MG, 31310-260, Brazil
- Department of Pharmaceuticals, School of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, MG, 31270-901, Brazil
| | - Natália Satchiko Hojo-Souza
- Centro de Tecnologia de Vacinas da, Universidade Federal de Minas Gerais, Belo Horizonte, Belo Horizonte, MG, 31310-260, Brazil
- Instituto René Rachou, Fundação Oswaldo Cruz-Minas, Belo Horizonte, MG, 30190-002, Brazil
| | - Bianca de Oliveira
- Centro de Tecnologia de Vacinas da, Universidade Federal de Minas Gerais, Belo Horizonte, Belo Horizonte, MG, 31310-260, Brazil
| | - Ana Paula S Moura Fernandes
- Centro de Tecnologia de Vacinas da, Universidade Federal de Minas Gerais, Belo Horizonte, Belo Horizonte, MG, 31310-260, Brazil
- Department of Clinical & Toxicological Analysis, School of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, MG, 31270-901, Brazil
| | - Flávio Guimarães da Fonseca
- Centro de Tecnologia de Vacinas da, Universidade Federal de Minas Gerais, Belo Horizonte, Belo Horizonte, MG, 31310-260, Brazil
- Department of Microbiology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, 31270-901, Brazil
| | - Ricardo Tostes Gazzinelli
- Centro de Tecnologia de Vacinas da, Universidade Federal de Minas Gerais, Belo Horizonte, Belo Horizonte, MG, 31310-260, Brazil
- Instituto René Rachou, Fundação Oswaldo Cruz-Minas, Belo Horizonte, MG, 30190-002, Brazil
- Department of Biochemistry & Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, 31270-901, Brazil
| | - Diego Dos Santos Ferreira
- Centro de Tecnologia de Vacinas da, Universidade Federal de Minas Gerais, Belo Horizonte, Belo Horizonte, MG, 31310-260, Brazil
- Department of Pharmaceuticals, School of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, MG, 31270-901, Brazil
| | - Santuza M Ribeiro Teixeira
- Centro de Tecnologia de Vacinas da, Universidade Federal de Minas Gerais, Belo Horizonte, Belo Horizonte, MG, 31310-260, Brazil.
- Department of Biochemistry & Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, 31270-901, Brazil.
| |
Collapse
|
9
|
Hashimoto M, Tsujii K, Nakajima-Yoshida H, Akiyama N, Yoshihara K, Dohi K, Yin Z, Ejima A, Yamamoto-Mizuno S, Nojiri Y, Saiki S, Baba K, Omoto S. A-910823, a squalene-based emulsion adjuvant, enhances robust and broad immune responses of quadrivalent influenza vaccine in ferrets. Vaccine 2025; 49:126780. [PMID: 39889536 DOI: 10.1016/j.vaccine.2025.126780] [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: 11/19/2024] [Revised: 01/10/2025] [Accepted: 01/20/2025] [Indexed: 02/03/2025]
Abstract
Robust and broad protective immunity is typically elicited by co-administration of an adjuvant with vaccine antigens. A-910823, a squalene-based emulsion adjuvant, has been approved in Japan. It effectively enhances the humoral and cellular immunity of S-268019-b, a recombinant COVID-19 vaccine; however, the adjuvant effects of A-910823 on other vaccines, including influenza vaccine, have not been evaluated. Here, a ferret model was employed to investigate the adjuvant effects of A-910823 when combined with split-inactivated quadrivalent influenza vaccine (QIV). We demonstrate that combination of A-910823 with QIV enhances the neutralizing antibody titer and its breadth against non-vaccine strains. Also, the protective efficacy of A-910823-adjuvanted QIV against virus challenge has been confirmed. Of note, QIV combined with A-910823 caused only minor detectable side effects, whereas a significant increase in fever was observed after two doses of mRNA-LNP in ferrets. This study provides information on the effectiveness and safety of A-910823-adjuvanted QIV and suggests the usefulness of the ferret model for evaluating vaccine-induced reactogenicity.
Collapse
Affiliation(s)
- Masayuki Hashimoto
- Shionogi & Co., Ltd., Vaccine Business Division, 1-1, Futaba-cho 3-chome, Toyonaka, Osaka 561-0825, Japan.
| | - Kenichiro Tsujii
- Shionogi & Co., Ltd., Vaccine Business Division, 1-1, Futaba-cho 3-chome, Toyonaka, Osaka 561-0825, Japan.
| | - Hitomi Nakajima-Yoshida
- Shionogi & Co., Ltd., Vaccine Business Division, 1-1, Futaba-cho 3-chome, Toyonaka, Osaka 561-0825, Japan.
| | - Nobuteru Akiyama
- Shionogi & Co., Ltd., Vaccine Business Division, 1-1, Futaba-cho 3-chome, Toyonaka, Osaka 561-0825, Japan.
| | - Ken Yoshihara
- Shionogi & Co., Ltd., Vaccine Business Division, 1-1, Futaba-cho 3-chome, Toyonaka, Osaka 561-0825, Japan.
| | - Keiji Dohi
- Shionogi & Co., Ltd., Vaccine Business Division, 1-1, Futaba-cho 3-chome, Toyonaka, Osaka 561-0825, Japan.
| | - Ziwei Yin
- Shionogi & Co., Ltd., Vaccine Business Division, 1-1, Futaba-cho 3-chome, Toyonaka, Osaka 561-0825, Japan.
| | - Aki Ejima
- Shionogi & Co., Ltd., Vaccine Business Division, 1-1, Futaba-cho 3-chome, Toyonaka, Osaka 561-0825, Japan.
| | - Saki Yamamoto-Mizuno
- Shionogi & Co., Ltd., Vaccine Business Division, 1-1, Futaba-cho 3-chome, Toyonaka, Osaka 561-0825, Japan.
| | - Yasushi Nojiri
- Shionogi TechnoAdvance Research Co., Ltd., 1-1, Futaba-cho 3-chome, Toyonaka, Osaka 561-0825, Japan.
| | - Shou Saiki
- Shionogi TechnoAdvance Research Co., Ltd., 1-1, Futaba-cho 3-chome, Toyonaka, Osaka 561-0825, Japan.
| | - Kaoru Baba
- Shionogi TechnoAdvance Research Co., Ltd., 1-1, Futaba-cho 3-chome, Toyonaka, Osaka 561-0825, Japan.
| | - Shinya Omoto
- Shionogi & Co., Ltd., Vaccine Business Division, 1-1, Futaba-cho 3-chome, Toyonaka, Osaka 561-0825, Japan.
| |
Collapse
|
10
|
Blizard GS, Dwivedi G, Pan YG, Hou C, Etersque JM, Said H, Chevrier A, Lavertu M, Ni H, Davis B, Tam Y, Cao Q, Mach RH, Weissman D, Alameh MG, Sellmyer MA. Monitoring mRNA vaccine antigen expression in vivo using PET/CT. Nat Commun 2025; 16:2234. [PMID: 40044669 PMCID: PMC11882883 DOI: 10.1038/s41467-025-57446-w] [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: 06/11/2024] [Accepted: 02/20/2025] [Indexed: 03/09/2025] Open
Abstract
Noninvasive visualization of the distribution and persistence of mRNA vaccine antigen expression in mammalian systems has implications for the development and evaluation of future mRNA vaccines. Here, we genetically fuse E. coli dihydrofolate reductase (eDHFR) to the delta furin diproline modified SARS-CoV-2 spike glycoprotein (S2P∆f) mRNA vaccine and image its expression in female mice and male non-human primates using [18F]fluoropropyl-trimethoprim ([18F]FP-TMP). Whole body positron emission tomography (PET) imaging revealed transient expression of the vaccine antigen in the injection site and draining lymph nodes (dLNs). Fusion of eDHFR did not impact S2P immunogenicity and no humoral or cellular immune response was detected against eDHFR in either species. In this work, we show that eDHFR can be used as an mRNA-encoded PET reporter gene to monitor the spatiotemporal dynamics of mRNA vaccine antigen expression in vivo. This technique could be applied in clinical translation of future mRNA vaccines or therapeutics.
Collapse
Affiliation(s)
- Gabrielle S Blizard
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Garima Dwivedi
- Division of Infectious Diseases, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn Institute for RNA Innovation, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yi-Gen Pan
- Division of Infectious Diseases, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Catherine Hou
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jean M Etersque
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Hooda Said
- Department of Pathology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Anik Chevrier
- Chemical Engineering Department, Polytechnique Montreal, Montreal, QC, Canada
| | - Marc Lavertu
- Chemical Engineering Department, Polytechnique Montreal, Montreal, QC, Canada
| | - Houping Ni
- Division of Infectious Diseases, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn Institute for RNA Innovation, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Benjamin Davis
- Division of Infectious Diseases, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn Institute for RNA Innovation, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ying Tam
- Acuitas Therapeutics, Vancouver, BC, Canada
| | - Quy Cao
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Robert H Mach
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Drew Weissman
- Division of Infectious Diseases, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn Institute for RNA Innovation, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mohamad-Gabriel Alameh
- Division of Infectious Diseases, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Penn Institute for RNA Innovation, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Pathology, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Mark A Sellmyer
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
11
|
Buckley M, Araínga M, Maiorino L, Pires IS, Kim BJ, Michaels KK, Dye J, Qureshi K, Zhang YJ, Mak H, Steichen JM, Schief WR, Villinger F, Irvine DJ. Visualizing lipid nanoparticle trafficking for mRNA vaccine delivery in non-human primates. Mol Ther 2025; 33:1105-1117. [PMID: 39797396 PMCID: PMC11897755 DOI: 10.1016/j.ymthe.2025.01.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 11/23/2024] [Accepted: 01/07/2025] [Indexed: 01/13/2025] Open
Abstract
mRNA delivered using lipid nanoparticles (LNPs) has become an important subunit vaccine modality, but mechanisms of action for mRNA vaccines remain incompletely understood. Here, we synthesized a metal chelator-lipid conjugate enabling positron emission tomography (PET) tracer labeling of LNP/mRNA vaccines for quantitative visualization of vaccine trafficking in live mice and non-human primates (NHPs). Following intramuscular injection, we observed LNPs distributing through injected muscle tissue, simultaneous with rapid trafficking to draining lymph nodes (dLNs). Deltoid injection of LNPs mimicking human vaccine administration led to stochastic LNP delivery to three different sets of dLNs. LNP uptake in dLNs was confirmed by histology, and cellular analysis of tissues via flow cytometry identified antigen-presenting cells as the primary immune cell type responsible for early LNP uptake and mRNA translation. These results provide insights into the biodistribution of mRNA vaccines administered at clinically relevant doses, injection volumes, and injection sites in an important large animal model for vaccine development.
Collapse
Affiliation(s)
- Maureen Buckley
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Mariluz Araínga
- New Iberia Research Center, University of Louisiana at Lafayette, Lafayette, LA 70560, USA
| | - Laura Maiorino
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| | - Ivan S Pires
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - B J Kim
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | | - Jonathan Dye
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Kashif Qureshi
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Yiming J Zhang
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Howard Mak
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Jon M Steichen
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA 92037, USA; Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA; IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - William R Schief
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA 92037, USA; Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA; IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Francois Villinger
- New Iberia Research Center, University of Louisiana at Lafayette, Lafayette, LA 70560, USA
| | - Darrell J Irvine
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, Harvard University, Cambridge, MA 02139, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA; Department of Materials Science of Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
12
|
Kirtane AR, Traverso G. Improving the Efficacy of Cancer mRNA Vaccines. Cancer J 2025; 31:e0764. [PMID: 40126883 DOI: 10.1097/ppo.0000000000000764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Accepted: 02/05/2025] [Indexed: 03/26/2025]
Abstract
mRNA vaccines consist of antigen-encoding mRNA, which produces the antigenic protein upon translation. Coupling antigen production with innate immune activation can generate a potent, antigen-specific T-cell response. Clinical reports have demonstrated the ability of mRNA vaccines to elicit an anticancer immune response against various tumor types. Here, we discuss strategies to enhance the potency of mRNA vaccines. We provide an overview of existing knowledge regarding the activation and trafficking mechanisms of mRNA vaccines and share optimization strategies to boost mRNA-mediated antigen production. In addition, we address methods to target mRNA vaccines to dendritic cells and lymph nodes, key initiators of the immune response. Finally, we review strategies for enhancing immune activation using adjuvants compatible with mRNA vaccines. mRNA vaccines offer unique advantages that can be utilized for oncology applications. However, significant work is needed to understand their underlying mechanisms and develop technologies to improve their effectiveness.
