1
|
Costa GL, Sautto GA. Towards an HCV vaccine: an overview of the immunization strategies for eliciting an effective B-cell response. Expert Rev Vaccines 2025; 24:96-120. [PMID: 39825640 DOI: 10.1080/14760584.2025.2452955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 10/26/2024] [Accepted: 01/09/2025] [Indexed: 01/20/2025]
Abstract
INTRODUCTION Fifty-eight million people worldwide are chronically infected with hepatitis C virus (HCV) and are at risk of developing cirrhosis and hepatocellular carcinoma (HCC). Direct-acting antivirals are highly effective; however, they are burdened by high costs and the unchanged risk of HCC and reinfection, making prophylactic countermeasures an urgent medical need. HCV high genetic diversity is one of the main obstacles to vaccine development. The protective role of the humoral response directed against the HCV E2 glycoprotein is well established, and broadly neutralizing antibodies play a crucial role in effective viral clearance. AREAS COVERED This review explores the HCV targets and the different vaccination approaches, encompassing different expression systems, antigen selection strategies, and delivery methods, focusing on those aimed at eliciting a broad and effective humoral response. Our search criteria included the keywords 'HCV,' 'Hepatitis C,' and 'vaccine' using publicly available databases. Following the screening, 54 papers were selected. EXPERT OPINION The investigation of novel vaccine platforms beyond traditional approaches is necessary. While progress has been made in this direction, continued investigations on the HCV virology, immunology, and vaccinology are essential to surmount associated obstacles, heling in the development of an HCV vaccine that can benefit the global public health.
Collapse
Affiliation(s)
- Gabriel L Costa
- Florida Research and Innovation Center, Cleveland Clinic, Port Saint Lucie, FL, USA
| | - Giuseppe A Sautto
- Florida Research and Innovation Center, Cleveland Clinic, Port Saint Lucie, FL, USA
| |
Collapse
|
2
|
Fougeroux C, Hagen SH, Goksøyr L, Aves KL, Okholm AK, Morin C, Lokras AG, Baghel SS, Foged C, van de Vegte-Bolmer M, van Gemert GJ, Jore MM, Vidal-Calvo EE, Gustavsson T, Salanti A, Theander TG, Nielsen MA, de Jongh WA, Sander Bertelsen AF. A modular mRNA vaccine platform encoding antigen-presenting capsid virus-like particles enhances the immunogenicity of the malaria antigen Pfs25. NATURE NANOTECHNOLOGY 2025:10.1038/s41565-025-01889-1. [PMID: 40369344 DOI: 10.1038/s41565-025-01889-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 02/10/2025] [Indexed: 05/16/2025]
Abstract
The COVID-19 pandemic has emphasized the potential of mRNA vaccines in fighting pandemics, owing to their rapid development, strong immunogenicity and adaptability. However, a drawback is their dose-limiting reactogenicity and inability to generate durable humoral immunity. Here we introduce a modular nucleotide vaccine platform combining the advantages of genetic and capsid virus-like-particle-based vaccines. This platform allows for the display of various antigens on different capsid virus-like particles, improving the magnitude, quality and longevity of the vaccine-induced immune responses. We applied this technology to enhance the immunogenicity of the Pfs25 antigen. Immunization with lipid-nanoparticle-formulated mRNA encoding Pfs25 capsid virus-like particles resulted in higher and potentially more durable anti-Pfs25 antibody responses, along with enhanced functional activity, compared with an mRNA vaccine encoding soluble Pfs25. By improving both humoral and cellular immune responses, this approach may reduce the dose and number of administrations required for effective protection. As a result, it can improve the feasibility of both DNA- and mRNA-based vaccines targeting pandemic and endemic infectious diseases.
Collapse
Affiliation(s)
| | | | | | - Kara-Lee Aves
- Centre for Translational Medicine and Parasitology, Department for Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Anna Kathrine Okholm
- Centre for Translational Medicine and Parasitology, Department for Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Candice Morin
- Centre for Translational Medicine and Parasitology, Department for Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Abhijeet Girish Lokras
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Saahil Sandeep Baghel
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Camilla Foged
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | | | - Matthijs M Jore
- Department of Medical Microbiology, Radboudumc, Nijmegen, The Netherlands
| | - Elena Ethel Vidal-Calvo
- Centre for Translational Medicine and Parasitology, Department for Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- VAR2 Pharmaceuticals, Copenhagen, Denmark
| | - Tobias Gustavsson
- Centre for Translational Medicine and Parasitology, Department for Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- VAR2 Pharmaceuticals, Copenhagen, Denmark
| | - Ali Salanti
- Centre for Translational Medicine and Parasitology, Department for Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- VAR2 Pharmaceuticals, Copenhagen, Denmark
| | - Thor Grundtvig Theander
- Centre for Translational Medicine and Parasitology, Department for Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Morten Agertoug Nielsen
- Centre for Translational Medicine and Parasitology, Department for Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Adam Frederik Sander Bertelsen
- AdaptVac Aps, Copenhagen, Denmark.
- Centre for Translational Medicine and Parasitology, Department for Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
3
|
Ling J, Chen H, Huang M, Wang J, Du X. An mRNA vaccine encoding proteasome-targeted antigen enhances CD8 + T cell immunity. J Control Release 2025; 381:113578. [PMID: 40015339 DOI: 10.1016/j.jconrel.2025.02.074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 02/22/2025] [Accepted: 02/24/2025] [Indexed: 03/01/2025]
Abstract
The efficient induction of antigen-specific CD8+ T cell activation is crucial in the development of mRNA tumor vaccines. Endogenous antigens are primarily degraded through the ubiquitin-proteasome system, followed by antigen presentation via major histocompatibility complex class I (MHC-I) molecules, leading to the activation of CD8+ T cells. Therefore, in this study, a novel mRNA vaccine was developed by fusing the mRNA sequence encoding the antigen with a proteasome-targeting peptide (PTP), aiming to enhance proteasomal targeting of the antigen and facilitate its degradation through the ubiquitin-proteasome system, thereby inducing a stronger CD8+ T cell immune response. This study confirmed a significant increase in antigen expression of the antigen-PTP fused mRNA vaccine upon treatment with a VHL inhibitor, as well as notable upregulation of genes associated with the MHC-I antigen-presenting pathway following treatment with the antigen-PTP fused mRNA vaccine. The intramuscular administration of the antigen-PTP fused mRNA vaccine significantly promoted the activation of dendritic cells, macrophages, and T cells in draining lymph nodes and spleens. Additionally, in TC-1 tumor-bearing mice, it markedly suppressed tumor growth, facilitated infiltration of intratumoral antigen-specific CD8+ T cells, and induced immune memory.
Collapse
Affiliation(s)
- Jin Ling
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, PR China
| | - Hongwei Chen
- School of Medicine, South China University of Technology, Guangzhou 510006, PR China
| | - Mengwen Huang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, PR China
| | - Jun Wang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, PR China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, PR China; Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou 510006, PR China.
| | - Xiaojiao Du
- School of Medicine, South China University of Technology, Guangzhou 510006, PR China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, PR China.
| |
Collapse
|
4
|
SU Y, QIAN X, QIN W. [Research advances of liposomes and exosomes in drug delivery and biomarker screening]. Se Pu 2025; 43:472-486. [PMID: 40331611 PMCID: PMC12059997 DOI: 10.3724/sp.j.1123.2024.08012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Indexed: 05/08/2025] Open
Abstract
Vesicles, are categorized as artificial (i.e., liposomes) or natural (i.e., extracellular vesicles (EVs)) and play significant roles in drug-delivery and biomarker-screening applications. Liposomes, as a representative form of artificial vesicle, are spherical lipid structures composed of one or more artificially synthesized phospholipid bilayers. Liposomes are highly biocompatible and bioavailable, very stable, and easily synthesized; hence, they are among the most commonly used and frequently applied nanocarriers in targeted drug-delivery systems (DDS). EVs are natural small membrane-bound vesicles actively secreted by cells and contain a variety of components, including nucleic acids, proteins, and lipids. They also serve as important mediators of intercellular communication. As the smallest EV subtype, with diameters of only 30-100 nm, exosomes contain unique biomolecules that are considered to be the fingerprints of the parent cells. In the pathological state, the content of exosomes will change; consequently, exosomes are potential disease-diagnosis biomarkers. Recent clinical trials have shown that exosomes are ideal nanocarriers in targeted drug-delivery therapies for a variety of diseases. Compared with traditional artificial liposomal carriers, exosomes display unique advantages and provide the DDS field with new possibilities. Liposomes and exosomes are receiving increasing levels of attention in the drug-delivery and biomarker-screening fields. This article introduces techniques for the preparation of liposomes, and the enrichment and separation of exosomes, and delves into research progress on their use in drug-delivery and biomarker-screening applications. Finally, challenges facing the use of liposomes and exosomes in clinical applications are discussed.
Collapse
|
5
|
Heiser BJ, Veyssi A, Ghosh D. Recent strategies for enhanced delivery of mRNA to the lungs. Nanomedicine (Lond) 2025; 20:1043-1069. [PMID: 40190037 PMCID: PMC12051540 DOI: 10.1080/17435889.2025.2485669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 03/25/2025] [Indexed: 05/02/2025] Open
Abstract
mRNA-based therapies have emerged as a transformative tool in modern medicine, gaining significant attention following their successful use in COVID-19 vaccines. Delivery to the lungs offers several compelling advantages for mRNA delivery. The lungs are one of the most vascularized organs in the body, which provides an extensive surface area that can facilitate efficient drug transport. Local delivery to the lungs bypasses gastrointestinal degradation, potentially enhancing therapeutic efficacy. In addition, the extensive capillary network of the lungs provides an ideal target for systemic delivery. However, developing effective mRNA therapies for the lungs presents significant challenges. The complex anatomy of the lungs and the body's immune response to foreign particles create barriers to delivery. This review discusses key approaches for overcoming these challenges and improving mRNA delivery to the lungs. It examines both local and systemic delivery strategies aimed at improving lung delivery while mitigating off-target effects. Although substantial progress has been made in lung-targeted mRNA therapies, challenges remain in optimizing cellular uptake and achieving therapeutic efficacy within pulmonary tissues. The continued refinement of delivery strategies that enhance lung-specific targeting while minimizing degradation is critical for the clinical success of mRNA-based pulmonary therapies.
Collapse
Affiliation(s)
- Brittany J. Heiser
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Arian Veyssi
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Debadyuti Ghosh
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX, USA
| |
Collapse
|
6
|
Yamada K, Suga K, Abe N, Hashimoto K, Tsutsumi S, Inagaki M, Hashiya F, Abe H, Hamada M. Multi-objective computational optimization of human 5' UTR sequences. Brief Bioinform 2025; 26:bbaf225. [PMID: 40413870 DOI: 10.1093/bib/bbaf225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 03/26/2025] [Accepted: 04/07/2025] [Indexed: 05/27/2025] Open
Abstract
The computational design of messenger RNA (mRNA) sequences is a critical technology for both scientific research and industrial applications. Recent advances in prediction and optimization models have enabled the automatic scoring and optimization of $5^\prime $ UTR sequences, key upstream elements of mRNA. However, fully automated design of $5^\prime $ UTR sequences with more than two objective scores has not yet been explored. In this study, we present a computational pipeline that optimizes human $5^\prime $ UTR sequences in a multi-objective framework, addressing up to four distinct and conflicting objectives. Our work represents an important advancement in the multi-objective computational design of mRNA sequences, paving the way for more sophisticated mRNA engineering.
Collapse
Affiliation(s)
- Keisuke Yamada
- Department of Electrical Engineering and Bioscience, Graduate School of Advanced Science and Engineering, Waseda University, 3-4-1, Okubo Shinjuku-ku, Tokyo 169-8555, Japan
- Department of Bioengineering, University of Pennsylvania, 210 South 33rd Street, Philadelphia, PA 19104, United States
| | - Kanta Suga
- Department of Electrical Engineering and Bioscience, Graduate School of Advanced Science and Engineering, Waseda University, 3-4-1, Okubo Shinjuku-ku, Tokyo 169-8555, Japan
| | - Naoko Abe
- Department of Chemistry, Graduate School of Science, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8602, Aichi, Japan
| | - Koji Hashimoto
- Department of Chemistry, Graduate School of Science, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8602, Aichi, Japan
- Graduate School of Arts and Sciences, The University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo 153-8902, Japan
| | - Susumu Tsutsumi
- Department of Chemistry, Graduate School of Science, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8602, Aichi, Japan
| | - Masahito Inagaki
- Department of Chemistry, Graduate School of Science, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8602, Aichi, Japan
| | - Fumitaka Hashiya
- Research Center for Materials Science, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8602, Aichi, Japan
| | - Hiroshi Abe
- Department of Chemistry, Graduate School of Science, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8602, Aichi, Japan
- Institute for Glyco-core Research (iGCORE), Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Aichi, Japan
| | - Michiaki Hamada
- Department of Electrical Engineering and Bioscience, Graduate School of Advanced Science and Engineering, Waseda University, 3-4-1, Okubo Shinjuku-ku, Tokyo 169-8555, Japan
- Cellular and Molecular Biotechnology Research Institute (CMB), National Institute of Advanced Industrial Science and Technology (AIST), 2-4-7, Aomi, Koto-ku, Tokyo 135-0064, Japan
- Graduate School of Medicine, Nippon Medical School, 1-1-5, Sendagi, Bunkyo-ku, Tokyo 113-8602, Japan
| |
Collapse
|
7
|
Zhang W, Wang C, Meng Y, He L, Dong M. EBV Vaccines in the Prevention and Treatment of Nasopharyngeal Carcinoma. Vaccines (Basel) 2025; 13:478. [PMID: 40432090 PMCID: PMC12115577 DOI: 10.3390/vaccines13050478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2025] [Revised: 04/21/2025] [Accepted: 04/22/2025] [Indexed: 05/29/2025] Open
Abstract
Epstein-Barr virus (EBV), a ubiquitous human herpesvirus, has been robustly linked to the pathogenesis of nasopharyngeal carcinoma (NPC). The mechanism of EBV-induced NPC involves complex interactions between viral proteins and host cell pathways. This review aims to comprehensively outline the mechanism of EBV-induced NPC and the latest advances in targeted EBV vaccines for prophylaxis and treatment. This review explores the intricate molecular mechanisms by which EBV contributes to NPC pathogenesis, highlighting viral latency, genetic and epigenetic alterations, and immune evasion strategies. It emphasizes the pivotal role of key viral proteins, including EBNA1, LMP1, and LMP2A, in carcinogenesis. Subsequently, the discussion shifts towards the development of targeted EBV vaccines, including preventive vaccines aimed at preventing primary EBV infection and therapeutic vaccines aimed at treating diagnosed EBV-related NPC. The review underscores the challenges and future directions in the field, stressing the importance of developing innovative vaccine strategies and combination therapies to improve efficacy. This review synthesizes current insights into the molecular mechanisms of EBV-induced NPC and the development of EBV-targeted vaccines, highlighting the potential use of mRNA vaccines for NPC treatment.
Collapse
Affiliation(s)
- Weiwei Zhang
- Department of Oncology, Cancer Prevention and Treatment Institute of Chengdu, Chengdu Fifth People’s Hospital/The Second Clinical Medical College, Affiliated Fifth People’s Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 611130, China; (W.Z.)
| | - Chuang Wang
- Chengdu Yunce Medical Biotechnology Co., Ltd., Chengdu 611135, China;
| | - Yousheng Meng
- Department of Oncology, Cancer Prevention and Treatment Institute of Chengdu, Chengdu Fifth People’s Hospital/The Second Clinical Medical College, Affiliated Fifth People’s Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 611130, China; (W.Z.)
| | - Lang He
- Department of Oncology, Cancer Prevention and Treatment Institute of Chengdu, Chengdu Fifth People’s Hospital/The Second Clinical Medical College, Affiliated Fifth People’s Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 611130, China; (W.Z.)
| | - Mingqing Dong
- Division of Pulmonary Medicine, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou Key Laboratory of Interdiscipline and Translational Medicine, Wenzhou Key Laboratory of Heart and Lung, Wenzhou 325000, China
| |
Collapse
|
8
|
Shen Y, Yang DQ, Liu Y, Lao JE, Liu CQ, Gao XH, He YR, Xia H. A review of advances in in vitro RNA preparation by ssRNAP. Int J Biol Macromol 2025; 304:141002. [PMID: 39952516 DOI: 10.1016/j.ijbiomac.2025.141002] [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: 09/06/2024] [Revised: 02/11/2025] [Accepted: 02/11/2025] [Indexed: 02/17/2025]
Abstract
In vitro transcription (IVT) based on single-subunit RNA polymerase (ssRNAP) has enhanced the widespread application of RNA drugs in the biomedical field, showcasing unprecedented potential for disease prevention and treatment. While the classical enzyme T7 RNA polymerase (T7 RNAP) has driven significant progress in RNA production, several challenges persist. These challenges include the selectivity of the initiation nucleotide, low incorporation efficiency of modified nucleotides, limited processivity on certain templates, heterogeneity at the 3' end of RNA products, and high level of double-stranded RNA (dsRNA) byproducts. No review has systematically addressed the efforts to overcome these challenges. To fill this gap, we reviewed recent advances in engineering T7 RNAP variants and the discovery of novel ssRNAPs aimed at addressing the shortcomings of T7 RNAP. We also discussed the underlying mechanisms of ssRNAP-mediated byproduct formation, strategies to mitigate dsRNA production using modified nucleotides, and for the first time to sorted out the application of artificial intelligence in IVT. Overall, this review summarizes the advances in RNA synthesis via IVT and provides potential strategies for improving RNA products. We believe that ssRNAPs with more excellent performance will be on the stage of RNA synthesis in the near future to meet the growing demands of both scientific research and pharmaceutical industry.
