1
|
Zheng X, Liu B, Ni P, Cai L, Shi X, Ke Z, Zhang S, Hu B, Yang B, Xu Y, Long W, Fang Z, Wang Y, Zhang W, Xu Y, Wang Z, Pan K, Zhou K, Wang H, Geng H, Hu H, Liu B. Development and application of an uncapped mRNA platform. Ann Med 2025; 57:2437046. [PMID: 39648715 PMCID: PMC11632943 DOI: 10.1080/07853890.2024.2437046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 11/28/2023] [Accepted: 06/01/2024] [Indexed: 12/10/2024] Open
Abstract
BACKGROUND A novel uncapped mRNA platform was developed. METHODS Five lipid nanoparticle (LNP)-encapsulated mRNA constructs were made to evaluate several aspects of our platform, including transfection efficiency and durability in vitro and in vivo and the activation of humoral and cellular immunity in several animal models. The constructs were eGFP-mRNA-LNP (for enhanced green fluorescence mRNA), Fluc-mRNA-LNP (for firefly luciferase mRNA), SδT-mRNA-LNP (for Delta strain SARS-CoV-2 spike protein trimer mRNA), gDED-mRNA-LNP (for truncated glycoprotein D mRNA coding ectodomain from herpes simplex virus type 2 (HSV2)) and gDFR-mRNA-LNP (for truncated HSV2 glycoprotein D mRNA coding amino acids 1-400). RESULTS Quantifiable target protein expression was achieved in vitro and in vivo with eGFP- and Fluc-mRNA-LNP. SδT-mRNA-LNP, gDED-mRNA-LNP and gDFR-mRNA-LNP induced both humoral and cellular immune responses comparable to those obtained by previously reported capped mRNA-LNP constructs. Notably, SδT-mRNA-LNP elicited neutralizing antibodies in hamsters against the Omicron and Delta strains. Additionally, gDED-mRNA-LNP and gDFR-mRNA-LNP induced potent neutralizing antibodies in rabbits and mice. The mRNA constructs with uridine triphosphate (UTP) outperformed those with N1-methylpseudouridine triphosphate (N1mψTP) in the induction of antibodies via SδT-mRNA-LNP. CONCLUSIONS Our uncapped, process-simplified and economical mRNA platform may have broad utility in vaccines and protein replacement drugs.KEY MESSAGESThe mRNA platform described in our paper uses internal ribosome entry site (IRES) (Rapid, Amplified, Capless and Economical, RACE; Register as BH-RACE platform) instead of caps and uridine triphosphate (UTP) instead of N1-methylpseudouridine triphosphate (N1mψTP) to synthesize mRNA.Through the self-developed packaging instrument and lipid nanoparticle (LNP) delivery system, mRNA can be expressed in cells more efficiently, quickly and economically.Particularly exciting is that potent neutralizing antibodies against Delta and Omicron real viruses were induced with the new coronavirus S protein mRNA vaccine from the BH-RACE platform.
Collapse
Affiliation(s)
- Xiaodi Zheng
- College of Bioengineering, National ‘‘111’’ Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, Hubei University of Technology, Wuhan, China
| | - Biao Liu
- College of Bioengineering, National ‘‘111’’ Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, Hubei University of Technology, Wuhan, China
| | - Peng Ni
- Wuhan Binhui Biopharmaceutical Co., Ltd., Wuhan, China
| | - Linkang Cai
- Wuhan Binhui Biopharmaceutical Co., Ltd., Wuhan, China
| | - Xiaotai Shi
- Wuhan Binhui Biopharmaceutical Co., Ltd., Wuhan, China
| | - Zonghuang Ke
- Wuhan Binhui Biopharmaceutical Co., Ltd., Wuhan, China
| | - Siqi Zhang
- Wuhan Binhui Biopharmaceutical Co., Ltd., Wuhan, China
| | - Bing Hu
- Hubei Provincial Centre for Disease Control and Prevention, Wuhan, China
| | - Binfeng Yang
- Wuhan Binhui Biopharmaceutical Co., Ltd., Wuhan, China
| | - Yiyan Xu
- College of Bioengineering, National ‘‘111’’ Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, Hubei University of Technology, Wuhan, China
| | - Wei Long
- Wuhan Binhui Biopharmaceutical Co., Ltd., Wuhan, China
| | - Zhizheng Fang
- Wuhan Binhui Biopharmaceutical Co., Ltd., Wuhan, China
| | - Yang Wang
- College of Bioengineering, National ‘‘111’’ Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, Hubei University of Technology, Wuhan, China
| | - Wen Zhang
- Department of Immunology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yan Xu
- Wuhan Binhui Biopharmaceutical Co., Ltd., Wuhan, China
| | - Zhong Wang
- Wuhan Binhui Biopharmaceutical Co., Ltd., Wuhan, China
| | - Kai Pan
- Hubei Provincial Centre for Disease Control and Prevention, Wuhan, China
| | - Kangping Zhou
- Hubei Provincial Centre for Disease Control and Prevention, Wuhan, China
| | - Hanming Wang
- Wuhan Binhui Biopharmaceutical Co., Ltd., Wuhan, China
| | - Hui Geng
- School of Life Science, Huazhong Normal University, Wuhan, China
| | - Han Hu
- College of Bioengineering, National ‘‘111’’ Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, Hubei University of Technology, Wuhan, China
| | - Binlei Liu
- College of Bioengineering, National ‘‘111’’ Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, Hubei University of Technology, Wuhan, China
- Wuhan Binhui Biopharmaceutical Co., Ltd., Wuhan, China
| |
Collapse
|
2
|
Bardach A, Brizuela M, Berrueta M, Ciapponi A, Sambade JM, Ballivian J, Ortega V, Castellana N, Comandé D, Parker EPK, Kampmann B, Stegelmann K, Xiong X, Stergachis A, Munoz FM, Buekens P, Mazzoni A. Umbrella review of the safety of Chikungunya vaccine platforms used in other vaccines. Hum Vaccin Immunother 2025; 21:2463191. [PMID: 39932481 PMCID: PMC11817526 DOI: 10.1080/21645515.2025.2463191] [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: 11/26/2024] [Revised: 01/20/2025] [Accepted: 02/02/2025] [Indexed: 02/14/2025] Open
Abstract
Chikungunya virus (CHIKV), transmitted through Aedes mosquitoes, is a significant global health concern. Various vaccine platforms have been explored to combat CHIKV, including formalin inactivation, live-attenuated strains, virus-like particles (VLPs), viral vectors, and mRNA technologies. This umbrella review synthesizes evidence on the safety profiles of vaccine platforms used in Chikungunya vaccines that have been applied in other vaccines, focusing on adverse events of special interest (AESI) in pregnant persons, children, and adolescents. A comprehensive overview of systematic reviews (SRs) was conducted. Results: Seven systematic reviews were included and complemented with primary studies. Vaccines like influenza, human papillomavirus (HPV), and COVID-19, which share platforms with Chikungunya vaccines, showed no significant increase in AESI. Moderate-to high-quality SRs supported favorable safety profiles. Vaccines sharing platforms with Chikungunya vaccines generally exhibit acceptable safety profiles in pregnant persons, children, and adolescents.
Collapse
Affiliation(s)
- Ariel Bardach
- Center for Research in Epidemiology and Public Health (CIESP-IECS), CONICET, Buenos Aires, Argentina
| | - Martin Brizuela
- Department of Health Technology Assessment and Economic Evaluations, Institute for Clinical Effectiveness and Health Policy (IECS), Buenos Aires, Argentina
| | - Mabel Berrueta
- Department of Mother and Child Health, Institute for Clinical Effectiveness and Health Policy (IECS), Buenos Aires, Argentina
| | - Agustín Ciapponi
- Argentine Cochrane Center, Institute for Clinical Effectiveness and Health Policy (IECS), Buenos Aires, Argentina
| | - Juan M. Sambade
- Department of Health Technology Assessment and Economic Evaluations, Institute for Clinical Effectiveness and Health Policy (IECS), Buenos Aires, Argentina
| | - Jamile Ballivian
- Department of Health Technology Assessment and Economic Evaluations, Institute for Clinical Effectiveness and Health Policy (IECS), Buenos Aires, Argentina
| | - Vanesa Ortega
- Department of Mother and Child Health, Institute for Clinical Effectiveness and Health Policy (IECS), Buenos Aires, Argentina
| | - Noelia Castellana
- Department of Health Technology Assessment and Economic Evaluations, Institute for Clinical Effectiveness and Health Policy (IECS), Buenos Aires, Argentina
| | - Daniel Comandé
- Department of Health Technology Assessment and Economic Evaluations, Institute for Clinical Effectiveness and Health Policy (IECS), Buenos Aires, Argentina
| | - Edward P. K. Parker
- The Vaccine Centre, London School of Hygiene & Tropical Medicine, Keppel Street, London, UK
| | - Beate Kampmann
- Health Protection Research Unit in Immunisation, London School of Hygiene & Tropical Medicine, London, UK
- Charite Centre for Global Health, Charité, Universitätsmedizin, Vaccine Centre, Berlin, Germany
| | - Katharina Stegelmann
- Department of Health Technology Assessment and Economic Evaluations, Institute for Clinical Effectiveness and Health Policy (IECS), Buenos Aires, Argentina
| | - Xu Xiong
- School of Public Health and Tropical Medicine, Tulane University, New Orleans, USA
| | - Andy Stergachis
- Schools of Pharmacy and Public Health, University of Washington, Seattle, USA
| | - Flor M. Munoz
- Departments of Pediatrics and Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Pierre Buekens
- School of Public Health and Tropical Medicine, Tulane University, New Orleans, USA
| | - Agustina Mazzoni
- Department of Health Technology Assessment and Economic Evaluations, Institute for Clinical Effectiveness and Health Policy (IECS), Buenos Aires, Argentina
| |
Collapse
|
3
|
Bahadir Z, Narayan P, Wolters R, Permar SR, Fouda G, Hessell AJ, Haigwood NL. Monoclonal Antibodies for Pediatric Viral Disease Prevention and Treatment. Pediatrics 2025; 155:e2024068690. [PMID: 40174915 DOI: 10.1542/peds.2024-068690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 02/27/2025] [Indexed: 04/04/2025] Open
Abstract
Medical advancements over the last century have improved our ability to treat pediatric infectious diseases, significantly reducing associated morbidity and mortality worldwide. Although vaccines have been pivotal in this progress, many viral pathogens still do not currently have effective vaccines. The COVID-19 pandemic highlighted the need for rapid responses to emerging viral pathogens and introduced new tools to combat them. This review addresses human monoclonal antibodies (mAbs) as a strategy for treating and preventing viral infections in pediatric populations. We discuss previously used and currently available mAbs and advancements in mAb discovery. We address the future of mAb therapy by describing novel approaches in drug production and delivery platforms in addition to alternative antibody classes. Finally, we review the challenges and limitations of mAb therapy development for newborns and children.
Collapse
Affiliation(s)
- Zeynep Bahadir
- Department of Pediatrics, Weill Cornell Medicine, Cornell University, New York, New York
| | - Priyanka Narayan
- Department of Pediatrics, Weill Cornell Medicine, Cornell University, New York, New York
| | - Rachael Wolters
- Division of Pathobiology & Immunology, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon
| | - Sallie R Permar
- Department of Pediatrics, Weill Cornell Medicine, Cornell University, New York, New York
| | - Genevieve Fouda
- Department of Pediatrics, Weill Cornell Medicine, Cornell University, New York, New York
| | - Ann J Hessell
- Division of Pathobiology & Immunology, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon
| | - Nancy L Haigwood
- Division of Pathobiology & Immunology, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon
| |
Collapse
|
4
|
Cheong DHJ, Yi B, Wong YH, Chu JJH. The Current Progress in the Quest for Vaccines Against the Semliki Forest Virus Complex. Med Res Rev 2025; 45:947-967. [PMID: 39757142 DOI: 10.1002/med.22097] [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: 05/26/2023] [Revised: 10/10/2024] [Accepted: 12/12/2024] [Indexed: 01/07/2025]
Abstract
The Semliki Forest virus (SFV) complex comprises of arboviruses that are transmitted by arthropod vectors and cause acute febrile illness in humans. In the last seven decades, re-emergence of these viruses has resulted in numerous outbreaks globally, affecting regions including Africa, Americas, Asia, Europe and the Caribbean. These viruses are transmitted to humans by the bite of infected mosquitoes. Symptoms of infection include high fever, severe joint pain, skin rash, muscle pain and headache. Fatal cases were reported, and mortality rate increased during the epidemic of these viruses. There is therefore a need to control the spread of these emerging arboviruses. Given that vaccination is one of the most effective ways to protect populations against viral outbreaks, efforts have been made to develop and test potential vaccine candidates. However, there are still no licensed vaccines available against the medically important viruses in the SFV complex. This review first summarizes the current knowledge of the SFV complex disease pathogenesis. Next, seven strategies that have been applied in vaccine development against these viruses are reviewed, indicating the immune response and efficacies of these vaccine candidates in in vivo models of infection. Finally, the more promising candidates that have entered clinical trials are discussed and insights into the future development of vaccines for viruses of the SFV complex are given.
Collapse
Affiliation(s)
- Dorothy Hui Juan Cheong
- Department of Microbiology and Immunology, Infectious Disease Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore City, Singapore
| | - Bowen Yi
- Biosafety Level 3 Core Facility, Yong Loo Lin School of Medicine, National University of Singapore, Singapore City, Singapore
| | - Yi Hao Wong
- Biosafety Level 3 Core Facility, Yong Loo Lin School of Medicine, National University of Singapore, Singapore City, Singapore
| | - Justin Jang Hann Chu
- Department of Microbiology and Immunology, Infectious Disease Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore City, Singapore
- Biosafety Level 3 Core Facility, Yong Loo Lin School of Medicine, National University of Singapore, Singapore City, Singapore
- Collaborative and Translation Unit for Hand, Foot and Mouth Disease (HFMD), Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore CIty, Singapore
| |
Collapse
|
5
|
Wang Y, Yuan S, Zhou L, Yang K, Jin Z, Lin A, Yang C, Tian W. Cutting-Edge Progress in the Acquisition, Modification and Therapeutic Applications of Exosomes for Drug Delivery. Int J Nanomedicine 2025; 20:5059-5080. [PMID: 40271148 PMCID: PMC12015628 DOI: 10.2147/ijn.s516840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Accepted: 04/08/2025] [Indexed: 04/25/2025] Open
Abstract
Exosomes are vesicles secreted by cells, typically ranging from 30 to 150 nm in diameter, and serve as crucial mediators of intercellular communication. Exosomes are capable of loading various therapeutic substances, such as small molecule compounds, proteins, and oligonucleotides, thereby making them an ideal vehicle for drug delivery. The distinctive biocompatibility, high stability, and targeting properties of exosomes render them highly valuable for future treatments of diseases like cancer and cardiovascular diseases. Despite the potential advantage of exosomes in delivering biologically active molecules, the techniques for the preparation, purification, preservation, and other aspects of stem cell exosomes are not yet mature enough. In this paper, we briefly introduce the composition, biogenesis, and benefits of exosomes, and primarily focus on summarizing the isolation and purification methods of exosomes, the preparation of engineered exosomes, and their clinical applications, to better provide new ideas for the development of exosome drug delivery systems.