Collapse
Affiliation(s)
- Ameya R Kirtane
- Department of Pharmaceutics, University of Minnesota, Minneapolis, MN
| | - Giovanni Traverso
- Department of Mechanical Engineering, Massachusetts Institute of Technology
- Broad Institute, Massachusetts Institute of Technology, Cambridge, MA
- Department of Gastroenterology, Hepatology and Endoscopy, Brigham and Women's Hospital, Boston, MA
| |
Collapse
|
13
|
Hou X, Zhai L, Fu L, Lu J, Guo P, Zhang Y, Zheng D, Ma G. Advances in Engineered Phages for Disease Treatment. SMALL METHODS 2025:e2401611. [PMID: 39935185 DOI: 10.1002/smtd.202401611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 01/15/2025] [Indexed: 02/13/2025]
Abstract
Phage therapy presents a promising solution for combating multidrug-resistant (MDR) bacterial infections and other bacteria-related diseases, attributed to their innate ability to target and lyse bacteria. Recent clinical successes, particularly in treating MDR-related respiratory and post-surgical infections, validated the therapeutic potential of phage therapy. However, the complex microenvironment within the human body poses significant challenges to phage activity and efficacy in vivo. To overcome these barriers, recent advances in phage engineering have aimed to enhance targeting accuracy, improve stability and survivability, and explore synergistic combinations with other therapeutic modalities. This review provides a comprehensive overview of phage therapy, emphasizing the application of engineered phages in antibacterial therapy, tumor therapy, and vaccine development. Furthermore, the review highlights the current challenges and future trends for advancing phage therapy toward broader clinical applications.
Collapse
Affiliation(s)
- Xiaolin Hou
- State Key Laboratory of Biopharmaceutical Preparation and Delivery Institute of Process Engineering, Chinese Academy of Sciences, Bejing, 100190, P. R. China
| | - Lin Zhai
- State Key Laboratory of Biopharmaceutical Preparation and Delivery Institute of Process Engineering, Chinese Academy of Sciences, Bejing, 100190, P. R. China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Laiying Fu
- State Key Laboratory of Biopharmaceutical Preparation and Delivery Institute of Process Engineering, Chinese Academy of Sciences, Bejing, 100190, P. R. China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Junna Lu
- State Key Laboratory of Biopharmaceutical Preparation and Delivery Institute of Process Engineering, Chinese Academy of Sciences, Bejing, 100190, P. R. China
| | - Peilin Guo
- State Key Laboratory of Biopharmaceutical Preparation and Delivery Institute of Process Engineering, Chinese Academy of Sciences, Bejing, 100190, P. R. China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Yu Zhang
- State Key Laboratory of Biopharmaceutical Preparation and Delivery Institute of Process Engineering, Chinese Academy of Sciences, Bejing, 100190, P. R. China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Diwei Zheng
- State Key Laboratory of Biopharmaceutical Preparation and Delivery Institute of Process Engineering, Chinese Academy of Sciences, Bejing, 100190, P. R. China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Guanghui Ma
- State Key Laboratory of Biopharmaceutical Preparation and Delivery Institute of Process Engineering, Chinese Academy of Sciences, Bejing, 100190, P. R. China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| |
Collapse
|
14
|
Gnazzo V, Saleh H, Castro ÍA, Boon ACM, Pinto AK, Brien JD, López CB. DDO-adjuvanted influenza A virus nucleoprotein mRNA vaccine induces robust humoral and cellular type 1 immune responses and protects mice from challenge. mBio 2025; 16:e0358924. [PMID: 39692514 PMCID: PMC11796404 DOI: 10.1128/mbio.03589-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Accepted: 11/25/2024] [Indexed: 12/19/2024] Open
Abstract
A challenge in viral vaccine development is to produce vaccines that generate both neutralizing antibodies to prevent infection and cytotoxic CD8+ T-cells that target conserved viral proteins and can eliminate infected cells to control virus spread. mRNA technology offers an opportunity to design vaccines based on conserved CD8-targeting epitopes, but achieving robust antigen-specific CD8+ T-cells remains a challenge. Here, we tested the viral-derived oligonucleotide DDO268 as an adjuvant in the context of a model influenza A virus (IAV) nucleoprotein (NP) mRNA vaccine in C57BL/6 mice. DDO268 when co-packaged with mRNA in lipid nanoparticles is sensed by RIG I-like receptors and safely induces local type I interferon (IFN) production followed by dendritic cells type 1 activation and migration to the draining lymph nodes. This early response triggered by DDO268 improved the generation of IgG2c antibodies and antigen-specific Th1 CD4+ and CD8+ T-cells (IFNγ+TNFα+IL2+) that provided enhanced protection against lethal IAV challenge. In addition, the inclusion of DDO268 reduced the antigen dose required to achieve protection. These results highlight the potential of DDO268 as an effective mRNA vaccine adjuvant and show that an IAV NP mRNA/DDO268 vaccine is a promising approach for generating protective immunity against conserved internal IAV epitopes.IMPORTANCEVaccines that generate neutralizing antibodies and cytotoxic CD8+ T-cells targeting conserved epitopes are ideal for effective protection against viruses. mRNA vaccines combined with the right adjuvant offer a promising solution to this challenge. We show that the virus-derived oligonucleotide DDO268 enhances antibody and T-cell responses to an influenza A virus (IAV) nucleoprotein mRNA vaccine in mice. DDO268 safely induces local type I interferon production and stimulates dendritic cell activation providing enhanced protection against IAV challenge. In addition, the adjuvant activity of DDO268 allows for the use of lower antigen doses during vaccination.
Collapse
MESH Headings
- Animals
- Influenza Vaccines/immunology
- Influenza Vaccines/administration & dosage
- Influenza Vaccines/genetics
- Mice
- Mice, Inbred C57BL
- Orthomyxoviridae Infections/prevention & control
- Orthomyxoviridae Infections/immunology
- Adjuvants, Immunologic/administration & dosage
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- Influenza A virus/immunology
- Influenza A virus/genetics
- CD8-Positive T-Lymphocytes/immunology
- mRNA Vaccines/immunology
- mRNA Vaccines/administration & dosage
- Female
- Immunity, Humoral
- Immunity, Cellular
- Antibodies, Neutralizing/blood
- Nucleoproteins/immunology
- Nucleoproteins/genetics
- Vaccines, Synthetic/immunology
- Vaccines, Synthetic/administration & dosage
- Vaccines, Synthetic/genetics
- Nucleocapsid Proteins
- Adjuvants, Vaccine/administration & dosage
- RNA-Binding Proteins/immunology
- RNA-Binding Proteins/genetics
- Viral Core Proteins/immunology
- Viral Core Proteins/genetics
- RNA, Messenger/immunology
- RNA, Messenger/genetics
- Th1 Cells/immunology
Collapse
Affiliation(s)
- Victoria Gnazzo
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri, USA
- Center for Women’s Infectious Disease Research, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Hanaa Saleh
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri, USA
- Center for Women’s Infectious Disease Research, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Ítalo A. Castro
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri, USA
- Center for Women’s Infectious Disease Research, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Adrianus C. M. Boon
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri, USA
- Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Amelia K. Pinto
- Department of Microbiology, Immunology & Molecular Genetics, University of Kentucky, Lexington, Kentucky, USA
| | - James D. Brien
- Department of Microbiology, Immunology & Molecular Genetics, University of Kentucky, Lexington, Kentucky, USA
| | - Carolina B. López
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri, USA
- Center for Women’s Infectious Disease Research, Washington University School of Medicine, Saint Louis, Missouri, USA
| |
Collapse
|
15
|
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
|
16
|
Yin D, Zhong Y, Ling S, Lu S, Wang X, Jiang Z, Wang J, Dai Y, Tian X, Huang Q, Wang X, Chen J, Li Z, Li Y, Xu Z, Jiang H, Wu Y, Shi Y, Wang Q, Xu J, Hong W, Xue H, Yang H, Zhang Y, Da L, Han ZG, Tao SC, Dong R, Ying T, Hong J, Cai Y. Dendritic-cell-targeting virus-like particles as potent mRNA vaccine carriers. Nat Biomed Eng 2025; 9:185-200. [PMID: 38714892 DOI: 10.1038/s41551-024-01208-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 03/31/2024] [Indexed: 02/21/2025]
Abstract
Messenger RNA vaccines lack specificity for dendritic cells (DCs)-the most effective cells at antigen presentation. Here we report the design and performance of a DC-targeting virus-like particle pseudotyped with an engineered Sindbis-virus glycoprotein that recognizes a surface protein on DCs, and packaging mRNA encoding for the Spike protein of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) or for the glycoproteins B and D of herpes simplex virus 1. Injection of the DC-targeting SARS-CoV-2 mRNA vaccine in the footpad of mice led to substantially higher and durable antigen-specific immunoglobulin-G titres and cellular immune responses than untargeted virus-like particles and lipid-nanoparticle formulations. The vaccines also protected the mice from infection with SARS-CoV-2 or with herpes simplex virus 1. Virus-like particles with preferential uptake by DCs may facilitate the development of potent prophylactic and therapeutic vaccines.
Collapse
Affiliation(s)
- Di Yin
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yiye Zhong
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
| | - Sikai Ling
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
- BDGENE Therapeutics, Shanghai, China
| | - Sicong Lu
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
| | | | - Zhuofan Jiang
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jie Wang
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yao Dai
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaolong Tian
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Qijing Huang
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xingbo Wang
- MOA Key Laboratory of Animal Virology, Zhejiang University Center for Veterinary Sciences, Hangzhou, China
| | - Junsong Chen
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ziying Li
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yang Li
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zhijue Xu
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hewei Jiang
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yuqing Wu
- Department of Ophthalmology and Vision Science, Shanghai Eye, Ear, Nose and Throat Hospital, Fudan University, Shanghai, China
| | - Yi Shi
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Shanghai Jiao Tong University, Shanghai, China
| | - Quanjun Wang
- National Beijing Center for Drug Safety Evaluation and Research, State Key Laboratory of Medical Countermeasures and Toxicology, Institute of Pharmacology and Toxicology, Academy of Military Sciences, Beijing, China
| | - Jianjiang Xu
- Department of Ophthalmology and Vision Science, Shanghai Eye, Ear, Nose and Throat Hospital, Fudan University, Shanghai, China
| | - Wei Hong
- CAS Key Laboratory of Special Pathogens and Biosafety, Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Heng Xue
- CAS Key Laboratory of Special Pathogens and Biosafety, Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Hang Yang
- CAS Key Laboratory of Special Pathogens and Biosafety, Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- University of Chinese Academy of Sciences, Beijing, China
- Hubei Jiangxia Laboratory, Wuhan, China
| | - Yan Zhang
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
| | - Lintai Da
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ze-Guang Han
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
| | - Sheng-Ce Tao
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ruijiao Dong
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
| | - Tianlei Ying
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Jiaxu Hong
- Department of Ophthalmology and Vision Science, Shanghai Eye, Ear, Nose and Throat Hospital, Fudan University, Shanghai, China.
| | - Yujia Cai
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
17
|
Lewin SR, Bansbach C, Kemps D, Mathae L, Das KT, McCune JM, Deeks SG, Ndung'u T. Target product profile for cell-based and gene-based therapies to achieve a cure for HIV. Lancet HIV 2025; 12:e154-e162. [PMID: 39761679 DOI: 10.1016/s2352-3018(24)00277-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 10/04/2024] [Accepted: 10/17/2024] [Indexed: 02/08/2025]
Abstract
This target product profile (TPP) highlights the minimal and optimal characteristics for ex-vivo and in-vivo cell and gene therapy-based products aimed at achieving an HIV cure (ie, durable antiretroviral-free viral control). The need for an effective, safe, scalable, affordable, accessible, and acceptable cure for HIV infection remains a major global priority. The possibilities for cell and gene therapy-based products for an HIV cure are rapidly expanding. In a multi-stakeholder consensus process of clinical experts and civil society, including representatives from low-income and middle-income countries, participants generally agreed on the optimal targets, whereas consensus on the minimal targets was not reached on every parameter. There was less agreement on the minimal targets for ex-vivo than in-vivo therapies given the complexity of ex-vivo interventions. The TPP is planned to be updated at regular intervals. Building a TPP, such as this one, is an important process for stakeholder engagement and aligning ambitions for the development of products that are acceptable to both clinicians and civil society.