Collapse
Affiliation(s)
- Yuan Shen
- School of Medicine, Sun Yat-sen University, Shenzhen, Guangdong 518107, China
| | - Dong-Qi Yang
- School of Medicine, Sun Yat-sen University, Shenzhen, Guangdong 518107, China
| | - Yuan Liu
- School of Medicine, Sun Yat-sen University, Shenzhen, Guangdong 518107, China
| | - Jia-En Lao
- School of Medicine, Sun Yat-sen University, Shenzhen, Guangdong 518107, China
| | - Chun-Qing Liu
- Eesy Time (Shenzhen) Technology Co., LTD., Bao An District, Shenzhen 518101, China
| | - Xing-Hong Gao
- School of Basic Medicine, Zunyi Medical University, West No. 6 Xuefu Road, Xinpu District, Zunyi 563006, Guizhou, China.
| | - Yun-Ru He
- Scientific Research Center of The Seventh Affiliated Hospital, Sun Yat-sen University, No. 628, Zhenyuan Road, Guangming District, Shenzhen 518107, China.
| | - Heng Xia
- Scientific Research Center of The Seventh Affiliated Hospital, Sun Yat-sen University, No. 628, Zhenyuan Road, Guangming District, Shenzhen 518107, China.
| |
Collapse
|
9
|
Sun N, Su Z, Zheng X. Research progress of mosquito-borne virus mRNA vaccines. Mol Ther Methods Clin Dev 2025; 33:101398. [PMID: 39834558 PMCID: PMC11743085 DOI: 10.1016/j.omtm.2024.101398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
In recent years, mRNA vaccines have emerged as a leading technology for preventing infectious diseases due to their rapid development and high immunogenicity. These vaccines encode viral antigens, which are translated into antigenic proteins within host cells, inducing both humoral and cellular immune responses. This review systematically examines the progress in mRNA vaccine research for major mosquito-borne viruses, including dengue virus, Zika virus, Japanese encephalitis virus, Chikungunya virus, yellow fever virus, Rift Valley fever virus, and Venezuelan equine encephalitis virus. Enhancements in mRNA vaccine design, such as improvements to the 5' cap structure, 5'UTR, open reading frame, 3'UTR, and polyadenylation tail, have significantly increased mRNA stability and translation efficiency. Additionally, the use of lipid nanoparticles and polymer nanoparticles has greatly improved the delivery efficiency of mRNA vaccines. Currently, mRNA vaccines against mosquito-borne viruses are under development and clinical trials, showing promising protective effects. Future research should continue to optimize vaccine design and delivery systems to achieve broad-spectrum and long-lasting protection against various mosquito-borne virus infections.
Collapse
Affiliation(s)
- Ningze Sun
- Beijing Institute of Tropical Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory for Research on Prevention and Treatment of Tropical Diseases, Beijing, China
| | - Zhiwei Su
- Beijing Institute of Tropical Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory for Research on Prevention and Treatment of Tropical Diseases, Beijing, China
| | - Xiaoyan Zheng
- Beijing Institute of Tropical Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory for Research on Prevention and Treatment of Tropical Diseases, Beijing, China
| |
Collapse
|
10
|
Wei Z, Zhang S, Wang X, Xue Y, Dang S, Zhai J. Technological breakthroughs and advancements in the application of mRNA vaccines: a comprehensive exploration and future prospects. Front Immunol 2025; 16:1524317. [PMID: 40103818 PMCID: PMC11913674 DOI: 10.3389/fimmu.2025.1524317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 02/17/2025] [Indexed: 03/20/2025] Open
Abstract
mRNA vaccines utilize single-stranded linear DNA as a template for in vitro transcription. The mRNA is introduced into the cytoplasm via the corresponding delivery system to express the target protein, which then performs its relevant biological function. mRNA vaccines are beneficial in various fields, including cancer vaccines, infectious disease vaccines, protein replacement therapy, and treatment of rare diseases. They offer advantages such as a simple manufacturing process, a quick development cycle, and ease of industrialization. Additionally, mRNA vaccines afford flexibility in adjusting antigen designs and combining sequences of multiple variants, thereby addressing the issue of frequent mutations in pathogenic microorganisms. This paper aims to provide an extensive review of the global development and current research status of mRNA vaccines, with a focus on immunogenicity, classification, design, delivery vector development, stability, and biomedical application. Moreover, the study highlights current challenges and offers insights into future directions for development.
Collapse
Affiliation(s)
- Zhimeng Wei
- School of Basic Medical Sciences, Inner Mongolia Minzu University, Tongliao, China
- Keerqin District First People’s Hospital, Tongliao, China
| | - Shuai Zhang
- School of Basic Medical Sciences, Inner Mongolia Minzu University, Tongliao, China
| | - Xingya Wang
- School of Basic Medical Sciences, Inner Mongolia Minzu University, Tongliao, China
| | - Ying Xue
- Keerqin District First People’s Hospital, Tongliao, China
| | - Sheng Dang
- Keerqin District First People’s Hospital, Tongliao, China
| | - Jingbo Zhai
- School of Basic Medical Sciences, Inner Mongolia Minzu University, Tongliao, China
- Brucellosis Prevention and Treatment Engineering Research Center of Inner Mongolia Autonomous Region, Tongliao, China
- Key Laboratory of Zoonose Prevention and Control at Universities of Inner Mongolia Autonomous Region, Tongliao, China
| |
Collapse
|
11
|
Fatima M, An T, Hong KJ. Revolutionizing mRNA Vaccines Through Innovative Formulation and Delivery Strategies. Biomolecules 2025; 15:359. [PMID: 40149895 PMCID: PMC11940278 DOI: 10.3390/biom15030359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 02/12/2025] [Accepted: 02/19/2025] [Indexed: 03/29/2025] Open
Abstract
Modernization of existing methods for the delivery of mRNA is vital in advanced therapeutics. Traditionally, mRNA has faced obstacles of poor stability due to enzymatic degradation. This work examines cutting-edge formulation and emerging techniques for safer delivery of mRNA vaccines. Inspired by the success of lipid nanoparticles (LNP) in delivering mRNA vaccines for COVID-19, a variety of other formulations have been developed to deliver mRNA vaccines for diverse infections. The meritorious features of nanoparticle-based mRNA delivery strategies, including LNP, polymeric, dendrimers, polysaccharide-based, peptide-derived, carbon and metal-based, DNA nanostructures, hybrid, and extracellular vesicles, have been examined. The impact of these delivery platforms on mRNA vaccine delivery efficacy, protection from enzymatic degradation, cellular uptake, controlled release, and immunogenicity has been discussed in detail. Even with significant developments, there are certain limitations to overcome, including toxicity concerns, limited information about immune pathways, the need to maintain a cold chain, and the necessity of optimizing administration methods. Continuous innovation is essential for improving delivery systems for mRNA vaccines. Future research directions have been proposed to address the existing challenges in mRNA delivery and to expand their potential prophylactic and therapeutic application.
Collapse
Affiliation(s)
- Munazza Fatima
- Department of Microbiology, Gachon University College of Medicine, Incheon 21936, Republic of Korea;
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Republic of Korea
| | - Timothy An
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Republic of Korea
| | - Kee-Jong Hong
- Department of Microbiology, Gachon University College of Medicine, Incheon 21936, Republic of Korea;
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Republic of Korea
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Republic of Korea
- Korea mRNA Vaccine Initiative, Gachon University, Seongnam 13120, Republic of Korea
| |
Collapse
|
12
|
Xu Y, Gong F, Golubovic A, Strilchuk A, Chen J, Zhou M, Dong S, Seto B, Li B. Rational design and modular synthesis of biodegradable ionizable lipids via the Passerini reaction for mRNA delivery. Proc Natl Acad Sci U S A 2025; 122:e2409572122. [PMID: 39883839 PMCID: PMC11804478 DOI: 10.1073/pnas.2409572122] [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: 05/13/2024] [Accepted: 01/02/2025] [Indexed: 02/01/2025] Open
Abstract
The ionizable lipid component of lipid nanoparticle (LNP) formulations is essential for mRNA delivery by facilitating endosomal escape. Conventionally, these lipids are synthesized through complex, multistep chemical processes that are both time-consuming and require significant engineering. Furthermore, the development of new ionizable lipids is hindered by a limited understanding of the structure-activity relationships essential for effective mRNA delivery. In this work, we have developed a modular platform utilizing the Passerini reaction to rapidly generate large, chemically diverse libraries of biodegradable ionizable lipids. This high-throughput approach enables the systematic exploration of various lipid components-head groups, tails, and spacers-and their impacts on mRNA delivery efficiency. By investigating the hydrogen bonding potential between the lipid's head groups and the mRNA's ribose phosphate complex, we found that optimizing the methylene units between the lipid's head groups and linkages could enhance endosomal escape and, consequently, mRNA delivery efficiencies. Leveraging this insight, our platform has led to the identification of the biodegradable ionizable lipid A4B4-S3, which outperforms the current clinical benchmark, SM-102, in gene editing efficacy in mouse liver following systemic administration and demonstrates the promise for repeat-dose protein replacement treatments. This work not only offers a rapid, scalable method for ionizable lipid synthesis but also deepens our understanding of their structure-activity relationships, paving the way for more effective mRNA therapeutics.
Collapse
Affiliation(s)
- Yue Xu
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ONM5S 3M2, Canada
| | - Fanglin Gong
- Institute of Biomedical Engineering, University of Toronto, Toronto, ONM5S 3G9, Canada
| | - Alex Golubovic
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ONM5S 3M2, Canada
| | - Amy Strilchuk
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ONM5S 3M2, Canada
| | - Jingan Chen
- Institute of Biomedical Engineering, University of Toronto, Toronto, ONM5S 3G9, Canada
| | - Muye Zhou
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ONM5S 3M2, Canada
| | - Songtao Dong
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ONM5S 3M2, Canada
| | - Breanna Seto
- Department of Chemistry, University of Toronto, Toronto, ONM5S 3H6, Canada
| | - Bowen Li
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ONM5S 3M2, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, ONM5S 3G9, Canada
- Department of Chemistry, University of Toronto, Toronto, ONM5S 3H6, Canada
- Princess Margaret Cancer Center, University Health Network, Toronto, ONM5G 2C1, Canada
| |
Collapse
|
13
|
Fatima M, Park PG, Hong KJ. Clinical advancements in mRNA vaccines against viral infections. Clin Immunol 2025; 271:110424. [PMID: 39734036 DOI: 10.1016/j.clim.2024.110424] [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: 11/18/2024] [Revised: 12/15/2024] [Accepted: 12/24/2024] [Indexed: 12/31/2024]
Abstract
Over the last decade, mRNA vaccines development has shown significant advancement, particularly during the COVID-19 pandemic. This comprehensive review examines the efficacy of pivotal vaccines against emerging COVID-19 variants and strategies for enhancing vaccine effectiveness. It also explores the versatility of mRNA technology in addressing other infectious diseases such as influenza, respiratory syncytial virus, HIV, cytomegalovirus, Ebola, Zika, Rabies, and Nipah viruses. The analysis includes safety and clinical progress of mRNA vaccines and evaluates their potential in combination vaccine strategies. Additionally, it addresses challenges related to delivery and scalability while highlighting opportunities for future advancements in the field. Recent advances in mRNA optimization, biomaterial-based delivery and thermostable designs offer promising solutions. It is essential to gain insights into the evolving landscape of mRNA vaccine technology to maximize its vital role in addressing diverse viral threats, advancing vaccinology and enhancing public health preparedness for future pandemic.
Collapse
Affiliation(s)
- Munazza Fatima
- Department of Microbiology, Gachon University College of Medicine, Incheon, Republic of Korea; Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Republic of Korea
| | - Pil-Gu Park
- Department of Microbiology, Gachon University College of Medicine, Incheon, Republic of Korea; Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Republic of Korea
| | - Kee-Jong Hong
- Department of Microbiology, Gachon University College of Medicine, Incheon, Republic of Korea; Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Republic of Korea; Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, Republic of Korea; Korea mRNA Vaccine Initiative, Gachon University, Seongnam, Republic of Korea.
| |
Collapse
|
14
|
Chen H, Liu D, Guo J, Aditham A, Zhou Y, Tian J, Luo S, Ren J, Hsu A, Huang J, Kostas F, Wu M, Liu DR, Wang X. Branched chemically modified poly(A) tails enhance the translation capacity of mRNA. Nat Biotechnol 2025; 43:194-203. [PMID: 38519719 PMCID: PMC11416571 DOI: 10.1038/s41587-024-02174-7] [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: 04/25/2023] [Accepted: 02/15/2024] [Indexed: 03/25/2024]
Abstract
Although messenger RNA (mRNA) has proved effective as a vaccine, its potential as a general therapeutic modality is limited by its instability and low translation capacity. To increase the duration and level of protein expression from mRNA, we designed and synthesized topologically and chemically modified mRNAs with multiple synthetic poly(A) tails. Here we demonstrate that the optimized multitailed mRNA yielded ~4.7-19.5-fold higher luminescence signals than the control mRNA from 24 to 72 h post transfection in cellulo and 14 days detectable signal versus <7 days signal from the control in vivo. We further achieve efficient multiplexed genome editing of the clinically relevant genes Pcsk9 and Angptl3 in mouse liver at a minimal mRNA dosage. Taken together, these results provide a generalizable approach to synthesize capped branched mRNA with markedly enhanced translation capacity.
Collapse
Affiliation(s)
- Hongyu Chen
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Dangliang Liu
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jianting Guo
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Abhishek Aditham
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Yiming Zhou
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jiakun Tian
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Shuchen Luo
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jingyi Ren
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Alvin Hsu
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
| | - Jiahao Huang
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Franklin Kostas
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Mingrui Wu
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - David R Liu
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
| | - Xiao Wang
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, 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
|
Ahn JH, Lee J, Roh G, Lee NY, Bae HJ, Kwon E, Han KM, Kim JE, Park HJ, Yoo S, Kwon SP, Bang EK, Keum G, Nam JH, Kang BC. Impact of administration routes and dose frequency on the toxicology of SARS-CoV-2 mRNA vaccines in mice model. Arch Toxicol 2025; 99:755-773. [PMID: 39656241 PMCID: PMC11775000 DOI: 10.1007/s00204-024-03912-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 11/20/2024] [Indexed: 01/30/2025]
Abstract
The increasing use of SARS-CoV-2 mRNA vaccines has raised concerns about their potential toxicological effects, necessitating further investigation to ensure their safety. To address this issue, we aimed to evaluate the toxicological effects of SARS-CoV-2 mRNA vaccine candidates formulated with four different types of lipid nanoparticles in ICR mice, focusing on repeated doses and administration routes. We conducted an extensive analysis in which mice received the mRNA vaccine candidates intramuscularly (50 μg/head) twice at 2-week intervals, followed by necropsy at 2 and 14 dpsi (days post-secondary injection). In addition, we performed a repeated dose toxicity test involving three, four, or five doses and compared the toxicological outcomes between intravenous and intramuscular routes. Our findings revealed that all vaccine candidates significantly induced SARS-CoV-2 spike protein-specific IgG and T cell responses. However, at 2 dpsi, there was a notable temporary decrease in lymphocyte and reticulocyte counts, anemia-related parameters, and significant increases in cardiac damage markers, troponin-I and NT-proBNP. Histopathological analysis revealed severe inflammation and necrosis at the injection site, decreased erythroid cells in bone marrow, cortical atrophy of the thymus, and increased spleen cellularity. While most toxicological changes observed at 2 dpsi had resolved by 14 dpsi, spleen enlargement and injection site damage persisted. Furthermore, repeated doses led to the accumulation of toxicity, and different administration routes resulted in distinct toxicological phenotypes. These findings highlight the potential toxicological risks associated with mRNA vaccines, emphasizing the necessity to carefully consider administration routes and dosage regimens in vaccine safety evaluations, particularly given the presence of bone marrow and immune organ toxicity, which, though eventually reversible, remains a serious concern.
Collapse
Affiliation(s)
- Jae-Hun Ahn
- Department of Experiment Animal Research, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Jisun Lee
- Department of Medical and Biological Sciences, The Catholic University of Korea, Bucheon, Republic of Korea
| | - Gahyun Roh
- Department of Medical and Biological Sciences, The Catholic University of Korea, Bucheon, Republic of Korea
- BK Four Department of Biotechnology, The Catholic University of Korea, Bucheon, Republic of Korea
| | - Na-Young Lee
- Department of Experiment Animal Research, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Veterinary Pathology and Research Institute of Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Hee-Jin Bae
- Department of Experiment Animal Research, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Euna Kwon
- Department of Experiment Animal Research, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Kang-Min Han
- Graduate School of Translational Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Pathology, CHA Ilsan Medical Center, CHA University, Goyang-si, Republic of Korea
| | - Ji-Eun Kim
- Department of Experiment Animal Research, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Hyo-Jung Park
- Department of Medical and Biological Sciences, The Catholic University of Korea, Bucheon, Republic of Korea
- BK Four Department of Biotechnology, The Catholic University of Korea, Bucheon, Republic of Korea
| | - Soyeon Yoo
- Center for Brain Technology, Brain Science Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Sung Pil Kwon
- Center for Brain Technology, Brain Science Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Eun-Kyoung Bang
- Center for Brain Technology, Brain Science Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Gyochang Keum
- Center for Brain Technology, Brain Science Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Jae-Hwan Nam
- Department of Medical and Biological Sciences, The Catholic University of Korea, Bucheon, Republic of Korea.