Collapse
Affiliation(s)
- Yuhao Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Engineering Research Center of Oral Translational Medicine, Ministry of Education & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, People’s Republic of China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, People’s Republic of China
| | - Shengmeng Yuan
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Engineering Research Center of Oral Translational Medicine, Ministry of Education & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, People’s Republic of China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, People’s Republic of China
| | - Lihua Zhou
- National Institute of Measurement and Testing Technology, Chengdu, Sichuan, 610041, People’s Republic of China
| | - Kexin Yang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Engineering Research Center of Oral Translational Medicine, Ministry of Education & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, People’s Republic of China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, People’s Republic of China
| | - Zhaorui Jin
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Engineering Research Center of Oral Translational Medicine, Ministry of Education & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, People’s Republic of China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, People’s Republic of China
| | - An Lin
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Engineering Research Center of Oral Translational Medicine, Ministry of Education & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, People’s Republic of China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, People’s Republic of China
| | - Chao Yang
- Chengdu Shiliankangjian Biotechnology Co., Ltd., Chengdu, Sichuan, 610041, People’s Republic of China
| | - Weidong Tian
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Engineering Research Center of Oral Translational Medicine, Ministry of Education & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, People’s Republic of China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, People’s Republic of China
| |
Collapse
|
6
|
Masum MHU, Mahdeen AA, Barua A. Revolutionizing Chikungunya Vaccines: mRNA Breakthroughs With Molecular and Immune Simulations. Bioinform Biol Insights 2025; 19:11779322251324859. [PMID: 40182080 PMCID: PMC11967231 DOI: 10.1177/11779322251324859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 02/14/2025] [Indexed: 04/05/2025] Open
Abstract
With the ability to cause massive epidemics that have consequences on millions of individuals globally, the Chikungunya virus (CHIKV) emerges as a severe menace. Developing an effective vaccine is urgent as no effective therapeutics are available for such viral infections. Therefore, we designed a novel mRNA vaccine against CHIKV with a combination of highly antigenic and potential MHC-I, MHC-II, and B-cell epitopes from the structural polyprotein. The vaccine demonstrated well-characterized physicochemical properties, indicating its solubility and potential functional stability within the body (GRAVY score of -0.639). Structural analyses of the vaccine revealed a well-stabilized secondary and tertiary structure (Ramachandran score of 82.8% and a Z-score of -4.17). Docking studies of the vaccine with TLR-2 (-1027.7 KJ/mol) and TLR-4 (-1212.4 KJ/mol) exhibited significant affinity with detailed hydrogen bond interactions. Molecular dynamics simulations highlighted distinct conformational dynamics among the vaccine, "vaccine-TLR-2" and "vaccine-TLR-4" complexes. The vaccine's ability to elicit both innate and adaptive immune responses, including the presence of memory B-cells and T-cells, persistent B-cell immunity for a year, and the activation of TH cells leading to the release of IFN-γ and IL-2, has significant implications for its potential effectiveness. The CHIKV vaccine developed in this study shows promise as a potential candidate for future vaccine production against CHIKV, suggesting its suitability for further clinical advancement, including in vitro and in vivo experiments.
Collapse
Affiliation(s)
- Md. Habib Ullah Masum
- Department of Genomics and Bioinformatics, Faculty of Biotechnology and Genetic Engineering, Chattogram Veterinary and Animal Sciences University, Khulshi, Chattogram, Bangladesh
| | - Ahmad Abdullah Mahdeen
- Department of Microbiology, Noakhali Science and Technology University, Noakhali, Bangladesh
| | - Abanti Barua
- Department of Microbiology, Noakhali Science and Technology University, Noakhali, Bangladesh
| |
Collapse
|
7
|
Chi H, Zhao SQ, Liu YP, Xiong XC, Zhong X, Suo XX, Huang XY, Yue C, Zhang JJ, Cong Z, Li N, Du FX, Chen T, Ma JR, Wang L, Ying B, Deng YQ, Yin JY, Wang X, Xue J, Qin CF. Efficacy and safety of an mRNA encoding neutralizing antibody against SARS-CoV-2 in non-human primates. Sci Bull (Beijing) 2025:S2095-9273(25)00338-X. [PMID: 40221320 DOI: 10.1016/j.scib.2025.03.055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 01/24/2025] [Accepted: 03/19/2025] [Indexed: 04/14/2025]
Affiliation(s)
- Hang Chi
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing 100071, China
| | - Suo-Qun Zhao
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing 100071, China
| | - Yun-Peng Liu
- State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Beijing 100021, China
| | - Xiao-Chuan Xiong
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing 100071, China
| | - Xia Zhong
- Suzhou Abogen Biosciences Co. Ltd, Suzhou 215123, China
| | - Xing-Xing Suo
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing 100071, China
| | - Xing-Yao Huang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing 100071, China
| | - Can Yue
- CAS Key Laboratory of Infection and Immunity, National Laboratory of Macromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Jing-Jing Zhang
- State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Beijing 100021, China
| | - Zhe Cong
- State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Beijing 100021, China
| | - Na Li
- State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Beijing 100021, China
| | - Fei-Xue Du
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing 100071, China
| | - Ting Chen
- State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Beijing 100021, China
| | - Jian-Rong Ma
- State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Beijing 100021, China
| | - Lei Wang
- CAS Key Laboratory of Infection and Immunity, National Laboratory of Macromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Bo Ying
- Suzhou Abogen Biosciences Co. Ltd, Suzhou 215123, China
| | - Yong-Qiang Deng
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing 100071, China
| | - Ji-Ye Yin
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Beijing 100089, China.
| | - Xiangxi Wang
- CAS Key Laboratory of Infection and Immunity, National Laboratory of Macromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China.
| | - Jing Xue
- State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Beijing 100021, China.
| | - Cheng-Feng Qin
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing 100071, China; Research Unit of Discovery and Tracing of Natural Focus Diseases, Chinese Academy of Medical Sciences, Beijing 100071, China.
| |
Collapse
|
8
|
Liang X, Zhou Y, Yang Y, Li Q, Wang J, Li B, Yang H, Tang C, Yu W, Wang H, Huang Q, Chen H, Yan Y, An R, Lin D, Quan W, Zhang Y, Li Y, Du X, Yuan Y, Yuan L, Zhou J, Sun Q, Wang Y, Lu S. CHIKV mRNA vaccines encoding conserved structural/envelope proteins confer broad cross-lineage protection against infection. Signal Transduct Target Ther 2025; 10:98. [PMID: 40148284 PMCID: PMC11950367 DOI: 10.1038/s41392-025-02182-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 02/03/2025] [Accepted: 02/17/2025] [Indexed: 03/29/2025] Open
Abstract
With the broad spread of the chikungunya virus (CHIKV), there is an increasing demand for more effective and broadly protective vaccines. Here, we designed CHIKV mRNA vaccines containing full-length structural proteins or part of structural proteins (envelope proteins) based on conserved sequences from 769 viral strains encompassing four lineages. The vaccine induced strong cellular and humoral immune responses in BALB/c mice and provided robust protection. Immunization of BALB/c mice with either of the two vaccines induced high levels of neutralizing antibodies against pseudoviruses from four distinct lineages, highlighting their potential for broad cross-lineage protective efficacy. Immunoglobulin repertoire analysis revealed two important BCR V-J gene combinations, IgHV1-4-IgHJ3 and IgHV1-4-IgHJ2, and lineage-specific immunity analysis revealed significant upregulation of TCRs containing V19 and V20. BCR and TCR immunodiversity may be a potential reason for the broad-spectrum protection against CHIKV afforded by the vaccine. In A129 mice, it elicited lower levels of neutralizing antibodies but prevented mouse mortality and cleared chronic infection. In the rhesus macaque model, both vaccines elicited a certain level of humoral and cellular immune responses and protected the rhesus macaques from the CHIKV challenge. In conclusion, the results from both mouse and rhesus macaque models indicate that the vaccine could be a candidate for clinical use against CHIKV.
Collapse
Affiliation(s)
- Xiaoming Liang
- Yunnan Key Laboratory of Cross-Border Infectious Disease Control and Prevention and Novel Drug Development, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
| | - Yanan Zhou
- Yunnan Key Laboratory of Cross-Border Infectious Disease Control and Prevention and Novel Drug Development, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
| | - Yun Yang
- Yunnan Key Laboratory of Cross-Border Infectious Disease Control and Prevention and Novel Drug Development, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
| | - Qianqian Li
- Yunnan Key Laboratory of Cross-Border Infectious Disease Control and Prevention and Novel Drug Development, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
| | - Junbin Wang
- Yunnan Key Laboratory of Cross-Border Infectious Disease Control and Prevention and Novel Drug Development, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
| | - Bai Li
- Yunnan Key Laboratory of Cross-Border Infectious Disease Control and Prevention and Novel Drug Development, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
| | - Hao Yang
- Yunnan Key Laboratory of Cross-Border Infectious Disease Control and Prevention and Novel Drug Development, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
| | - Cong Tang
- Yunnan Key Laboratory of Cross-Border Infectious Disease Control and Prevention and Novel Drug Development, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
| | - Wenhai Yu
- Yunnan Key Laboratory of Cross-Border Infectious Disease Control and Prevention and Novel Drug Development, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
| | - Haixuan Wang
- Yunnan Key Laboratory of Cross-Border Infectious Disease Control and Prevention and Novel Drug Development, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
| | - Qing Huang
- Yunnan Key Laboratory of Cross-Border Infectious Disease Control and Prevention and Novel Drug Development, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
| | - Hongyu Chen
- Yunnan Key Laboratory of Cross-Border Infectious Disease Control and Prevention and Novel Drug Development, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
| | - Yuhuan Yan
- Yunnan Key Laboratory of Cross-Border Infectious Disease Control and Prevention and Novel Drug Development, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
| | - Ran An
- Yunnan Key Laboratory of Cross-Border Infectious Disease Control and Prevention and Novel Drug Development, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
| | - Dongdong Lin
- Yunnan Key Laboratory of Cross-Border Infectious Disease Control and Prevention and Novel Drug Development, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
| | - Wenqi Quan
- Yunnan Key Laboratory of Cross-Border Infectious Disease Control and Prevention and Novel Drug Development, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
| | - Yong Zhang
- Yunnan Key Laboratory of Cross-Border Infectious Disease Control and Prevention and Novel Drug Development, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
| | - Yanwen Li
- Yunnan Key Laboratory of Cross-Border Infectious Disease Control and Prevention and Novel Drug Development, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
| | - Xuena Du
- Yunnan Key Laboratory of Cross-Border Infectious Disease Control and Prevention and Novel Drug Development, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
| | - Yuxia Yuan
- Yunnan Key Laboratory of Cross-Border Infectious Disease Control and Prevention and Novel Drug Development, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
| | - Longhai Yuan
- Yunnan Key Laboratory of Cross-Border Infectious Disease Control and Prevention and Novel Drug Development, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
| | - Jian Zhou
- Yunnan Key Laboratory of Cross-Border Infectious Disease Control and Prevention and Novel Drug Development, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
| | - Qiangming Sun
- Yunnan Key Laboratory of Cross-Border Infectious Disease Control and Prevention and Novel Drug Development, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China.
- State Key Laboratory of Respiratory Health and Multimorbidity, Beijing, China.
- Key Laboratory of Pathogen Infection Prevention and Control (Peking Union Medical College), Ministry of Education, Beijing, China.
- Yunnan Provincial Key Laboratory of Vector-borne Diseases Control and Research, Kunming, China.
| | - Youchun Wang
- Yunnan Key Laboratory of Cross-Border Infectious Disease Control and Prevention and Novel Drug Development, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China.
- State Key Laboratory of Respiratory Health and Multimorbidity, Beijing, China.
- Key Laboratory of Pathogen Infection Prevention and Control (Peking Union Medical College), Ministry of Education, Beijing, China.
| | - Shuaiyao Lu
- Yunnan Key Laboratory of Cross-Border Infectious Disease Control and Prevention and Novel Drug Development, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China.
- State Key Laboratory of Respiratory Health and Multimorbidity, Beijing, China.
- Key Laboratory of Pathogen Infection Prevention and Control (Peking Union Medical College), Ministry of Education, Beijing, China.
- Yunnan Provincial Key Laboratory of Vector-borne Diseases Control and Research, Kunming, China.
| |
Collapse
|
9
|
Żak MM, Zangi L. Clinical development of therapeutic mRNA applications. Mol Ther 2025:S1525-0016(25)00208-4. [PMID: 40143545 DOI: 10.1016/j.ymthe.2025.03.034] [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: 12/04/2024] [Revised: 02/21/2025] [Accepted: 03/21/2025] [Indexed: 03/28/2025] Open
Abstract
mRNA therapeutics are emerging as a transformative approach in modern medicine, providing innovative, highly adaptable solutions for a wide range of diseases, from viral infections to cancer. Since the approval of the first mRNA therapeutic-the coronavirus disease 2019 vaccines in 2021-we have identified more than 70 current clinical trials utilizing mRNA for various diseases. We propose classifying mRNA therapeutics into four main categories: vaccines, protein replacement therapies, antibodies, and mRNA-based cell and gene therapies. Each category can be further divided into subcategories. Vaccines include those targeting viral antigens, bacterial or parasitic antigens, general and individualized cancer antigens, and self-antigens. Protein replacement therapies include maintenance therapeutics designed to treat genetic disorders and interventional therapeutics, where delivering therapeutic proteins could improve patient outcomes, such as vascular endothelial growth factor A for ischemic heart disease or proinflammatory cytokines in cancer. Therapeutic antibodies are based on mRNA sequences encoding the heavy and light chains of clinically relevant antibodies, enabling patient cells to produce them directly, bypassing the costly and complex process of manufacturing protein-ready antibodies. Another category of mRNA-based therapeutics encompasses cell and gene therapies, including CRISPR with mRNA-mediated delivery of Cas9 and the in vivo generation of cells expressing CAR through mRNA. We discuss examples of mRNA therapeutics currently in clinical trials within each category, providing a comprehensive overview of the field's progress and highlighting key advancements as of the end of 2024.