Collapse
Affiliation(s)
- Sharon R Lewin
- Department of Infectious Diseases, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia.
| | | | | | - Lauren Mathae
- Center for Global Health Practice and Impact, Georgetown University, Washington, DC, USA
| | - Kumitaa Theva Das
- Advanced Medical and Dental Institute, Universiti Sains Malaysia, Penang, Malaysia
| | - Joseph M McCune
- HIV Frontiers, Global Health Accelerator, Bill & Melinda Gates Foundation, Seattle, WA, USA
| | - Steven G Deeks
- Division of HIV, Infectious Diseases and Global Medicine, University of California, San Francisco, CA, USA
| | - Thumbi Ndung'u
- Africa Health Research Institute, Durban, South Africa; HIV Pathogenesis Programme, Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa; Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA; University College London, London, UK
| |
Collapse
|
18
|
Zan Q, Fan L, Lu W, Zhang Y, Huang Y, Yu X, Han Y, Zhang R, Dong C, Shuang S. Early Diagnosis of Liver Injury and Real-Time Evaluation of Photothermal Therapy Efficacy with a Viscosity-Responsive NIR-II Smart Molecule. Adv Healthc Mater 2025; 14:e2402614. [PMID: 39440592 DOI: 10.1002/adhm.202402614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 10/14/2024] [Indexed: 10/25/2024]
Abstract
The early diagnosis of liver injury and in situ real-time monitoring of tumor therapy efficacy are important for the enhancement of personalized precision therapy but remain challenging due to the lack of reliable in vivo visualization tools with integrated diagnostic, therapeutic, and efficacy monitoring functions. Herein, a smart second near-infrared window (NIR-II) molecule (BITX-OH) is rationally designed for diagnosis and therapy by vinyl-bridging hydroxyl diphenyl xanthine unit and benzo[cd]indolium skeleton. BITX-OH exhibits high selectivity and sensitivity toward viscosity, exhibiting a significant enhancement (1167-fold) in NIR-II fluorescence at 962 nm. With the assistance of BITX-OH and NIR-II fluorescence imaging, early diagnosis and therapeutic evaluation of non-alcoholic fatty liver (NAFL), as well as in-site real-time monitoring of hepatic fibrosis (HF) in live mice have been successfully achieved, which is at least several hours earlier than the typical clinical test. Notably, BITX-OH displays excellent photothermal conversion efficiency when exposed to an 808 nm laser, which can induce tumor ablation and increase viscosity, thereby enhancing NIR-II fluorescence for the real-time evaluation of photothermal therapy (PTT). This viscosity-based "self-monitoring" strategy provides a convenient and reliable platform for timely obtaining therapeutic feedback to avoid over- or under-treatment, thus enabling personalized precision therapy.
Collapse
Affiliation(s)
- Qi Zan
- School of Chemistry and Chemical Engineering, Institute of Environmental Science, Shanxi University, Taiyuan, 030006, China
| | - Li Fan
- School of Chemistry and Chemical Engineering, Institute of Environmental Science, Shanxi University, Taiyuan, 030006, China
| | - Wenjing Lu
- School of Chemistry and Chemical Engineering, Institute of Environmental Science, Shanxi University, Taiyuan, 030006, China
| | - Yuewei Zhang
- School of Chemistry and Pharmaceutical Engineering, Jilin Institute of Chemical Technology, Jilin, 132022, China
| | - Yunong Huang
- School of Chemistry and Pharmaceutical Engineering, Jilin Institute of Chemical Technology, Jilin, 132022, China
| | - Xue Yu
- School of Chemistry and Pharmaceutical Engineering, Jilin Institute of Chemical Technology, Jilin, 132022, China
| | - Yahong Han
- Shanxi Medical University, Taiyuan, 030032, China
| | - Ruiping Zhang
- Shanxi Provincial People's Hospital, Taiyuan, 030001, China
| | - Chuan Dong
- School of Chemistry and Chemical Engineering, Institute of Environmental Science, Shanxi University, Taiyuan, 030006, China
| | - Shaomin Shuang
- School of Chemistry and Chemical Engineering, Institute of Environmental Science, Shanxi University, Taiyuan, 030006, China
| |
Collapse
|
19
|
Cheng S, Long X, Zhang Y, Lan X, Jiang D. Advancing Cancer Vaccines with Radionuclide Imaging. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2406950. [PMID: 39530610 DOI: 10.1002/smll.202406950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 10/27/2024] [Indexed: 11/16/2024]
Abstract
Cancer vaccines are emerged as a beacon of hope in the fight against cancer. However, the lack of effective methods to directly observe their in vivo behavior and monitor therapeutic responses hinders their translation into clinical settings. Radionuclide imaging allows for non-invasive and real-time visualization of vaccine biodistribution and immunological response, offering valuable insights into the effectiveness of cancer vaccines and aiding in patient stratification. In this review, the latest advances and potential applications of radionuclide imaging in cancer vaccines are discussed, with a specific focus on strategies for visualizing the spatiotemporal distribution of vaccines in vivo and monitoring treatment efficacy. The challenges and considerations for implementing these techniques in clinical practice are also highlighted, aiming to inform and guide future research in this field.
Collapse
Affiliation(s)
- Sixuan Cheng
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xingru Long
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yongxue Zhang
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Xiaoli Lan
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, Wuhan, 430022, China
| | - Dawei Jiang
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, Wuhan, 430022, China
| |
Collapse
|
20
|
Zhang C, Wang Y, Peng J, Wen X, Zhang Y, Li K, Du H, Hu X. Decoding trends in mRNA vaccine research: A comprehensive bibliometric study. Hum Vaccin Immunother 2024; 20:2355037. [PMID: 38813652 PMCID: PMC11141478 DOI: 10.1080/21645515.2024.2355037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 05/10/2024] [Indexed: 05/31/2024] Open
Abstract
BACKGROUND In recent years, infectious diseases like COVID-19 have had profound global socio-economic impacts. mRNA vaccines have gained prominence due to their rapid development, industrial adaptability, simplicity, and responsiveness to new variants. Notably, the 2023 Nobel Prize in Physiology or Medicine recognized significant contributions to mRNA vaccine research. METHODS Our study employed a comprehensive bibliometric analysis using the Web of Science Core Collection (WoSCC) database, encompassing 5,512 papers on mRNA vaccines from 2003 to 2023. We generated cooperation maps, co-citation analyses, and keyword clustering to evaluate the field's developmental history and achievements. RESULTS The analysis yielded knowledge maps highlighting countries/institutions, influential authors, frequently published and highly cited journals, and seminal references. Ongoing research hotspots encompass immune responses, stability enhancement, applications in cancer prevention and treatment, and combating infectious diseases using mRNA technology. CONCLUSIONS mRNA vaccines represent a transformative development in infectious disease prevention. This study provides insights into the field's growth and identifies key research priorities, facilitating advancements in vaccine technology and addressing future challenges.
Collapse
Affiliation(s)
- Chaobin Zhang
- Southwest Eye Hospital, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yuhang Wang
- School of Basic Medicine, Capital Medical University, Beijing, China
| | - Jianding Peng
- School of Basic Medicine, Capital Medical University, Beijing, China
| | - Xiaotian Wen
- School of Basic Medicine, Capital Medical University, Beijing, China
| | - Youwen Zhang
- School of Law, City University of Hongkong, Hong Kong, China
| | - Kejun Li
- Department of Library, Chongqing Vocational Institute of Engineering, Chongqing, China
| | - Hanjian Du
- Department of Neurosurgery, Chongqing Research Center for Glioma Precision Medicine, Chongqing General Hospital, Chongqing University, Chongqing, China
| | - Xiaofei Hu
- Department of Nuclear Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| |
Collapse
|
21
|
Huang S, Que H, Wang M, Wei X. mRNA vaccines as cancer therapies. Chin Med J (Engl) 2024; 137:2979-2995. [PMID: 39668413 PMCID: PMC11706586 DOI: 10.1097/cm9.0000000000003455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Indexed: 12/14/2024] Open
Abstract
ABSTRACT Cancer remains a major global health challenge, with conventional treatments like chemotherapy and radiotherapy often hindered by significant side effects, lack of specificity, and limited efficacy in advanced cases. Among emerging therapeutic strategies, mRNA vaccines have shown remarkable potential due to their adaptability, rapid production, and capability for personalized cancer treatment. This review provides an in-depth analysis of messenger RNA (mRNA) vaccines as a therapeutic approach for cancer immunotherapy, focusing on their molecular biology, classification, mechanisms, and clinical studies. Derived from reported literature and data on clinicaltrials.gov, it examines studies on mRNA vaccines encoding tumor-specific antigens (TSAs), tumor-associated antigens (TAAs), immunomodulators, and chimeric antigen receptors (CARs) across various cancer types. The review highlights the ability of mRNA vaccines to encode TSAs and TAAs, enabling personalized cancer treatments, and classifies these vaccines into non-replicating and self-amplifying types. It further explores their mechanisms of action, including antigen presentation and immune activation, while emphasizing findings from clinical studies that demonstrate the potential of mRNA vaccines in cancer therapy. Despite their promise, challenges remain in enhancing delivery systems, improving immunogenicity, and addressing tumor heterogeneity. Overcoming these obstacles will require further investigation to fully harness the potential of mRNA vaccines in personalized cancer treatment.
Collapse
Affiliation(s)
- Shaoxiong Huang
- Laboratory of Aging Research and Cancer Drug Target, National/State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan 610041, China
| | - Haiying Que
- Laboratory of Aging Research and Cancer Drug Target, National/State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan 610041, China
| | - Manni Wang
- Laboratory of Aging Research and Cancer Drug Target, National/State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan 610041, China
- Department of Biotherapy, West China Hospital and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, National/State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
22
|
Madden PJ, Marina-Zárate E, Rodrigues KA, Steichen JM, Shil M, Ni K, Michaels KK, Maiorino L, Upadhyay AA, Saha S, Pradhan A, Kalyuzhiny O, Liguori A, Lopez PG, Phung I, Phelps N, Georgeson E, Alavi N, Kubitz M, Lu D, Eskandarzadeh S, Metz A, Rodriguez OL, Shields K, Schultze S, Smith ML, Healy BS, Lim D, Lewis VR, Ben-Akiva E, Pinney W, Gregory J, Xiao S, Carnathan DG, Kasturi SP, Watson CT, Bosinger SE, Silvestri G, Schief WR, Irvine DJ, Crotty S. Diverse priming outcomes under conditions of very rare precursor B cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.21.624746. [PMID: 39651117 PMCID: PMC11623517 DOI: 10.1101/2024.11.21.624746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
Rare B cells can have special pathogen-recognition features giving them the potential to make outsized contributions to protective immunity. However, rare naive B cells infrequently participate in immune responses. We investigated how germline-targeting vaccine antigen delivery and adjuvant selection affect priming of exceptionally rare BG18-like HIV broadly neutralizing antibody-precursor B cells (~1 in 50 million) in non-human primates. Only escalating dose (ED) priming immunization using the saponin adjuvant SMNP elicited detectable BG18-like cells in germinal centers (GCs). All groups had strong GC responses, but only ED+SMNP and bolus+SMNP induced BG18-like memory B cells in >50% of animals. One group had vaccine-specific GC responses equivalent to ED+SMNP, but BG18-like memory B cells were rarely detected. Following homologous boosting, BG18-like memory B cells were more frequent in a bolus priming group, but had lower somatic hypermutation and affinities. This outcome was inversely associated with post-prime antibody titers, suggesting antibody feedback can significantly influence rare precursor B cell responses.
Collapse
|
23
|
Federico M. The Immunologic Downsides Associated with the Powerful Translation of Current COVID-19 Vaccine mRNA Can Be Overcome by Mucosal Vaccines. Vaccines (Basel) 2024; 12:1281. [PMID: 39591184 PMCID: PMC11599006 DOI: 10.3390/vaccines12111281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 11/11/2024] [Accepted: 11/13/2024] [Indexed: 11/28/2024] Open
Abstract
The action of mRNA-based vaccines requires the expression of the antigen in cells targeted by lipid nanoparticle-mRNA complexes. When the vaccine antigen is not fully retained by the producer cells, its local and systemic diffusion can have consequences depending on both the levels of antigen expression and its biological activity. A peculiarity of mRNA-based COVID-19 vaccines is the extraordinarily high amounts of the Spike antigen expressed by the target cells. In addition, vaccine Spike can be shed and bind to ACE-2 cell receptors, thereby inducing responses of pathogenetic significance including the release of soluble factors which, in turn, can dysregulate key immunologic processes. Moreover, the circulatory immune responses triggered by the vaccine Spike is quite powerful, and can lead to effective anti-Spike antibody cross-binding, as well as to the emergence of both auto- and anti-idiotype antibodies. In this paper, the immunologic downsides of the strong efficiency of the translation of the mRNA associated with COVID-19 vaccines are discussed together with the arguments supporting the idea that most of them can be avoided with the advent of next-generation, mucosal COVID-19 vaccines.
Collapse
Affiliation(s)
- Maurizio Federico
- National Center for Global Health, Istituto Superiore di Sanità, 00161 Rome, Italy
| |
Collapse
|
24
|
Omata D, Kawahara E, Munakata L, Tanaka H, Akita H, Yoshioka Y, Suzuki R. Effect of Anti-PEG Antibody on Immune Response of mRNA-Loaded Lipid Nanoparticles. Mol Pharm 2024; 21:5672-5680. [PMID: 39324825 PMCID: PMC11539066 DOI: 10.1021/acs.molpharmaceut.4c00628] [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: 06/07/2024] [Revised: 09/10/2024] [Accepted: 09/10/2024] [Indexed: 09/27/2024]
Abstract
Lipid nanoparticle-encapsulated mRNA (mRNA-LNP) vaccines have been approved for use to combat coronavirus disease 2019 (COVID-19). The mRNA-LNPs contain PEG-conjugated lipids. Clinical studies have reported that mRNA-LNPs induce the production of anti-PEG antibodies, but the anti-PEG antibodies do not affect the production of neutralizing antibodies. However, the detailed influence of anti-PEG antibodies on mRNA-LNP vaccines remains unclear. Therefore, in this study, we prepared ovalbumin (OVA) as a model antigen-encoding mRNA-loaded LNP (mRNA-OVA-LNP), and we determined whether anti-PEG antibodies could affect the antigen-specific immune response of mRNA-OVA-LNP vaccination in mice pretreated with PEG-modified liposomes to induce the production of anti-PEG antibodies. After intramuscular (i.m.) injection of the mRNA-LNP, the anti-PEG antibodies did not change the expression of protein or induction of cytokine and cellular immune response but did slightly increase the induction of antigen-specific antibodies. Furthermore, repeated mRNA-LNP i.m. injection induced the production of anti-PEG IgM and anti-PEG IgG. Our results suggest that mRNA-LNP induces the production of anti-PEG antibodies, but the priming of the antigen-specific immune response of mRNA-LNP vaccination is not notably affected by anti-PEG antibodies.