- BK Four Department of Biotechnology, The Catholic University of Korea, Bucheon, Republic of Korea.
| | - Byeong-Cheol Kang
- Department of Experiment Animal Research, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea.
- Graduate School of Translational Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea.
- Department of Veterinary Pathology and Research Institute of Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
17
|
Boomgarden AC, Upadhyay C. Progress and Challenges in HIV-1 Vaccine Research: A Comprehensive Overview. Vaccines (Basel) 2025; 13:148. [PMID: 40006695 PMCID: PMC11860913 DOI: 10.3390/vaccines13020148] [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: 12/30/2024] [Revised: 01/20/2025] [Accepted: 01/28/2025] [Indexed: 02/27/2025] Open
Abstract
The development of an effective HIV-1 vaccine remains a formidable challenge in biomedical research. Despite significant advancements in our understanding of HIV biology and pathogenesis, progress has been impeded by factors such as the virus's genetic diversity, high mutation rates, and its ability to establish latent reservoirs. Recent innovative approaches, including mosaic vaccines and mRNA technology to induce broadly neutralizing antibodies, have shown promise. However, the efficacy of these vaccines has been modest, with the best results achieving approximately 30% effectiveness. Ongoing research emphasizes the necessity of a multifaceted strategy to overcome these obstacles and achieve a breakthrough in HIV-1 vaccine development. This review summarizes current approaches utilized to further understand HIV-1 biology and to create a global vaccine. We discuss the impact of these approaches on vaccine development for other diseases, including COVID-19, influenza, and Zika virus. Additionally, we highlight the specific limitations faced with each approach and present the methods researchers employ to overcome these challenges. These innovative techniques, which have demonstrated preclinical and clinical success, have advanced the field closer to the ultimate goal of developing a global HIV-1 vaccine. Leveraging these advancements will enable significant strides in combating HIV-1 and other infectious diseases, ultimately improving global health outcomes.
Collapse
Affiliation(s)
| | - Chitra Upadhyay
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
| |
Collapse
|
18
|
Wang Z, Tian C, Zhu J, Wang S, Ao X, He Y, Chen H, Liao X, Kong D, Zhou Y, Tai W, Liao M, Fan H. Avian influenza mRNA vaccine encoding hemagglutinin provides complete protection against divergent H5N1 viruses in specific-pathogen-free chickens. J Nanobiotechnology 2025; 23:55. [PMID: 39881325 PMCID: PMC11776166 DOI: 10.1186/s12951-025-03156-w] [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: 05/27/2024] [Accepted: 01/22/2025] [Indexed: 01/31/2025] Open
Abstract
BACKGROUND The rapid mutation of avian influenza virus (AIV) poses a significant threat to both the poultry industry and public health. Herein, we have successfully developed an mRNA-LNPs candidate vaccine for H5 subtype highly pathogenic avian influenza and evaluated its immunogenicity and protective efficacy. RESULTS In experiments on BALB/c mice, the vaccine candidate elicited strong humoral and a certain cellular immune responses and protected mice from the heterologous AIV challenge. Antibody and splenocyte passive transfer assays in mice suggested that antibodies played a crucial role in providing protection. Experiments involving SPF chickens have revealed that two doses of the 5 µg vaccine candidate in this study provided 100% complete protection against homologous strains, but only 50% complete protection against heterologous strains. Even immunization with two doses of the 15 µg vaccine candidate resulted in 90% complete protection against heterologous strains. To enhance the immune efficacy of the candidate vaccine, we designed 6 sequences with different secondary structures and screened out the candidate sequence with the highest expression (SY2-HA mRNA). Experiments on SPF chickens showed that two doses of 5 µg SY2-HA mRNA-LNP vaccine provided 100% complete protection against homologous and heterologous H5N1 AIV strains. Immunization tests with the SY2-HA mRNA-LNP vaccine were repeated in the SPF chicken model, inducing antibody production levels that are consistent with previous tests and providing 100% complete protection against both homologous and heterologous strains of the virus, indicating that the vaccine has a stable immune efficacy. CONCLUSIONS The vaccine developed in this study provides complete protection against divergent H5N1 AIV strains in chickens, offering a promising approach for the future development of mRNA vaccines against multivalent avian influenza subtypes.
Collapse
Affiliation(s)
- Zhaoyang Wang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou, 510642, China
- Key Laboratory of Veterinary Vaccine Innovation of the Ministry of Agriculture and Rural Affairs, Guangzhou, 510642, China
| | - Chongyu Tian
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen, 518132, China
| | - Jiahang Zhu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou, 510642, China
- Key Laboratory of Veterinary Vaccine Innovation of the Ministry of Agriculture and Rural Affairs, Guangzhou, 510642, China
| | - Shiqian Wang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou, 510642, China
- Key Laboratory of Veterinary Vaccine Innovation of the Ministry of Agriculture and Rural Affairs, Guangzhou, 510642, China
| | - Xiang Ao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou, 510642, China
- Key Laboratory of Veterinary Vaccine Innovation of the Ministry of Agriculture and Rural Affairs, Guangzhou, 510642, China
| | - Yanjuan He
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou, 510642, China
- Key Laboratory of Veterinary Vaccine Innovation of the Ministry of Agriculture and Rural Affairs, Guangzhou, 510642, China
| | - Huixin Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou, 510642, China
- Key Laboratory of Veterinary Vaccine Innovation of the Ministry of Agriculture and Rural Affairs, Guangzhou, 510642, China
| | - Xiuying Liao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou, 510642, China
- Key Laboratory of Veterinary Vaccine Innovation of the Ministry of Agriculture and Rural Affairs, Guangzhou, 510642, China
| | - Deming Kong
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou, 510642, China
- Key Laboratory of Veterinary Vaccine Innovation of the Ministry of Agriculture and Rural Affairs, Guangzhou, 510642, China
| | - Yongfei Zhou
- Institute of Hemu Biotechnology, Beijing Hemu Biotechnology Co., Ltd, Beijing, 102206, China
| | - Wanbo Tai
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen, 518132, China.
| | - Ming Liao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China.
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou, 510642, China.
- Key Laboratory of Veterinary Vaccine Innovation of the Ministry of Agriculture and Rural Affairs, Guangzhou, 510642, China.
| | - Huiying Fan
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China.
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou, 510642, China.
- Key Laboratory of Veterinary Vaccine Innovation of the Ministry of Agriculture and Rural Affairs, Guangzhou, 510642, China.
| |
Collapse
|
19
|
Kisakov DN, Karpenko LI, Kisakova LA, Sharabrin SV, Borgoyakova MB, Starostina EV, Taranov OS, Ivleva EK, Pyankov OV, Zaykovskaya AV, Dmitrienko EV, Yakovlev VA, Tigeeva EV, Bauer IA, Krasnikova SI, Rudometova NB, Rudometov AP, Sergeev AA, Ilyichev AA. Jet Injection of Naked mRNA Encoding the RBD of the SARS-CoV-2 Spike Protein Induces a High Level of a Specific Immune Response in Mice. Vaccines (Basel) 2025; 13:65. [PMID: 39852844 PMCID: PMC11769039 DOI: 10.3390/vaccines13010065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 12/26/2024] [Accepted: 01/03/2025] [Indexed: 01/26/2025] Open
Abstract
Background: Although mRNA vaccines encapsulated in lipid nanoparticles (LNPs) have demonstrated a safety profile with minimal serious adverse events in clinical trials, there is opportunity to further reduce mRNA reactogenicity. The development of naked mRNA vaccines could improve vaccine tolerability. Naked nucleic acid delivery using the jet injection method may be a solution. Methods: In the first part of the study, the optimal conditions providing low traumatization and high expression of the model mRNA-GFP molecule in the tissues of laboratory animals were determined. Then, we used the selected protocol to immunize BALB/c mice with mRNA-RBD encoding the SARS-CoV-2 receptor-binding domain (RBD). It was demonstrated that mice vaccinated with naked mRNA-RBD developed a high level of specific antibodies with virus-neutralizing activity. The vaccine also induced a strong RBD-specific T-cell response and reduced the viral load in the lungs of the animals after infection with the SARS-CoV-2 virus. The level of immune response in mice immunized with mRNA-RBD using a spring-loaded jet injector was comparable to that in animals immunized with mRNA-RBD encapsulated in LNPs. Results: In this study, the efficacy of an inexpensive, simple, and safe method of mRNA delivery using a spring-loaded jet injector was evaluated and validated. Conclusions: Our findings suggest that the jet injection method may be a possible alternative to LNPs for delivering mRNA vaccines against SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Denis N. Kisakov
- State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, World-Class Genomic Research Center for Biological Safety and Technological Independence, Federal Scientific and Technical Program on the Development of Genetic Technologies, 630559 Koltsovo, Russia; (L.I.K.); (L.A.K.); (S.V.S.); (M.B.B.); (E.V.S.); (O.S.T.); (E.K.I.); (O.V.P.); (A.V.Z.); (V.A.Y.); (E.V.T.); (S.I.K.); (N.B.R.); (A.P.R.); (A.A.S.); (A.A.I.)
| | - Larisa I. Karpenko
- State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, World-Class Genomic Research Center for Biological Safety and Technological Independence, Federal Scientific and Technical Program on the Development of Genetic Technologies, 630559 Koltsovo, Russia; (L.I.K.); (L.A.K.); (S.V.S.); (M.B.B.); (E.V.S.); (O.S.T.); (E.K.I.); (O.V.P.); (A.V.Z.); (V.A.Y.); (E.V.T.); (S.I.K.); (N.B.R.); (A.P.R.); (A.A.S.); (A.A.I.)
| | - Lyubov A. Kisakova
- State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, World-Class Genomic Research Center for Biological Safety and Technological Independence, Federal Scientific and Technical Program on the Development of Genetic Technologies, 630559 Koltsovo, Russia; (L.I.K.); (L.A.K.); (S.V.S.); (M.B.B.); (E.V.S.); (O.S.T.); (E.K.I.); (O.V.P.); (A.V.Z.); (V.A.Y.); (E.V.T.); (S.I.K.); (N.B.R.); (A.P.R.); (A.A.S.); (A.A.I.)
| | - Sergey V. Sharabrin
- State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, World-Class Genomic Research Center for Biological Safety and Technological Independence, Federal Scientific and Technical Program on the Development of Genetic Technologies, 630559 Koltsovo, Russia; (L.I.K.); (L.A.K.); (S.V.S.); (M.B.B.); (E.V.S.); (O.S.T.); (E.K.I.); (O.V.P.); (A.V.Z.); (V.A.Y.); (E.V.T.); (S.I.K.); (N.B.R.); (A.P.R.); (A.A.S.); (A.A.I.)
| | - Mariya B. Borgoyakova
- State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, World-Class Genomic Research Center for Biological Safety and Technological Independence, Federal Scientific and Technical Program on the Development of Genetic Technologies, 630559 Koltsovo, Russia; (L.I.K.); (L.A.K.); (S.V.S.); (M.B.B.); (E.V.S.); (O.S.T.); (E.K.I.); (O.V.P.); (A.V.Z.); (V.A.Y.); (E.V.T.); (S.I.K.); (N.B.R.); (A.P.R.); (A.A.S.); (A.A.I.)
| | - Ekaterina V. Starostina
- State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, World-Class Genomic Research Center for Biological Safety and Technological Independence, Federal Scientific and Technical Program on the Development of Genetic Technologies, 630559 Koltsovo, Russia; (L.I.K.); (L.A.K.); (S.V.S.); (M.B.B.); (E.V.S.); (O.S.T.); (E.K.I.); (O.V.P.); (A.V.Z.); (V.A.Y.); (E.V.T.); (S.I.K.); (N.B.R.); (A.P.R.); (A.A.S.); (A.A.I.)
| | - Oleg S. Taranov
- State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, World-Class Genomic Research Center for Biological Safety and Technological Independence, Federal Scientific and Technical Program on the Development of Genetic Technologies, 630559 Koltsovo, Russia; (L.I.K.); (L.A.K.); (S.V.S.); (M.B.B.); (E.V.S.); (O.S.T.); (E.K.I.); (O.V.P.); (A.V.Z.); (V.A.Y.); (E.V.T.); (S.I.K.); (N.B.R.); (A.P.R.); (A.A.S.); (A.A.I.)
| | - Elena K. Ivleva
- State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, World-Class Genomic Research Center for Biological Safety and Technological Independence, Federal Scientific and Technical Program on the Development of Genetic Technologies, 630559 Koltsovo, Russia; (L.I.K.); (L.A.K.); (S.V.S.); (M.B.B.); (E.V.S.); (O.S.T.); (E.K.I.); (O.V.P.); (A.V.Z.); (V.A.Y.); (E.V.T.); (S.I.K.); (N.B.R.); (A.P.R.); (A.A.S.); (A.A.I.)
| | - Oleg V. Pyankov
- State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, World-Class Genomic Research Center for Biological Safety and Technological Independence, Federal Scientific and Technical Program on the Development of Genetic Technologies, 630559 Koltsovo, Russia; (L.I.K.); (L.A.K.); (S.V.S.); (M.B.B.); (E.V.S.); (O.S.T.); (E.K.I.); (O.V.P.); (A.V.Z.); (V.A.Y.); (E.V.T.); (S.I.K.); (N.B.R.); (A.P.R.); (A.A.S.); (A.A.I.)
| | - Anna V. Zaykovskaya
- State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, World-Class Genomic Research Center for Biological Safety and Technological Independence, Federal Scientific and Technical Program on the Development of Genetic Technologies, 630559 Koltsovo, Russia; (L.I.K.); (L.A.K.); (S.V.S.); (M.B.B.); (E.V.S.); (O.S.T.); (E.K.I.); (O.V.P.); (A.V.Z.); (V.A.Y.); (E.V.T.); (S.I.K.); (N.B.R.); (A.P.R.); (A.A.S.); (A.A.I.)
| | - Elena V. Dmitrienko
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch, Russian Academy of Sciences, 630090 Novosibirsk, Russia; (E.V.D.); (I.A.B.)
| | - Vladimir A. Yakovlev
- State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, World-Class Genomic Research Center for Biological Safety and Technological Independence, Federal Scientific and Technical Program on the Development of Genetic Technologies, 630559 Koltsovo, Russia; (L.I.K.); (L.A.K.); (S.V.S.); (M.B.B.); (E.V.S.); (O.S.T.); (E.K.I.); (O.V.P.); (A.V.Z.); (V.A.Y.); (E.V.T.); (S.I.K.); (N.B.R.); (A.P.R.); (A.A.S.); (A.A.I.)
| | - Elena V. Tigeeva
- State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, World-Class Genomic Research Center for Biological Safety and Technological Independence, Federal Scientific and Technical Program on the Development of Genetic Technologies, 630559 Koltsovo, Russia; (L.I.K.); (L.A.K.); (S.V.S.); (M.B.B.); (E.V.S.); (O.S.T.); (E.K.I.); (O.V.P.); (A.V.Z.); (V.A.Y.); (E.V.T.); (S.I.K.); (N.B.R.); (A.P.R.); (A.A.S.); (A.A.I.)
| | - Irina Alekseevna Bauer
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch, Russian Academy of Sciences, 630090 Novosibirsk, Russia; (E.V.D.); (I.A.B.)
| | - Svetlana I. Krasnikova
- State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, World-Class Genomic Research Center for Biological Safety and Technological Independence, Federal Scientific and Technical Program on the Development of Genetic Technologies, 630559 Koltsovo, Russia; (L.I.K.); (L.A.K.); (S.V.S.); (M.B.B.); (E.V.S.); (O.S.T.); (E.K.I.); (O.V.P.); (A.V.Z.); (V.A.Y.); (E.V.T.); (S.I.K.); (N.B.R.); (A.P.R.); (A.A.S.); (A.A.I.)
| | - Nadezhda B. Rudometova
- State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, World-Class Genomic Research Center for Biological Safety and Technological Independence, Federal Scientific and Technical Program on the Development of Genetic Technologies, 630559 Koltsovo, Russia; (L.I.K.); (L.A.K.); (S.V.S.); (M.B.B.); (E.V.S.); (O.S.T.); (E.K.I.); (O.V.P.); (A.V.Z.); (V.A.Y.); (E.V.T.); (S.I.K.); (N.B.R.); (A.P.R.); (A.A.S.); (A.A.I.)
| | - Andrey P. Rudometov
- State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, World-Class Genomic Research Center for Biological Safety and Technological Independence, Federal Scientific and Technical Program on the Development of Genetic Technologies, 630559 Koltsovo, Russia; (L.I.K.); (L.A.K.); (S.V.S.); (M.B.B.); (E.V.S.); (O.S.T.); (E.K.I.); (O.V.P.); (A.V.Z.); (V.A.Y.); (E.V.T.); (S.I.K.); (N.B.R.); (A.P.R.); (A.A.S.); (A.A.I.)