Collapse
Affiliation(s)
- Magdalena M Żak
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Lior Zangi
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
10
|
Cheung TH, Shoichet MS. The Interplay of Endosomal Escape and RNA Release from Polymeric Nanoparticles. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2025; 41:7174-7190. [PMID: 40080875 DOI: 10.1021/acs.langmuir.4c05176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/15/2025]
Abstract
Ribonucleic acid (RNA) nanocarriers, specifically lipid nanoparticles and polymeric nanoparticles, enable RNA transfection both in vitro and in vivo; however, only a small percentage of RNA endocytosed by a cell is delivered to the cytosolic machinery, minimizing its effect. RNA nanocarriers face two major obstacles after endocytosis: endosomal escape and RNA release. Overcoming both obstacles simultaneously is challenging because endosomal escape is usually achieved by using high positive charge to disrupt the endosomal membrane. However, this high positive charge typically also inhibits RNA release because anionic RNA is strongly bound to the nanocarrier by electrostatic interactions. Many nanocarriers address one over the other despite a growing body of evidence demonstrating that both are crucial for RNA transfection. In this review, we survey the various strategies that have been employed to accomplish both endosomal escape and RNA release with a focus on polymeric nanomaterials. We first consider the various requirements a nanocarrier must achieve for RNA delivery including protection from degradation, cellular internalization, endosomal escape, and RNA release. We then discuss current polymers used for RNA delivery and examine the strategies for achieving both endosomal escape and RNA release. Finally, we review various stimuli-responsive strategies for RNA release. While RNA release continues to be a challenge in achieving efficient RNA transfection, many new innovations in polymeric materials have elucidated promising strategies.
Collapse
Affiliation(s)
- Timothy H Cheung
- Department of Chemistry, University of Toronto, 80 St. George Street, Toronto, Ontario M5S 3H6, Canada
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 160 College Street, Toronto, Ontario M5S 3E1, Canada
| | - Molly S Shoichet
- Department of Chemistry, University of Toronto, 80 St. George Street, Toronto, Ontario M5S 3H6, Canada
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 160 College Street, Toronto, Ontario M5S 3E1, Canada
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, Ontario M5S 3E5, Canada
- Institute of Biomedical Engineering, University of Toronto, 164 College Street, Toronto, Ontario M5S 3G9, Canada
| |
Collapse
|
11
|
Zhou J, Rao R, Shapiro ME, Tania N, Herron C, Musante CJ, Hughes JH. Model-Informed Drug Development Applications and Opportunities in mRNA-LNP Therapeutics. Clin Pharmacol Ther 2025. [PMID: 40083288 DOI: 10.1002/cpt.3641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 03/03/2025] [Indexed: 03/16/2025]
Abstract
The utilization of lipid nanoparticles (LNP) for encapsulating mRNA has revolutionized the field of therapeutics, enabling the rapid development of COVID-19 vaccines and cancer vaccines. However, the clinical development of mRNA-LNP therapeutics faces numerous challenges due to their complex mechanisms of action and limited clinical experience. To overcome these hurdles, Model-Informed Drug Development (MIDD) emerges as a valuable tool that can be applied to mRNA-LNP therapeutics, facilitating the evaluation of their safety and efficacy through the integration of data from all stages into appropriate modeling and simulation techniques. In this review, we provide an overview of current MIDD applications in mRNA-LNP therapeutics clinical development using in vivo data. A variety of modeling methods are reviewed, including quantitative system pharmacology (QSP), physiologically based pharmacokinetics (PBPK), mechanistic pharmacokinetics/pharmacodynamics (PK/PD), population PK/PD, and model-based meta-analysis (MBMA). Additionally, we compare the differences between mRNA-based therapeutics, small interfering RNA, and adeno-associated virus-based gene therapies in terms of their clinical pharmacology, and discuss the potential for mutual sharing of MIDD knowledge between these therapeutics. Furthermore, we highlight the promising future opportunities for applying MIDD approaches in the development of mRNA-LNP drugs. By emphasizing the importance of applying MIDD knowledge throughout mRNA-LNP therapeutics development, this review aims to encourage stakeholders to recognize the value of MIDD and its potential to enhance the safety and efficacy evaluation of mRNA-LNP therapeutics.
Collapse
Affiliation(s)
| | - Rohit Rao
- Pfizer Inc, Cambridge, Massachusetts, USA
| | | | | | | | | | | |
Collapse
|
12
|
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
|
13
|
Leong KY, Tham SK, Poh CL. Revolutionizing immunization: a comprehensive review of mRNA vaccine technology and applications. Virol J 2025; 22:71. [PMID: 40075519 PMCID: PMC11900334 DOI: 10.1186/s12985-025-02645-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Accepted: 01/30/2025] [Indexed: 03/14/2025] Open
Abstract
Messenger RNA (mRNA) vaccines have emerged as a transformative platform in modern vaccinology. mRNA vaccine is a powerful alternative to traditional vaccines due to their high potency, safety, and efficacy, coupled with the ability for rapid clinical development, scalability and cost-effectiveness in manufacturing. Initially conceptualized in the 1970s, the first study about the effectiveness of a mRNA vaccine against influenza was conducted in 1993. Since then, the development of mRNA vaccines has rapidly gained significance, especially in combating the COVID-19 pandemic. Their unprecedented success during the COVID-19 pandemic, as demonstrated by the Pfizer-BioNTech and Moderna vaccines, highlighted their transformative potential. This review provides a comprehensive analysis of the mRNA vaccine technology, detailing the structure of the mRNA vaccine and its mechanism of action in inducing immunity. Advancements in nanotechnology, particularly lipid nanoparticles (LNPs) as delivery vehicles, have revolutionized the field. The manufacturing processes, including upstream production, downstream purification, and formulation are also reviewed. The clinical progress of mRNA vaccines targeting viruses causing infectious diseases is discussed, emphasizing their versatility and therapeutic potential. Despite their success, the mRNA vaccine platform faces several challenges, including improved stability to reduce dependence on cold chain logistics in transport, enhanced delivery mechanisms to target specific tissues or cells, and addressing the risk of rare adverse events. High costs associated with encapsulation in LNPs and the potential for unequal global access further complicate their widespread adoption. As the world continues to confront emerging viral threats, overcoming these challenges will be essential to fully harness the potential of mRNA vaccines. It is anticipated that mRNA vaccines will play a major role in defining and shaping the future of global health.
Collapse
Affiliation(s)
- Kai Yuan Leong
- MyGenome, ALPS Global Holding Berhad, Kuala Lumpur, Malaysia
| | - Seng Kong Tham
- MyGenome, ALPS Global Holding Berhad, Kuala Lumpur, Malaysia
| | - Chit Laa Poh
- MyGenome, ALPS Global Holding Berhad, Kuala Lumpur, Malaysia.
| |
Collapse
|
14
|
Verwimp S, Wagoner J, Arenas EG, De Coninck L, Abdelnabi R, Hyde JL, Schiffer JT, White JM, Matthijnssens J, Neyts J, Polyak SJ, Delang L. Combinations of approved oral nucleoside analogues confer potent suppression of alphaviruses in vitro and in vivo. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.24.633564. [PMID: 39896535 PMCID: PMC11785157 DOI: 10.1101/2025.01.24.633564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Background Alphaviruses, including chikungunya virus (CHIKV), pose a significant global health threat, yet specific antiviral therapies remain unavailable. Methods We evaluated combinations of three oral directly acting antiviral drugs (sofosbuvir (SOF), molnupiravir (MPV), and favipiravir (FAV)), which are approved for other indications, against CHIKV, Semliki Forest virus (SFV), Sindbis virus (SINV), and Venezuelan Equine Encephalitis virus (VEEV) in vitro and in vivo . We assessed antiviral efficacy in human skin fibroblasts and liver cells, as well as in a mouse model of CHIKV-induced arthritis. Findings In human skin fibroblasts, synergistic antiviral effects were observed for combinations of MPV + SOF and FAV + SOF against CHIKV, and for FAV + SOF against SFV. In human liver cells, FAV + MPV conferred additive to synergistic activity against VEEV and SINV, while SOF synergized with FAV against SINV. In mice, MPV improved CHIKV-induced foot swelling and reduced systemic infectious virus titres. Combination treatment with MPV and SOF significantly reduced swelling and infectious virus titres compared to monotherapies of each drug. Sequencing of CHIKV RNA from joint tissue revealed that MPV caused dose- dependent increases in mutations in the CHIKV genome. Upon combination therapy of MPV with SOF, the number of mutations was significantly lower compared to monotherapy with several higher doses of MPV. Interpretation Combining these approved oral nucleoside analogues confers potent suppression of multiple alphaviruses in vitro and in vivo with enhanced control of viral genetic evolution in face of antiviral pressure. These drug combinations may ultimately lead to the development of potent combinations of pan-family alphavirus inhibitors. Funding This work was supported by a PhD fellowship granted to S.V. by the Research Foundation - Flanders (FWO) (11D5923N). L.D.C. was also supported by Research Foundation - Flanders (FWO) PhD fellowship (11L1325N). Dr. Polyak and Schiffer are partially supported by R01AI121129. Research in Context Evidence before this study: Alphaviruses such as chikungunya virus (CHIKV), Sindbis virus (SINV), and Venezuelan Equine Encephalitis virus (VEEV) are a major threat for global health. Alphaviruses are responsible for debilitating diseases with major public health implications, yet no antiviral drugs are currently approved for treating these virus infections. Existing treatment options are limited to supportive care and are unlikely to provide protection against future outbreaks of other alphaviruses. Previous studies have shown that oral approved nucleoside analogues such as favipiravir (FAV), molnupiravir (MPV), and sofosbuvir (SOF) have antiviral activity against certain RNA viruses, including alphaviruses. However, systematic in vivo evaluations of these drugs and testing of drug combinations in both in vitro and in vivo settings are limited. Added value of this study: This study provides a comprehensive evaluation of combinations of FAV, MPV and SOF against multiple alphaviruses in two human cell lines and a CHIKV mouse model. We demonstrate that certain combinations of these drugs confer synergistic antiviral activity, effectively suppressing CHIKV, SFV, SINV, and VEEV replication in vitro . Moreover, in vivo , we show for the first time that MPV treatment results in reduced CHIKV-induced foot swelling and systemic virus replication. Combining MPV with SOF enhances antiviral activity in mice as compared to monotherapy. By sequencing the viral genome, we show that MPV increases the number of mutations in a dose-dependent manner. Combination therapy of MPV and SOF reduces the number of mutations compared to higher doses of MPV. These findings highlight the potential of nucleoside analogue combinations as a promising therapeutic strategy against alphavirus infections. Implications of all the available evidence: The results of this study suggest that combination therapy with approved nucleoside analogues could provide an effective treatment strategy for alphavirus infections. The observed increased efficacy of drug combinations supports the potential for dose optimization to enhance efficacy while reducing toxicity and development of resistance. Future research should focus on clinical evaluation of these drug combinations, pharmacokinetic studies, and further exploration of their impact on viral evolution. Given the expanding geographical distribution of alphaviruses and the lack of available treatments, these findings provide a foundation for developing pan-alphavirus antiviral therapies that could improve patient outcomes and global outbreak preparedness.
Collapse
|
15
|
Suzuki Y, Miyazaki T, Ida Y, Suzuki T, Itoh Y, Nakao S, Kondo K, Kubara K, Nishioka K, Muto H, Watari R, Hirayama T, Kakiuchi D, Sato S, Inoue S, Uemoto Y, Mukai Y, Hoshino A, Okamoto T, Matsui J. In vivo production of engineered ACE2 decoy protects lungs from SARS-CoV-2 infection. MOLECULAR THERAPY. NUCLEIC ACIDS 2025; 36:102467. [PMID: 40027884 PMCID: PMC11869860 DOI: 10.1016/j.omtn.2025.102467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 01/24/2025] [Indexed: 03/05/2025]
Abstract
Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) variants repeatedly evade the immune system within short periods. Thus, next-generation therapeutics that are resistant to mutations and can be rapidly supplied to individuals in an emergency are required. Here, we designed an mRNA encoding an engineered angiotensin-converting enzyme 2 (ACE2) decoy, 3N39v4, composed of high-affinity ACE2 and a human immunoglobulin G Fc domain. The 3N39v4-encoded mRNA was encapsulated in lipid nanoparticles for efficient in vivo delivery. Systemic delivery of mRNA in mice resulted in a dose-dependent expression of 3N39v4 in plasma (20-261 μg/mL at 1-10 mg/kg) with sufficient tolerability. An improved pharmacokinetic profile of the produced protein was compared to injection of the 3N39v4 protein. In vivo-expressed 3N39v4 exhibited broad neutralization against nine SARS-CoV-2 variants and other sarbecoviruses, including the currently circulating Omicron subvariants JN.1 and BA.2.86. A single intravenous injection of 3N39v4-encoded mRNA resulted in a robust, dose-dependent improvement in the outcomes of mice infected with SARS-CoV-2. The mRNA treatment in monkeys produced 3N39v4 in sera, which inhibited the replication of the authentic viruses. The rapid development of mRNA drugs highlights the potential of mRNA-encoded ACE2 decoys in emergencies to combat diverse SARS-CoV-2 variants, including future variants.