Collapse
Affiliation(s)
- Daiki Omata
- Laboratory
of Drug and Gene Delivery Research, Faculty of Pharmaceutical Sciences, Teikyo University, Tokyo 173-8605, Japan
| | - Eigo Kawahara
- Laboratory
of Nano-design for Innovative Drug Development, Graduate School of
Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
- Vaccine
Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories,
Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Lisa Munakata
- Laboratory
of Drug and Gene Delivery Research, Faculty of Pharmaceutical Sciences, Teikyo University, Tokyo 173-8605, Japan
| | - Hiroki Tanaka
- Laboratory
of DDS Design and Drug Disposition, Graduate School of Pharmaceutical
Sciences, Tohoku University, 6-3 Aoba, Aramaki, Aoba-ku, Sendai City, Miyagi 980-8578, Japan
| | - Hidetaka Akita
- Laboratory
of DDS Design and Drug Disposition, Graduate School of Pharmaceutical
Sciences, Tohoku University, 6-3 Aoba, Aramaki, Aoba-ku, Sendai City, Miyagi 980-8578, Japan
- Center
for Advanced Modalities and DDS, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Yasuo Yoshioka
- Laboratory
of Nano-design for Innovative Drug Development, Graduate School of
Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
- Vaccine
Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories,
Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
- Vaccine Creation
Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Institute
for Open and Transdisciplinary Research Initiatives, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
- Vaccine
Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, The Research Foundation for Microbial Diseases of
Osaka University, 3-1
Yamadaoka, Suita, Osaka 565-0871, Japan
- Center
for Advanced Modalities and DDS, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Ryo Suzuki
- Laboratory
of Drug and Gene Delivery Research, Faculty of Pharmaceutical Sciences, Teikyo University, Tokyo 173-8605, Japan
| |
Collapse
|
25
|
Pardi N, Krammer F. mRNA vaccines for infectious diseases - advances, challenges and opportunities. Nat Rev Drug Discov 2024; 23:838-861. [PMID: 39367276 DOI: 10.1038/s41573-024-01042-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/27/2024] [Indexed: 10/06/2024]
Abstract
The concept of mRNA-based vaccines emerged more than three decades ago. Groundbreaking discoveries and technological advancements over the past 20 years have resolved the major roadblocks that initially delayed application of this new vaccine modality. The rapid development of nucleoside-modified COVID-19 mRNA vaccines demonstrated that this immunization platform is easy to develop, has an acceptable safety profile and can be produced at a large scale. The flexibility and ease of antigen design have enabled mRNA vaccines to enter development for a wide range of viruses as well as for various bacteria and parasites. However, gaps in our knowledge limit the development of next-generation mRNA vaccines with increased potency and safety. A deeper understanding of the mechanisms of action of mRNA vaccines, application of novel technologies enabling rational antigen design, and innovative vaccine delivery strategies and vaccination regimens will likely yield potent novel vaccines against a wide range of pathogens.
Collapse
Affiliation(s)
- Norbert Pardi
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Ignaz Semmelweis Institute, Interuniversity Institute for Infection Research, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
26
|
Gnazzo V, Saleh H, Castro Í, Boon AC, Pinto AK, Brien JD, López CB. DDO-adjuvanted influenza A virus nucleoprotein mRNA vaccine induces robust humoral and cellular type 1 immune responses and protects mice from challenge. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.27.620508. [PMID: 39553933 PMCID: PMC11565765 DOI: 10.1101/2024.10.27.620508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
A challenge in viral vaccine development is to produce vaccines that generate both neutralizing antibodies to prevent infection and cytotoxic CD8+ T-cells that target conserved viral proteins and can eliminate infected cells to control virus spread. mRNA vaccines offer an opportunity to design vaccines based on conserved CD8-targeting epitopes, but achieving robust antigen-specific CD8+ T-cells remains a challenge. Here we tested the viral-derived oligonucleotide DDO268 as an adjuvant in the context of a model influenza A virus (IAV) nucleoprotein (NP) mRNA vaccine in C57BL/6 mice. DDO268 safely induced local type I interferon (IFN) production, stimulated dendritic cells type 1 (DC1) activation and migration to the draining lymph nodes, and improved the generation of IgG2c antibodies and antigen-specific effector Th1 CD4+ and CD8+ T-cells (IFNγ+TNFα+IL2+) when co-packaged with NP mRNA. The DDO268 adjuvanted vaccine provided enhanced protection against lethal IAV challenge and reduced the antigen dose required to achieve this protection. These results highlight the potential of DDO268 as an effective mRNA vaccine adjuvant and show that an IAV NP mRNA/DDO268 vaccine is a promising approach for generating protective immunity against conserved IAV epitopes.
Collapse
Affiliation(s)
- Victoria Gnazzo
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, MO
- Center for Women’s Infectious Disease Research, Washington University School of Medicine, Saint Louis, MO
| | - Hanaa Saleh
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, MO
- Center for Women’s Infectious Disease Research, Washington University School of Medicine, Saint Louis, MO
| | - Ítalo Castro
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, MO
- Center for Women’s Infectious Disease Research, Washington University School of Medicine, Saint Louis, MO
| | - Adrianus C.M. Boon
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, MO
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO
| | - Amelia K Pinto
- Department of Microbiology, Immunology & Molecular Genetics, University of Kentucky, Lexington KY
| | - James D. Brien
- Department of Microbiology, Immunology & Molecular Genetics, University of Kentucky, Lexington KY
| | - Carolina B. López
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, MO
- Center for Women’s Infectious Disease Research, Washington University School of Medicine, Saint Louis, MO
| |
Collapse
|
27
|
McIntire KM, Meng H, Lin TH, Kim W, Moore NE, Han J, McMahon M, Wang M, Malladi SK, Mohammed BM, Zhou JQ, Schmitz AJ, Hoehn KB, Carreño JM, Yellin T, Suessen T, Middleton WD, Teefey SA, Presti RM, Krammer F, Turner JS, Ward AB, Wilson IA, Kleinstein SH, Ellebedy AH. Maturation of germinal center B cells after influenza virus vaccination in humans. J Exp Med 2024; 221:e20240668. [PMID: 38935072 PMCID: PMC11211068 DOI: 10.1084/jem.20240668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 05/20/2024] [Accepted: 05/22/2024] [Indexed: 06/28/2024] Open
Abstract
Germinal centers (GC) are microanatomical lymphoid structures where affinity-matured memory B cells and long-lived bone marrow plasma cells are primarily generated. It is unclear how the maturation of B cells within the GC impacts the breadth and durability of B cell responses to influenza vaccination in humans. We used fine needle aspiration of draining lymph nodes to longitudinally track antigen-specific GC B cell responses to seasonal influenza vaccination. Antigen-specific GC B cells persisted for at least 13 wk after vaccination in two out of seven individuals. Monoclonal antibodies (mAbs) derived from persisting GC B cell clones exhibit enhanced binding affinity and breadth to influenza hemagglutinin (HA) antigens compared with related GC clonotypes isolated earlier in the response. Structural studies of early and late GC-derived mAbs from one clonal lineage in complex with H1 and H5 HAs revealed an altered binding footprint. Our study shows that inducing sustained GC reactions after influenza vaccination in humans supports the maturation of responding B cells.
Collapse
Affiliation(s)
- Katherine M. McIntire
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - Hailong Meng
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
| | - Ting-Hui Lin
- Department of Integrative Structural and Computational Biology, Scripps Research, La Jolla, CA, USA
| | - Wooseob Kim
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
- Department of Microbiology, Korea University College of Medicine, Seoul, Korea
| | - Nina E. Moore
- Department of Integrative Structural and Computational Biology, Scripps Research, La Jolla, CA, USA
| | - Julianna Han
- Department of Integrative Structural and Computational Biology, Scripps Research, La Jolla, CA, USA
| | - Meagan McMahon
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Meng Wang
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
- Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA
| | - Sameer Kumar Malladi
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - Bassem M. Mohammed
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - Julian Q. Zhou
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - Aaron J. Schmitz
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - Kenneth B. Hoehn
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
| | - Juan Manuel Carreño
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Temima Yellin
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Teresa Suessen
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, MO, USA
| | - William D. Middleton
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, MO, USA
| | - Sharlene A. Teefey
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, MO, USA
| | - Rachel M. Presti
- Department of Internal Medicine-Infectious Diseases, Washington University School of Medicine, St Louis, MO, USA
- Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine, St. Louis, MO, USA
- The Andrew M. and Jane M. Bursky Center for Human Immunology & Immunotherapy Programs, Washington University School of Medicine, St Louis, MO, USA
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jackson S. Turner
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - Andrew B. Ward
- Department of Integrative Structural and Computational Biology, Scripps Research, La Jolla, CA, USA
| | - Ian A. Wilson
- Department of Integrative Structural and Computational Biology, Scripps Research, La Jolla, CA, USA
| | - Steven H. Kleinstein
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
- Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Ali H. Ellebedy
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
- Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine, St. Louis, MO, USA
- The Andrew M. and Jane M. Bursky Center for Human Immunology & Immunotherapy Programs, Washington University School of Medicine, St Louis, MO, USA
| |
Collapse
|
28
|
Li J, Zhang Y, Yang YG, Sun T. Advancing mRNA Therapeutics: The Role and Future of Nanoparticle Delivery Systems. Mol Pharm 2024; 21:3743-3763. [PMID: 38953708 DOI: 10.1021/acs.molpharmaceut.4c00276] [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: 07/04/2024]
Abstract
The coronavirus (COVID-19) pandemic has underscored the critical role of mRNA-based vaccines as powerful, adaptable, readily manufacturable, and safe methodologies for prophylaxis. mRNA-based treatments are emerging as a hopeful avenue for a plethora of conditions, encompassing infectious diseases, cancer, autoimmune diseases, genetic diseases, and rare disorders. Nonetheless, the in vivo delivery of mRNA faces challenges due to its instability, suboptimal delivery, and potential for triggering undesired immune reactions. In this context, the development of effective drug delivery systems, particularly nanoparticles (NPs), is paramount. Tailored with biophysical and chemical properties and susceptible to surface customization, these NPs have demonstrated enhanced mRNA delivery in vivo and led to the approval of several NPs-based formulations for clinical use. Despite these advancements, the necessity for developing a refined, targeted NP delivery system remains imperative. This review comprehensively surveys the biological, translational, and clinical progress in NPs-mediated mRNA therapeutics for both the prevention and treatment of diverse diseases. By addressing critical factors for enhancing existing methodologies, it aims to inform the future development of precise and efficacious mRNA-based therapeutic interventions.
Collapse
Affiliation(s)
- Jiaxuan Li
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital of Jilin University, Changchun, Jilin 130021, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin 130021, China
| | - Yuning Zhang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital of Jilin University, Changchun, Jilin 130021, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin 130021, China
| | - Yong-Guang Yang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital of Jilin University, Changchun, Jilin 130021, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin 130021, China
- International Center of Future Science, Jilin University, Changchun, Jilin 130021, China
| | - Tianmeng Sun
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital of Jilin University, Changchun, Jilin 130021, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin 130021, China
- International Center of Future Science, Jilin University, Changchun, Jilin 130021, China
- State Key Laboratory of Supramolecular Structure and Materials, Jilin University, Changchun, Jilin 130021, China
| |
Collapse
|
29
|
Song Z, Jiao L, Wang D, Qiu Y, Miao J, Zhu T, Yu R, Wang Z, Zhou Y, Cai T, Zhang S, Liu H, Sun H, Sun Y, Liu Z. Controlling the speed of antigens transport in dendritic cells improves humoral and cellular immunity for vaccine. Biomed Pharmacother 2024; 177:117036. [PMID: 38941888 DOI: 10.1016/j.biopha.2024.117036] [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: 04/16/2024] [Revised: 06/20/2024] [Accepted: 06/24/2024] [Indexed: 06/30/2024] Open
Abstract
Vaccines are an effective intervention for preventing infectious diseases. Currently many vaccine strategies are designed to improve vaccine efficacy by controlling antigen release, typically involving various approaches at the injection site. Yet, strategies for intracellular slow-release of antigens in vaccines are still unexplored. Our study showed that controlling the degradation of antigens in dendritic cells and slowing their transport from early endosomes to lysosomes markedly enhances both antigen-specific T-cell immune responses and germinal center B cell responses. This leads to the establishment of sustained humoral and cellular immunity in vivo imaging and flow cytometry indicated this method not only prolongs antigen retention at the injection site but also enhances antigen concentration in lymph nodes, surpassing traditional Aluminium (Alum) adjuvants. Additionally, we demonstrated that the slow antigen degradation induces stronger follicular helper T cell responses and increases proportions of long-lived plasma cells and memory B cells. Overall, these findings propose that controlling the speed of antigens transport in dendritic cells can significantly boost vaccine efficacy, offering an innovative avenue for developing highly immunogenic next-generation vaccines.