| | - Artemiy A. Sergeev
- State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, World-Class Genomic Research Center for Biological Safety and Technological Independence, Federal Scientific and Technical Program on the Development of Genetic Technologies, 630559 Koltsovo, Russia; (L.I.K.); (L.A.K.); (S.V.S.); (M.B.B.); (E.V.S.); (O.S.T.); (E.K.I.); (O.V.P.); (A.V.Z.); (V.A.Y.); (E.V.T.); (S.I.K.); (N.B.R.); (A.P.R.); (A.A.S.); (A.A.I.)
| | - Alexander A. Ilyichev
- State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, World-Class Genomic Research Center for Biological Safety and Technological Independence, Federal Scientific and Technical Program on the Development of Genetic Technologies, 630559 Koltsovo, Russia; (L.I.K.); (L.A.K.); (S.V.S.); (M.B.B.); (E.V.S.); (O.S.T.); (E.K.I.); (O.V.P.); (A.V.Z.); (V.A.Y.); (E.V.T.); (S.I.K.); (N.B.R.); (A.P.R.); (A.A.S.); (A.A.I.)
| |
Collapse
|
20
|
Rahman MM, Masum MHU, Parvin R, Das SC, Talukder A. Designing of an mRNA vaccine against high-risk human papillomavirus targeting the E6 and E7 oncoproteins exploiting immunoinformatics and dynamic simulation. PLoS One 2025; 20:e0313559. [PMID: 39761277 PMCID: PMC11703113 DOI: 10.1371/journal.pone.0313559] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 10/25/2024] [Indexed: 05/01/2025] Open
Abstract
Human papillomavirus 16 and human papillomavirus 18 have been associated with different life-threatening cancers, including cervical, lung, penal, vulval, vaginal, anal, and oropharyngeal cancers, while cervical cancer is the most prominent one. Several research studies have suggested that the oncoproteins E6 and E7 are the leading cause of cancers associated with the human papillomavirus infection. Therefore, we developed two mRNA vaccines (V1 and V2) targeting these oncoproteins. We used several bioinformatics tools to predict helper T lymphocyte, cytotoxic T lymphocyte, and B-cell epitopes derived from the proteins and assessed their antigenicity, allergenicity, and toxicity. Both vaccines were constructed using selected epitopes, linkers, and adjuvants. After that, the vaccines were applied for physicochemical properties, secondary and tertiary structure predictions, and subsequent docking and simulation analyses. Accordingly, vaccine 1 (V1) and vaccine 2 (V2) showed better hydrophilicity with the grand average hydropathicity score of -0.811 and -0.648, respectively. The secondary and tertiary structures of the vaccines were also deemed satisfactory, with high stability indicated by the Ramachandran plot (V1:94.5% and V2:87.1%) and Z scores (V1: -5.15 and V2: -4.1). Docking analysis revealed substantial affinity of the vaccines towards the toll-like receptor-2 (V1: -1159.3, V2: -1246.3) and toll-like receptor-4 (V1: -1109.3, V2: -1244.8) receptors. Molecular dynamic simulation validated structural integrity and indicated varying stability throughout the simulation. Codon optimization showed significant expression of the vaccines (V1:51.88% and V2:51.63%) in E. coli vectors. Furthermore, regarding immune stimulation, the vaccines elicited significant B-cell and T-cell responses, including sustained adaptive and innate immune responses. Finally, thermodynamic predictions indicated stable mRNA structures of the vaccines (V1: -502.60 kcal/mol and V2: -450.90 kcal/mol). The proposed vaccines designed effectively targeting human papillomavirus oncoproteins have demonstrated promising results via robust immune responses, suggesting their suitability for further clinical advancement, including in vitro and in vivo experiments.
Collapse
Affiliation(s)
- Md. Mijanur Rahman
- Department of Microbiology, Noakhali Science and Technology University, Noakhali, Bangladesh
- Microbiology, Cancer and Bioinformatics Research Group, Noakhali Science and Technology University, Noakhali, Bangladesh
- School of Pharmacy and Medical Sciences, Griffith University, Queensland, Australia
| | - Md. Habib Ullah Masum
- Department of Microbiology, Noakhali Science and Technology University, Noakhali, Bangladesh
- Microbiology, Cancer and Bioinformatics Research Group, Noakhali Science and Technology University, Noakhali, Bangladesh
- Department of Genomics and Bioinformatics, Faculty of Biotechnology and Genetic Engineering, Chattogram Veterinary and Animal Sciences University, Khulshi, Chittagong, Bangladesh
| | - Rehana Parvin
- Department of Pathology and Parasitology, Faculty of Veterinary Medicine, Chattogram Veterinary and Animal Sciences University, Khulshi, Chittagong, Bangladesh
| | - Shuvo Chandra Das
- Department of Biotechnology and Genetic Engineering, Noakhali Science and Technology University, Noakhali, Bangladesh
| | - Asma Talukder
- Microbiology, Cancer and Bioinformatics Research Group, Noakhali Science and Technology University, Noakhali, Bangladesh
- School of Pharmacy and Medical Sciences, Griffith University, Queensland, Australia
- Department of Biotechnology and Genetic Engineering, Noakhali Science and Technology University, Noakhali, Bangladesh
| |
Collapse
|
21
|
Xu L, Li C, Liao R, Xiao Q, Wang X, Zhao Z, Zhang W, Ding X, Cao Y, Cai L, Rosenecker J, Guan S, Tang J. From Sequence to System: Enhancing IVT mRNA Vaccine Effectiveness through Cutting-Edge Technologies. Mol Pharm 2025; 22:81-102. [PMID: 39601789 DOI: 10.1021/acs.molpharmaceut.4c00863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
The COVID-19 pandemic has spotlighted the potential of in vitro transcribed (IVT) mRNA vaccines with their demonstrated efficacy, safety, cost-effectiveness, and rapid manufacturing. Numerous IVT mRNA vaccines are now under clinical trials for a range of targets, including infectious diseases, cancers, and genetic disorders. Despite their promise, IVT mRNA vaccines face hurdles such as limited expression levels, nonspecific targeting beyond the liver, rapid degradation, and unintended immune activation. Overcoming these challenges is crucial to harnessing the full therapeutic potential of IVT mRNA vaccines for global health advancement. This review provides a comprehensive overview of the latest research progress and optimization strategies for IVT mRNA molecules and delivery systems, including the application of artificial intelligence (AI) models and deep learning techniques for IVT mRNA structure optimization and mRNA delivery formulation design. We also discuss recent development of the delivery platforms, such as lipid nanoparticles (LNPs), polymers, and exosomes, which aim to address challenges related to IVT mRNA protection, cellular uptake, and targeted delivery. Lastly, we offer insights into future directions for improving IVT mRNA vaccines, with the hope to spur further progress in IVT mRNA vaccine research and development.
Collapse
Affiliation(s)
- Lifeng Xu
- National Engineering Research Center of Immunological Products, Third Military Medical University, Chongqing 400038, China
| | - Chao Li
- National Engineering Research Center of Immunological Products, Third Military Medical University, Chongqing 400038, China
| | - Rui Liao
- National Engineering Research Center of Immunological Products, Third Military Medical University, Chongqing 400038, China
| | - Qin Xiao
- National Engineering Research Center of Immunological Products, Third Military Medical University, Chongqing 400038, China
| | - Xiaoran Wang
- Department of Pharmacy, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830000, China
| | - Zhuo Zhao
- National Engineering Research Center of Immunological Products, Third Military Medical University, Chongqing 400038, China
| | - Weijun Zhang
- National Engineering Research Center of Immunological Products, Third Military Medical University, Chongqing 400038, China
| | - Xiaoyan Ding
- Department of Pediatrics, Ludwig-Maximilians University of Munich, Munich 80337, Germany
| | - Yuxue Cao
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Larry Cai
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Joseph Rosenecker
- Department of Pediatrics, Ludwig-Maximilians University of Munich, Munich 80337, Germany
| | - Shan Guan
- National Engineering Research Center of Immunological Products, Third Military Medical University, Chongqing 400038, China
| | - Jie Tang
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| |
Collapse
|
22
|
Parr MK, Keiler AM. Oligonucleotide therapeutics in sports? An antidoping perspective. Arch Pharm (Weinheim) 2025; 358:e2400404. [PMID: 39449227 PMCID: PMC11704058 DOI: 10.1002/ardp.202400404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 09/26/2024] [Accepted: 09/30/2024] [Indexed: 10/26/2024]
Abstract
Within the last two decades, the European Medicines Agency and the US Food and Drug Administration have approved several gene therapies. One category is oligonucleotide therapeutics, which allow for the regulation of the expression of target genes. Besides already approved therapeutics, there are several preclinical and clinical trials ongoing. The World Anti-Doping Agency prohibits the use of "nucleic acids or nucleic acid analogs that may alter genome sequences and/or alter gene expression by any mechanism" as a nonspecified method at all times. Hence, the administration of nucleic acids or analogs by athletes would cause an Anti-Doping Rule Violation. Herein, we discuss types of oligonucleotide therapeutics, their potential to be misused in sports, and considerations to sample preparation and mass spectrometric approaches with regard to antidoping analysis.
Collapse
Affiliation(s)
- Maria K. Parr
- Institute of Pharmacy, Pharmaceutical and Medicinal ChemistryFreie Universität BerlinBerlinGermany
| | - Annekathrin M. Keiler
- Institute of Doping Analysis & Sports BiochemistryKreischaGermany
- Environmental Monitoring & Endocrinology, Faculty of BiologyTechnische Universität DresdenDresdenGermany
| |
Collapse
|
23
|
Shariati A, Khani P, Nasri F, Afkhami H, Khezrpour A, Kamrani S, Shariati F, Alavimanesh S, Modarressi MH. mRNA cancer vaccines from bench to bedside: a new era in cancer immunotherapy. Biomark Res 2024; 12:157. [PMID: 39696625 DOI: 10.1186/s40364-024-00692-9] [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: 09/10/2024] [Accepted: 11/15/2024] [Indexed: 12/20/2024] Open
Abstract
Harnessing the power of the immune system to target cancer cells is one of the most appealing approaches for cancer therapy. Among these immunotherapies, messenger ribonucleic acid (mRNA) cancer vaccines are worthy of consideration, as they have demonstrated promising results in clinical trials. These vaccines have proven to be safe and well-tolerated. They can be easily mass-produced in a relatively short time and induce a systemic immune response effective against both the primary tumor and metastases. Transcripts encoding immunomodulatory molecules can also be incorporated into the mRNA, enhancing its efficacy. On the other hand, there are some challenges associated with their application, including mRNA instability, insufficient uptake by immune cells, and intrinsic immunogenicity, which can block mRNA translation. Many innovations have been suggested to overcome these obstacles, including structural modification (such as 5' cap modification), optimizing delivery vehicles (especially dendritic cells (DCs) and nanoparticles), and using antigens that can enhance immunogenicity by circumventing tolerance mechanisms. A popular approach is to combine mRNA cancer vaccines with traditional and novel cancer treatments like chemotherapy, radiotherapy, and immune checkpoint blockade (ICB). They are most efficacious when combined with other therapies like ICBs. There is still a long way to go before these vaccines enter the standard of care for cancer patients, but with the incredible pace of development in this field, their clinical application will soon be witnessed. This review highlights the recent advances and challenges of mRNA cancer vaccines. Finally, some of the most prominent clinical applications of these vaccines will be reviewed.
Collapse
Affiliation(s)
- Alireza Shariati
- School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Pouria Khani
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Farzad Nasri
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Hamed Afkhami
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran
- Department of Medical Microbiology, Faculty of Medicine, Shahed University, Tehran, Iran
| | - Arya Khezrpour
- School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Sina Kamrani
- Department of Orthopedic, Faculty of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Fatemeh Shariati
- Department of Genetics, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Sajad Alavimanesh
- Student Research Committee, Shahrekord University of Medical Sciences, Shahrekord, Iran.
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran.
| | - Mohammad Hossein Modarressi
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran.
| |
Collapse
|
24
|
Cheng Z, Ma J, Zhao C. Advantages of Broad-Spectrum Influenza mRNA Vaccines and Their Impact on Pulmonary Influenza. Vaccines (Basel) 2024; 12:1382. [PMID: 39772044 PMCID: PMC11680418 DOI: 10.3390/vaccines12121382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 11/28/2024] [Accepted: 12/06/2024] [Indexed: 01/11/2025] Open
Abstract
Influenza poses a significant global health challenge due to its rapid mutation and antigenic variability, which often leads to seasonal epidemics and frequent outbreaks. Traditional vaccines struggle to offer comprehensive protection because of mismatches with circulating viral strains. The development of a broad-spectrum vaccine is therefore crucial. This paper explores the potential of mRNA vaccine technology to address these challenges by providing a swift, adaptable, and broad protective response against evolving influenza strains. We detail the mechanisms of antigenic variation in influenza viruses and discuss the rapid design and production, enhanced immunogenicity, encoding of multiple antigens, and safety and stability of mRNA vaccines compared to traditional methods. By leveraging these advantages, mRNA vaccines represent a revolutionary approach in influenza prevention, potentially offering broad-spectrum protection and significantly improving global influenza management and response strategies.
Collapse
Affiliation(s)
- Ziqi Cheng
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China;
- Division of HIV/AIDS and Sex-Transmitted Virus Vaccines, National Institutes for Food and Drug Control (NIFDC), Beijing 102629, China
| | - Junfeng Ma
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China;
| | - Chenyan Zhao
- Division of HIV/AIDS and Sex-Transmitted Virus Vaccines, National Institutes for Food and Drug Control (NIFDC), Beijing 102629, China
| |
Collapse
|
25
|
Fan P, Sun B, Liu Z, Fang T, Ren Y, Zhao X, Song Z, Yang Y, Li J, Yu C, Chen W. A pan-orthoebolavirus neutralizing antibody encoded by mRNA effectively prevents virus infection. Emerg Microbes Infect 2024; 13:2432366. [PMID: 39560055 PMCID: PMC11590195 DOI: 10.1080/22221751.2024.2432366] [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: 09/09/2024] [Revised: 10/24/2024] [Accepted: 11/17/2024] [Indexed: 11/20/2024]
Abstract
Orthoebolavirus is a genus of hazardous pathogens that has caused over 30 outbreaks. However, currently approved therapies are limited in scope, as they are only effective against the Ebola virus and lack cross-protection against other orthoebolaviruses. Here, we demonstrate that a previously isolated human-derived antibody, 2G1, can recognize the glycoprotein (GP) of every orthoebolavirus species. The cryo-electron microscopy structure of 2G1 Fab in complex with the GPΔMucin trimer reveals that 2G1 binds a quaternary pocket formed by three subunits from two GP protomers. 2G1 recognizes highly conserved epitopes among filoviruses and achieves neutralization by blocking GP proteolysis. We designed an efficient mRNA module capable of producing test antibodies at expression levels exceeding 1500 ng/mL in vitro. The lipid nanoparticle (LNP)-encapsulated mRNA-2G1 exhibited potent neutralizing activities against the HIV-pseudotyped Ebola and Sudan viruses that were 19.8 and 12.5 times that of IgG format, respectively. In mice, the antibodies encoded by the mRNA-2G1-LNP peaked within 24 h, effectively blocking the invasion of pseudoviruses with no apparent liver toxicity. This study suggests that the 2G1 antibody and its mRNA formulation represent promising candidate interventions for orthoebolavirus disease, and it provides an efficient mRNA framework applicable to antibody-based therapies.
Collapse
Affiliation(s)
- Pengfei Fan
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, People’s Republic of China
| | - Bingjie Sun
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, People’s Republic of China
| | - Zixuan Liu
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, People’s Republic of China
| | - Ting Fang
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, People’s Republic of China
| | - Yi Ren
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, People’s Republic of China
| | - Xiaofan Zhao
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, People’s Republic of China
| | - Zhenwei Song
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, People’s Republic of China
| | - Yilong Yang
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, People’s Republic of China
| | - Jianmin Li
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, People’s Republic of China
| | - Changming Yu
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, People’s Republic of China
| | - Wei Chen
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, People’s Republic of China
| |
Collapse
|
26
|
Lloren KKS, Senevirathne A, Lee JH. Advancing vaccine technology through the manipulation of pathogenic and commensal bacteria. Mater Today Bio 2024; 29:101349. [PMID: 39850273 PMCID: PMC11754135 DOI: 10.1016/j.mtbio.2024.101349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 10/30/2024] [Accepted: 11/15/2024] [Indexed: 01/25/2025] Open
Abstract
Advancements in vaccine technology are increasingly focused on leveraging the unique properties of both pathogenic and commensal bacteria. This revolutionary approach harnesses the diverse immune modulatory mechanisms and bacterial biology inherent in different bacterial species enhancing vaccine efficacy and safety. Pathogenic bacteria, known for their ability to induce robust immune responses, are being studied for their potential to be engineered into safe, attenuated vectors that can target specific diseases with high precision. Concurrently, commensal bacteria, which coexist harmlessly with their hosts and contribute to immune system regulation, are also being explored as novel delivery systems and in microbiome-based therapy. These bacteria can modulate immune responses, offering a promising avenue for developing effective and personalized vaccines. Integrating the distinctive characteristics of pathogenic and commensal bacteria with advanced bacterial engineering techniques paves the way for innovative vaccine and therapeutic platforms that could address a wide range of infectious diseases and potentially non-infectious conditions. This holistic approach signifies a paradigm shift in vaccine development and immunotherapy, emphasizing the intricate interplay between the bacteria and the immune systems to achieve optimal immunological outcomes.