Collapse
Affiliation(s)
- Yuta Suzuki
- Tsukuba Research Laboratories, Eisai Co., Ltd., Ibaraki 300-2635, Japan
| | - Takayuki Miyazaki
- Tsukuba Research Laboratories, Eisai Co., Ltd., Ibaraki 300-2635, Japan
| | - Yoko Ida
- Kobe Research Laboratories, Eisai Co., Ltd., Kobe 650-0047, Japan
| | - Tatsuya Suzuki
- Department of Microbiology, Juntendo University School of Medicine, Tokyo 113-8421, Japan
- Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
| | - Yumi Itoh
- Department of Microbiology, Juntendo University School of Medicine, Tokyo 113-8421, Japan
- Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
| | - Shuto Nakao
- Department of Microbiology, Juntendo University School of Medicine, Tokyo 113-8421, Japan
| | - Keita Kondo
- Tsukuba Research Laboratories, Eisai Co., Ltd., Ibaraki 300-2635, Japan
| | - Kenji Kubara
- Tsukuba Research Laboratories, Eisai Co., Ltd., Ibaraki 300-2635, Japan
| | - Keisuke Nishioka
- Department of Infectious Diseases, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Hiroki Muto
- Tsukuba Research Laboratories, Eisai Co., Ltd., Ibaraki 300-2635, Japan
| | - Ryuji Watari
- Tsukuba Research Laboratories, Eisai Co., Ltd., Ibaraki 300-2635, Japan
| | | | - Dai Kakiuchi
- Tsukuba Research Laboratories, Eisai Co., Ltd., Ibaraki 300-2635, Japan
| | - Shinya Sato
- Tsukuba Research Laboratories, Eisai Co., Ltd., Ibaraki 300-2635, Japan
| | - Satoshi Inoue
- Tsukuba Research Laboratories, Eisai Co., Ltd., Ibaraki 300-2635, Japan
| | - Yoshifumi Uemoto
- Tsukuba Research Laboratories, Eisai Co., Ltd., Ibaraki 300-2635, Japan
| | - Yohei Mukai
- Kobe Research Laboratories, Eisai Co., Ltd., Kobe 650-0047, Japan
| | - Atsushi Hoshino
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Toru Okamoto
- Department of Microbiology, Juntendo University School of Medicine, Tokyo 113-8421, Japan
- Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
| | - Junji Matsui
- Tsukuba Research Laboratories, Eisai Co., Ltd., Ibaraki 300-2635, Japan
| |
Collapse
|
16
|
Ostrowsky JT, Katzelnick LC, Bourne N, Barrett ADT, Thomas SJ, Diamond MS, Beasley DWC, Harris E, Wilder-Smith A, Leighton T, Mehr AJ, Moua NM, Ulrich AK, Cehovin A, Fay PC, Golding JP, Moore KA, Osterholm MT, Lackritz EM. Zika virus vaccines and monoclonal antibodies: a priority agenda for research and development. THE LANCET. INFECTIOUS DISEASES 2025:S1473-3099(24)00750-3. [PMID: 40024262 DOI: 10.1016/s1473-3099(24)00750-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 11/06/2024] [Accepted: 11/08/2024] [Indexed: 03/04/2025]
Abstract
The 2015-16 Zika virus epidemic in the Americas drew global attention to Zika virus infection as a cause of microcephaly and Guillain-Barré syndrome. The epidemic highlighted the urgent need for preventive measures, including vaccines and monoclonal antibodies (mAbs). However, nearly 9 years later, no licensed Zika virus vaccines or mAbs are available, leaving the world's populations unprotected from ongoing disease transmission and future epidemics. The current low Zika virus incidence and unpredictability of future outbreaks complicates prospects for evaluation, licensure, and commercial viability of Zika virus vaccines and mAbs. We conducted an extensive review of Zika virus vaccines and mAbs in development, identifying 16 vaccines in phase 1 or phase 2 trials and three mAbs in phase 1 trials, and convened a 2-day meeting of 130 global Zika virus experts to discuss research priorities to advance their development. This Series paper summarises a priority research agenda to address key knowledge gaps and accelerate the licensure of Zika virus vaccines and mAbs for global use.
Collapse
Affiliation(s)
- Julia T Ostrowsky
- Center for Infectious Disease Research and Policy, University of Minnesota, Minneapolis, MN, USA
| | - Leah C Katzelnick
- Viral Epidemiology and Immunity Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Nigel Bourne
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX, USA; Department of Pediatrics, University of Texas Medical Branch, Galveston, TX, USA
| | - Alan D T Barrett
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX, USA; Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
| | - Stephen J Thomas
- Department of Microbiology and Immunology, State University of New York (SUNY) Upstate Medical University, Syracuse, NY, USA; Institute for Global Health and Translational Sciences, State University of New York (SUNY) Upstate Medical University, Syracuse, NY, USA
| | - Michael S Diamond
- Department of Pathology and Immunology and Center for Genome Sciences, Lab and Genomic Medicine, Washington University School of Medicine, St Louis, MO, USA
| | - David W C Beasley
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX, USA; Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Eva Harris
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California Berkeley, Berkeley, CA, USA
| | - Annelies Wilder-Smith
- Immunization, Vaccines, and Biologicals, World Health Organization, Geneva, Switzerland
| | - Tabitha Leighton
- Center for Infectious Disease Research and Policy, University of Minnesota, Minneapolis, MN, USA
| | - Angela J Mehr
- Center for Infectious Disease Research and Policy, University of Minnesota, Minneapolis, MN, USA
| | - Nicolina M Moua
- Center for Infectious Disease Research and Policy, University of Minnesota, Minneapolis, MN, USA
| | - Angela K Ulrich
- Center for Infectious Disease Research and Policy, University of Minnesota, Minneapolis, MN, USA
| | - Ana Cehovin
- Infectious Disease Strategic Programme, Wellcome Trust, London, UK
| | - Petra C Fay
- Infectious Disease Strategic Programme, Wellcome Trust, London, UK
| | | | - Kristine A Moore
- Center for Infectious Disease Research and Policy, University of Minnesota, Minneapolis, MN, USA
| | - Michael T Osterholm
- Center for Infectious Disease Research and Policy, University of Minnesota, Minneapolis, MN, USA
| | - Eve M Lackritz
- Center for Infectious Disease Research and Policy, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
17
|
Lantz AM, Baxter VK. Neuropathogenesis of Old World Alphaviruses: Considerations for the Development of Medical Countermeasures. Viruses 2025; 17:261. [PMID: 40007016 PMCID: PMC11860675 DOI: 10.3390/v17020261] [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: 01/17/2025] [Revised: 02/08/2025] [Accepted: 02/12/2025] [Indexed: 02/27/2025] Open
Abstract
Chikungunya virus (CHIKV) and other alphaviruses that primarily induce arthritogenic disease in humans, known as "Old World" alphaviruses, present an emerging public health concern as geographic ranges of mosquito vectors expand due to climate change. While a vaccine against CHIKV has recently been approved by several countries in North America and Europe, access to effective preventative countermeasures against disease induced by Old World alphaviruses remains elusive for the most vulnerable populations. Furthermore, treatment options continue to be limited to supportive care. Atypical neurological disease manifestations caused by Old World alphaviruses, which make up as many as 25% of the cases in some CHIKV outbreaks, present special challenges when considering strategies for developing effective countermeasures. This review focuses on Old World alphaviruses, specifically CHIKV, Ross River virus, O'nyoug-nyoug virus, and Mayaro virus, concentrating on the atypical neurological disease manifestations they may cause. Our current understanding of Old World alphavirus neuropathogenesis, gained from human cases and preclinical animal models, is discussed, including viral and host factors' roles in disease development. The current state of alphavirus preventatives and treatments, both virus-targeting and host-directed therapies, is then summarized and discussed in the context of addressing neurological disease induced by Old World alphaviruses.
Collapse
|
18
|
Nguyen T, Gebo C, Lu J, Popoola DO, Thomas SJ, Li Y, Waickman AT. Development and optimization of an mRNA-vectored single-chain IgA1 isotype monoclonal antibody with potential to treat or prevent dengue virus infection. Antiviral Res 2025; 234:106078. [PMID: 39778815 DOI: 10.1016/j.antiviral.2025.106078] [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: 12/18/2024] [Accepted: 01/06/2025] [Indexed: 01/11/2025]
Abstract
Dengue virus (DENV) is a rapidly expanding infectious disease threat that causes an estimated 100 million symptomatic infections every year. A barrier to preventing DENV infections with traditional vaccines or prophylactic monoclonal antibody (mAb) therapies is the phenomenon of Antibody-Dependent Enhancement (ADE), wherein sub-neutralizing levels of DENV-specific IgG antibodies can enhance infection and pathogenesis rather than providing protection from disease. Fortunately, IgG is not the only antibody isotype capable of binding and neutralizing DENV, as DENV-specific IgA1 isotype mAbs can bind and neutralize DENV while without exhibiting any ADE activity. However, the development of IgA1-based mAb therapies is currently hindered by inefficient in vitro expression systems and the lack of saleable purification platforms. Accordingly, alternative delivery modalities are required to realize the therapeutic potential of IgA-based infectious-disease therapies. In this study we describe the development and optimization of a DENV-specific single-chain IgA construct that retains the desirable biological properties of the parental IgA mAb yet is compatible with efficient in vivo delivery with a novel/liver-tropic lipid nanoparticle. We propose that this platform is uniquely and exceptionally well suited for preventing and/or treating DENV infections and may have broad applicability in the greater infectious disease space in situations where the use of IgG isotype mAbs may be counterindicated.
Collapse
MESH Headings
- Dengue/prevention & control
- Dengue/therapy
- Dengue/drug therapy
- Dengue/immunology
- Dengue Virus/immunology
- Animals
- Antibodies, Viral/immunology
- Antibodies, Viral/genetics
- Antibodies, Viral/therapeutic use
- Antibodies, Monoclonal/genetics
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/therapeutic use
- Humans
- Immunoglobulin A/genetics
- Immunoglobulin A/immunology
- Immunoglobulin A/therapeutic use
- Antibody-Dependent Enhancement
- Antibodies, Neutralizing/immunology
- Antibodies, Neutralizing/therapeutic use
- Antibodies, Neutralizing/genetics
- RNA, Messenger/genetics
- Single-Chain Antibodies/genetics
- Single-Chain Antibodies/immunology
- Single-Chain Antibodies/therapeutic use
- Mice
- Nanoparticles
Collapse
Affiliation(s)
- Thanh Nguyen
- Department of Microbiology and Immunology, State University of New York Upstate Medical University, Syracuse, NY, 13210, USA
| | - Chad Gebo
- Department of Microbiology and Immunology, State University of New York Upstate Medical University, Syracuse, NY, 13210, USA
| | - Joseph Lu
- Department of Microbiology and Immunology, State University of New York Upstate Medical University, Syracuse, NY, 13210, USA; Institute for Global Health and Translational Sciences, State University of New York Upstate Medical University, Syracuse, NY, 13210, USA
| | - David O Popoola
- Department of Pharmacology, State University of New York Upstate Medical University, Syracuse, NY, 13210, USA
| | - Stephen J Thomas
- Department of Microbiology and Immunology, State University of New York Upstate Medical University, Syracuse, NY, 13210, USA; Institute for Global Health and Translational Sciences, State University of New York Upstate Medical University, Syracuse, NY, 13210, USA
| | - Yamin Li
- Department of Pharmacology, State University of New York Upstate Medical University, Syracuse, NY, 13210, USA.
| | - Adam T Waickman
- Department of Microbiology and Immunology, State University of New York Upstate Medical University, Syracuse, NY, 13210, USA; Institute for Global Health and Translational Sciences, State University of New York Upstate Medical University, Syracuse, NY, 13210, USA.
| |
Collapse
|
19
|
Howard EL, Goens MM, Susta L, Patel A, Wootton SK. Anti-Drug Antibody Response to Therapeutic Antibodies and Potential Mitigation Strategies. Biomedicines 2025; 13:299. [PMID: 40002712 PMCID: PMC11853408 DOI: 10.3390/biomedicines13020299] [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: 12/28/2024] [Revised: 01/21/2025] [Accepted: 01/22/2025] [Indexed: 02/27/2025] Open
Abstract
The development of anti-drug antibodies (ADAs) against therapeutic monoclonal antibodies (mAbs) poses significant challenges in the efficacy and safety of these treatments. ADAs can lead to adverse immune reactions, reduced drug efficacy, and increased clearance of therapeutic antibodies. This paper reviews the formation and mechanisms of ADAs, explores factors contributing to their development, and discusses potential strategies to mitigate ADA responses. Current and emerging strategies to reduce ADA formation include in silico and in vitro prediction tools, deimmunization techniques, antibody engineering, and various drug delivery methods. Additionally, novel approaches such as tolerogenic nanoparticles, oral tolerance, and in vivo delivery of therapeutic proteins via viral vectors and synthetic mRNA or DNA are explored. These strategies have the potential to enhance clinical outcomes of mAb therapies by minimizing immunogenicity and improving patient safety. Further research and innovation in this field are critical to overcoming the ongoing challenges of ADA responses in therapeutic antibody development.
Collapse
Affiliation(s)
- Erin L. Howard
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada; (E.L.H.)
| | - Melanie M. Goens
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada; (E.L.H.)
| | - Leonardo Susta
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada; (E.L.H.)
| | - Ami Patel
- The Wistar Institute, Philadelphia, PA 19104, USA
| | - Sarah K. Wootton
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada; (E.L.H.)
| |
Collapse
|
20
|
Ullah A, Khan M, Zhang Y, Shafiq M, Ullah M, Abbas A, Xianxiang X, Chen G, Diao Y. Advancing Therapeutic Strategies with Polymeric Drug Conjugates for Nucleic Acid Delivery and Treatment. Int J Nanomedicine 2025; 20:25-52. [PMID: 39802382 PMCID: PMC11717654 DOI: 10.2147/ijn.s429279] [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: 09/13/2024] [Accepted: 11/26/2024] [Indexed: 01/16/2025] Open
Abstract
The effective clinical translation of messenger RNA (mRNA), small interfering RNA (siRNA), and microRNA (miRNA) for therapeutic purposes hinges on the development of efficient delivery systems. Key challenges include their susceptibility to degradation, limited cellular uptake, and inefficient intracellular release. Polymeric drug conjugates (PDCs) offer a promising solution, combining the benefits of polymeric carriers and therapeutic agents for targeted delivery and treatment. This comprehensive review explores the clinical translation of nucleic acid therapeutics, focusing on polymeric drug conjugates. It investigates how these conjugates address delivery obstacles, enhance systemic circulation, reduce immunogenicity, and provide controlled release, improving safety profiles. The review delves into the conjugation strategies, preparation methods, and various classes of PDCs, as well as strategic design, highlighting their role in nucleic acid delivery. Applications of PDCs in treating diseases such as cancer, immune disorders, and fibrosis are also discussed. Despite significant advancements, challenges in clinical adoption persist. The review concludes with insights into future directions for this transformative technology, underscoring the potential of PDCs to advance nucleic acid-based therapies and combat infectious diseases significantly.