Collapse
Affiliation(s)
- Zuchen Song
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, PR China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, PR China
| | - Lina Jiao
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, PR China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, PR China
| | - Deyun Wang
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, PR China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, PR China
| | - Yawei Qiu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, PR China
| | - Jinfeng Miao
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, PR China
| | - Tianyu Zhu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, PR China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, PR China
| | - Ruihong Yu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, PR China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, PR China
| | - Zheng Wang
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, PR China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, PR China
| | - Yantong Zhou
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, PR China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, PR China
| | - Ting Cai
- Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, Zhejiang, PR China
| | - Shun Zhang
- Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, Zhejiang, PR China
| | - Huina Liu
- Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, Zhejiang, PR China
| | - Haifeng Sun
- Key Laboratory of Bacteriology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, PR China
| | - Yuechao Sun
- Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, Zhejiang, PR China
| | - Zhenguang Liu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, PR China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, PR China.
| |
Collapse
|
30
|
Li B, Liu H, Zhao M, Zhang X, Huang P, Chen X, Lin J. Carboxylesterase Activatable Molecular Probe for Personalized Treatment Guidance by Analyte-Induced Molecular Transformation. Angew Chem Int Ed Engl 2024; 63:e202404093. [PMID: 38727540 DOI: 10.1002/anie.202404093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Indexed: 06/28/2024]
Abstract
Accurate visualization of tumor microenvironment is of great significance for personalized medicine. Here, we develop a near-infrared (NIR) fluorescence/photoacoustic (FL/PA) dual-mode molecular probe (denoted as NIR-CE) for distinguishing tumors based on carboxylesterase (CE) level by an analyte-induced molecular transformation (AIMT) strategy. The recognition moiety for CE activity is the acetyl unit of NIR-CE, generating the pre-product, NIR-CE-OH, which undergoes spontaneous hydrogen atom exchange between the nitrogen atoms in the indole group and the phenol hydroxyl group, eventually transforming into NIR-CE-H. In cellular experiments and in vivo blind studies, the human hepatoma cells and tumors with high level of CE were successfully distinguished by both NIR FL and PA imaging. Our findings provide a new molecular imaging strategy for personalized treatment guidance.
Collapse
Affiliation(s)
- Benhao Li
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518055, China
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore, 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
- Nanomedicine Translational Research Program, NUS Centre for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Hengke Liu
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518055, China
| | - Mengyao Zhao
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore, 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
- Nanomedicine Translational Research Program, NUS Centre for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Xinming Zhang
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518055, China
| | - Peng Huang
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518055, China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore, 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
- Nanomedicine Translational Research Program, NUS Centre for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Theranostics Center of Excellence (TCE), Yong Loo Lin School of Medicine, National University of Singapore, 11 Biopolis Way, Helios, Singapore, 138667
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), Singapore, 138673, Singapore
| | - Jing Lin
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518055, China
| |
Collapse
|
31
|
Mandolesi M, Das H, de Vries L, Yang Y, Kim C, Dhinakaran M, Castro Dopico X, Fischbach J, Kim S, Guryleva MV, Àdori M, Chernyshev M, Stålmarck A, Hanke L, McInerney GM, Sheward DJ, Corcoran M, Hällberg BM, Murrell B, Karlsson Hedestam GB. Multi-compartmental diversification of neutralizing antibody lineages dissected in SARS-CoV-2 spike-immunized macaques. Nat Commun 2024; 15:6338. [PMID: 39068149 PMCID: PMC11283548 DOI: 10.1038/s41467-024-50286-0] [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: 02/15/2024] [Accepted: 07/03/2024] [Indexed: 07/30/2024] Open
Abstract
The continued evolution of SARS-CoV-2 underscores the need to understand qualitative aspects of the humoral immune response elicited by spike immunization. Here, we combine monoclonal antibody (mAb) isolation with deep B cell receptor (BCR) repertoire sequencing of rhesus macaques immunized with prefusion-stabilized spike glycoprotein. Longitudinal tracing of spike-sorted B cell lineages in multiple immune compartments demonstrates increasing somatic hypermutation and broad dissemination of vaccine-elicited B cells in draining and non-draining lymphoid compartments, including the bone marrow, spleen and, most notably, periaortic lymph nodes. Phylogenetic analysis of spike-specific monoclonal antibody lineages identified through deep repertoire sequencing delineates extensive intra-clonal diversification that shaped neutralizing activity. Structural analysis of the spike in complex with a broadly neutralizing mAb provides a molecular basis for the observed differences in neutralization breadth between clonally related antibodies. Our findings highlight that immunization leads to extensive intra-clonal B cell evolution where members of the same lineage can both retain the original epitope specificity and evolve to recognize additional spike variants not previously encountered.
Collapse
MESH Headings
- Animals
- Spike Glycoprotein, Coronavirus/immunology
- Spike Glycoprotein, Coronavirus/genetics
- Macaca mulatta
- Antibodies, Neutralizing/immunology
- SARS-CoV-2/immunology
- SARS-CoV-2/genetics
- B-Lymphocytes/immunology
- Antibodies, Viral/immunology
- Phylogeny
- Antibodies, Monoclonal/immunology
- Epitopes/immunology
- COVID-19/immunology
- COVID-19/virology
- Humans
- COVID-19 Vaccines/immunology
- Receptors, Antigen, B-Cell/immunology
- Receptors, Antigen, B-Cell/genetics
- Somatic Hypermutation, Immunoglobulin
- Immunization
Collapse
Affiliation(s)
- Marco Mandolesi
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden.
| | - Hrishikesh Das
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Liset de Vries
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Yiqiu Yang
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Changil Kim
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Manojj Dhinakaran
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Xaquin Castro Dopico
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Julian Fischbach
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Sungyong Kim
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Mariia V Guryleva
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Monika Àdori
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Mark Chernyshev
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Aron Stålmarck
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Leo Hanke
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Gerald M McInerney
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Daniel J Sheward
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Martin Corcoran
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - B Martin Hällberg
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Ben Murrell
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | | |
Collapse
|
32
|
Mohanty B, Ahmad Mir R, Priyadarshini A, Ahmad Bhat K, Barati S, Roshani Asl E, Choi JR, Rasmi Y. Potential use of
CRISPR/Cas13
system for vaccine development against various RNA-viral infections. Future Virol 2024; 19:401-418. [DOI: 10.1080/17460794.2024.2403253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 09/09/2024] [Indexed: 03/07/2025]
Affiliation(s)
- Barsha Mohanty
- Centre for Biotechnology, Siksha‘O’Anusandhan (Deemed to be University), Bhubaneswar, India
| | - Rakeeb Ahmad Mir
- Department of Biotechnology, School of Life Sciences, Central University of Kashmir, Ganderbal, J&K, India
| | - Ankita Priyadarshini
- Centre for Biotechnology, Siksha‘O’Anusandhan (Deemed to be University), Bhubaneswar, India
| | - Kaisar Ahmad Bhat
- Department of Biotechnology, BGSB University, Rajouri, J&K, 185234, India
| | - Shirin Barati
- Department of Anatomy, Saveh University of Medical Sciences, Saveh, Iran
| | - Elmira Roshani Asl
- Department of Biochemistry, Saveh University of Medical Sciences, Saveh, Iran
| | - Jane Ru Choi
- Life Science Centre, University of British Columbia, Vancouver, Canada
| | - Yousef Rasmi
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran
- Department of Biochemistry, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
33
|
Rousculp MD, Hollis K, Ziemiecki R, Odom D, Marchese AM, Montazeri M, Odak S, Jackson L, Beyhaghi H, Toback S. Reactogenicity Differences between Adjuvanted, Protein-Based and Messenger Ribonucleic Acid (mRNA)-Based COVID-19 Vaccines. Vaccines (Basel) 2024; 12:802. [PMID: 39066440 PMCID: PMC11281689 DOI: 10.3390/vaccines12070802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 07/16/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
Participants in studies investigating COVID-19 vaccines commonly report reactogenicity events, and concerns about side effects may lead to a reluctance to receive updated COVID-19 vaccinations. A real-world, post hoc analysis, observational 2019nCoV-406 study was conducted to examine reactogenicity within the first 2 days after vaccination with either a protein-based vaccine (NVX-CoV2373) or an mRNA vaccine (BNT162b2 or mRNA-1273) in individuals who previously completed a primary series. Propensity score adjustments were conducted to address potential confounding. The analysis included 1130 participants who received a booster dose of NVX-CoV2373 (n = 303) or an mRNA vaccine (n = 827) during the study period. Within the first 2 days after vaccination, solicited systemic reactogenicity events (adjusted) were reported in 60.5% of participants who received NVX-CoV2373 compared with 84.3% of participants who received an mRNA vaccine; moreover, 33.9% and 61.4%, respectively, reported ≥3 systemic reactogenicity symptoms. The adjusted mean (95% CI) number of systemic symptoms was 1.8 (1.6-2.0) and 3.2 (3.0-3.4), respectively. Local reactogenicity events (adjusted) were reported in 73.4% and 91.7% of participants who received NVX-CoV2373 and mRNA vaccines, respectively; the adjusted mean (95% CI) number of local symptoms was 1.5 (1.33-1.61) and 2.4 (2.31-2.52), respectively. These results support the use of adjuvanted, protein-based NVX-CoV2373 as an immunization option with lower reactogenicity than mRNAs.
Collapse
Affiliation(s)
- Matthew D. Rousculp
- Novavax, Inc., Gaithersburg, MD 20878, USA; (A.M.M.); (M.M.); (H.B.); (S.T.)
| | - Kelly Hollis
- RTI Health Solutions, Research Triangle Park, NC 27709, USA; (K.H.); (R.Z.); (D.O.); (S.O.); (L.J.)
| | - Ryan Ziemiecki
- RTI Health Solutions, Research Triangle Park, NC 27709, USA; (K.H.); (R.Z.); (D.O.); (S.O.); (L.J.)
| | - Dawn Odom
- RTI Health Solutions, Research Triangle Park, NC 27709, USA; (K.H.); (R.Z.); (D.O.); (S.O.); (L.J.)
| | - Anthony M. Marchese
- Novavax, Inc., Gaithersburg, MD 20878, USA; (A.M.M.); (M.M.); (H.B.); (S.T.)
| | - Mitra Montazeri
- Novavax, Inc., Gaithersburg, MD 20878, USA; (A.M.M.); (M.M.); (H.B.); (S.T.)
| | - Shardul Odak
- RTI Health Solutions, Research Triangle Park, NC 27709, USA; (K.H.); (R.Z.); (D.O.); (S.O.); (L.J.)
| | - Laurin Jackson
- RTI Health Solutions, Research Triangle Park, NC 27709, USA; (K.H.); (R.Z.); (D.O.); (S.O.); (L.J.)
| | - Hadi Beyhaghi
- Novavax, Inc., Gaithersburg, MD 20878, USA; (A.M.M.); (M.M.); (H.B.); (S.T.)
| | - Seth Toback
- Novavax, Inc., Gaithersburg, MD 20878, USA; (A.M.M.); (M.M.); (H.B.); (S.T.)
| |
Collapse
|
34
|
Yu Y, Wei D, Bing T, Wang Y, Liu C, Xiao H. A Polyplatin with Hands-Holding Near-Infrared-II Fluorophores and Prodrugs at a Precise Ratio for Tracking Drug Fate with Realtime Readout and Treatment Feedback. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2402452. [PMID: 38691849 DOI: 10.1002/adma.202402452] [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: 02/16/2024] [Revised: 04/16/2024] [Indexed: 05/03/2024]
Abstract
The in vivo fate of chemotherapeutic drugs plays a vital role in understanding the therapeutic outcome, side effects, and the mechanism. However, the lack of imaging abilities of drugs, tedious labeling processes, and premature leakage of imaging agents result in loss of fidelity between the drugs and imaging signals. Herein, an amphiphilic polymer is created by copolymerization of a near-infrared-II (NIR-II) fluorophore tracer (T) and an anticancer Pt(IV) prodrug (D) of cisplatin in a hand-holding manner into one polymer chain for the first time. The obtained PolyplatinDT is capable of delivering the drugs and the fluorophores concomitantly at a precise D/T ratio, thereby resulting in tracking the platinum drugs and even readout of them in real-time via NIR-II imaging. PolyplatinDT can self-assemble into nanoparticles, referred to as NanoplatinDT. Furthermore, a caspase-3 cleavable peptide that serves as an apoptosis reporter is attached to NanoplatinDT, resulting in NanoplatinDTR that are capable of simultaneously tracking platinum drugs and evaluating the therapeutic efficacy. Overall, it is reported here the design of the first theranostic polymer with anticancer drugs, drug tracers, and drug efficacy reporters that can work in concert to provide insight into the drug fate and mechanism of action.