Collapse
Affiliation(s)
- Khristine Kaith S. Lloren
- College of Veterinary Medicine, Jeonbuk National University, 79 Gobong-ro, Iksan City, Jeollabuk-do, 54596, Republic of Korea
| | - Amal Senevirathne
- College of Veterinary Medicine, Jeonbuk National University, 79 Gobong-ro, Iksan City, Jeollabuk-do, 54596, Republic of Korea
| | - John Hwa Lee
- College of Veterinary Medicine, Jeonbuk National University, 79 Gobong-ro, Iksan City, Jeollabuk-do, 54596, Republic of Korea
| |
Collapse
|
27
|
Seephetdee C, Kiss DL. Codon optimality modulates cellular stress and innate immune responses triggered by exogenous RNAs. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.26.625518. [PMID: 39651201 PMCID: PMC11623643 DOI: 10.1101/2024.11.26.625518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
The COVID-19 mRNA vaccines demonstrated the power of mRNA medicines. Despite advancements in sequence design, evidence regarding the preferential use of synonymous codons on cellular stress and innate immune responses is lacking. To this end, we developed a proprietary codon optimality matrix to re-engineer the coding sequences of three luciferase reporters. We demonstrate that optimal mRNAs elicited dramatic increases in luciferase activities compared to non-optimal sequences. Notably, transfecting an optimal RNA affects the translation of other RNAs in the cell including control transcripts in dual luciferase assays. This held true in multiple cell lines and for an unrelated reporter. Further, non-optimal mRNAs preferentially activated innate immune pathways and the phosphorylation of the translation initiation factor eIF2α, a central event of the integrated stress response. Using nucleoside-modified or circular RNAs partially or fully abrogated these responses. Finally, we show that circularizing RNAs enhances both RNA lifespan and durability of protein expression. Our results show that RNA sequence, composition, and structure all govern RNA translatability. However, we also show that RNA sequences with poor codon optimality are immunogenic and induce cellular stress. Hence, RNA sequence engineering, chemical, and topological modifications must all be combined to elicit favorable therapeutic outcomes.
Collapse
|
28
|
Laila UE, An W, Xu ZX. Emerging prospects of mRNA cancer vaccines: mechanisms, formulations, and challenges in cancer immunotherapy. Front Immunol 2024; 15:1448489. [PMID: 39654897 PMCID: PMC11625737 DOI: 10.3389/fimmu.2024.1448489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 10/18/2024] [Indexed: 12/12/2024] Open
Abstract
Cancer continues to pose an alarming threat to global health, necessitating the need for the development of efficient therapeutic solutions despite massive advances in the treatment. mRNA cancer vaccines have emerged as a hopeful avenue, propelled by the victory of mRNA technology in COVID-19 vaccines. The article delves into the intricate mechanisms and formulations of cancer vaccines, highlighting the ongoing efforts to strengthen mRNA stability and ensure successful translation inside target cells. Moreover, it discusses the design and mechanism of action of mRNA, showcasing its potential as a useful benchmark for developing efficacious cancer vaccines. The significance of mRNA therapy and selecting appropriate tumor antigens for the personalized development of mRNA vaccines are emphasized, providing insights into the immune mechanism. Additionally, the review explores the integration of mRNA vaccines with other immunotherapies and the utilization of progressive delivery platforms, such as lipid nanoparticles, to improve immune responses and address challenges related to immune evasion and tumor heterogeneity. While underscoring the advantages of mRNA vaccines, the review also addresses the challenges associated with the susceptibility of RNA to degradation and the difficulty in identifying optimum tumor-specific antigens, along with the potential solutions. Furthermore, it provides a comprehensive overview of the ongoing research efforts aimed at addressing these hurdles and enhancing the effectiveness of mRNA-based cancer vaccines. Overall, this review is a focused and inclusive impression of the present state of mRNA cancer vaccines, outlining their possibilities, challenges, and future predictions in the fight against cancer, ultimately aiding in the development of more targeted therapies against cancer.
Collapse
Affiliation(s)
| | | | - Zhi-Xiang Xu
- School of Life Sciences, Henan University, Kaifeng, Henan, China
| |
Collapse
|
29
|
Barazesh M, Abbasi M, Mohammadi M, Nasiri MN, Rezaei F, Mohammadi S, Kavousipour S. Bioinformatics analysis to design a multi-epitope mRNA vaccine against S. agalactiae exploiting pathogenic proteins. Sci Rep 2024; 14:28294. [PMID: 39550419 PMCID: PMC11569170 DOI: 10.1038/s41598-024-79503-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 11/11/2024] [Indexed: 11/18/2024] Open
Abstract
Antibiotic resistance in bacterial pathogen infections is a growing global issue that occurs due to their adaptation to changing environmental conditions. Therefore, producing an efficient vaccine as an alternative approach can improve the immune system, eradicate related pathogens, and overcome this growing problem. Streptococcus agalactiae belongs to group B Streptococcus (GBS). Colonization of GBS during pregnancy is a significant risk factor for infants and young children. S. agalactiae infected population exhibits resistance to beta-lactams, including penicillin and the second-line antibiotics erythromycin and clindamycin. On the other hand, there are currently no commercial vaccines against this pathogen. Vaccination of pregnant women is a highly effective method to protect newborns and infants from S. agalactiae infection, and it has been identified as an urgent demand by the World Health Organization. This study employed various immunoinformatic tools to develop an effective vaccine that could trigger both humoral and cell-mediated immunity and prevent disease. For this purpose, three conserved antigenic proteins of the main pathogenic strains of S. agalactiae were utilized to predict CTL, HTL, and B-cell epitopes for producing an mRNA vaccine against different strains of S. agalactiae. The selected epitopes were fused using proper linkers. The Resuscitation promoting factor E (RpfE) sequence was incorporated in the designed vaccine construct as an adjuvant to boost its immune response. Different physicochemical characteristics of the final designed vaccine, modeling of the three-dimensional structure, molecular docking, molecular dynamics simulation, and immunological response simulation were screened following vaccine administration in an in vivo model. Computational immune simulation data identified that IgG1, IgM, INF γ, IL-2, T helper, and B-cell populations increased significantly after vaccination. These findings suggested that the vaccine candidate may provide good protection against S. agalactiae infection. However, experimental and animal model studies are required for additional validation and implementation in human vaccination programs.
Collapse
Affiliation(s)
- Mahdi Barazesh
- Department of Medical Biotechnology, School of Paramedical, Gerash University of Medical Sciences, Gerash, Iran
| | - Maryam Abbasi
- Endocrinology and Metabolism Research Center, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
- Department of Medicinal Chemistry, Faculty of Pharmacy, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Mohsen Mohammadi
- Hepatitis Research Center and Department of Pharmacognosy and Pharmaceutical Biotechnology, Faculty of Pharmacy, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Mohammad Naser Nasiri
- Department of Clinical pharmacy, School of Pharmacy, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Faranak Rezaei
- Razi Herbal Medicines Research Center, School of Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Shiva Mohammadi
- Hepatitis Research Center, Department of Medical Biotechnology, School of Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran.
| | - Soudabeh Kavousipour
- Molecular Medicine Research Center, Hormozgan Health Institute, Hormozgan University of Medical Sciences, BandarAbbas, Iran
| |
Collapse
|
30
|
Qin Q, Yan H, Gao W, Cao R, Liu G, Zhang X, Wang N, Zuo W, Yuan L, Gao P, Liu Q. Engineered mRNAs With Stable Structures Minimize Double-stranded RNA Formation and Increase Protein Expression. J Mol Biol 2024; 436:168822. [PMID: 39427983 DOI: 10.1016/j.jmb.2024.168822] [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: 09/23/2024] [Revised: 10/11/2024] [Accepted: 10/13/2024] [Indexed: 10/22/2024]
Abstract
The therapeutic use of synthetic message RNA (mRNA) has been validated in COVID-19 vaccines and shows enormous potential in developing infectious and oncological vaccines. However, double-stranded RNA (dsRNA) byproducts generated during the in vitro transcription (IVT) process can diminish the efficacy of mRNA-based therapeutics and provoke innate immune responses. Existing methods to eliminate dsRNA byproducts are often cumbersome and labor-intensive. In this study, we revealed that a loose mRNA secondary structure and more unpaired U bases in the sequence generally lead to the formation of more dsRNA byproducts during the IVT process. We further developed a predictive model for dsRNA byproducts formation based on sequence characteristics to guide the optimization of mRNA sequences, helping to minimize unwanted immune response and improve the protein expression of mRNA products. Collectively, our study provides novel clues and methodologies for developing effective mRNA therapeutics with minimized dsRNA byproducts and increased protein expression.
Collapse
Affiliation(s)
- Qianshan Qin
- State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China; Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Orthopaedic Department of Tongji Hospital, Frontier Science Center for Stem Cell Research, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China; Suzhou Abogen Biosciences Co., Ltd., Suzhou, Jiangsu 215123, China
| | - Huayuan Yan
- Suzhou Abogen Biosciences Co., Ltd., Suzhou, Jiangsu 215123, China
| | - Weixiang Gao
- Suzhou Abogen Biosciences Co., Ltd., Suzhou, Jiangsu 215123, China
| | - Ruyin Cao
- Suzhou Abogen Biosciences Co., Ltd., Suzhou, Jiangsu 215123, China
| | - Guopeng Liu
- Suzhou Abogen Biosciences Co., Ltd., Suzhou, Jiangsu 215123, China
| | - Xiaojing Zhang
- Suzhou Abogen Biosciences Co., Ltd., Suzhou, Jiangsu 215123, China
| | - Niangang Wang
- Suzhou Abogen Biosciences Co., Ltd., Suzhou, Jiangsu 215123, China
| | - Wenjie Zuo
- Suzhou Abogen Biosciences Co., Ltd., Suzhou, Jiangsu 215123, China
| | - Lei Yuan
- Suzhou Abogen Biosciences Co., Ltd., Suzhou, Jiangsu 215123, China
| | - Peng Gao
- Suzhou Abogen Biosciences Co., Ltd., Suzhou, Jiangsu 215123, China.
| | - Qi Liu
- State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China; Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Orthopaedic Department of Tongji Hospital, Frontier Science Center for Stem Cell Research, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China.
| |
Collapse
|
31
|
Peng Y, Bai J, Li W, Su Z, Cheng X. Advancements in p53-Based Anti-Tumor Gene Therapy Research. Molecules 2024; 29:5315. [PMID: 39598704 PMCID: PMC11596491 DOI: 10.3390/molecules29225315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 11/05/2024] [Accepted: 11/07/2024] [Indexed: 11/29/2024] Open
Abstract
The p53 gene is one of the genes most closely associated with human tumors and has become a popular target for tumor drug design. Currently, p53-based gene therapy techniques have been developed, but these therapies face challenges such as immaturity, high safety hazards, limited efficacy, and low patient acceptance. However, researchers are no less enthusiastic about the treatment because of its theoretical potential to treat cancer. In this paper, the advances in p53-based gene therapy and related nucleic acid delivery technologies were reviewed and prospected in order to support further development in this field.
Collapse
Affiliation(s)
- Yuanwan Peng
- Institute of Modern Fermentation Engineering and Future Foods, School of Light Industry and Food Engineering, Guangxi University, No. 100, Daxuedong Road, Nanning 530004, China; (Y.P.); (J.B.); (W.L.)
| | - Jinping Bai
- Institute of Modern Fermentation Engineering and Future Foods, School of Light Industry and Food Engineering, Guangxi University, No. 100, Daxuedong Road, Nanning 530004, China; (Y.P.); (J.B.); (W.L.)
| | - Wang Li
- Institute of Modern Fermentation Engineering and Future Foods, School of Light Industry and Food Engineering, Guangxi University, No. 100, Daxuedong Road, Nanning 530004, China; (Y.P.); (J.B.); (W.L.)
| | - Zhengding Su
- School of Pharmaceutical Sciences and Institute of Materia Medica, Xinjiang University, Urumqi 830017, China
| | - Xiyao Cheng
- Institute of Modern Fermentation Engineering and Future Foods, School of Light Industry and Food Engineering, Guangxi University, No. 100, Daxuedong Road, Nanning 530004, China; (Y.P.); (J.B.); (W.L.)
| |
Collapse
|
32
|
Standley A, Xie J, Lau AW, Grote L, Gifford AJ. Working with Miraculous Mice: Mus musculus as a Model Organism. Curr Protoc 2024; 4:e70021. [PMID: 39435766 DOI: 10.1002/cpz1.70021] [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: 10/23/2024]
Abstract
The laboratory mouse has been described as a "miracle" model organism, providing a window by which we may gain an understanding of ourselves. Since the first recorded mouse experiment in 1664, the mouse has become the most used animal model in biomedical research. Mice are ideally suited as a model organism because of their small size, short gestation period, large litter size, and genetic similarity to humans. This article provides a broad overview of the laboratory mouse as a model organism and is intended for undergraduates and those new to working with mice. We delve into the history of the laboratory mouse and outline important terminology to accurately describe research mice. The types of laboratory mice available to researchers are reviewed, including outbred stocks, inbred strains, immunocompromised mice, and genetically engineered mice. The critical role mice have played in advancing knowledge in the areas of oncology, immunology, and pharmacology is highlighted by examining the significant contribution of mice to Nobel Prize winning research. International mouse mutagenesis programs and accurate phenotyping of mouse models are outlined. We also explain important considerations for working with mice, including animal ethics; the welfare principles of replacement, refinement, and reduction; and the choice of mouse model in experimental design. Finally, we present practical advice for maintaining a mouse colony, which involves adequate training of staff, the logistics of mouse housing, monitoring colony health, and breeding strategies. Useful resources for working with mice are also listed. The aim of this overview is to equip the reader with a broad appreciation of the enormous potential and some of the complexities of working with the laboratory mouse in a quest to improve human health. © 2024 The Author(s). Current Protocols published by Wiley Periodicals LLC.
Collapse
Affiliation(s)
- Anick Standley
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia
| | - Jinhan Xie
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia
| | - Angelica Wy Lau
- Garvan Institute of Medical Research, St Vincent's Clinical School, Darlinghurst, NSW, Australia
| | - Lauren Grote
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia
| | - Andrew J Gifford
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia
- Anatomical Pathology, NSW Heath Pathology, Prince of Wales Hospital, Randwick, NSW, Australia
- School of Clinical Medicine, UNSW Medicine & Health, UNSW Sydney, Sydney, NSW, Australia
| |
Collapse
|
33
|
Chandra S, Wilson JC, Good D, Wei MQ. mRNA vaccines: a new era in vaccine development. Oncol Res 2024; 32:1543-1564. [PMID: 39308511 PMCID: PMC11413818 DOI: 10.32604/or.2024.043987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 07/02/2024] [Indexed: 09/25/2024] Open
Abstract
The advent of RNA therapy, particularly through the development of mRNA cancer vaccines, has ushered in a new era in the field of oncology. This article provides a concise overview of the key principles, recent advancements, and potential implications of mRNA cancer vaccines as a groundbreaking modality in cancer treatment. mRNA cancer vaccines represent a revolutionary approach to combatting cancer by leveraging the body's innate immune system. These vaccines are designed to deliver specific mRNA sequences encoding cancer-associated antigens, prompting the immune system to recognize and mount a targeted response against malignant cells. This personalized and adaptive nature of mRNA vaccines holds immense potential for addressing the heterogeneity of cancer and tailoring treatments to individual patients. Recent breakthroughs in the development of mRNA vaccines, exemplified by the success of COVID-19 vaccines, have accelerated their application in oncology. The mRNA platform's versatility allows for the rapid adaptation of vaccine candidates to various cancer types, presenting an agile and promising avenue for therapeutic intervention. Clinical trials of mRNA cancer vaccines have demonstrated encouraging results in terms of safety, immunogenicity, and efficacy. Pioneering candidates, such as BioNTech's BNT111 and Moderna's mRNA-4157, have exhibited promising outcomes in targeting melanoma and solid tumors, respectively. These successes underscore the potential of mRNA vaccines to elicit robust and durable anti-cancer immune responses. While the field holds great promise, challenges such as manufacturing complexities and cost considerations need to be addressed for widespread adoption. The development of scalable and cost-effective manufacturing processes, along with ongoing clinical research, will be pivotal in realizing the full potential of mRNA cancer vaccines. Overall, mRNA cancer vaccines represent a cutting-edge therapeutic approach that holds the promise of transforming cancer treatment. As research progresses, addressing challenges and refining manufacturing processes will be crucial in advancing these vaccines from clinical trials to mainstream oncology practice, offering new hope for patients in the fight against cancer.