Collapse
Affiliation(s)
- Aftab Ullah
- School of Medicine, Huaqiao University, Quanzhou, Fujian, People’s Republic of China
| | - Marina Khan
- Department of Biotechnology and Genetic Engineering, Kohat University of Science and Technology, Kohat, Pakistan
| | - Yibang Zhang
- School of Pharmacy, Jiangsu University, Zhenjiang, Jiangsu, People’s Republic of China
| | - Muhammad Shafiq
- Research Institute of Clinical Pharmacy, Department of Pharmacology, Shantou University Medical College, Shantou, Guangdong, People’s Republic of China
| | - Mohsan Ullah
- School of Medicine, Huaqiao University, Quanzhou, Fujian, People’s Republic of China
| | - Azar Abbas
- Institute of Medicine, Shenzhen Institute of Advanced Technology, Shenzhen, Guangdong, People’s Republic of China
| | - Xu Xianxiang
- School of Medicine, Huaqiao University, Quanzhou, Fujian, People’s Republic of China
| | - Gang Chen
- School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao, Shandong, People’s Republic of China
- Qingdao Central Hospital, University of Health and Rehabilitation Sciences (Qingdao central Medical Group), Qingdao, Shandong, People’s Republic of China
| | - Yong Diao
- School of Medicine, Huaqiao University, Quanzhou, Fujian, People’s Republic of China
| |
Collapse
|
21
|
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
|
22
|
Patel A, Rosenke K, Parzych EM, Feldmann F, Bharti S, Griffin AJ, Schouest B, Lewis M, Choi J, Chokkalingam N, Machado V, Smith BJ, Frase D, Ali AR, Lovaglio J, Nguyen B, Hanley PW, Walker SN, Gary EN, Kulkarni A, Generotti A, Francica JR, Rosenthal K, Kulp DW, Esser MT, Smith TRF, Shaia C, Weiner DB, Feldmann H. In vivo delivery of engineered synthetic DNA-encoded SARS-CoV-2 monoclonal antibodies for pre-exposure prophylaxis in non-human primates. Emerg Microbes Infect 2024; 13:2294860. [PMID: 38165394 PMCID: PMC10903752 DOI: 10.1080/22221751.2023.2294860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 12/11/2023] [Indexed: 01/03/2024]
Abstract
COVID-19 remains a major public health concern. Monoclonal antibodies have received emergency use authorization (EUA) for pre-exposure prophylaxis against COVID-19 among high-risk groups for treatment of mild to moderate COVID-19. In addition to recombinant biologics, engineered synthetic DNA-encoded antibodies (DMAb) are an important strategy for direct in vivo delivery of protective mAb. A DMAb cocktail was synthetically engineered to encode the immunoglobulin heavy and light chains of two different two different Fc-engineered anti-SARS-CoV-2 antibodies. The DMAbs were designed to enhance in vivo expression and delivered intramuscularly to cynomolgus and rhesus macaques with a modified in vivo delivery regimen. Serum levels were detected in macaques, along with specific binding to SARS-CoV-2 spike receptor binding domain protein and neutralization of multiple SARS-CoV-2 variants of concern in pseudovirus and authentic live virus assays. Prophylactic administration was protective in rhesus macaques against signs of SARS-CoV-2 (USA-WA1/2020) associated disease in the lungs. Overall, the data support further study of DNA-encoded antibodies as an additional delivery mode for prevention of COVID-19 severe disease. These data have implications for human translation of gene-encoded mAbs for emerging infectious diseases and low dose mAb delivery against COVID-19.
Collapse
Affiliation(s)
- Ami Patel
- Vaccine and Immunotherapy Center, The Wistar Institute of Anatomy and Biology, Philadelphia, PA, USA
| | - Kyle Rosenke
- Rocky Mountain Laboratories, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Elizabeth M. Parzych
- Vaccine and Immunotherapy Center, The Wistar Institute of Anatomy and Biology, Philadelphia, PA, USA
| | - Friederike Feldmann
- Rocky Mountain Laboratories, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Suman Bharti
- Vaccine and Immunotherapy Center, The Wistar Institute of Anatomy and Biology, Philadelphia, PA, USA
| | - Amanda J. Griffin
- Rocky Mountain Laboratories, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | | | - Matt Lewis
- Rocky Mountain Laboratories, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Jihae Choi
- Vaccine and Immunotherapy Center, The Wistar Institute of Anatomy and Biology, Philadelphia, PA, USA
| | - Neethu Chokkalingam
- Vaccine and Immunotherapy Center, The Wistar Institute of Anatomy and Biology, Philadelphia, PA, USA
| | | | - Brian J. Smith
- Rocky Mountain Laboratories, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Drew Frase
- Vaccine and Immunotherapy Center, The Wistar Institute of Anatomy and Biology, Philadelphia, PA, USA
| | - Ali R. Ali
- Vaccine and Immunotherapy Center, The Wistar Institute of Anatomy and Biology, Philadelphia, PA, USA
| | - Jamie Lovaglio
- Rocky Mountain Laboratories, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Brian Nguyen
- Inovio Pharmaceuticals, Plymouth Meeting, PA, USA
| | - Patrick W. Hanley
- Rocky Mountain Laboratories, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Susanne N. Walker
- Vaccine and Immunotherapy Center, The Wistar Institute of Anatomy and Biology, Philadelphia, PA, USA
| | - Ebony N. Gary
- Vaccine and Immunotherapy Center, The Wistar Institute of Anatomy and Biology, Philadelphia, PA, USA
| | - Abhijeet Kulkarni
- Vaccine and Immunotherapy Center, The Wistar Institute of Anatomy and Biology, Philadelphia, PA, USA
| | | | - Joseph R. Francica
- Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Kim Rosenthal
- Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Daniel W. Kulp
- Vaccine and Immunotherapy Center, The Wistar Institute of Anatomy and Biology, Philadelphia, PA, USA
| | - Mark T. Esser
- Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | | | - Carl Shaia
- Rocky Mountain Laboratories, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - David B. Weiner
- Vaccine and Immunotherapy Center, The Wistar Institute of Anatomy and Biology, Philadelphia, PA, USA
| | - Heinz Feldmann
- Rocky Mountain Laboratories, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| |
Collapse
|
23
|
Haque MA, Shrestha A, Mikelis CM, Mattheolabakis G. Comprehensive analysis of lipid nanoparticle formulation and preparation for RNA delivery. Int J Pharm X 2024; 8:100283. [PMID: 39309631 PMCID: PMC11415597 DOI: 10.1016/j.ijpx.2024.100283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 08/21/2024] [Accepted: 09/07/2024] [Indexed: 09/25/2024] Open
Abstract
Nucleic acid-based therapeutics are a common approach that is increasingly popular for a wide spectrum of diseases. Lipid nanoparticles (LNPs) are promising delivery carriers that provide RNA stability, with strong transfection efficiency, favorable and tailorable pharmacokinetics, limited toxicity, and established translatability. In this review article, we describe the lipid-based delivery systems, focusing on lipid nanoparticles, the need of their use, provide a comprehensive analysis of each component, and highlight the advantages and disadvantages of the existing manufacturing processes. We further summarize the ongoing and completed clinical trials utilizing LNPs, indicating important aspects/questions worth of investigation, and analyze the future perspectives of this significant and promising therapeutic approach.
Collapse
Affiliation(s)
- Md. Anamul Haque
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, USA
| | - Archana Shrestha
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, USA
| | - Constantinos M. Mikelis
- Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Patras 26504, Greece
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - George Mattheolabakis
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, USA
| |
Collapse
|
24
|
Bader J, Brigger F, Leroux JC. Extracellular vesicles versus lipid nanoparticles for the delivery of nucleic acids. Adv Drug Deliv Rev 2024; 215:115461. [PMID: 39490384 DOI: 10.1016/j.addr.2024.115461] [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: 10/21/2024] [Accepted: 10/23/2024] [Indexed: 11/05/2024]
Abstract
Extracellular vesicles (EVs) are increasingly investigated for delivering nucleic acid (NA) therapeutics, leveraging their natural role in transporting NA and protein-based cargo in cell-to-cell signaling. Their synthetic counterparts, lipid nanoparticles (LNPs), have been developed over the past decades as NA carriers, culminating in the approval of several marketed formulations such as patisiran/Onpattro® and the mRNA-1273/BNT162 COVID-19 vaccines. The success of LNPs has sparked efforts to develop innovative technologies to target extrahepatic organs, and to deliver novel therapeutic modalities, such as tools for in vivo gene editing. Fueled by the recent advancements in both fields, this review aims to provide a comprehensive overview of the basic characteristics of EV and LNP-based NA delivery systems, from EV biogenesis to structural properties of LNPs. It addresses the primary challenges encountered in utilizing these nanocarriers from a drug formulation and delivery perspective. Additionally, biodistribution profiles, in vitro and in vivo transfection outcomes, as well as their status in clinical trials are compared. Overall, this review provides insights into promising research avenues and potential dead ends for EV and LNP-based NA delivery systems.
Collapse
Affiliation(s)
- Johannes Bader
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, 8093 Zurich, Switzerland
| | - Finn Brigger
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, 8093 Zurich, Switzerland
| | - Jean-Christophe Leroux
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, 8093 Zurich, Switzerland.
| |
Collapse
|
25
|
Freppel W, Silva LA, Stapleford KA, Herrero LJ. Pathogenicity and virulence of chikungunya virus. Virulence 2024; 15:2396484. [PMID: 39193780 PMCID: PMC11370967 DOI: 10.1080/21505594.2024.2396484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 08/09/2024] [Accepted: 08/14/2024] [Indexed: 08/29/2024] Open
Abstract
Chikungunya virus (CHIKV) is a mosquito-transmitted, RNA virus that causes an often-severe musculoskeletal illness characterized by fever, joint pain, and a range of debilitating symptoms. The virus has re-emerged as a global health threat in recent decades, spreading from its origin in Africa across Asia and the Americas, leading to widespread outbreaks impacting millions of people. Despite more than 50 years of research into the pathogenesis of CHIKV, there is still no curative treatment available. Current management of CHIKV infections primarily involves providing supportive care to alleviate symptoms and improve the patient's quality of life. Given the ongoing threat of CHIKV, there is an urgent need to better understand its pathogenesis. This understanding is crucial for deciphering the mechanisms underlying the disease and for developing effective strategies for both prevention and management. This review aims to provide a comprehensive overview of CHIKV and its pathogenesis, shedding light on the complex interactions of viral genetics, host factors, immune responses, and vector-related factors. By exploring these intricate connections, the review seeks to contribute to the knowledge base surrounding CHIKV, offering insights that may ultimately lead to more effective prevention and management strategies for this re-emerging global health threat.
Collapse
Affiliation(s)
- Wesley Freppel
- Institute for Biomedicine and Glycomics, Gold Coast Campus, Griffith University, Southport, Australia
| | - Laurie A. Silva
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Kenneth A. Stapleford
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, USA
| | - Lara J. Herrero
- Institute for Biomedicine and Glycomics, Gold Coast Campus, Griffith University, Southport, Australia
| |
Collapse
|
26
|
Sun X, Setrerrahmane S, Li C, Hu J, Xu H. Nucleic acid drugs: recent progress and future perspectives. Signal Transduct Target Ther 2024; 9:316. [PMID: 39609384 PMCID: PMC11604671 DOI: 10.1038/s41392-024-02035-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 09/20/2024] [Accepted: 10/25/2024] [Indexed: 11/30/2024] Open
Abstract
High efficacy, selectivity and cellular targeting of therapeutic agents has been an active area of investigation for decades. Currently, most clinically approved therapeutics are small molecules or protein/antibody biologics. Targeted action of small molecule drugs remains a challenge in medicine. In addition, many diseases are considered 'undruggable' using standard biomacromolecules. Many of these challenges however, can be addressed using nucleic therapeutics. Nucleic acid drugs (NADs) are a new generation of gene-editing modalities characterized by their high efficiency and rapid development, which have become an active research topic in new drug development field. However, many factors, including their low stability, short half-life, high immunogenicity, tissue targeting, cellular uptake, and endosomal escape, hamper the delivery and clinical application of NADs. Scientists have used chemical modification techniques to improve the physicochemical properties of NADs. In contrast, modified NADs typically require carriers to enter target cells and reach specific intracellular locations. Multiple delivery approaches have been developed to effectively improve intracellular delivery and the in vivo bioavailability of NADs. Several NADs have entered the clinical trial recently, and some have been approved for therapeutic use in different fields. This review summarizes NADs development and evolution and introduces NADs classifications and general delivery strategies, highlighting their success in clinical applications. Additionally, this review discusses the limitations and potential future applications of NADs as gene therapy candidates.
Collapse
Affiliation(s)
- Xiaoyi Sun
- Jiangsu Province Engineering Research Center of Synthetic Peptide Drug Discovery and Evaluation, China Pharmaceutical University, Nanjing, 210009, China
| | | | - Chencheng Li
- Jiangsu Province Engineering Research Center of Synthetic Peptide Drug Discovery and Evaluation, China Pharmaceutical University, Nanjing, 210009, China
| | - Jialiang Hu
- Jiangsu Province Engineering Research Center of Synthetic Peptide Drug Discovery and Evaluation, China Pharmaceutical University, Nanjing, 210009, China
| | - Hanmei Xu
- Jiangsu Province Engineering Research Center of Synthetic Peptide Drug Discovery and Evaluation, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
27
|
Luo F, Xu C, Zhang C, Tan A, Lu D, Luo P, Cheng P, Zhang W, Bai L, Yu C, Sun S, Zeng H, Zou Q. mRNA-based platform for preventing and treating Staphylococcus aureus by targeted staphylococcal enterotoxin B. Front Immunol 2024; 15:1490044. [PMID: 39640268 PMCID: PMC11617584 DOI: 10.3389/fimmu.2024.1490044] [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: 09/02/2024] [Accepted: 10/29/2024] [Indexed: 12/07/2024] Open
Abstract
Staphylococcus aureus (S. aureus) possesses numerous virulence factors, with the increasing prevalence of drug-resistant strains heightening the threat posed by this pathogen. Staphylococcal enterotoxin B (SEB), a highly conserved toxin secreted by S. aureus, is also recognized as a potential bioweapon with super-antigenic activity. SEB represents a promising target in efforts to combat infections caused by S. aureus. We developed mRNA-based vaccine and antibody targeting SEB for both prophylactic and therapeutic purposes in varying S. aureus infection conditions. The mSEB mRNA vaccine (10 μg per mouse) induces more robust and persistent immune responses, including higher antibody titers and specific cellular immune responses, compared to immunization with 30 μg of mSEB protein adjuvanted with aluminum phosphate. Additionally, the anti-SEB mRNA antibody maintains secretion of anti-SEB monoclonal antibody (mAb) with a dosage that is 10 times lower than purified protein administration. The mRNA-based antibody exhibits superior pharmacokinetic profiles compared to its protein counterparts, efficiently neutralizing SEB and clearing S. aureus from circulation. Both the mRNA vaccine and mRNA antibody demonstrate preventive and therapeutic effects by eliciting specific immune responses and generating high-affinity antibodies in mice. We have laid the groundwork for the development and evaluation of mRNA-based vaccines and antibodies targeting SEB produced by S. aureus. Our studies demonstrate that these approaches are more effective than traditional protein-based vaccines and antibodies in terms of inducing immune responses, pharmacokinetics, and their prophylactic or therapeutic efficacy against S. aureus infections.