Collapse
Affiliation(s)
- Yingjie Yu
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Dengshuai Wei
- Beijing National Laboratory for Molecular Sciences, Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Science, Beijing, 100190, China
- University of Chinese Academy of Science, Beijing, 100049, China
| | - Tiejun Bing
- Immunology and Oncology Center, ICE Bioscience, Beijing, 100176, China
| | - Yongheng Wang
- Beijing National Laboratory for Molecular Sciences, Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Science, Beijing, 100190, China
- University of Chinese Academy of Science, Beijing, 100049, China
| | - Chaoyong Liu
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Haihua Xiao
- Beijing National Laboratory for Molecular Sciences, Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Science, Beijing, 100190, China
| |
Collapse
|
35
|
Buckley M, Araínga M, Maiorino L, Pires IS, Kim BJ, Michaels KK, Dye J, Qureshi K, Zhang Y, Mak H, Steichen JM, Schief WR, Villinger F, Irvine DJ. Visualizing lipid nanoparticle trafficking for mRNA vaccine delivery in non-human primates. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.21.600088. [PMID: 38979236 PMCID: PMC11230287 DOI: 10.1101/2024.06.21.600088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
mRNA delivered using lipid nanoparticles (LNPs) has become an important subunit vaccine modality, but mechanisms of action for mRNA vaccines remain incompletely understood. Here, we synthesized a metal chelator-lipid conjugate enabling positron emission tomography (PET) tracer labeling of LNP/mRNA vaccines for quantitative visualization of vaccine trafficking in live non-human primates (NHPs). Following i.m. injection, we observed LNPs distributing through injected muscle tissue, simultaneous with rapid trafficking to draining lymph nodes (dLNs). Deltoid injection of LNPs mimicking human vaccine administration led to stochastic LNP delivery to 3 different sets of dLNs. LNP uptake in dLNs was confirmed by histology, and cellular analysis of tissues via flow cytometry identified antigen-presenting cells as the primary cell type responsible for early LNP uptake and mRNA translation. These results provide insights into the biodistribution of mRNA vaccines administered at clinically relevant doses, injection volumes, and injection sites in an important large animal model for vaccine development.
Collapse
|
36
|
Raheem MA, Rahim MA, Gul I, Reyad-Ul-Ferdous M, Zhang CY, Yu D, Pandey V, Du K, Wang R, Han S, Han Y, Qin P. COVID-19: Post infection implications in different age groups, mechanism, diagnosis, effective prevention, treatment, and recommendations. Life Sci 2024:122861. [PMID: 38925222 DOI: 10.1016/j.lfs.2024.122861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 05/28/2024] [Accepted: 06/18/2024] [Indexed: 06/28/2024]
Abstract
SARS-CoV-2 is a highly contagious pathogen that predominantly caused the COVID-19 pandemic. The persistent effects of COVID-19 are defined as an inflammatory or host response to the virus that begins four weeks after initial infection and persists for an undetermined length of time. Chronic effects are more harmful than acute ones thus, this review explored the long-term effects of the virus on various human organs, including the pulmonary, cardiovascular, and neurological, reproductive, gastrointestinal, musculoskeletal, endocrine, and lymphoid systems and found that SARS-CoV-2 adversely affects these organs of older adults. Regarding diagnosis, the RT-PCR is a gold standard method of diagnosing COVID-19; however, it requires specialized equipment and personnel for performing assays and a long time for results production. Therefore, to overcome these limitations, artificial intelligence employed in imaging and microfluidics technologies is the most promising in diagnosing COVID-19. Pharmacological and non-pharmacological strategies are the most effective treatment for reducing the persistent impacts of COVID-19 by providing immunity to post-COVID-19 patients by reducing cytokine release syndrome, improving the T cell response, and increasing the circulation of activated natural killer and CD8 T cells in blood and tissues, which ultimately reduces fever, nausea, fatigue, and muscle weakness and pain. Vaccines such as inactivated viral, live attenuated viral, protein subunit, viral vectored, mRNA, DNA, or nanoparticle vaccines significantly reduce the adverse long-term virus effects in post-COVID-19 patients; however, no vaccine was reported to provide lifetime protection against COVID-19; consequently, protective measures such as physical separation, mask use, and hand cleansing are promising strategies. This review provides a comprehensive knowledge of the persistent effects of COVID-19 on people of varying ages, as well as diagnosis, treatment, vaccination, and future preventative measures against the spread of SARS-CoV-2.
Collapse
Affiliation(s)
- Muhammad Akmal Raheem
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, PR China; Center of Precision Medicine and Healthcare, Tsinghua-Berkeley Shenzhen Institute, Shenzhen, Guangdong Province 518055, PR China
| | - Muhammad Ajwad Rahim
- College of Animal Science and Technology, Ahnui Agricultural University, Hefei, PR China
| | - Ijaz Gul
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, PR China; Center of Precision Medicine and Healthcare, Tsinghua-Berkeley Shenzhen Institute, Shenzhen, Guangdong Province 518055, PR China
| | - Md Reyad-Ul-Ferdous
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, PR China; Center of Precision Medicine and Healthcare, Tsinghua-Berkeley Shenzhen Institute, Shenzhen, Guangdong Province 518055, PR China
| | - Can Yang Zhang
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, PR China; Center of Precision Medicine and Healthcare, Tsinghua-Berkeley Shenzhen Institute, Shenzhen, Guangdong Province 518055, PR China
| | - Dongmei Yu
- School of Mechanical, Electrical & Information Engineering, Shandong University
| | - Vijay Pandey
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, PR China; Center of Precision Medicine and Healthcare, Tsinghua-Berkeley Shenzhen Institute, Shenzhen, Guangdong Province 518055, PR China
| | - Ke Du
- Department of Chemical and Environmental Engineering, University of California, Riverside, CA, USA
| | - Runming Wang
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, PR China; Center of Precision Medicine and Healthcare, Tsinghua-Berkeley Shenzhen Institute, Shenzhen, Guangdong Province 518055, PR China
| | - Sanyang Han
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, PR China; Center of Precision Medicine and Healthcare, Tsinghua-Berkeley Shenzhen Institute, Shenzhen, Guangdong Province 518055, PR China
| | - Yuxing Han
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, PR China; Center of Precision Medicine and Healthcare, Tsinghua-Berkeley Shenzhen Institute, Shenzhen, Guangdong Province 518055, PR China
| | - Peiwu Qin
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, PR China; Center of Precision Medicine and Healthcare, Tsinghua-Berkeley Shenzhen Institute, Shenzhen, Guangdong Province 518055, PR China.
| |
Collapse
|
37
|
Lemdani K, Marlin R, Mayet C, Perkov V, Pascal Q, Ripoll M, Relouzat F, Dhooge N, Bossevot L, Dereuddre-Bosquet N, Dargazanli G, Thibaut-Duprey K, Haensler J, Chapon C, Prost C, Le Grand R. Distinct dynamics of mRNA LNPs in mice and nonhuman primates revealed by in vivo imaging. NPJ Vaccines 2024; 9:113. [PMID: 38902327 PMCID: PMC11189915 DOI: 10.1038/s41541-024-00900-5] [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: 12/07/2023] [Accepted: 06/03/2024] [Indexed: 06/22/2024] Open
Abstract
The characterization of vaccine distribution to relevant tissues after in vivo administration is critical to understanding their mechanisms of action. Vaccines based on mRNA lipid nanoparticles (LNPs) are now being widely considered against infectious diseases and cancer. Here, we used in vivo imaging approaches to compare the trafficking of two LNP formulations encapsulating mRNA following intramuscular administration: DLin-MC3-DMA (MC3) and the recently developed DOG-IM4. The mRNA formulated in DOG-IM4 LNPs persisted at the injection site, whereas mRNA formulated in MC3 LNPs rapidly migrated to the draining lymph nodes. Furthermore, MC3 LNPs induced the fastest increase in blood neutrophil counts after injection and greater inflammation, as shown by IL-1RA, IL-15, CCL-1, and IL-6 concentrations in nonhuman primate sera. These observations highlight the influence of the nature of the LNP on mRNA vaccine distribution and early immune responses.
Collapse
Affiliation(s)
- Katia Lemdani
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-Immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses & Le Kremlin-Bicêtre, France
- Sanofi, Marcy-L'étoile, France
| | - Romain Marlin
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-Immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses & Le Kremlin-Bicêtre, France
| | - Céline Mayet
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-Immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses & Le Kremlin-Bicêtre, France
| | | | - Quentin Pascal
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-Immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses & Le Kremlin-Bicêtre, France
| | | | - Francis Relouzat
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-Immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses & Le Kremlin-Bicêtre, France
| | - Nina Dhooge
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-Immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses & Le Kremlin-Bicêtre, France
| | - Laetitia Bossevot
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-Immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses & Le Kremlin-Bicêtre, France
| | - Nathalie Dereuddre-Bosquet
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-Immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses & Le Kremlin-Bicêtre, France
| | | | | | | | - Catherine Chapon
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-Immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses & Le Kremlin-Bicêtre, France
| | | | - Roger Le Grand
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-Immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses & Le Kremlin-Bicêtre, France.
| |
Collapse
|
38
|
Pitard B, Pitard I. [« ReNAissance » of biotherapies with RNA]. Med Sci (Paris) 2024; 40:525-533. [PMID: 38986097 DOI: 10.1051/medsci/2024079] [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: 07/12/2024] Open
Abstract
Many diseases originate from either the absence or defective expression of a given protein. For some of them, the lacking protein is secreted or can be taken up by cells when delivered exogenously. In such cases, therapies initially involved administering the physiological protein extracted from human tissues. Subsequently, genetic engineering enabled the production of proteins through cell fermentation after introducing the corresponding gene. For many other pathologies, the deficient protein cannot be delivered exogenously. Thus, an endogenous production of the therapeutic protein by the cells themselves is necessary. Messenger RNA (mRNA) technology, like its predecessor DNA, aims to supplement the genetic information needed to produce the therapeutic protein within the cells. However, unlike DNA-based therapies, mRNA transfer allows for transient expression of the protein of interest, which offers an advantage in numerous pathologies. Nonetheless, mastering the quantity, quality, and spatio-temporal regulation of protein production encoded by therapeutic mRNA remains a significant challenge for the development of this approach.
Collapse
Affiliation(s)
- Bruno Pitard
- Nantes Université, Université d'Angers, Immunology and New Concepts in Immunotherapy (INCIT), Inserm UMR-S 1302, CNRS EMR6001, Nantes, France
| | | |
Collapse
|
39
|
Bettini E, Chudnovskiy A, Protti G, Nakadakari-Higa S, Ceglia S, Castaño D, Chiu J, Muramatsu H, Mdluli T, Abraham E, Lipinszki Z, Maillard I, Tam YK, Reboldi A, Pardi N, Spreafico R, Victora GD, Locci M. Distinct components of nucleoside-modified messenger RNA vaccines cooperate to instruct efficient germinal center responses. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.17.594726. [PMID: 38798523 PMCID: PMC11118742 DOI: 10.1101/2024.05.17.594726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Nucleoside-modified mRNA vaccines elicit protective antibodies through their ability to promote T follicular helper (Tfh) cells. The lipid nanoparticle (LNP) component of mRNA vaccines possesses inherent adjuvant activity. However, to what extent the nucleoside-modified mRNA can be sensed and contribute to Tfh cell responses remains largely undefined. Herein, we deconvoluted the signals induced by LNP and mRNA that instruct dendritic cells (DCs) to promote Tfh cell differentiation. We demonstrated that the nucleoside-modified mRNA drives the production of type I interferons that act on DCs to induce their maturation and the induction of Th1-biased Tfh responses. Conversely, LNP favors the acquisition of a Tfh cell-inducing program in DCs, a stronger Th2 polarization in Tfh cells, and allows for rapid mRNA translation by DCs within the draining lymph node. Our work unravels distinct adjuvant features of mRNA and LNP necessary for the induction of Tfh cells, with implications for vaccine design.
Collapse
|
40
|
Hellgren F, Rosdahl A, Arcoverde Cerveira R, Lenart K, Ols S, Gwon YD, Kurt S, Delis AM, Joas G, Evander M, Normark J, Ahlm C, Forsell MN, Cajander S, Loré K. Modulation of innate immune response to mRNA vaccination after SARS-CoV-2 infection or sequential vaccination in humans. JCI Insight 2024; 9:e175401. [PMID: 38716734 PMCID: PMC11141904 DOI: 10.1172/jci.insight.175401] [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: 09/05/2023] [Accepted: 03/22/2024] [Indexed: 06/02/2024] Open
Abstract
mRNA vaccines are likely to become widely used for the prevention of infectious diseases in the future. Nevertheless, a notable gap exists in mechanistic data, particularly concerning the potential effects of sequential mRNA immunization or preexisting immunity on the early innate immune response triggered by vaccination. In this study, healthy adults, with or without documented prior SARS-CoV-2 infection, were vaccinated with the BNT162b2/Comirnaty mRNA vaccine. Prior infection conferred significantly stronger induction of proinflammatory and type I IFN-related gene signatures, serum cytokines, and monocyte expansion after the prime vaccination. The response to the second vaccination further increased the magnitude of the early innate response in both study groups. The third vaccination did not further increase vaccine-induced inflammation. In vitro stimulation of PBMCs with TLR ligands showed no difference in cytokine responses between groups, or before or after prime vaccination, indicating absence of a trained immunity effect. We observed that levels of preexisting antigen-specific CD4 T cells, antibody, and memory B cells correlated with elements of the early innate response to the first vaccination. Our data thereby indicate that preexisting memory formed by infection may augment the innate immune activation induced by mRNA vaccines.