Collapse
Affiliation(s)
- Shubhra Chandra
- School of Pharmacy & Medical Sciences, Gold Coast campus, Griffith University, Brisbane, QLD-4222, Australia
- Menzies Health Institute Queensland (MHIQ), Gold Coast Campus, Griffith University, Brisbane, QLD-4215, Australia
| | - Jennifer C Wilson
- School of Pharmacy & Medical Sciences, Gold Coast campus, Griffith University, Brisbane, QLD-4222, Australia
- Menzies Health Institute Queensland (MHIQ), Gold Coast Campus, Griffith University, Brisbane, QLD-4215, Australia
| | - David Good
- School of Allied Health, Australian Catholic University, Brisbane, QLD-4014, Australia
| | - Ming Q Wei
- School of Pharmacy & Medical Sciences, Gold Coast campus, Griffith University, Brisbane, QLD-4222, Australia
- Menzies Health Institute Queensland (MHIQ), Gold Coast Campus, Griffith University, Brisbane, QLD-4215, Australia
| |
Collapse
|
34
|
Wang Y, Ma Q, Li M, Mai Q, Ma L, Zhang H, Zhong H, Mai K, Cheng N, Feng P, Guan P, Wu S, Zhang L, Dai J, Zhang B, Pan W, Yang Z. A decavalent composite mRNA vaccine against both influenza and COVID-19. mBio 2024; 15:e0066824. [PMID: 39105586 PMCID: PMC11389412 DOI: 10.1128/mbio.00668-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Accepted: 07/02/2024] [Indexed: 08/07/2024] Open
Abstract
The COVID-19 pandemic caused by SARS-CoV-2 has had a persistent and significant impact on global public health for 4 years. Recently, there has been a resurgence of seasonal influenza transmission worldwide. The co-circulation of SARS-CoV-2 and seasonal influenza viruses results in a dual burden on communities. Additionally, the pandemic potential of zoonotic influenza viruses, such as avian Influenza A/H5N1 and A/H7N9, remains a concern. Therefore, a combined vaccine against all these respiratory diseases is in urgent need. mRNA vaccines, with their superior efficacy, speed in development, flexibility, and cost-effectiveness, offer a promising solution for such infectious diseases and potential future pandemics. In this study, we present FLUCOV-10, a novel 10-valent mRNA vaccine created from our proven platform. This vaccine encodes hemagglutinin (HA) proteins from four seasonal influenza viruses and two avian influenza viruses with pandemic potential, as well as spike proteins from four SARS-CoV-2 variants. A two-dose immunization with the FLUCOV-10 elicited robust immune responses in mice, producing IgG antibodies, neutralizing antibodies, and antigen-specific cellular immune responses against all the vaccine-matched viruses of influenza and SARS-CoV-2. Remarkably, the FLUCOV-10 immunization provided complete protection in mouse models against both homologous and heterologous strains of influenza and SARS-CoV-2. These results highlight the potential of FLUCOV-10 as an effective vaccine candidate for the prevention of influenza and COVID-19.IMPORTANCEAmidst the ongoing and emerging respiratory viral threats, particularly the concurrent and sequential spread of SARS-CoV-2 and influenza, our research introduces FLUCOV-10. This novel mRNA-based combination vaccine, designed to counteract both influenza and COVID-19, by incorporating genes for surface glycoproteins from various influenza viruses and SARS-CoV-2 variants. This combination vaccine was highly effective in preclinical trials, generating strong immune responses and ensuring protection against both matching and heterologous strains of influenza viruses and SARS-CoV-2. FLUCOV-10 represents a significant step forward in our ability to address respiratory viral threats, showcasing potential as a singular, adaptable vaccine solution for global health challenges.
Collapse
MESH Headings
- Animals
- COVID-19/prevention & control
- COVID-19/immunology
- Mice
- SARS-CoV-2/immunology
- SARS-CoV-2/genetics
- COVID-19 Vaccines/immunology
- COVID-19 Vaccines/administration & dosage
- mRNA Vaccines
- Humans
- Influenza Vaccines/immunology
- Influenza Vaccines/administration & dosage
- Influenza Vaccines/genetics
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- Influenza, Human/prevention & control
- Influenza, Human/immunology
- Influenza, Human/virology
- Vaccines, Synthetic/immunology
- Vaccines, Synthetic/genetics
- Vaccines, Synthetic/administration & dosage
- Mice, Inbred BALB C
- Female
- Antibodies, Neutralizing/blood
- Antibodies, Neutralizing/immunology
- Orthomyxoviridae Infections/prevention & control
- Orthomyxoviridae Infections/immunology
- Orthomyxoviridae Infections/virology
- Hemagglutinin Glycoproteins, Influenza Virus/immunology
- Hemagglutinin Glycoproteins, Influenza Virus/genetics
- Spike Glycoprotein, Coronavirus/immunology
- Spike Glycoprotein, Coronavirus/genetics
- Influenza A Virus, H5N1 Subtype/immunology
- Influenza A Virus, H5N1 Subtype/genetics
- Influenza A virus/immunology
- Influenza A virus/genetics
Collapse
Affiliation(s)
- Yang Wang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Guangzhou National Laboratory, Guangzhou, China
| | - Qinhai Ma
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Man Li
- Argorna Pharmaceuticals Co., Ltd., Guangzhou, China
| | - Qianyi Mai
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Lin Ma
- Guangzhou RiboBio Co., Ltd, Guangzhou, China
| | - Hong Zhang
- Argorna Pharmaceuticals Co., Ltd., Guangzhou, China
| | | | - Kailin Mai
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Nan Cheng
- Guangzhou RiboBio Co., Ltd, Guangzhou, China
| | - Pei Feng
- Respiratory Disease AI Laboratory on Epidemic and Medical Big Data Instrument Applications, Faculty of Innovation Engineering, Macau University of Science and Technology, Macau SAR, China
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Taipa, Macau SAR, China
| | - Peikun Guan
- Guangzhou National Laboratory, Guangzhou, China
| | - Shengzhen Wu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Lu Zhang
- Technology Centre, Guangzhou Customs, Guangzhou, China
| | - Jun Dai
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Guangzhou National Laboratory, Guangzhou, China
- Technology Centre, Guangzhou Customs, Guangzhou, China
| | - Biliang Zhang
- Argorna Pharmaceuticals Co., Ltd., Guangzhou, China
- State Key Laboratory of Respiratory Disease, Laboratory of Computational Biomedicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Weiqi Pan
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Respiratory Disease AI Laboratory on Epidemic and Medical Big Data Instrument Applications, Faculty of Innovation Engineering, Macau University of Science and Technology, Macau SAR, China
| | - Zifeng Yang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Guangzhou National Laboratory, Guangzhou, China
- Respiratory Disease AI Laboratory on Epidemic and Medical Big Data Instrument Applications, Faculty of Innovation Engineering, Macau University of Science and Technology, Macau SAR, China
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Taipa, Macau SAR, China
| |
Collapse
|
35
|
Biscaia-Caleiras M, Fonseca NA, Lourenço AS, Moreira JN, Simões S. Rational formulation and industrial manufacturing of lipid-based complex injectables: Landmarks and trends. J Control Release 2024; 373:617-639. [PMID: 39002799 DOI: 10.1016/j.jconrel.2024.07.021] [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: 04/05/2024] [Revised: 07/02/2024] [Accepted: 07/08/2024] [Indexed: 07/15/2024]
Abstract
Lipid-based complex injectables are renowned for their effectiveness in delivering drugs, with many approved products. While significant strides have been made in formulating nanosystems for small molecular weight drugs, a pivotal breakthrough emerged with the recognition of lipid nanoparticles as a promising platform for delivering nucleic acids. This finding has paved the way for tackling long-standing challenges in molecular and delivery aspects (e.g., mRNA stability, intracellular delivery) that have impeded the clinical translation of gene therapy, especially in the realm of immunotherapy. Nonetheless, developing and implementing new lipid-based delivery systems pose significant challenges, as industrial manufacturing of these formulations often involves complex, multi-batch processes, giving rise to issues related to scalability, stability, sterility, and regulatory compliance. To overcome these obstacles, embracing the principles of quality-by-design (QbD) is imperative. Furthermore, adopting cutting-edge manufacturing and process analytical tools (PAT) that facilitate the transition from batch to continuous production is essential. Herein, the key milestones and insights derived from the development of currently approved lipid- nanosystems will be explored. Additionally, a comprehensive and critical overview of the latest technologies and regulatory guidelines that underpin the creation of more efficient, scalable, and flexible manufacturing processes for complex lipid-based nanoformulations will be provided.
Collapse
Affiliation(s)
- Mariana Biscaia-Caleiras
- CNC - Center for Neurosciences and Cell Biology, Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Faculty of Medicine (Polo 1), Rua Larga, 3004-504 Coimbra, Portugal; Bluepharma-Indústria Farmacêutica, S.A., São Martinho do Bispo, 3045-016 Coimbra, Portugal; Univ Coimbra-University of Coimbra, CIBB, Faculty of Pharmacy, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| | - Nuno A Fonseca
- Bluepharma-Indústria Farmacêutica, S.A., São Martinho do Bispo, 3045-016 Coimbra, Portugal
| | - Ana Sofia Lourenço
- Bluepharma-Indústria Farmacêutica, S.A., São Martinho do Bispo, 3045-016 Coimbra, Portugal
| | - João Nuno Moreira
- CNC - Center for Neurosciences and Cell Biology, Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Faculty of Medicine (Polo 1), Rua Larga, 3004-504 Coimbra, Portugal; Univ Coimbra-University of Coimbra, CIBB, Faculty of Pharmacy, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| | - Sérgio Simões
- CNC - Center for Neurosciences and Cell Biology, Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Faculty of Medicine (Polo 1), Rua Larga, 3004-504 Coimbra, Portugal; Bluepharma-Indústria Farmacêutica, S.A., São Martinho do Bispo, 3045-016 Coimbra, Portugal; Univ Coimbra-University of Coimbra, CIBB, Faculty of Pharmacy, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal.
| |
Collapse
|
36
|
Shoja Doost J, Fazel F, Boodhoo N, Sharif S. mRNA Vaccination: An Outlook on Innate Sensing and Adaptive Immune Responses. Viruses 2024; 16:1404. [PMID: 39339880 PMCID: PMC11437395 DOI: 10.3390/v16091404] [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: 08/06/2024] [Revised: 08/29/2024] [Accepted: 08/30/2024] [Indexed: 09/30/2024] Open
Abstract
Vaccination has led to significant dismantling of infectious diseases worldwide. Since the dawn of the SARS-CoV-2 pandemic, there has been increased popularity in the usage and study of the mRNA vaccine platform. Here, we highlight fundamental knowledge on mRNA vaccine pharmacology, followed by the immunity conferred by innate sensing and adaptive responses resulting from exposure to the mRNA vaccine construct and encapsulation materials. A better understanding of these immune mechanisms will shed light on further improvements in mRNA vaccine design, aiming to improve efficiency and optimize immune responses upon inoculation.
Collapse
Affiliation(s)
| | | | | | - Shayan Sharif
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada; (J.S.D.); (F.F.); (N.B.)
| |
Collapse
|
37
|
Cullis PR, Felgner PL. The 60-year evolution of lipid nanoparticles for nucleic acid delivery. Nat Rev Drug Discov 2024; 23:709-722. [PMID: 38965378 DOI: 10.1038/s41573-024-00977-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/17/2024] [Indexed: 07/06/2024]
Abstract
Delivery of genetic information to the interior of target cells in vivo has been a major challenge facing gene therapies. This barrier is now being overcome, owing in part to dramatic advances made by lipid-based systems that have led to lipid nanoparticles (LNPs) that enable delivery of nucleic acid-based vaccines and therapeutics. Examples include the clinically approved COVID-19 LNP mRNA vaccines and Onpattro (patisiran), an LNP small interfering RNA therapeutic to treat transthyretin-induced amyloidosis (hATTR). In addition, a host of promising LNP-enabled vaccines and gene therapies are in clinical development. Here, we trace this success to two streams of research conducted over the past 60 years: the discovery of the transfection properties of lipoplexes composed of positively charged cationic lipids complexed with nucleic acid cargos and the development of lipid nanoparticles using ionizable cationic lipids. The fundamental insights gained from these two streams of research offer potential delivery solutions for most forms of gene therapies.
Collapse
Affiliation(s)
- P R Cullis
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada.
| | - P L Felgner
- Department of Physiology & Biophysics, University of California, Irvine, CA, USA.
| |
Collapse
|
38
|
Oladipo EK, Oyelakin OD, Aiyelabegan AO, Olajide EO, Olatayo VO, Owolabi KP, Shittu YB, Olugbodi RO, Ajala HA, Rukayat RA, Olayiwola DO, Irewolede BA, Jimah EM, Oloke JK, Ojo TO, Ajani OF, Iwalokun BA, Kolawole OM, Ariyo OE, Adediran DA, Olufemi SE, Onyeaka H. Exploring computational approaches to design mRNA Vaccine against vaccinia and Mpox viruses. Immun Inflamm Dis 2024; 12:e1360. [PMID: 39150224 PMCID: PMC11328121 DOI: 10.1002/iid3.1360] [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: 08/02/2023] [Revised: 07/15/2024] [Accepted: 07/19/2024] [Indexed: 08/17/2024] Open
Abstract
BACKGROUND Messenger RNA (mRNA) vaccines emerged as a powerful tool in the fight against infections. Unlike traditional vaccines, this unique type of vaccine elicits robust and persistent innate and humoral immune response with a unique host cell-mediated pathogen gene expression and antigen presentation. METHODS This offers a novel approach to combat poxviridae infections. From the genome of vaccinia and Mpox viruses, three key genes (E8L, E7R, and H3L) responsible for virus attachment and virulence were selected and employed for designing the candidate mRNA vaccine against vaccinia and Mpox viral infection. Various bioinformatics tools were employed to generate (B cell, CTL, and HTL) epitopes, of which 28 antigenic and immunogenic epitopes were selected and are linked to form the mRNA vaccine construct. Additional components, including a 5' cap, 5' UTR, adjuvant, 3' UTR, and poly(A) tail, were incorporated to enhance stability and effectiveness. Safety measures such as testing for human homology and in silico immune simulations were implemented to avoid autoimmunity and to mimics the immune response of human host to the designed mRNA vaccine, respectively. The mRNA vaccine's binding affinity was evaluated by docking it with TLR-2, TLR-3, TLR-4, and TLR-9 receptors which are subsequently followed by molecular dynamics simulations for the highest binding one to predict the stability of the binding complex. RESULTS With a 73% population coverage, the mRNA vaccine looks promising, boasting a molecular weight of 198 kDa and a molecular formula of C8901H13609N2431O2611S48 and it is said to be antigenic, nontoxic and nonallergic, making it safe and effective in preventing infections with Mpox and vaccinia viruses, in comparison with other insilico-designed vaccine for vaccinia and Mpox viruses. CONCLUSIONS However, further validation through in vivo and in vitro techniques is underway to fully assess its potential.
Collapse
Affiliation(s)
- Elijah K. Oladipo
- Division of Vaccine Design and DevelopmentHelix Biogen InstituteOgbomosoOyo StateNigeria
- Laboratory of Molecular Biology, Immunology and Bioinformatics, Department of MicrobiologyAdeleke UniversityEdeOsun StateNigeria
| | - Olanrewaju D. Oyelakin
- Division of Vaccine Design and DevelopmentHelix Biogen InstituteOgbomosoOyo StateNigeria
| | | | - Elizabeth O. Olajide
- Division of Vaccine Design and DevelopmentHelix Biogen InstituteOgbomosoOyo StateNigeria
- Molecular Biology and Biotechnology DepartmentNigeria Institute of Medical ResearchLagosNigeria
| | - Victoria O. Olatayo
- Division of Vaccine Design and DevelopmentHelix Biogen InstituteOgbomosoOyo StateNigeria
| | - Kaothar P. Owolabi
- Division of Vaccine Design and DevelopmentHelix Biogen InstituteOgbomosoOyo StateNigeria
| | - Yewande B. Shittu
- Division of Vaccine Design and DevelopmentHelix Biogen InstituteOgbomosoOyo StateNigeria
| | - Rhoda O. Olugbodi
- Division of Vaccine Design and DevelopmentHelix Biogen InstituteOgbomosoOyo StateNigeria
| | - Hezekiah A. Ajala
- Division of Vaccine Design and DevelopmentHelix Biogen InstituteOgbomosoOyo StateNigeria
| | - Raji A. Rukayat
- Division of Vaccine Design and DevelopmentHelix Biogen InstituteOgbomosoOyo StateNigeria
| | - Deborah O. Olayiwola
- Division of Vaccine Design and DevelopmentHelix Biogen InstituteOgbomosoOyo StateNigeria
| | | | - Esther M. Jimah
- Division of Vaccine Design and DevelopmentHelix Biogen InstituteOgbomosoOyo StateNigeria
| | - Julius K. Oloke
- Department of Natural SciencesPrecious Cornerstone UniversityIbadanOyo StateNigeria
| | - Taiwo O. Ojo
- Division of Vaccine Design and DevelopmentHelix Biogen InstituteOgbomosoOyo StateNigeria
| | | | - Bamidele A. Iwalokun
- Molecular Biology and Biotechnology DepartmentNigeria Institute of Medical ResearchLagosNigeria
| | | | - Olumuyiwa E. Ariyo
- Department of Medicine, Infectious Disease and Tropical Medicine UnitFederal Teaching HospitalIdo EkitiEkiti StateNigeria
| | - Daniel A. Adediran
- Division of Vaccine Design and DevelopmentHelix Biogen InstituteOgbomosoOyo StateNigeria
- Department of BiochemistryLadoke Akintola University of TechnologyOgbomosoOyo StateNigeria
| | - Seun E. Olufemi
- Division of Vaccine Design and DevelopmentHelix Biogen InstituteOgbomosoOyo StateNigeria
- Department of BiochemistryLadoke Akintola University of TechnologyOgbomosoOyo StateNigeria
| | - Helen Onyeaka
- School of Chemical EngineeringUniversity of BirminghamBirminghamUK
| |
Collapse
|
39
|
Fazel F, Doost JS, Raj S, Boodhoo N, Karimi K, Sharif S. The mRNA vaccine platform for veterinary species. Vet Immunol Immunopathol 2024; 274:110803. [PMID: 39003921 DOI: 10.1016/j.vetimm.2024.110803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 06/27/2024] [Indexed: 07/16/2024]
Abstract
Vaccination has proven to be an effective means of controlling pathogens in animals. Since the introduction of veterinary vaccines in the 19th century, several generations of vaccines have been introduced. These vaccines have had a positive impact on global animal health and production. Despite, the success of veterinary vaccines, there are still some pathogens for which there are no effective vaccines available, such as African swine fever. Further, animal health is under the constant threat of emerging and re-emerging pathogens, some of which are zoonotic and can pose a threat to human health. The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic has highlighted the need for new vaccine platforms that are safe and efficacious, but also importantly, are adaptable and can be modified rapidly to match the circulating pathogens. mRNA vaccines have been shown to be an effective vaccine platform against various viral and bacterial pathogens. This review will cover some of the recent advances in the field of mRNA vaccines for veterinary species. Moreover, various mRNA vaccines and their delivery methods, as well as their reported efficacy, will be discussed. Current limitations and future prospects of this vaccine platform in veterinary medicine will also be discussed.