Collapse
Affiliation(s)
- Fumei Luo
- School of Pharmacy, University of South China, Hunan, China
- National Engineering Research Center of Immunological Products, Third Military Medical University, Chongqing, China
| | - Chuanfei Xu
- National Engineering Research Center of Immunological Products, Third Military Medical University, Chongqing, China
| | - Chengwen Zhang
- Medical Research Institute, Southwest University, Chongqing, China
| | - Aomo Tan
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, China
| | - Dongshui Lu
- National Engineering Research Center of Immunological Products, Third Military Medical University, Chongqing, China
| | - Ping Luo
- National Engineering Research Center of Immunological Products, Third Military Medical University, Chongqing, China
| | - Ping Cheng
- National Engineering Research Center of Immunological Products, Third Military Medical University, Chongqing, China
| | - Weijun Zhang
- National Engineering Research Center of Immunological Products, Third Military Medical University, Chongqing, China
| | - Lijuan Bai
- College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Cuiyun Yu
- School of Pharmacy, University of South China, Hunan, China
| | - Si Sun
- National Engineering Research Center of Immunological Products, Third Military Medical University, Chongqing, China
| | - Hao Zeng
- National Engineering Research Center of Immunological Products, Third Military Medical University, Chongqing, China
| | - Quanming Zou
- National Engineering Research Center of Immunological Products, Third Military Medical University, Chongqing, China
| |
Collapse
|
28
|
Shi Y, Shi M, Wang Y, You J. Progress and prospects of mRNA-based drugs in pre-clinical and clinical applications. Signal Transduct Target Ther 2024; 9:322. [PMID: 39543114 PMCID: PMC11564800 DOI: 10.1038/s41392-024-02002-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 09/03/2024] [Accepted: 09/26/2024] [Indexed: 11/17/2024] Open
Abstract
In the last decade, messenger ribonucleic acid (mRNA)-based drugs have gained great interest in both immunotherapy and non-immunogenic applications. This surge in interest can be largely attributed to the demonstration of distinct advantages offered by various mRNA molecules, alongside the rapid advancements in nucleic acid delivery systems. It is noteworthy that the immunogenicity of mRNA drugs presents a double-edged sword. In the context of immunotherapy, extra supplementation of adjuvant is generally required for induction of robust immune responses. Conversely, in non-immunotherapeutic scenarios, immune activation is unwanted considering the host tolerability and high expression demand for mRNA-encoded functional proteins. Herein, mainly focused on the linear non-replicating mRNA, we overview the preclinical and clinical progress and prospects of mRNA medicines encompassing vaccines and other therapeutics. We also highlight the importance of focusing on the host-specific variations, including age, gender, pathological condition, and concurrent medication of individual patient, for maximized efficacy and safety upon mRNA administration. Furthermore, we deliberate on the potential challenges that mRNA drugs may encounter in the realm of disease treatment, the current endeavors of improvement, as well as the application prospects for future advancements. Overall, this review aims to present a comprehensive understanding of mRNA-based therapies while illuminating the prospective development and clinical application of mRNA drugs.
Collapse
Affiliation(s)
- Yingying Shi
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, P. R. China
| | - Meixing Shi
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, P. R. China
| | - Yi Wang
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, P. R. China.
| | - Jian You
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, P. R. China.
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, 79 Qingchun Road, Shangcheng District, Hangzhou, Zhejiang, P. R. China.
- The First Affiliated Hospital, College of Medicine, Zhejiang University, 79 QingChun Road, Hangzhou, Zhejiang, P. R. China.
- Jinhua Institute of Zhejiang University, 498 Yiwu Street, Jinhua, Zhejiang, P. R. China.
| |
Collapse
|
29
|
Chaudhary N, Kasiewicz LN, Newby AN, Arral ML, Yerneni SS, Melamed JR, LoPresti ST, Fein KC, Strelkova Petersen DM, Kumar S, Purwar R, Whitehead KA. Amine headgroups in ionizable lipids drive immune responses to lipid nanoparticles by binding to the receptors TLR4 and CD1d. Nat Biomed Eng 2024; 8:1483-1498. [PMID: 39363106 PMCID: PMC11863198 DOI: 10.1038/s41551-024-01256-w] [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: 11/04/2022] [Accepted: 09/05/2024] [Indexed: 10/05/2024]
Abstract
Lipid nanoparticles (LNPs) are the most clinically advanced delivery vehicle for RNA therapeutics, partly because of established lipid structure-activity relationships focused on formulation potency. Yet such knowledge has not extended to LNP immunogenicity. Here we show that the innate and adaptive immune responses elicited by LNPs are linked to their ionizable lipid chemistry. Specifically, we show that the amine headgroups in ionizable lipids drive LNP immunogenicity by binding to Toll-like receptor 4 and CD1d and by promoting lipid-raft formation. Immunogenic LNPs favour a type-1 T-helper-cell-biased immune response marked by increases in the immunoglobulins IgG2c and IgG1 and in the pro-inflammatory cytokines tumour necrosis factor, interferon γ and the interleukins IL-6 and IL-2. Notably, the inflammatory signals originating from these receptors inhibit the production of anti-poly(ethylene glycol) IgM antibodies, preventing the often-observed loss of efficacy in the LNP-mediated delivery of siRNA and mRNA. Moreover, we identified computational methods for the prediction of the structure-dependent innate and adaptive responses of LNPs. Our findings may help accelerate the discovery of well-tolerated ionizable lipids suitable for repeated dosing.
Collapse
Affiliation(s)
- Namit Chaudhary
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Lisa N Kasiewicz
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Alexandra N Newby
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Mariah L Arral
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA
| | | | - Jilian R Melamed
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Samuel T LoPresti
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Katherine C Fein
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA
| | | | - Sushant Kumar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Rahul Purwar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Kathryn A Whitehead
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA.
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA.
| |
Collapse
|
30
|
Pardi N, Krammer F. mRNA vaccines for infectious diseases - advances, challenges and opportunities. Nat Rev Drug Discov 2024; 23:838-861. [PMID: 39367276 DOI: 10.1038/s41573-024-01042-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/27/2024] [Indexed: 10/06/2024]
Abstract
The concept of mRNA-based vaccines emerged more than three decades ago. Groundbreaking discoveries and technological advancements over the past 20 years have resolved the major roadblocks that initially delayed application of this new vaccine modality. The rapid development of nucleoside-modified COVID-19 mRNA vaccines demonstrated that this immunization platform is easy to develop, has an acceptable safety profile and can be produced at a large scale. The flexibility and ease of antigen design have enabled mRNA vaccines to enter development for a wide range of viruses as well as for various bacteria and parasites. However, gaps in our knowledge limit the development of next-generation mRNA vaccines with increased potency and safety. A deeper understanding of the mechanisms of action of mRNA vaccines, application of novel technologies enabling rational antigen design, and innovative vaccine delivery strategies and vaccination regimens will likely yield potent novel vaccines against a wide range of pathogens.
Collapse
Affiliation(s)
- Norbert Pardi
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Ignaz Semmelweis Institute, Interuniversity Institute for Infection Research, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
31
|
Eftekhari Z, Zohrabi H, Oghalaie A, Ebrahimi T, Shariati FS, Behdani M, Kazemi-Lomedasht F. Advancements and challenges in mRNA and ribonucleoprotein-based therapies: From delivery systems to clinical applications. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102313. [PMID: 39281702 PMCID: PMC11402252 DOI: 10.1016/j.omtn.2024.102313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/18/2024]
Abstract
The use of mRNA and ribonucleoproteins (RNPs) as therapeutic agents is a promising strategy for treating diseases such as cancer and infectious diseases. This review provides recent advancements and challenges in mRNA- and RNP-based therapies, focusing on delivery systems such as lipid nanoparticles (LNPs), which ensure efficient delivery to target cells. Strategies such as microfluidic devices are employed to prepare LNPs loaded with mRNA and RNPs, demonstrating effective genome editing and protein expression in vitro and in vivo. These applications extend to cancer treatment and infectious disease management, with promising results in genome editing for cancer therapy using LNPs encapsulating Cas9 mRNA and single-guide RNA. In addition, tissue-specific targeting strategies offer potential for improved therapeutic outcomes and reduced off-target effects. Despite progress, challenges such as optimizing delivery efficiency and targeting remain. Future research should enhance delivery efficiency, explore tissue-specific targeting, investigate combination therapies, and advance clinical translation. In conclusion, mRNA- and RNP-based therapies offer a promising avenue for treating various diseases and have the potential to revolutionize medicine, providing new hope for patients worldwide.
Collapse
Affiliation(s)
- Zohre Eftekhari
- Venom and Biotherapeutics Molecules Laboratory, Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran 1316943551, Iran
| | - Horieh Zohrabi
- Venom and Biotherapeutics Molecules Laboratory, Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran 1316943551, Iran
| | - Akbar Oghalaie
- Venom and Biotherapeutics Molecules Laboratory, Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran 1316943551, Iran
| | - Tahereh Ebrahimi
- Department of Nanobiotechnology, New Technologies Research Group, Pasteur Institute of Iran, Tehran 1316943551, Iran
| | - Fatemeh Sadat Shariati
- Department of Influenza and other Respiratory Viruses, Pasteur Institute of Iran, Tehran 1316943551, Iran
| | - Mahdi Behdani
- Venom and Biotherapeutics Molecules Laboratory, Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran 1316943551, Iran
| | - Fatemeh Kazemi-Lomedasht
- Venom and Biotherapeutics Molecules Laboratory, Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran 1316943551, Iran
| |
Collapse
|
32
|
Tkaczyk C, Newton M, Patnaik MM, Thom G, Strain M, Gamson A, Daramola O, Murthy A, Douthwaite J, Stepanov O, Boger E, Yang H, Esser MT, Lidwell A, DiGiandomenico A, Santos L, Sellman BR. In vivo mRNA expression of a multi-mechanistic mAb combination protects against Staphylococcus aureus infection. Mol Ther 2024; 32:2505-2518. [PMID: 38822525 PMCID: PMC11405172 DOI: 10.1016/j.ymthe.2024.05.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 04/30/2024] [Accepted: 05/29/2024] [Indexed: 06/03/2024] Open
Abstract
Single monoclonal antibodies (mAbs) can be expressed in vivo through gene delivery of their mRNA formulated with lipid nanoparticles (LNPs). However, delivery of a mAb combination could be challenging due to the risk of heavy and light variable chain mispairing. We evaluated the pharmacokinetics of a three mAb combination against Staphylococcus aureus first in single chain variable fragment scFv-Fc and then in immunoglobulin G 1 (IgG1) format in mice. Intravenous delivery of each mRNA/LNP or the trio (1 mg/kg each) induced functional antibody expression after 24 h (10-100 μg/mL) with 64%-78% cognate-chain paired IgG expression after 3 days, and an absence of non-cognate chain pairing for scFv-Fc. We did not observe reduced neutralizing activity for each mAb compared with the level of expression of chain-paired mAbs. Delivery of the trio mRNA protected mice in an S. aureus-induced dermonecrosis model. Intravenous administration of the three mRNA in non-human primates achieved peak serum IgG levels ranging between 2.9 and 13.7 μg/mL with a half-life of 11.8-15.4 days. These results suggest nucleic acid delivery of mAb combinations holds promise and may be a viable option to streamline the development of therapeutic antibodies.
Collapse
Affiliation(s)
- Christine Tkaczyk
- AstraZeneca, Early Vaccines & Immune Therapies, Gaithersburg, MD 20878, USA.
| | - Michael Newton
- AstraZeneca, BioPharmaceutical Development, BioPharmaceuticals R&D, Gaithersburg, MD 20878, USA
| | - Mun Mun Patnaik
- AstraZeneca, Early Vaccines & Immune Therapies, Gaithersburg, MD 20878, USA
| | - George Thom
- AstraZeneca, Discovery Sciences, BioPharmaceuticals R&D, Cambridge CB21 6GH, UK
| | - Martin Strain
- AstraZeneca, Biologics Engineering, BioPharmaceuticals R&D, Cambridge CB216GH, UK
| | - Adam Gamson
- AstraZeneca, Early Vaccines & Immune Therapies, Gaithersburg, MD 20878, USA
| | - Olalekan Daramola
- AstraZeneca, BioPharmaceutical Development, BioPharmaceuticals R&D, Cambridge CB21 6GH, UK
| | - Andal Murthy
- AstraZeneca, BioPharmaceutical Development, BioPharmaceuticals R&D, Cambridge CB21 6GH, UK
| | - Julie Douthwaite
- AstraZeneca, Discovery Sciences, BioPharmaceuticals R&D, Cambridge CB21 6GH, UK
| | - Oleg Stepanov
- Clinical Pharmacology and Pharmacometrics, BioPharmaceuticals R&D, AstraZeneca, Cambridge CB2 8PA, UK
| | - Elin Boger
- Drug Metabolism and Pharmacokinetics, Research and Early Development, Respirator & immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Haitao Yang
- Clinical Pharmacology and Pharmacometrics, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD 20878, USA
| | - Mark T Esser
- AstraZeneca, Early Vaccines & Immune Therapies, Gaithersburg, MD 20878, USA
| | - Ashley Lidwell
- AstraZeneca, Early Vaccines & Immune Therapies, Gaithersburg, MD 20878, USA
| | | | - Luis Santos
- AstraZeneca, BioPharmaceutical Development, BioPharmaceuticals R&D, Gaithersburg, MD 20878, USA
| | - Bret R Sellman
- AstraZeneca, Early Vaccines & Immune Therapies, Gaithersburg, MD 20878, USA
| |
Collapse
|
33
|
Wang C, Yuan F. A comprehensive comparison of DNA and RNA vaccines. Adv Drug Deliv Rev 2024; 210:115340. [PMID: 38810703 PMCID: PMC11181159 DOI: 10.1016/j.addr.2024.115340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 05/06/2024] [Accepted: 05/18/2024] [Indexed: 05/31/2024]
Abstract
Nucleic acid technology has revolutionized vaccine development, enabling rapid design and production of RNA and DNA vaccines for prevention and treatment of diseases. The successful deployment of mRNA and plasmid DNA vaccines against COVID-19 has further validated the technology. At present, mRNA platform is prevailing due to its higher efficacy, while DNA platform is undergoing rapid evolution because it possesses unique advantages that can potentially overcome the problems associated with the mRNA platform. To help understand the recent performances of the two vaccine platforms and recognize their clinical potentials in the future, this review compares the advantages and drawbacks of mRNA and DNA vaccines that are currently known in the literature, in terms of development timeline, financial cost, ease of distribution, efficacy, safety, and regulatory approval of products. Additionally, the review discusses the ongoing clinical trials, strategies for improvement, and alternative designs of RNA and DNA platforms for vaccination.