Collapse
Affiliation(s)
- Fredrika Hellgren
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden & Karolinska University Hospital, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Anja Rosdahl
- Department of Infectious Diseases and
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Rodrigo Arcoverde Cerveira
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden & Karolinska University Hospital, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Klara Lenart
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden & Karolinska University Hospital, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Sebastian Ols
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden & Karolinska University Hospital, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Yong-Dae Gwon
- Department of Clinical Microbiology, Umeå University, Umeå, Sweden
| | - Seta Kurt
- Department of Clinical Research Laboratory, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Anna Maria Delis
- Department of Clinical Research Laboratory, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Gustav Joas
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden & Karolinska University Hospital, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Magnus Evander
- Department of Clinical Microbiology, Umeå University, Umeå, Sweden
| | - Johan Normark
- Department of Clinical Microbiology, Umeå University, Umeå, Sweden
| | - Clas Ahlm
- Department of Clinical Microbiology, Umeå University, Umeå, Sweden
| | | | - Sara Cajander
- Department of Infectious Diseases and
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Karin Loré
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden & Karolinska University Hospital, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
41
|
Abbasi S, Matsui-Masai M, Yasui F, Hayashi A, Tockary TA, Mochida Y, Akinaga S, Kohara M, Kataoka K, Uchida S. Carrier-free mRNA vaccine induces robust immunity against SARS-CoV-2 in mice and non-human primates without systemic reactogenicity. Mol Ther 2024; 32:1266-1283. [PMID: 38569556 PMCID: PMC11081875 DOI: 10.1016/j.ymthe.2024.03.022] [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: 10/29/2023] [Revised: 02/21/2024] [Accepted: 03/11/2024] [Indexed: 04/05/2024] Open
Abstract
Carrier-free naked mRNA vaccines may reduce the reactogenicity associated with delivery carriers; however, their effectiveness against infectious diseases has been suboptimal. To boost efficacy, we targeted the skin layer rich in antigen-presenting cells (APCs) and utilized a jet injector. The jet injection efficiently introduced naked mRNA into skin cells, including APCs in mice. Further analyses indicated that APCs, after taking up antigen mRNA in the skin, migrated to the lymph nodes (LNs) for antigen presentation. Additionally, the jet injection provoked localized lymphocyte infiltration in the skin, serving as a physical adjuvant for vaccination. Without a delivery carrier, our approach confined mRNA distribution to the injection site, preventing systemic mRNA leakage and associated systemic proinflammatory reactions. In mouse vaccination, the naked mRNA jet injection elicited robust antigen-specific antibody production over 6 months, along with germinal center formation in LNs and the induction of both CD4- and CD8-positive T cells. By targeting the SARS-CoV-2 spike protein, this approach provided protection against viral challenge. Furthermore, our approach generated neutralizing antibodies against SARS-CoV-2 in non-human primates at levels comparable to those observed in mice. In conclusion, our approach offers a safe and effective option for mRNA vaccines targeting infectious diseases.
Collapse
Affiliation(s)
- Saed Abbasi
- Innovation Center of NanoMedicine (iCONM), Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
| | - Miki Matsui-Masai
- Department of Research, NANO MRNA Co., Ltd., 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
| | - Fumihiko Yasui
- Department of Diseases and Infection, Tokyo Metropolitan Institute of Medical Science, 2-1-6, Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan
| | - Akimasa Hayashi
- Department of Pathology, Kyorin University School of Medicine, 6-20-2 Shinkawa, Mitaka-shi, Tokyo 181-8611, Japan
| | - Theofilus A Tockary
- Innovation Center of NanoMedicine (iCONM), Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
| | - Yuki Mochida
- Innovation Center of NanoMedicine (iCONM), Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan; Department of Advanced Nanomedical Engineering, Medical Research Institute, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Shiro Akinaga
- Department of Research, NANO MRNA Co., Ltd., 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
| | - Michinori Kohara
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, 2-1-6, Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan
| | - Kazunori Kataoka
- Innovation Center of NanoMedicine (iCONM), Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan.
| | - Satoshi Uchida
- Innovation Center of NanoMedicine (iCONM), Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan; Department of Advanced Nanomedical Engineering, Medical Research Institute, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan.
| |
Collapse
|
42
|
Kim J, Eygeris Y, Ryals RC, Jozić A, Sahay G. Strategies for non-viral vectors targeting organs beyond the liver. NATURE NANOTECHNOLOGY 2024; 19:428-447. [PMID: 38151642 DOI: 10.1038/s41565-023-01563-4] [Citation(s) in RCA: 40] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Accepted: 11/01/2023] [Indexed: 12/29/2023]
Abstract
In recent years, nanoparticles have evolved to a clinical modality to deliver diverse nucleic acids. Rising interest in nanomedicines comes from proven safety and efficacy profiles established by continuous efforts to optimize physicochemical properties and endosomal escape. However, despite their transformative impact on the pharmaceutical industry, the clinical use of non-viral nucleic acid delivery is limited to hepatic diseases and vaccines due to liver accumulation. Overcoming liver tropism of nanoparticles is vital to meet clinical needs in other organs. Understanding the anatomical structure and physiological features of various organs would help to identify potential strategies for fine-tuning nanoparticle characteristics. In this Review, we discuss the source of liver tropism of non-viral vectors, present a brief overview of biological structure, processes and barriers in select organs, highlight approaches available to reach non-liver targets, and discuss techniques to accelerate the discovery of non-hepatic therapies.
Collapse
Affiliation(s)
- Jeonghwan Kim
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, OR, USA
- College of Pharmacy, Yeungnam University, Gyeongsan, South Korea
| | - Yulia Eygeris
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, OR, USA
| | - Renee C Ryals
- Department of Ophthalmology, Casey Eye Institute, Oregon Health and Science University, Portland, OR, USA
| | - Antony Jozić
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, OR, USA
| | - Gaurav Sahay
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, OR, USA.
- Department of Ophthalmology, Casey Eye Institute, Oregon Health and Science University, Portland, OR, USA.
- Department of Biomedical Engineering, Robertson Life Sciences Building, Oregon Health and Science University, Portland, OR, USA.
| |
Collapse
|
43
|
Zhou F, Huang L, Li S, Yang W, Chen F, Cai Z, Liu X, Xu W, Lehto V, Lächelt U, Huang R, Shi Y, Lammers T, Tao W, Xu ZP, Wagner E, Xu Z, Yu H. From structural design to delivery: mRNA therapeutics for cancer immunotherapy. EXPLORATION (BEIJING, CHINA) 2024; 4:20210146. [PMID: 38855617 PMCID: PMC11022630 DOI: 10.1002/exp.20210146] [Citation(s) in RCA: 32] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 10/15/2023] [Indexed: 06/11/2024]
Abstract
mRNA therapeutics have emerged as powerful tools for cancer immunotherapy in accordance with their superiority in expressing all sequence-known proteins in vivo. In particular, with a small dosage of delivered mRNA, antigen-presenting cells (APCs) can synthesize mutant neo-antigens and multi-antigens and present epitopes to T lymphocytes to elicit antitumor effects. In addition, expressing receptors like chimeric antigen receptor (CAR), T-cell receptor (TCR), CD134, and immune-modulating factors including cytokines, interferons, and antibodies in specific cells can enhance immunological response against tumors. With the maturation of in vitro transcription (IVT) technology, large-scale and pure mRNA encoding specific proteins can be synthesized quickly. However, the clinical translation of mRNA-based anticancer strategies is restricted by delivering mRNA into target organs or cells and the inadequate endosomal escape efficiency of mRNA. Recently, there have been some advances in mRNA-based cancer immunotherapy, which can be roughly classified as modifications of the mRNA structure and the development of delivery systems, especially the lipid nanoparticle platforms. In this review, the latest strategies for overcoming the limitations of mRNA-based cancer immunotherapies and the recent advances in delivering mRNA into specific organs and cells are summarized. Challenges and opportunities for clinical applications of mRNA-based cancer immunotherapy are also discussed.
Collapse
Affiliation(s)
- Feng Zhou
- State Key Laboratory of Chemical Biology and Center of Pharmaceutics, Shanghai Institute of Materia MedicaChinese Academy of SciencesShanghaiChina
- University of Chinese Academy of SciencesBeijingChina
| | - Lujia Huang
- State Key Laboratory of Chemical Biology and Center of Pharmaceutics, Shanghai Institute of Materia MedicaChinese Academy of SciencesShanghaiChina
- University of Chinese Academy of SciencesBeijingChina
| | - Shiqin Li
- State Key Laboratory of Chemical Biology and Center of Pharmaceutics, Shanghai Institute of Materia MedicaChinese Academy of SciencesShanghaiChina
| | - Wenfang Yang
- State Key Laboratory of Chemical Biology and Center of Pharmaceutics, Shanghai Institute of Materia MedicaChinese Academy of SciencesShanghaiChina
| | - Fangmin Chen
- State Key Laboratory of Chemical Biology and Center of Pharmaceutics, Shanghai Institute of Materia MedicaChinese Academy of SciencesShanghaiChina
- University of Chinese Academy of SciencesBeijingChina
| | - Zhixiong Cai
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian ProvinceMengchao Hepatobiliary Hospital of Fujian Medical UniversityFuzhouChina
| | - Xiaolong Liu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian ProvinceMengchao Hepatobiliary Hospital of Fujian Medical UniversityFuzhouChina
| | - Wujun Xu
- Department of Applied PhysicsUniversity of Eastern FinlandKuopioFinland
| | - Vesa‐Pekka Lehto
- Department of Applied PhysicsUniversity of Eastern FinlandKuopioFinland
| | - Ulrich Lächelt
- Department of Pharmaceutical SciencesUniversity of ViennaViennaAustria
| | - Rongqin Huang
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug DeliveryMinistry of Education, Fudan UniversityShanghaiChina
| | - Yang Shi
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular ImagingRWTH Aachen University ClinicAachenGermany
| | - Twan Lammers
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular ImagingRWTH Aachen University ClinicAachenGermany
| | - Wei Tao
- Center for Nanomedicine and Department of Anaesthesiology, Brigham and Women's HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | - Zhi Ping Xu
- Institute of Biomedical Health Technology and Engineering and Institute of Systems and Physical BiologyShenzhen Bay LaboratoryShenzhenChina
| | - Ernst Wagner
- Pharmaceutical Biotechnology, Center for NanoscienceLudwig‐Maximilians‐UniversitätMunichGermany
| | - Zhiai Xu
- School of Chemistry and Molecular EngineeringEast China Normal UniversityShanghaiChina
| | - Haijun Yu
- State Key Laboratory of Chemical Biology and Center of Pharmaceutics, Shanghai Institute of Materia MedicaChinese Academy of SciencesShanghaiChina
- University of Chinese Academy of SciencesBeijingChina
| |
Collapse
|
44
|
Zhang H, Gao X, Sun Q, Dong X, Zhu Z, Yang C. Incorporation of poly(γ-glutamic acid) in lipid nanoparticles for enhanced mRNA delivery efficiency in vitro and in vivo. Acta Biomater 2024; 177:361-376. [PMID: 38342193 DOI: 10.1016/j.actbio.2024.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 01/26/2024] [Accepted: 02/04/2024] [Indexed: 02/13/2024]
Abstract
Messenger RNA (mRNA)-based therapy shows immense potential for broad biomedical applications. However, the development of safe and efficacious mRNA delivery vectors remains challenging due to delivery barriers and inefficient intracellular payload release. Herein, we presented a simple strategy to boost the mRNA intracellular release by incorporation of anionic poly(γ-glutamic acid) (PGA) into an ionizable lipid-based LNP/mRNA. We systematically investigated the impact of PGA incorporation on mRNA transfection both in vitro and in vivo. The molecular weights and formulation ratios of PGA greatly affected the transfection efficacy of LNP/mRNA. From in vitro study, the optimized LNP/mRNA/PGA was formulated by incorporation of PGA with the molecular weight of 80 kDa or 200 kDa and the charge ratio (N/P/C) of 25/1/1. The optimized formulation achieved around 3-fold mRNA expression in HeLa cells compared to the bare LNP/mRNA. The intracellular releasing study using specific DNA probe revealed that this enhancement of transfection efficacy was attributed to the elevated mRNA release into cytoplasm. Moreover, the optimized LNP/mRNA/PGA achieved up to 5-fold or 3-fold increase of luciferase mRNA expression in vivo after being injected into mice systematically or intramuscularly, respectively. In addition, the incorporation of PGA did not significantly alter the biodistribution profile of the complexes on both organ and cellular levels. Therefore, our work provides a simple strategy to boost mRNA delivery, which holds great promise to improve the efficacy of mRNA therapeutics for various biomedical applications. STATEMENT OF SIGNIFICANCE: The process of designing and screening potent mRNA carriers is complicated and time-consuming, while the efficacy is not always satisfying due to the delivery barriers and inefficient mRNA release. This work presented an alternative strategy to boost the mRNA delivery efficacy by incorporating an anionic natural polymer poly(γ-glutamic acid) (PGA) into LNP/mRNA complexes. The optimized LNP/mRNA/PGA achieved up to 3-fold and 5-fold increase in transfection efficacy in vitro and in vivo, respectively. Intracellular releasing analysis revealed that the enhancement of transfection efficacy was mainly attributed to the elevated intracellular release of mRNA. In addition, the incorporation of PGA did not alter the biodistribution or the biosafety profile of the complexes. These findings indicate that PGA incorporation is a promising strategy to improve the efficacy of mRNA therapeutics.