Collapse
Affiliation(s)
- Fatemeh Fazel
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Janan Shoja Doost
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Sugandha Raj
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Nitish Boodhoo
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Khalil Karimi
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Shayan Sharif
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario N1G 2W1, Canada.
| |
Collapse
|
40
|
Miyazato P, Noguchi T, Ogawa F, Sugimoto T, Fauzyah Y, Sasaki R, Ebina H. 1mΨ influences the performance of various positive-stranded RNA virus-based replicons. Sci Rep 2024; 14:17634. [PMID: 39085360 PMCID: PMC11292005 DOI: 10.1038/s41598-024-68617-y] [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: 05/16/2024] [Accepted: 07/25/2024] [Indexed: 08/02/2024] Open
Abstract
Self-amplifying RNAs (saRNAs) are versatile vaccine platforms that take advantage of a viral RNA-dependent RNA polymerase (RdRp) to amplify the messenger RNA (mRNA) of an antigen of interest encoded within the backbone of the viral genome once inside the target cell. In recent years, more saRNA vaccines have been clinically tested with the hope of reducing the vaccination dose compared to the conventional mRNA approach. The use of N1-methyl-pseudouridine (1mΨ), which enhances RNA stability and reduces the innate immune response triggered by RNAs, is among the improvements included in the current mRNA vaccines. In the present study, we evaluated the effects of this modified nucleoside on various saRNA platforms based on different viruses. The results showed that different stages of the replication process were affected depending on the backbone virus. For TNCL, an insect virus of the Alphanodavirus genus, replication was impaired by poor recognition of viral RNA by RdRp. In contrast, the translation step was severely abrogated in coxsackievirus B3 (CVB3), a member of the Picornaviridae family. Finally, the effects of 1mΨ on Semliki forest virus (SFV), were not detrimental in in vitro studies, but no advantages were observed when immunogenicity was tested in vivo.
Collapse
Affiliation(s)
- Paola Miyazato
- The Research Foundation for Microbial Diseases of Osaka University (BIKEN), Suita, Osaka, Japan
- Virus Vaccine Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Osaka, Japan
| | - Takafumi Noguchi
- The Research Foundation for Microbial Diseases of Osaka University (BIKEN), Suita, Osaka, Japan
- Virus Vaccine Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Osaka, Japan
| | - Fumiyo Ogawa
- The Research Foundation for Microbial Diseases of Osaka University (BIKEN), Suita, Osaka, Japan
- Virus Vaccine Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Osaka, Japan
| | - Takeshi Sugimoto
- The Research Foundation for Microbial Diseases of Osaka University (BIKEN), Suita, Osaka, Japan
- Virus Vaccine Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Osaka, Japan
| | - Yuzy Fauzyah
- The Research Foundation for Microbial Diseases of Osaka University (BIKEN), Suita, Osaka, Japan
- Virus Vaccine Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Osaka, Japan
| | - Ryo Sasaki
- The Research Foundation for Microbial Diseases of Osaka University (BIKEN), Suita, Osaka, Japan
| | - Hirotaka Ebina
- The Research Foundation for Microbial Diseases of Osaka University (BIKEN), Suita, Osaka, Japan.
- Virus Vaccine Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Osaka, Japan.
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan.
- Center for Advanced Modalities and DDS (CAMaD), Osaka University, Suita, Osaka, Japan.
- Center for Infectious Disease Education and Research (CiDER), Osaka University, Suita, Osaka, Japan.
| |
Collapse
|
41
|
Tadic S, Martínez A. Nucleic acid cancer vaccines targeting tumor related angiogenesis. Could mRNA vaccines constitute a game changer? Front Immunol 2024; 15:1433185. [PMID: 39081320 PMCID: PMC11286457 DOI: 10.3389/fimmu.2024.1433185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 07/01/2024] [Indexed: 08/02/2024] Open
Abstract
Tumor related angiogenesis is an attractive target in cancer therapeutic research due to its crucial role in tumor growth, invasion, and metastasis. Different agents were developed aiming to inhibit this process; however they had limited success. Cancer vaccines could be a promising tool in anti-cancer/anti-angiogenic therapy. Cancer vaccines aim to initiate an immune response against cancer cells upon presentation of tumor antigens which hopefully will result in the eradication of disease and prevention of its recurrence by inducing an efficient and long-lasting immune response. Different vaccine constructs have been developed to achieve this and they could include either protein-based or nucleic acid-based vaccines. Nucleic acid vaccines are simple and relatively easy to produce, with high efficiency and safety, thus prompting a high interest in the field. Different DNA vaccines have been developed to target crucial regulators of tumor angiogenesis. Most of them were successful in pre-clinical studies, mostly when used in combination with other therapeutics, but had limited success in the clinic. Apparently, different tumor evasion mechanisms and reduced immunogenicity still limit the potential of these vaccines and there is plenty of room for improvement. Nowadays, mRNA cancer vaccines are making remarkable progress due to improvements in the manufacturing technology and represent a powerful potential alternative. Apart from their efficiency, mRNA vaccines are simple and cheap to produce, can encompass multiple targets simultaneously, and can be quickly transferred from bench to bedside. mRNA vaccines have already accomplished amazing results in cancer clinical trials, thus ensuring a bright future in the field, although no anti-angiogenic mRNA vaccines have been described yet. This review aims to describe recent advances in anti-angiogenic DNA vaccine therapy and to provide perspectives for use of revolutionary approaches such are mRNA vaccines for anti-angiogenic treatments.
Collapse
Affiliation(s)
| | - Alfredo Martínez
- Angiogenesis Unit, Oncology Area, Center for Biomedical Research of La Rioja (CIBIR), Logroño, Spain
| |
Collapse
|
42
|
Guasp P, Reiche C, Sethna Z, Balachandran VP. RNA vaccines for cancer: Principles to practice. Cancer Cell 2024; 42:1163-1184. [PMID: 38848720 DOI: 10.1016/j.ccell.2024.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 04/29/2024] [Accepted: 05/06/2024] [Indexed: 06/09/2024]
Abstract
Vaccines are the most impactful medicines to improve health. Though potent against pathogens, vaccines for cancer remain an unfulfilled promise. However, recent advances in RNA technology coupled with scientific and clinical breakthroughs have spurred rapid discovery and potent delivery of tumor antigens at speed and scale, transforming cancer vaccines into a tantalizing prospect. Yet, despite being at a pivotal juncture, with several randomized clinical trials maturing in upcoming years, several critical questions remain: which antigens, tumors, platforms, and hosts can trigger potent immunity with clinical impact? Here, we address these questions with a principled framework of cancer vaccination from antigen detection to delivery. With this framework, we outline features of emergent RNA technology that enable rapid, robust, real-time vaccination with somatic mutation-derived neoantigens-an emerging "ideal" antigen class-and highlight latent features that have sparked the belief that RNA could realize the enduring vision for vaccines against cancer.
Collapse
Affiliation(s)
- Pablo Guasp
- Immuno-Oncology Service, Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Hepatopancreatobiliary Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Charlotte Reiche
- Immuno-Oncology Service, Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Hepatopancreatobiliary Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Zachary Sethna
- Immuno-Oncology Service, Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Hepatopancreatobiliary Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Vinod P Balachandran
- Immuno-Oncology Service, Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Hepatopancreatobiliary Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA; David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
43
|
Masum MHU, Wajed S, Hossain MI, Moumi NR, Talukder A, Rahman MM. An mRNA vaccine for pancreatic cancer designed by applying in silico immunoinformatics and reverse vaccinology approaches. PLoS One 2024; 19:e0305413. [PMID: 38976715 PMCID: PMC11230540 DOI: 10.1371/journal.pone.0305413] [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: 03/16/2024] [Accepted: 05/30/2024] [Indexed: 07/10/2024] Open
Abstract
Pancreatic ductal adenocarcinoma is the most prevalent pancreatic cancer, which is considered a significant global health concern. Chemotherapy and surgery are the mainstays of current pancreatic cancer treatments; however, a few cases are suitable for surgery, and most of the cases will experience recurrent episodes. Compared to DNA or peptide vaccines, mRNA vaccines for pancreatic cancer have more promise because of their delivery, enhanced immune responses, and lower proneness to mutation. We constructed an mRNA vaccine by analyzing S100 family proteins, which are all major activators of receptors for advanced glycation end products. We applied immunoinformatic approaches, including physicochemical properties analysis, structural prediction and validation, molecular docking study, in silico cloning, and immune simulations. The designed mRNA vaccine was estimated to have a molecular weight of 165023.50 Da and was highly soluble (grand average of hydropathicity of -0.440). In the structural assessment, the vaccine seemed to be a well-stable and functioning protein (Z score of -8.94). Also, the docking analysis suggested that the vaccine had a high affinity for TLR-2 and TLR-4 receptors. Additionally, the molecular mechanics with generalized Born and surface area solvation analysis of the "Vaccine-TLR-2" (-141.07 kcal/mol) and "Vaccine-TLR-4" (-271.72 kcal/mol) complexes also suggests a strong binding affinity for the receptors. Codon optimization also provided a high expression level with a GC content of 47.04% and a codon adaptation index score 1.0. The appearance of memory B-cells and T-cells was also observed over a while, with an increased level of helper T-cells and immunoglobulins (IgM and IgG). Moreover, the minimum free energy of the mRNA vaccine was predicted at -1760.00 kcal/mol, indicating the stability of the vaccine following its entry, transcription, and expression. This hypothetical vaccine offers a groundbreaking tool for future research and therapeutic development of pancreatic cancer.
Collapse
Affiliation(s)
- Md Habib Ullah Masum
- Department of Microbiology, Noakhali Science and Technology University, Noakhali, Bangladesh
- Microbiology, Cancer and Bioinformatics Research Group, Noakhali Science and Technology University, Noakhali, Bangladesh
| | - Shah Wajed
- Department of Microbiology, Noakhali Science and Technology University, Noakhali, Bangladesh
- Microbiology, Cancer and Bioinformatics Research Group, Noakhali Science and Technology University, Noakhali, Bangladesh
- Infectiology: Biology of Infectious Diseases, Universite Paris-Saclay, Gif-sur-Yvette, France
| | - Md Imam Hossain
- Department of Microbiology, Noakhali Science and Technology University, Noakhali, Bangladesh
| | - Nusrat Rahman Moumi
- Medical Sciences, University of Central Lancashire, Preston, Lancashire, United Kingdom
| | - Asma Talukder
- Microbiology, Cancer and Bioinformatics Research Group, Noakhali Science and Technology University, Noakhali, Bangladesh
- Department of Biotechnology and Genetic Engineering, Noakhali Science and Technology University, Noakhali, Bangladesh
- School of Pharmacy and Medical Sciences, and Menzies Health Institute Queensland, Griffith University, Brisbane, Queensland, Australia
| | - Md Mijanur Rahman
- Department of Microbiology, Noakhali Science and Technology University, Noakhali, Bangladesh
- Microbiology, Cancer and Bioinformatics Research Group, Noakhali Science and Technology University, Noakhali, Bangladesh
- School of Pharmacy and Medical Sciences, and Menzies Health Institute Queensland, Griffith University, Brisbane, Queensland, Australia
| |
Collapse
|
44
|
Makhijani S, Elossaily GM, Rojekar S, Ingle RG. mRNA-based vaccines - global approach, challenges, and could be a promising wayout for future pandemics. Pharm Dev Technol 2024; 29:559-565. [PMID: 38814266 DOI: 10.1080/10837450.2024.2361656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 05/27/2024] [Accepted: 05/27/2024] [Indexed: 05/31/2024]
Abstract
mRNA-based vaccines are assured to significantly boost biopharmaceuticals since outbreak of coronavirus disease- 2019. Respiratory infections, such as influenza, SARS, MERS, COVID-19, and respiratory syncytial virus, often have high transmission rates due to their airborne spread. Respiratory infections can lead to severe illness and death. These outbreaks can cause substantial economic and social disruption, as seen with the COVID-19 pandemic. In our interconnected world, respiratory diseases can spread rapidly across borders. mRNA-based vaccines (e.g. mRNA-1283) can reduce the transmission by creating immunity in the population, thus lowering the incidence and spread of these diseases. Vaccines are crucial for global health security, helping to prevent local outbreaks from becoming global pandemics. Nevertheless, various concerns remain such as intracellular delivery, susceptibility to degradation by catalytic hydrolysis, and instability due to several physiological conditions. Therefore, an hour needed to address these challenges and opportunities for attaining high-quality and stable mRNA-based vaccines with novel drug delivery systems. The authors contributed an extensive review of the mRNA-based clinical development, progress in stability, and delivery challenges to mitigate market needs. In addition, the authors discuss crucial advances in the growth of mRNA-based vaccines to date; which dominate an extensive scope of therapeutic implementation. Finally, recent mRNA-based vaccines in clinical trials, adjuvant benefits, and prospects are discussed.
Collapse
Affiliation(s)
- Shivani Makhijani
- Datta Meghe College of Pharmacy, Datta Meghe Institute of Higher Education and Research (DMIHER), Deemed to be University, Wardha, Maharashtra, India
| | - Gehan M Elossaily
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, Riyadh, Saudi Arabia
| | | | - Rahul G Ingle
- Datta Meghe College of Pharmacy, Datta Meghe Institute of Higher Education and Research (DMIHER), Deemed to be University, Wardha, Maharashtra, India
| |
Collapse
|
45
|
Esih H, Mezgec K, Billmeier M, Malenšek Š, Benčina M, Grilc B, Vidmar S, Gašperlin M, Bele M, Zidarn M, Zupanc TL, Morgan T, Jordan I, Sandig V, Schrödel S, Thirion C, Protzer U, Wagner R, Lainšček D, Jerala R. Mucoadhesive film for oral delivery of vaccines for protection of the respiratory tract. J Control Release 2024; 371:179-192. [PMID: 38795814 DOI: 10.1016/j.jconrel.2024.05.041] [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: 02/05/2024] [Revised: 05/20/2024] [Accepted: 05/21/2024] [Indexed: 05/28/2024]
Abstract
The delivery of vaccines plays a pivotal role in influencing the strength and longevity of the immune response and controlling reactogenicity. Mucosal immunization, as compared to parenteral vaccination, could offer greater protection against respiratory infections while being less invasive. While oral vaccination has been presumed less effective and believed to target mainly the gastrointestinal tract, trans-buccal delivery using mucoadhesive films (MAF) may allow targeted delivery to the mucosa. Here we present an effective strategy for mucosal delivery of several vaccine platforms incorporated in MAF, including DNA plasmids, viral vectors, and lipid nanoparticles incorporating mRNA (mRNA/LNP). The mRNA/LNP vaccine formulation targeting SARS-CoV-2 as a proof of concept remained stable within MAF consisting of slowly releasing water-soluble polymers and an impermeable backing layer, facilitating enhanced penetration into the oral mucosa. This formulation elicited antibody and cellular responses comparable to the intramuscular injection, but also induced the production of mucosal IgAs, highlighting its efficacy, particularly for use as a booster vaccine and the potential advantage for protection against respiratory infections. The MAF vaccine preparation demonstrates significant advantages, such as efficient delivery, stability, and simple noninvasive administration with the potential to alleviate vaccine hesitancy.