Collapse
Affiliation(s)
- Chunxi Wang
- Department of Biomedical Engineering, Duke University, Durham, NC 27705, United States
| | - Fan Yuan
- Department of Biomedical Engineering, Duke University, Durham, NC 27705, United States.
| |
Collapse
|
34
|
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
|
35
|
Pitard B, Pitard I. [« ReNAissance » of biotherapies with RNA]. Med Sci (Paris) 2024; 40:525-533. [PMID: 38986097 DOI: 10.1051/medsci/2024079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/12/2024] Open
Abstract
Many diseases originate from either the absence or defective expression of a given protein. For some of them, the lacking protein is secreted or can be taken up by cells when delivered exogenously. In such cases, therapies initially involved administering the physiological protein extracted from human tissues. Subsequently, genetic engineering enabled the production of proteins through cell fermentation after introducing the corresponding gene. For many other pathologies, the deficient protein cannot be delivered exogenously. Thus, an endogenous production of the therapeutic protein by the cells themselves is necessary. Messenger RNA (mRNA) technology, like its predecessor DNA, aims to supplement the genetic information needed to produce the therapeutic protein within the cells. However, unlike DNA-based therapies, mRNA transfer allows for transient expression of the protein of interest, which offers an advantage in numerous pathologies. Nonetheless, mastering the quantity, quality, and spatio-temporal regulation of protein production encoded by therapeutic mRNA remains a significant challenge for the development of this approach.
Collapse
Affiliation(s)
- Bruno Pitard
- Nantes Université, Université d'Angers, Immunology and New Concepts in Immunotherapy (INCIT), Inserm UMR-S 1302, CNRS EMR6001, Nantes, France
| | | |
Collapse
|
36
|
Irfan H, Ahmed A. Advancements in chikungunya virus management: FDA approval of ixchiq vaccine and global perspectives. Health Sci Rep 2024; 7:e2183. [PMID: 38912367 PMCID: PMC11192840 DOI: 10.1002/hsr2.2183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 04/03/2024] [Accepted: 05/17/2024] [Indexed: 06/25/2024] Open
Affiliation(s)
- Hamza Irfan
- Department of MedicineShaikh Khalifa Bin Zayed Al Nahyan Medical and Dental CollegeLahorePakistan
| | - Aliza Ahmed
- Department of MedicineJinnah Sindh Medical UniversityKarachiPakistan
| |
Collapse
|
37
|
Shuptrine CW, Chen Y, Miriyala J, Lenz K, Moffett D, Nguyen TA, Michaux J, Campbell K, Smith C, Morra M, Rivera-Molina Y, Murr N, Cooper S, McGuire A, Makani V, Oien N, Zugates JT, de Silva S, Schreiber TH, de Picciotto S, Fromm G. Lipid-Encapsulated mRNAs Encoding Complex Fusion Proteins Potentiate Antitumor Immune Responses. Cancer Res 2024; 84:1550-1559. [PMID: 38381555 PMCID: PMC11094416 DOI: 10.1158/0008-5472.can-23-2875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 12/22/2023] [Accepted: 02/14/2024] [Indexed: 02/23/2024]
Abstract
Lipid nanoparticle (LNP)-encapsulated mRNA has been used for in vivo production of several secreted protein classes, such as IgG, and has enabled the development of personalized vaccines in oncology. Establishing the feasibility of delivering complex multispecific modalities that require higher-order structures important for their function could help expand the use of mRNA/LNP biologic formulations. Here, we evaluated whether in vivo administration of mRNA/LNP formulations of SIRPα-Fc-CD40L and TIGIT-Fc-LIGHT could achieve oligomerization and extend exposure, on-target activity, and antitumor responses comparable with that of the corresponding recombinant fusion proteins. Intravenous infusion of the formulated LNP-encapsulated mRNAs led to rapid and sustained production of functional hexameric proteins in vivo, which increased the overall exposure relative to the recombinant protein controls by ∼28 to 140 fold over 96 hours. High concentrations of the mRNA-encoded proteins were also observed in secondary lymphoid organs and within implanted tumors, with protein concentrations in tumors up to 134-fold greater than with the recombinant protein controls 24 hours after treatment. In addition, SIRPα-Fc-CD40L and TIGIT-Fc-LIGHT mRNAs induced a greater increase in antigen-specific CD8+ T cells in the tumors. These mRNA/LNP formulations were well tolerated and led to a rapid increase in serum and intratumoral IL2, delayed tumor growth, extended survival, and outperformed the activities of benchmark mAb controls. Furthermore, the mRNA/LNPs demonstrated improved efficacy in combination with anti-PD-L1 relative to the recombinant fusion proteins. These data support the delivery of complex oligomeric biologics as mRNA/LNP formulations, where high therapeutic expression and exposure could translate into improved patient outcomes. SIGNIFICANCE Lipid nanoparticle-encapsulated mRNA can efficiently encode complex fusion proteins encompassing immune checkpoint blockers and costimulators that functionally oligomerize in vivo with extended pharmacokinetics and durable exposure to induce potent antitumor immunity.
Collapse
|
38
|
Sun W, Wu Y, Ying T. Progress in novel delivery technologies to improve efficacy of therapeutic antibodies. Antiviral Res 2024; 225:105867. [PMID: 38521465 DOI: 10.1016/j.antiviral.2024.105867] [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: 11/23/2023] [Revised: 03/06/2024] [Accepted: 03/11/2024] [Indexed: 03/25/2024]
Abstract
Monoclonal antibody-based therapeutics have achieved remarkable success in treating a wide range of human diseases. However, conventional systemic delivery methods have limitations in insufficient target tissue permeability, high costs, repeated administrations, etc. Novel technologies have been developed to address these limitations and further enhance antibody therapy. Local antibody delivery via respiratory tract, gastrointestinal tract, eye and blood-brain barrier have shown promising results in increasing local concentrations and overcoming barriers. Nucleic acid-encoded antibodies expressed from plasmid DNA, viral vectors or mRNA delivery platforms also offer advantages over recombinant proteins such as sustained expression, rapid onset, and lower costs. This review summarizes recent advances in antibody delivery methods and highlights innovative technologies that have potential to expand therapeutic applications of antibodies.
Collapse
Affiliation(s)
- Wenli Sun
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Institute of Infectious Disease and Biosecurity, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yanling Wu
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Institute of Infectious Disease and Biosecurity, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Engineering Research Center for Synthetic Immunology, Shanghai 200032, China.
| | - Tianlei Ying
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Institute of Infectious Disease and Biosecurity, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Engineering Research Center for Synthetic Immunology, Shanghai 200032, China.
| |
Collapse
|
39
|
Wu DH, Senyo SE. Scratching beneath the surface: IL4Rα blockade and the progression from myocardial ischemic injury to heart failure. Am J Physiol Heart Circ Physiol 2024; 326:H1177-H1179. [PMID: 38517226 DOI: 10.1152/ajpheart.00150.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 03/15/2024] [Accepted: 03/15/2024] [Indexed: 03/23/2024]
Affiliation(s)
- Douglas H Wu
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, United States
- Medical Scientist Training Program, Case Western Reserve University, Cleveland, Ohio, United States
| | - Samuel E Senyo
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, United States
| |
Collapse
|
40
|
Parhiz H, Atochina-Vasserman EN, Weissman D. mRNA-based therapeutics: looking beyond COVID-19 vaccines. Lancet 2024; 403:1192-1204. [PMID: 38461842 DOI: 10.1016/s0140-6736(23)02444-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 07/06/2023] [Accepted: 10/30/2023] [Indexed: 03/12/2024]
Abstract
Recent advances in mRNA technology and its delivery have enabled mRNA-based therapeutics to enter a new era in medicine. The rapid, potent, and transient nature of mRNA-encoded proteins, without the need to enter the nucleus or the risk of genomic integration, makes them desirable tools for treatment of a range of diseases, from infectious diseases to cancer and monogenic disorders. The rapid pace and ease of mass-scale manufacturability of mRNA-based therapeutics supported the global response to the COVID-19 pandemic. Nonetheless, challenges remain with regards to mRNA stability, duration of expression, delivery efficiency, and targetability, to broaden the applicability of mRNA therapeutics beyond COVID-19 vaccines. By learning from the rapidly expanding preclinical and clinical studies, we can optimise the mRNA platform to meet the clinical needs of each disease. Here, we will summarise the recent advances in mRNA technology; its use in vaccines, immunotherapeutics, protein replacement therapy, and genomic editing; and its delivery to desired specific cell types and organs for development of a new generation of targeted mRNA-based therapeutics.
Collapse
Affiliation(s)
- Hamideh Parhiz
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Drew Weissman
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
41
|
Chuong C, Cereghino C, Rai P, Bates TA, Oberer M, Weger-Lucarelli J. Enhanced attenuation of chikungunya vaccines expressing antiviral cytokines. NPJ Vaccines 2024; 9:59. [PMID: 38472211 PMCID: PMC10933427 DOI: 10.1038/s41541-024-00843-x] [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: 01/03/2023] [Accepted: 02/21/2024] [Indexed: 03/14/2024] Open
Abstract
Alphaviruses are vector-borne, medically relevant, positive-stranded RNA viruses that cause disease in animals and humans worldwide. Of this group, chikungunya virus (CHIKV) is the most significant human pathogen, responsible for generating millions of infections leading to severe febrile illness and debilitating chronic joint pain. Currently, there are limited treatments to protect against alphavirus disease; thus, there is a tremendous need to generate safe and effective vaccines. Live-attenuated vaccines (LAVs) are cost-effective and potent immunization strategies capable of generating long-term protection in a single dose. However, LAVs often produce systemic viral replication, which can lead to unwanted post-vaccination side effects and pose a risk of reversion to a pathogenic phenotype and transmission to mosquitoes. Here, we utilized a chimeric infectious clone of CHIKV engineered with the domain C of the E2 gene of Semliki Forest virus (SFV) to express IFNγ and IL-21-two potent antiviral and immunomodulatory cytokines-in order to improve the LAV's attenuation while maintaining immunogenicity. The IFNγ- and IL-21-expressing vaccine candidates were stable during passage and significantly attenuated post-vaccination, as mice experienced reduced footpad swelling with minimal systemic replication and dissemination capacity compared to the parental vaccine. Additionally, these candidates provided complete protection to mice challenged with WT CHIKV. Our dual attenuation strategy represents an innovative way to generate safe and effective alphavirus vaccines that could be applied to other viruses.
Collapse
Affiliation(s)
- Christina Chuong
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, VA-MD Regional College of Veterinary Medicine, Blacksburg, VA, USA
- Center for Emerging, Zoonotic, and Arthropod-borne Pathogens, Virginia Tech, Blacksburg, VA, USA
| | - Chelsea Cereghino
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, VA-MD Regional College of Veterinary Medicine, Blacksburg, VA, USA
- Center for Emerging, Zoonotic, and Arthropod-borne Pathogens, Virginia Tech, Blacksburg, VA, USA
| | - Pallavi Rai
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, VA-MD Regional College of Veterinary Medicine, Blacksburg, VA, USA
- Center for Emerging, Zoonotic, and Arthropod-borne Pathogens, Virginia Tech, Blacksburg, VA, USA
| | - Tyler A Bates
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, VA-MD Regional College of Veterinary Medicine, Blacksburg, VA, USA
| | - Megan Oberer
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, VA-MD Regional College of Veterinary Medicine, Blacksburg, VA, USA
| | - James Weger-Lucarelli
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, VA-MD Regional College of Veterinary Medicine, Blacksburg, VA, USA.
- Center for Emerging, Zoonotic, and Arthropod-borne Pathogens, Virginia Tech, Blacksburg, VA, USA.
| |
Collapse
|
42
|
Wang S, Zhu Z, Li J. Pharmacokinetic Analyses of a Lipid Nanoparticle-Encapsulated mRNA-Encoded Antibody against Rift Valley Fever Virus. Mol Pharm 2024; 21:1342-1352. [PMID: 38295278 DOI: 10.1021/acs.molpharmaceut.3c01016] [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: 02/02/2024]
Abstract
Rift Valley fever virus (RVFV) could cause an emergency illness characterized by fever, muscle pain, and even death in humans or ruminants. However, there are no approved antiviral drugs that prevent or treat RVFV infection. While therapeutic antibodies have shown promising potential for prevention or treatment in several studies, many studies are ongoing, especially in the field of infectious diseases. Among these studies, the mRNA-LNP platform shows great potential for application, following the COVID-19 pandemic. Previously, we have obtained a neutralizing antibody against RVFV, which was named A38 protein and verified to have a high binding and neutralization ability. In this study, we aimed to identify an effectively optimized sequence and expressed the prioritized mRNA-encoded antibody in vitro. Notably, we effectively expressed mRNA-encoded protein and used the mRNA-LNP platform to generate A38-mRNA-LNP. Pharmacokinetic experiments were conducted in vivo and set up in two groups of mRNA-A38 group and A38 protein group, which were derived from mRNA-LNP and plasmid DNA-expressed proteins, respectively. A38-mRNA-LNPs were administrated by intramuscular injection, A38 proteins were administrated by intravenous administration, and their unique ability to maintain long-lasting protein concentrations by mRNA-encoded protein was demonstrated with the mRNA-encoded protein providing a longer circulating half-life compared to injection of the free A38 protein. These preclinical data on the mRNA-encoded antibody highlighted its potential to prevent infectious diseases in the future.
Collapse
Affiliation(s)
- Shuo Wang
- Institute of Bioengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou 311215, China
| | - Ziling Zhu
- School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Jianmin Li
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou 311215, China
- Laboratory of Vaccine and Antibody Engineering, Beijing Institute of Biotechnology, Beijing 100071, China
| |
Collapse
|
43
|
Shi Y, Zhen X, Zhang Y, Li Y, Koo S, Saiding Q, Kong N, Liu G, Chen W, Tao W. Chemically Modified Platforms for Better RNA Therapeutics. Chem Rev 2024; 124:929-1033. [PMID: 38284616 DOI: 10.1021/acs.chemrev.3c00611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2024]
Abstract
RNA-based therapies have catalyzed a revolutionary transformation in the biomedical landscape, offering unprecedented potential in disease prevention and treatment. However, despite their remarkable achievements, these therapies encounter substantial challenges including low stability, susceptibility to degradation by nucleases, and a prominent negative charge, thereby hindering further development. Chemically modified platforms have emerged as a strategic innovation, focusing on precise alterations either on the RNA moieties or their associated delivery vectors. This comprehensive review delves into these platforms, underscoring their significance in augmenting the performance and translational prospects of RNA-based therapeutics. It encompasses an in-depth analysis of various chemically modified delivery platforms that have been instrumental in propelling RNA therapeutics toward clinical utility. Moreover, the review scrutinizes the rationale behind diverse chemical modification techniques aiming at optimizing the therapeutic efficacy of RNA molecules, thereby facilitating robust disease management. Recent empirical studies corroborating the efficacy enhancement of RNA therapeutics through chemical modifications are highlighted. Conclusively, we offer profound insights into the transformative impact of chemical modifications on RNA drugs and delineates prospective trajectories for their future development and clinical integration.