Collapse
Affiliation(s)
- Hongqian Zhang
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, and School of Chemistry and Chemical Engineering, Shandong University, Jinan 25010, China
| | - Xue Gao
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, and School of Chemistry and Chemical Engineering, Shandong University, Jinan 25010, China
| | - Qian Sun
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, and School of Chemistry and Chemical Engineering, Shandong University, Jinan 25010, China
| | - Xiaoxue Dong
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, and School of Chemistry and Chemical Engineering, Shandong University, Jinan 25010, China
| | - Zongwei Zhu
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, and School of Chemistry and Chemical Engineering, Shandong University, Jinan 25010, China
| | - Chuanxu Yang
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, and School of Chemistry and Chemical Engineering, Shandong University, Jinan 25010, China.
| |
Collapse
|
45
|
Hassett KJ, Rajlic IL, Bahl K, White R, Cowens K, Jacquinet E, Burke KE. mRNA vaccine trafficking and resulting protein expression after intramuscular administration. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102083. [PMID: 38161733 PMCID: PMC10755037 DOI: 10.1016/j.omtn.2023.102083] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 11/20/2023] [Indexed: 01/03/2024]
Abstract
The mRNA vaccine route from injection site to critical immunologic tissues, as well as the localization of protein antigen following intramuscular (i.m.) administration, is crucial to generating an effective immune response. Here, we quantified mRNA at the injection site, lymph nodes, and in select tissues. mRNA was primarily present 24 h after administration and then rapidly degraded from local and systemic tissues. Histological analyses of mRNA and expressed protein at the site of administration and in the lymph nodes following i.m. administration of our vaccine in rodents and nonhuman primates (NHPs) were completed, and mRNA and protein expression were detected in tissue resident and infiltrating immune cells at the injection site. In addition, high levels of protein expression were observed within subcapsular and medullary sinus macrophages in draining lymph nodes. More important, results were similar between rodents and NHPs, indicating cross-species similarities.
Collapse
Affiliation(s)
| | | | - Kapil Bahl
- Orbital Therapeutics, 21 Erie Street, Cambridge, MA 02139, USA
| | - Rebecca White
- ReNAgade Therapeutics, 640 Memorial Drive, Suite 2300, Cambridge, MA 02139, USA
| | - Kristen Cowens
- Moderna, Inc., 200 Technology Square, Cambridge, MA 02139, USA
| | - Eric Jacquinet
- Moderna, Inc., 200 Technology Square, Cambridge, MA 02139, USA
| | | |
Collapse
|
46
|
Chaudhary N, Newby AN, Arral ML, Yerneni SS, LoPresti ST, Doerfler R, Petersen DMS, Montoya C, Kim JS, Fox B, Coon T, Malaney A, Sadovsky Y, Whitehead KA. Lipid nanoparticle structure and delivery route during pregnancy dictate mRNA potency, immunogenicity, and maternal and fetal outcomes. Proc Natl Acad Sci U S A 2024; 121:e2307810121. [PMID: 38437545 PMCID: PMC10945816 DOI: 10.1073/pnas.2307810121] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 11/02/2023] [Indexed: 03/06/2024] Open
Abstract
Treating pregnancy-related disorders is exceptionally challenging because the threat of maternal and/or fetal toxicity discourages the use of existing medications and hinders new drug development. One potential solution is the use of lipid nanoparticle (LNP) RNA therapies, given their proven efficacy, tolerability, and lack of fetal accumulation. Here, we describe LNPs for efficacious mRNA delivery to maternal organs in pregnant mice via several routes of administration. In the placenta, our lead LNP transfected trophoblasts, endothelial cells, and immune cells, with efficacy being structurally dependent on the ionizable lipid polyamine headgroup. Next, we show that LNP-induced maternal inflammatory responses affect mRNA expression in the maternal compartment and hinder neonatal development. Specifically, pro-inflammatory LNP structures and routes of administration curtailed efficacy in maternal lymphoid organs in an IL-1β-dependent manner. Further, immunogenic LNPs provoked the infiltration of adaptive immune cells into the placenta and restricted pup growth after birth. Together, our results provide mechanism-based structural guidance on the design of potent LNPs for safe use during pregnancy.
Collapse
Affiliation(s)
- Namit Chaudhary
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA15213
| | - Alexandra N. Newby
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA15213
| | - Mariah L. Arral
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA15213
| | | | - Samuel T. LoPresti
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA15213
| | - Rose Doerfler
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA15213
| | | | - Catalina Montoya
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA15213
| | - Julie S. Kim
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA15213
| | - Bethany Fox
- Mellon Institute Centralized Vivarium, Carnegie Mellon University, Pittsburgh, PA15213
| | - Tiffany Coon
- Department of Obstetrics, Gynecology and Reproductive Sciences, Magee-Womens Research Institute, University of Pittsburgh, Pittsburgh, PA15213
| | - Angela Malaney
- Mellon Institute Centralized Vivarium, Carnegie Mellon University, Pittsburgh, PA15213
| | - Yoel Sadovsky
- Department of Obstetrics, Gynecology and Reproductive Sciences, Magee-Womens Research Institute, University of Pittsburgh, Pittsburgh, PA15213
| | - Kathryn A. Whitehead
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA15213
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA15213
| |
Collapse
|
47
|
Wan J, Wang Z, Wang L, Wu L, Zhang C, Zhou M, Fu ZF, Zhao L. Circular RNA vaccines with long-term lymph node-targeting delivery stability after lyophilization induce potent and persistent immune responses. mBio 2024; 15:e0177523. [PMID: 38078742 PMCID: PMC10790773 DOI: 10.1128/mbio.01775-23] [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: 07/09/2023] [Accepted: 10/26/2023] [Indexed: 01/17/2024] Open
Abstract
IMPORTANCE messenger RNA (mRNA) vaccines are a key technology in combating existing and emerging infectious diseases. However, the inherent instability of mRNA and the nonspecificity of lipid nanoparticle-encapsulated (LNP) delivery systems result in the need for cold storage and a relatively short-duration immune response to mRNA vaccines. Herein, we develop a novel vaccine in the form of circRNAs encapsulated in LNPs, and the circular structure of the circRNAs enhances their stability. Lyophilization is considered the most effective method for the long-term preservation of RNA vaccines. However, this process may result in irreversible damage to the nanoparticles, particularly the potential disruption of targeting modifications on LNPs. During the selection of lymph node-targeting ligands, we found that LNPs modified with mannose maintained their physical properties almost unchanged after lyophilization. Additionally, the targeting specificity and immunogenicity remained unaffected. In contrast, even with the addition of cryoprotectants such as sucrose, the physical properties of LNPs were impaired, leading to an obvious decrease in immunogenicity. This may be attributed to the protective role of mannose on the surface of LNPs during lyophilization. Freshly prepared and lyophilized mLNP-circRNA vaccines elicited comparable immune responses in both the rabies virus model and the SARS-CoV-2 model. Our data demonstrated that mLNP-circRNA vaccines elicit robust immune responses while improving stability after lyophilization, with no compromise in tissue targeting specificity. Therefore, mannose-modified LNP-circRNA vaccines represent a promising vaccine design strategy.
Collapse
Affiliation(s)
- Jiawu Wan
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- Hubei Hongshan Laboratory, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Zongmei Wang
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- Hubei Hongshan Laboratory, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Lingli Wang
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- Hubei Hongshan Laboratory, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Liqin Wu
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- Hubei Hongshan Laboratory, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Chengguang Zhang
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- Hubei Hongshan Laboratory, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Ming Zhou
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- Hubei Hongshan Laboratory, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Zhen F. Fu
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- Hubei Hongshan Laboratory, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Ling Zhao
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- Hubei Hongshan Laboratory, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
48
|
Jones CH, Androsavich JR, So N, Jenkins MP, MacCormack D, Prigodich A, Welch V, True JM, Dolsten M. Breaking the mold with RNA-a "RNAissance" of life science. NPJ Genom Med 2024; 9:2. [PMID: 38195675 PMCID: PMC10776758 DOI: 10.1038/s41525-023-00387-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 12/07/2023] [Indexed: 01/11/2024] Open
Abstract
In the past decade, RNA therapeutics have gone from being a promising concept to one of the most exciting frontiers in healthcare and pharmaceuticals. The field is now entering what many call a renaissance or "RNAissance" which is being fueled by advances in genetic engineering and delivery systems to take on more ambitious development efforts. However, this renaissance is occurring at an unprecedented pace, which will require a different way of thinking if the field is to live up to its full potential. Recognizing this need, this article will provide a forward-looking perspective on the field of RNA medical products and the potential long-term innovations and policy shifts enabled by this revolutionary and game-changing technological platform.
Collapse
Affiliation(s)
| | | | - Nina So
- Pfizer, 66 Hudson Boulevard, New York, NY, 10018, USA
| | | | | | | | - Verna Welch
- Pfizer, 66 Hudson Boulevard, New York, NY, 10018, USA
| | - Jane M True
- Pfizer, 66 Hudson Boulevard, New York, NY, 10018, USA.
| | | |
Collapse
|
49
|
Mochida Y, Uchida S. mRNA vaccine designs for optimal adjuvanticity and delivery. RNA Biol 2024; 21:1-27. [PMID: 38528828 PMCID: PMC10968337 DOI: 10.1080/15476286.2024.2333123] [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] [Revised: 02/26/2024] [Accepted: 03/15/2024] [Indexed: 03/27/2024] Open
Abstract
Adjuvanticity and delivery are crucial facets of mRNA vaccine design. In modern mRNA vaccines, adjuvant functions are integrated into mRNA vaccine nanoparticles, allowing the co-delivery of antigen mRNA and adjuvants in a unified, all-in-one formulation. In this formulation, many mRNA vaccines utilize the immunostimulating properties of mRNA and vaccine carrier components, including lipids and polymers, as adjuvants. However, careful design is necessary, as excessive adjuvanticity and activation of improper innate immune signalling can conversely hinder vaccination efficacy and trigger adverse effects. mRNA vaccines also require delivery systems to achieve antigen expression in antigen-presenting cells (APCs) within lymphoid organs. Some vaccines directly target APCs in the lymphoid organs, while others rely on APCs migration to the draining lymph nodes after taking up mRNA vaccines. This review explores the current mechanistic understanding of these processes and the ongoing efforts to improve vaccine safety and efficacy based on this understanding.
Collapse
Affiliation(s)
- Yuki Mochida
- Department of Advanced Nanomedical Engineering, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
- Innovation Center of NanoMedicine (iCONM), Kawasaki Institute of Industrial Promotion, Kawasaki, Japan
| | - Satoshi Uchida
- Department of Advanced Nanomedical Engineering, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
- Innovation Center of NanoMedicine (iCONM), Kawasaki Institute of Industrial Promotion, Kawasaki, Japan
| |
Collapse
|
50
|
Metkar M, Pepin CS, Moore MJ. Tailor made: the art of therapeutic mRNA design. Nat Rev Drug Discov 2024; 23:67-83. [PMID: 38030688 DOI: 10.1038/s41573-023-00827-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/09/2023] [Indexed: 12/01/2023]
Abstract
mRNA medicine is a new and rapidly developing field in which the delivery of genetic information in the form of mRNA is used to direct therapeutic protein production in humans. This approach, which allows for the quick and efficient identification and optimization of drug candidates for both large populations and individual patients, has the potential to revolutionize the way we prevent and treat disease. A key feature of mRNA medicines is their high degree of designability, although the design choices involved are complex. Maximizing the production of therapeutic proteins from mRNA medicines requires a thorough understanding of how nucleotide sequence, nucleotide modification and RNA structure interplay to affect translational efficiency and mRNA stability. In this Review, we describe the principles that underlie the physical stability and biological activity of mRNA and emphasize their relevance to the myriad considerations that factor into therapeutic mRNA design.
Collapse
|