Collapse
Affiliation(s)
- Hana Esih
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, 1000 Ljubljana, Slovenia; Graduate School of Biomedicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Klemen Mezgec
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, 1000 Ljubljana, Slovenia; Graduate School of Biomedicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Martina Billmeier
- Institute of Medical Microbiology & Hygiene, Molecular Microbiology (Virology), University of Regensburg, Regensburg, Germany
| | - Špela Malenšek
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, 1000 Ljubljana, Slovenia; Graduate School of Biomedicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Mojca Benčina
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, 1000 Ljubljana, Slovenia; Centre for Technologies of Gene and Cell Therapy, 1000 Ljubljana, Slovenia
| | - Blaž Grilc
- University of Ljubljana, Faculty of Pharmacy, Department of Pharmaceutical Technology, Ljubljana 1000, Slovenia
| | - Sara Vidmar
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, 1000 Ljubljana, Slovenia; Graduate School of Biomedicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Mirjana Gašperlin
- University of Ljubljana, Faculty of Pharmacy, Department of Pharmaceutical Technology, Ljubljana 1000, Slovenia
| | - Marjan Bele
- Department of Materials Chemistry, National Institute of Chemistry, Ljubljana 1000, Slovenia
| | - Mihaela Zidarn
- University Clinic of Pulmonary and Allergic Diseases Golnik, Golnik, Slovenia
| | | | - Tina Morgan
- University Clinic of Pulmonary and Allergic Diseases Golnik, Golnik, Slovenia
| | - Ingo Jordan
- Applied Science & Technologies, ProBioGen AG, Berlin, Germany
| | - Volker Sandig
- Applied Science & Technologies, ProBioGen AG, Berlin, Germany
| | - Silke Schrödel
- SIRION Biotech GmbH, Am Klopferspitz 19, 82152 Martinsried, Germany
| | | | - Ulrike Protzer
- Institute of Virology, School of Medicine, Technical University of Munich, Helmholtz Zentrum München, Munich, Germany
| | - Ralf Wagner
- Institute of Medical Microbiology & Hygiene, Molecular Microbiology (Virology), University of Regensburg, Regensburg, Germany; Institute of Clinical Microbiology & Hygiene, University Hospital, Regensburg, Germany
| | - Duško Lainšček
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, 1000 Ljubljana, Slovenia; Centre for Technologies of Gene and Cell Therapy, 1000 Ljubljana, Slovenia.
| | - Roman Jerala
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, 1000 Ljubljana, Slovenia; Centre for Technologies of Gene and Cell Therapy, 1000 Ljubljana, Slovenia.
| |
Collapse
|
46
|
Smith AR, Rizvi F, Everton E, Adeagbo A, Wu S, Tam Y, Muramatsu H, Pardi N, Weissman D, Gouon-Evans V. Transient growth factor expression via mRNA in lipid nanoparticles promotes hepatocyte cell therapy in mice. Nat Commun 2024; 15:5010. [PMID: 38866762 PMCID: PMC11169405 DOI: 10.1038/s41467-024-49332-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 06/03/2024] [Indexed: 06/14/2024] Open
Abstract
Primary human hepatocyte (PHH) transplantation is a promising alternative to liver transplantation, whereby liver function could be restored by partial repopulation of the diseased organ with healthy cells. However, currently PHH engraftment efficiency is low and benefits are not maintained long-term. Here we refine two male mouse models of human chronic and acute liver diseases to recapitulate compromised hepatocyte proliferation observed in nearly all human liver diseases by overexpression of p21 in hepatocytes. In these clinically relevant contexts, we demonstrate that transient, yet robust expression of human hepatocyte growth factor and epidermal growth factor in the liver via nucleoside-modified mRNA in lipid nanoparticles, whose safety was validated with mRNA-based COVID-19 vaccines, drastically improves PHH engraftment, reduces disease burden, and improves overall liver function. This strategy may overcome the critical barriers to clinical translation of cell therapies with primary or stem cell-derived hepatocytes for the treatment of liver diseases.
Collapse
Affiliation(s)
- Anna R Smith
- Department of Medicine, Section of Gastroenterology, Center for Regenerative Medicine, Boston University Chobanian & Avedisian School of Medicine & Boston Medical Center, Boston, MA, USA
| | - Fatima Rizvi
- Department of Medicine, Section of Gastroenterology, Center for Regenerative Medicine, Boston University Chobanian & Avedisian School of Medicine & Boston Medical Center, Boston, MA, USA
| | - Elissa Everton
- Department of Medicine, Section of Gastroenterology, Center for Regenerative Medicine, Boston University Chobanian & Avedisian School of Medicine & Boston Medical Center, Boston, MA, USA
| | - Anisah Adeagbo
- Department of Medicine, Section of Gastroenterology, Center for Regenerative Medicine, Boston University Chobanian & Avedisian School of Medicine & Boston Medical Center, Boston, MA, USA
| | - Susan Wu
- Department of Medicine, Section of Gastroenterology, Center for Regenerative Medicine, Boston University Chobanian & Avedisian School of Medicine & Boston Medical Center, Boston, MA, USA
| | - Ying Tam
- Acuitas Therapeutics, Vancouver, BC, Canada
| | - Hiromi Muramatsu
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Norbert Pardi
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Drew Weissman
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Valerie Gouon-Evans
- Department of Medicine, Section of Gastroenterology, Center for Regenerative Medicine, Boston University Chobanian & Avedisian School of Medicine & Boston Medical Center, Boston, MA, USA.
| |
Collapse
|
47
|
Androsavich JR. Frameworks for transformational breakthroughs in RNA-based medicines. Nat Rev Drug Discov 2024; 23:421-444. [PMID: 38740953 DOI: 10.1038/s41573-024-00943-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/04/2024] [Indexed: 05/16/2024]
Abstract
RNA has sparked a revolution in modern medicine, with the potential to transform the way we treat diseases. Recent regulatory approvals, hundreds of new clinical trials, the emergence of CRISPR gene editing, and the effectiveness of mRNA vaccines in dramatic response to the COVID-19 pandemic have converged to create tremendous momentum and expectation. However, challenges with this relatively new class of drugs persist and require specialized knowledge and expertise to overcome. This Review explores shared strategies for developing RNA drug platforms, including layering technologies, addressing common biases and identifying gaps in understanding. It discusses the potential of RNA-based therapeutics to transform medicine, as well as the challenges associated with improving applicability, efficacy and safety profiles. Insights gained from RNA modalities such as antisense oligonucleotides (ASOs) and small interfering RNAs are used to identify important next steps for mRNA and gene editing technologies.
Collapse
Affiliation(s)
- John R Androsavich
- RNA Accelerator, Pfizer Inc, Cambridge, MA, USA.
- Ginkgo Bioworks, Boston, MA, USA.
| |
Collapse
|
48
|
Drzeniek NM, Kahwaji N, Picht S, Dimitriou IM, Schlickeiser S, Moradian H, Geissler S, Schmueck-Henneresse M, Gossen M, Volk HD. In Vitro Transcribed mRNA Immunogenicity Induces Chemokine-Mediated Lymphocyte Recruitment and Can Be Gradually Tailored by Uridine Modification. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308447. [PMID: 38491873 DOI: 10.1002/advs.202308447] [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: 11/06/2023] [Revised: 02/28/2024] [Indexed: 03/18/2024]
Abstract
Beyond SARS-CoV2 vaccines, mRNA drugs are being explored to overcome today's greatest healthcare burdens, including cancer and cardiovascular disease. Synthetic mRNA triggers immune responses in transfected cells, which can be reduced by chemically modified nucleotides. However, the side effects of mRNA-triggered immune activation on cell function and how different nucleotides, such as the N1-methylpseudouridine (m1Ψ) used in SARS-CoV2 vaccines, can modulate cellular responses is not fully understood. Here, cellular responses toward a library of uridine-modified mRNAs are investigated in primary human cells. Targeted proteomics analyses reveal that unmodified mRNA induces a pro-inflammatory paracrine pattern marked by the secretion of chemokines, which recruit T and B lymphocytes toward transfected cells. Importantly, the magnitude of mRNA-induced changes in cell function varies quantitatively between unmodified, Ψ-, m1Ψ-, and 5moU-modified mRNA and can be gradually tailored, with implications for deliberately exploiting this effect in mRNA drug design. Indeed, both the immunosuppressive effect of stromal cells on T-cell proliferation, and the anti-inflammatory effect of IL-10 mRNA are enhanced by appropriate uridine modification. The results provide new insights into the effects of mRNA drugs on cell function and cell-cell communication and open new possibilities to tailor mRNA-triggered immune activation to the desired pro- or anti-inflammatory application.
Collapse
Affiliation(s)
- Norman M Drzeniek
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universitaet Berlin and Humboldt-Universitaet zu Berlin, Institute of Medical Immunology, Augustenburger Platz 1, 13353, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Föhrer Straße 15, 13353, Berlin, Germany
| | - Nourhan Kahwaji
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Föhrer Straße 15, 13353, Berlin, Germany
| | - Samira Picht
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Föhrer Straße 15, 13353, Berlin, Germany
- Berlin-Brandenburg School for Regenerative Therapies (BSRT; graduate school 203 of the German Excellence Initiative), Augustenburger Platz 1, 13353, Berlin, Germany
| | - Ioanna Maria Dimitriou
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Föhrer Straße 15, 13353, Berlin, Germany
- Berlin-Brandenburg School for Regenerative Therapies (BSRT; graduate school 203 of the German Excellence Initiative), Augustenburger Platz 1, 13353, Berlin, Germany
- Julius Wolff Institute (JWI), Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
- Department of Biology, Chemistry, Pharmacy, Institute of Chemistry and Biochemistry, Freie Universität Berlin, Thielallee 63, 14195, Berlin, Germany
| | - Stephan Schlickeiser
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universitaet Berlin and Humboldt-Universitaet zu Berlin, Institute of Medical Immunology, Augustenburger Platz 1, 13353, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Föhrer Straße 15, 13353, Berlin, Germany
- CheckImmune GmbH, Campus Virchow Klinikum, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Hanieh Moradian
- Institute of Active Polymers, Helmholtz-Zentrum Hereon, Kantstraße 55, 14513, Teltow, Germany
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Augustenburger Platz 1, 13353, Berlin, Germany
| | - Sven Geissler
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Föhrer Straße 15, 13353, Berlin, Germany
- Julius Wolff Institute (JWI), Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin Center for Advanced Therapies (BeCAT), Augustenburger Platz 1, 13353, Berlin, Germany
| | - Michael Schmueck-Henneresse
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Föhrer Straße 15, 13353, Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin Center for Advanced Therapies (BeCAT), Augustenburger Platz 1, 13353, Berlin, Germany
| | - Manfred Gossen
- Institute of Active Polymers, Helmholtz-Zentrum Hereon, Kantstraße 55, 14513, Teltow, Germany
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Augustenburger Platz 1, 13353, Berlin, Germany
| | - Hans-Dieter Volk
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universitaet Berlin and Humboldt-Universitaet zu Berlin, Institute of Medical Immunology, Augustenburger Platz 1, 13353, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Föhrer Straße 15, 13353, Berlin, Germany
- CheckImmune GmbH, Campus Virchow Klinikum, Augustenburger Platz 1, 13353, Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin Center for Advanced Therapies (BeCAT), Augustenburger Platz 1, 13353, Berlin, Germany
| |
Collapse
|
49
|
Movahed F, Darzi S, Mahdavi P, Salih Mahdi M, Qutaiba B Allela O, Naji Sameer H, Adil M, Zarkhah H, Yasamineh S, Gholizadeh O. The potential use of therapeutics and prophylactic mRNA vaccines in human papillomavirus (HPV). Virol J 2024; 21:124. [PMID: 38822328 PMCID: PMC11143593 DOI: 10.1186/s12985-024-02397-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 05/23/2024] [Indexed: 06/02/2024] Open
Abstract
Cervical cancer (CC) and other malignant malignancies are acknowledged to be primarily caused by persistent human papillomavirus (HPV) infection. Historically, vaccinations against viruses that produce neutralizing antibodies unique to the virus have been an affordable way to manage viral diseases. CC risk is decreased, but not eliminated, by HPV vaccinations. Since vaccinations have been made available globally, almost 90% of HPV infections have been successfully avoided. On the lesions and diseases that are already present, however, no discernible treatment benefit has been shown. As a result, therapeutic vaccines that elicit immune responses mediated by cells are necessary for the treatment of established infections and cancers. mRNA vaccines possess remarkable potential in combating viral diseases and malignancy as a result of their superior industrial production, safety, and efficacy. Furthermore, considering the expeditiousness of production, the mRNA vaccine exhibits promise as a therapeutic approach targeting HPV. Given that the HPV-encoded early proteins, including oncoproteins E6 and E7, are consistently present in HPV-related cancers and pre-cancerous lesions and have crucial functions in the progression and persistence of HPV-related diseases, they serve as ideal targets for therapeutic HPV vaccines. The action mechanism of HPV and HPV-related cancer mRNA vaccines, their recent advancements in clinical trials, and the potential for their therapeutic applications are highlighted in this study, which also offers a quick summary of the present state of mRNA vaccines. Lastly, we highlight a few difficulties with mRNA HPV vaccination clinical practice and provide our thoughts on further advancements in this quickly changing sector. It is expected that mRNA vaccines will soon be produced quickly for clinical HPV prevention and treatment.
Collapse
Affiliation(s)
- Fatemeh Movahed
- Department of Gynecology and Obstetrics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Satinik Darzi
- Department Of Obstetrics and Gynecology, Abnormal Uterine Bleeding Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | - Parya Mahdavi
- Department of Obstetrics and Gynecology, Faculty of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | | | | | - Hayder Naji Sameer
- Collage of Pharmacy, National University of Science and Technology, Dhi Qar, 64001, Iraq
| | - Mohaned Adil
- Pharmacy college, Al-Farahidi University, Baghdad, Iraq
| | - Hasna Zarkhah
- Department of Obstetrics and Gynaecology, Tabriz University of Medical Siences, Tabriz, Iran.
| | - Saman Yasamineh
- Young Researchers and Elite Club, Tabriz Branch, Islamic Azad University, Tabriz, Iran.
| | | |
Collapse
|
50
|
Johnson Y, Shakri AR, Pond-Tor S, Jnawali A, Najrana T, Wu H, Badhai J, Alameh MG, Weissman D, Kabyemela E, Duffy P, Fried M, Kurtis J, Raj DK. Immunization with PfGBP130 generates antibodies that inhibit RBC invasion by P. falciparum parasites. Front Immunol 2024; 15:1350560. [PMID: 38863702 PMCID: PMC11165087 DOI: 10.3389/fimmu.2024.1350560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 04/30/2024] [Indexed: 06/13/2024] Open
Abstract
Background Despite decades of effort, Plasmodium falciparum malaria remains a leading killer of children. The absence of a highly effective vaccine and the emergence of parasites resistant to both diagnosis as well as treatment hamper effective public health interventions. Methods and results To discover new vaccine candidates, we used our whole proteome differential screening method and identified PfGBP130 as a parasite protein uniquely recognized by antibodies from children who had developed resistance to P. falciparum infection but not from those who remained susceptible. We formulated PfGBP130 as lipid encapsulated mRNA, DNA plasmid, and recombinant protein-based immunogens and evaluated the efficacy of murine polyclonal anti-PfGBP130 antisera to inhibit parasite growth in vitro. Immunization of mice with PfGBP130-A (aa 111-374), the region identified in our differential screen, formulated as a DNA plasmid or lipid encapsulated mRNA, but not as a recombinant protein, induced antibodies that inhibited RBC invasion in vitro. mRNA encoding the full ectodomain of PfGBP130 (aa 89-824) also generated parasite growth-inhibitory antibodies. Conclusion We are currently advancing PfGBP130-A formulated as a lipid-encapsulated mRNA for efficacy evaluation in non-human primates.
Collapse
Affiliation(s)
- Yannick Johnson
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, RI, United States
| | - Ahmad Rushdi Shakri
- Department of Internal Medicine, University of South Florida, Tampa, FL, United States
| | - Sunthorn Pond-Tor
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, RI, United States
- Center for International Health Research, Rhode Island Hospital, Warren Alpert Medical School, Brown University, Providence, RI, United States
| | - Anup Jnawali
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, RI, United States
- Center for International Health Research, Rhode Island Hospital, Warren Alpert Medical School, Brown University, Providence, RI, United States
| | - Tanbir Najrana
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, RI, United States
- Center for International Health Research, Rhode Island Hospital, Warren Alpert Medical School, Brown University, Providence, RI, United States
| | - Haiwei Wu
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, RI, United States
- Center for International Health Research, Rhode Island Hospital, Warren Alpert Medical School, Brown University, Providence, RI, United States
| | - Jhasketan Badhai
- Department of Internal Medicine, University of South Florida, Tampa, FL, United States
| | | | - Drew Weissman
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Edward Kabyemela
- Department of Pathology, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Patrick Duffy
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, United States
| | - Michal Fried
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, United States
| | - Jonathan Kurtis
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, RI, United States
- Center for International Health Research, Rhode Island Hospital, Warren Alpert Medical School, Brown University, Providence, RI, United States
| | - Dipak Kumar Raj
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, RI, United States
- Department of Internal Medicine, University of South Florida, Tampa, FL, United States
- Center for International Health Research, Rhode Island Hospital, Warren Alpert Medical School, Brown University, Providence, RI, United States
| |
Collapse
|