Collapse
Affiliation(s)
- Yesi Shi
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Xueyan Zhen
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Yiming Zhang
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Yongjiang Li
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Seyoung Koo
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Qimanguli Saiding
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Na Kong
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou 310058, China
| | - Gang Liu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Wei Chen
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
- Genomics Research Center, Academia Sinica, Taipei 11529, Taiwan
| | - Wei Tao
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| |
Collapse
|
44
|
Sharma P, Hoorn D, Aitha A, Breier D, Peer D. The immunostimulatory nature of mRNA lipid nanoparticles. Adv Drug Deliv Rev 2024; 205:115175. [PMID: 38218350 DOI: 10.1016/j.addr.2023.115175] [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: 10/25/2023] [Revised: 12/25/2023] [Accepted: 12/28/2023] [Indexed: 01/15/2024]
Abstract
mRNA-Lipid nanoparticles (LNPs) are at the forefront of global medical research. With the development of mRNA-LNP vaccines to combat the COVID-19 pandemic, the clinical potential of this platform was unleashed. Upon administering 16 billion doses that protected billions of people, it became clear that a fraction of them witnessed mild and in some cases even severe adverse effects. Therefore, it is paramount to define the safety along with the therapeutic efficacy of the mRNA-LNP platform for the successful translation of new genetic medicines based on this technology. While mRNA was the effector molecule of this platform, the ionizable lipid component of the LNPs played an indispensable role in its success. However, both of these components possess the ability to induce undesired immunostimulation, which is an area that needs to be addressed systematically. The immune cell agitation caused by this platform is a two-edged sword as it may prove beneficial for vaccination but detrimental to other applications. Therefore, a key challenge in advancing the mRNA-LNP drug delivery platform from bench to bedside is understanding the immunostimulatory behavior of these components. Herein, we provide a detailed overview of the structural modifications and immunogenicity of synthetic mRNA. We discuss the effect of ionizable lipid structure on LNP functionality and offer a mechanistic overview of the ability of LNPs to elicit an immune response. Finally, we shed some light on the current status of this technology in clinical trials and discuss a few challenges to be addressed to advance the field.
Collapse
Affiliation(s)
- Preeti Sharma
- Laboratory of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel; Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel; Cancer Biology Research Center, Tel Aviv University, Tel Aviv, Israel
| | - Daniek Hoorn
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Anjaiah Aitha
- Laboratory of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel; Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel; Cancer Biology Research Center, Tel Aviv University, Tel Aviv, Israel
| | - Dor Breier
- Laboratory of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel; Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel; Cancer Biology Research Center, Tel Aviv University, Tel Aviv, Israel
| | - Dan Peer
- Laboratory of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel; Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel; Cancer Biology Research Center, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
45
|
Principi N, Esposito S. Development of Vaccines against Emerging Mosquito-Vectored Arbovirus Infections. Vaccines (Basel) 2024; 12:87. [PMID: 38250900 PMCID: PMC10818606 DOI: 10.3390/vaccines12010087] [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: 10/19/2023] [Revised: 12/27/2023] [Accepted: 12/29/2023] [Indexed: 01/23/2024] Open
Abstract
Among emergent climate-sensitive infectious diseases, some mosquito-vectored arbovirus infections have epidemiological, social, and economic effects. Dengue virus (DENV), West Nile virus (WNV), and Chikungunya virus (CHIKV) disease, previously common only in the tropics, currently pose a major risk to global health and are expected to expand dramatically in the near future if adequate containment measures are not implemented. The lack of safe and effective vaccines is critical as it seems likely that emerging mosquito-vectored arbovirus infections will be con-trolled only when effective and safe vaccines against each of these infections become available. This paper discusses the clinical characteristics of DENV, WNV, and CHIKV infections and the state of development of vaccines against these viruses. An ideal vaccine should be able to evoke with a single administration a prompt activation of B and T cells, adequate concentrations of protecting/neutralizing antibodies, and the creation of a strong immune memory capable of triggering an effective secondary antibody response after new infection with a wild-type and/or mutated infectious agent. Moreover, the vaccine should be well tolerated, safe, easily administrated, cost-effective, and widely available throughout the world. However, the development of vaccines against emerging mosquito-vectored arbovirus diseases is far from being satisfactory, and it seems likely that it will take many years before effective and safe vaccines for all these infections are made available worldwide.
Collapse
Affiliation(s)
| | - Susanna Esposito
- Pediatric Clinic, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| |
Collapse
|
46
|
de Jong HK, Grobusch MP. Monoclonal antibody applications in travel medicine. Trop Dis Travel Med Vaccines 2024; 10:2. [PMID: 38221606 PMCID: PMC10789029 DOI: 10.1186/s40794-023-00212-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 11/21/2023] [Indexed: 01/16/2024] Open
Abstract
For decades, immunoglobulin preparations have been used to prevent or treat infectious diseases. Since only a few years, monoclonal antibody applications (mAbs) are taking flight and are increasingly dominating this field. In 2014, only two mAbs were registered; end of October 2023, more than ten mAbs are registered or have been granted emergency use authorization, and many more are in (pre)clinical phases. Especially the COVID-19 pandemic has generated this surge in licensed monoclonal antibodies, although multiple phase 1 studies were already underway in 2019 for other infectious diseases such as malaria and yellow fever. Monoclonal antibodies could function as prophylaxis (i.e., for the prevention of malaria), or could be used to treat (tropical) infections (i.e., rabies, dengue fever, yellow fever). This review focuses on the discussion of the prospects of, and obstacles for, using mAbs in the prevention and treatment of (tropical) infectious diseases seen in the returning traveler; and provides an update on the mAbs currently being developed for infectious diseases, which could potentially be of interest for travelers.
Collapse
Affiliation(s)
- Hanna K de Jong
- Centre of Tropical Medicine and Travel Medicine, Department of Infectious Diseases, Amsterdam University Medical Centers, Location AMC, Amsterdam Infection and Immunity, Amsterdam Public Health, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.
| | - Martin P Grobusch
- Centre of Tropical Medicine and Travel Medicine, Department of Infectious Diseases, Amsterdam University Medical Centers, Location AMC, Amsterdam Infection and Immunity, Amsterdam Public Health, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
- Institute of Tropical Medicine & Deutsches Zentrum Für Infektionsforschung, University of Tübingen, Tübingen, Germany
- Centre de Recherches Médicales, (CERMEL), Lambaréné, Gabon
- Masanga Medical Research Unit (MMRU), Masanga, Sierra Leone
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
47
|
Paneerselvam N, Khan A, Lawson BR. Broadly neutralizing antibodies targeting HIV: Progress and challenges. Clin Immunol 2023; 257:109809. [PMID: 37852345 PMCID: PMC10872707 DOI: 10.1016/j.clim.2023.109809] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 10/05/2023] [Accepted: 10/11/2023] [Indexed: 10/20/2023]
Abstract
Anti-HIV broadly neutralizing antibodies (bNAbs) offer a novel approach to treating, preventing, or curing HIV. Pre-clinical models and clinical trials involving the passive transfer of bNAbs have demonstrated that they can control viremia and potentially serve as alternatives or complement antiretroviral therapy (ART). However, antibody decay, persistent latent reservoirs, and resistance impede bNAb treatment. This review discusses recent advancements and obstacles in applying bNAbs and proposes strategies to enhance their therapeutic potential. These strategies include multi-epitope targeting, antibody half-life extension, combining with current and newer antiretrovirals, and sustained antibody secretion.
Collapse
Affiliation(s)
| | - Amber Khan
- The Scintillon Research Institute, 6868 Nancy Drive, San Diego, CA 92121, USA
| | - Brian R Lawson
- The Scintillon Research Institute, 6868 Nancy Drive, San Diego, CA 92121, USA.
| |
Collapse
|
48
|
Zong Y, Lin Y, Wei T, Cheng Q. Lipid Nanoparticle (LNP) Enables mRNA Delivery for Cancer Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2303261. [PMID: 37196221 DOI: 10.1002/adma.202303261] [Citation(s) in RCA: 143] [Impact Index Per Article: 71.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 05/13/2023] [Indexed: 05/19/2023]
Abstract
Messenger RNA (mRNA) has received great attention in the prevention and treatment of various diseases due to the success of coronavirus disease 2019 (COVID-19) mRNA vaccines (Comirnaty and Spikevax). To meet the therapeutic purpose, it is required that mRNA must enter the target cells and express sufficient proteins. Therefore, the development of effective delivery systems is necessary and crucial. Lipid nanoparticle (LNP) represents a remarkable vehicle that has indeed accelerated mRNA applications in humans, as several mRNA-based therapies have already been approved or are in clinical trials. In this review, the focus is on mRNA-LNP-mediated anticancer therapy. It summarizes the main development strategies of mRNA-LNP formulations, discusses representative therapeutic approaches in cancer, and points out current challenges and possible future directions of this research field. It is hoped that these delivered messages can help further improve the application of mRNA-LNP technology in cancer therapy.
Collapse
Affiliation(s)
- Yan Zong
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing, 100871, China
| | - Yi Lin
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing, 100871, China
| | - Tuo Wei
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Qiang Cheng
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing, 100871, China
| |
Collapse
|
49
|
Deal CE, Richards AF, Yeung T, Maron MJ, Wang Z, Lai YT, Fritz BR, Himansu S, Narayanan E, Liu D, Koleva R, Licht S, Hsiao CJ, Rajlic IL, Koch H, Kleyman M, Pulse ME, Weiss WJ, Doering JE, Lindberg SK, Mantis NJ, Carfi A, Plante OJ. An mRNA-based platform for the delivery of pathogen-specific IgA into mucosal secretions. Cell Rep Med 2023; 4:101253. [PMID: 37918405 PMCID: PMC10694625 DOI: 10.1016/j.xcrm.2023.101253] [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: 04/06/2023] [Revised: 07/28/2023] [Accepted: 09/29/2023] [Indexed: 11/04/2023]
Abstract
Colonization of the gut and airways by pathogenic bacteria can lead to local tissue destruction and life-threatening systemic infections, especially in immunologically compromised individuals. Here, we describe an mRNA-based platform enabling delivery of pathogen-specific immunoglobulin A (IgA) monoclonal antibodies into mucosal secretions. The platform consists of synthetic mRNA encoding IgA heavy, light, and joining (J) chains, packaged in lipid nanoparticles (LNPs) that express glycosylated, dimeric IgA with functional activity in vitro and in vivo. Importantly, mRNA-derived IgA had a significantly greater serum half-life and a more native glycosylation profile in mice than did a recombinantly produced IgA. Expression of an mRNA encoded Salmonella-specific IgA in mice resulted in intestinal localization and limited Peyer's patch invasion. The same mRNA-LNP technology was used to express a Pseudomonas-specific IgA that protected from a lung challenge. Leveraging the mRNA antibody technology as a means to intercept bacterial pathogens at mucosal surfaces opens up avenues for prophylactic and therapeutic interventions.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Ding Liu
- Moderna, Inc., Cambridge, MA 02139, USA
| | | | | | | | | | | | | | - Mark E Pulse
- HSC College of Pharmacy, University of North Texas, Fort Worth, TX 76132, USA
| | - William J Weiss
- HSC College of Pharmacy, University of North Texas, Fort Worth, TX 76132, USA
| | - Jennifer E Doering
- Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, Albany, NY 12211, USA
| | - Samantha K Lindberg
- Department of Biomedical Sciences, University at Albany School of Public Health, Rensselaer, NY 12144, USA
| | - Nicholas J Mantis
- Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, Albany, NY 12211, USA; Department of Biomedical Sciences, University at Albany School of Public Health, Rensselaer, NY 12144, USA
| | | | | |
Collapse
|
50
|
Kon E, Ad-El N, Hazan-Halevy I, Stotsky-Oterin L, Peer D. Targeting cancer with mRNA-lipid nanoparticles: key considerations and future prospects. Nat Rev Clin Oncol 2023; 20:739-754. [PMID: 37587254 DOI: 10.1038/s41571-023-00811-9] [Citation(s) in RCA: 112] [Impact Index Per Article: 56.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/25/2023] [Indexed: 08/18/2023]
Abstract
Harnessing mRNA-lipid nanoparticles (LNPs) to treat patients with cancer has been an ongoing research area that started before these versatile nanoparticles were successfully used as COVID-19 vaccines. Currently, efforts are underway to harness this platform for oncology therapeutics, mainly focusing on cancer vaccines targeting multiple neoantigens or direct intratumoural injections of mRNA-LNPs encoding pro-inflammatory cytokines. In this Review, we describe the opportunities of using mRNA-LNPs in oncology applications and discuss the challenges for successfully translating the findings of preclinical studies of these nanoparticles into the clinic. We critically appraise the potential of various mRNA-LNP targeting and delivery strategies, considering physiological, technological and manufacturing challenges. We explore these approaches in the context of the potential clinical applications best suited to each approach and highlight the obstacles that currently need to be addressed to achieve these applications. Finally, we provide insights from preclinical and clinical studies that are leading to this powerful platform being considered the next frontier in oncology treatment.
Collapse
Affiliation(s)
- Edo Kon
- Laboratory of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
- Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel
- Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel
- Cancer Biology Research Center, Tel Aviv University, Tel Aviv, Israel
| | - Nitay Ad-El
- Laboratory of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
- Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel
- Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel
- Cancer Biology Research Center, Tel Aviv University, Tel Aviv, Israel
| | - Inbal Hazan-Halevy
- Laboratory of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
- Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel
- Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel
- Cancer Biology Research Center, Tel Aviv University, Tel Aviv, Israel
| | - Lior Stotsky-Oterin
- Laboratory of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
- Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel
- Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel
- Cancer Biology Research Center, Tel Aviv University, Tel Aviv, Israel
| | - Dan Peer
- Laboratory of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel.
- Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel.
- Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel.
- Cancer Biology Research Center, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|