1
|
Wei L, Yu P, Wang H, Liu J. Adeno-associated viral vectors deliver gene vaccines. Eur J Med Chem 2025; 281:117010. [PMID: 39488197 DOI: 10.1016/j.ejmech.2024.117010] [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: 09/02/2024] [Revised: 10/24/2024] [Accepted: 10/27/2024] [Indexed: 11/04/2024]
Abstract
Adeno-associated viruses (AAVs) are leading platforms for in vivo delivery of gene therapies, with six licensed AAV-based therapeutics attributed to their non-pathogenic nature, low immunogenicity, and high efficiency. In the realm of gene-based vaccines, one of the most vital therapeutic areas, AAVs are also emerging as promising delivery tools. We scrutinized AAVs, focusing on their virological properties, as well as bioengineering and chemical modifications to demonstrate their significant potential in gene vaccine delivery, and detailing the preparation of AAV particles. Additionally, we summarized the use of AAV vectors in vaccines for both infectious and non-infectious diseases, such as influenza, COVID-19, Alzheimer's disease, and cancer. Furthermore, this review, along with the latest clinical trial updates, provides a comprehensive overview of studies on the potential of using AAV vectors for gene vaccine delivery. It aims to deepen our understanding of the challenges and limitations in nucleic acid delivery and pave the way for future clinical success.
Collapse
Affiliation(s)
- Lai Wei
- College of Life Science and Technology, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Peng Yu
- College of Biotechnology, Tianjin University of Science & Technology, 300457 Tianjin, China
| | - Haomeng Wang
- CanSino (Shanghai) Biological Research Co., Ltd, 201208, Shanghai, China.
| | - Jiang Liu
- Rosalind Franklin Institute, Harwell Campus, OX11 0QS, Oxford, United Kingdom; Department of Pharmacology, University of Oxford, Mansfield Road, OX1 3QT, Oxford, United Kingdom.
| |
Collapse
|
2
|
Ma G, Xu Z, Li C, Zhou F, Hu B, Guo J, Ke C, Chen L, Zhang G, Lau H, Pan H, Chen X, Li R, Liu L. Induction of neutralizing antibody responses by AAV5-based vaccine for respiratory syncytial virus in mice. Front Immunol 2024; 15:1451433. [PMID: 39469716 PMCID: PMC11513327 DOI: 10.3389/fimmu.2024.1451433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 09/25/2024] [Indexed: 10/30/2024] Open
Abstract
Introduction Respiratory Syncytial Virus (RSV) is a significant cause of respiratory illnesses worldwide, particularly in infants and elderly individuals. Despite the burden RSV imposes, effective preventive measures are limited. The research application of adeno-associated virus (AAV) in vaccine platforms has been expanding, and its potential in prevention and treatment has garnered much attention. Methods In this study, we explored the potential application of a recombinant adeno-associated virus 5 (rAAV5) vector-based RSV vaccine, focusing on the expression of the pre-fusion (Pre-F) protein structure. Through intramuscular immunization in mice. The immunogenicity of the vaccine was evaluated in Balb/c mice immunized intramuscularly and intranasal, respectively. Results The rAAV5-RSV-Fm vaccine demonstrated positive humoral and induced antibody titers against RSV strains A and B for up to 120 days post-immunization. Notably, intranasal administration also elicited protective antibodies. Characterization studies confirmed the ability of the vac-cine to express the Pre-F protein and its superior immunogenicity compared to that of full-length F protein. Conclusion These findings underscore the potential application of rAAV5 vector platforms in RSV vaccine development and further investigation into their protective efficacy is warranted.
Collapse
Affiliation(s)
- Gangyuan Ma
- Guangzhou National Laboratory, Guangzhou, China
- Guangzhou Medical University, Guangzhou, China
| | - Zeping Xu
- Guangdong Keguanda Pharmaceutical Technology Co., Ltd, Guangzhou, China
| | - Chinyu Li
- Guangdong Keguanda Pharmaceutical Technology Co., Ltd, Guangzhou, China
| | - Feng Zhou
- Guangzhou National Laboratory, Guangzhou, China
| | - Bobo Hu
- Guangzhou National Laboratory, Guangzhou, China
| | - Junwei Guo
- Guangdong Keguanda Pharmaceutical Technology Co., Ltd, Guangzhou, China
| | - Changwen Ke
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, China
| | - Liqing Chen
- Guangzhou National Laboratory, Guangzhou, China
| | - Guilin Zhang
- Guangdong Keguanda Pharmaceutical Technology Co., Ltd, Guangzhou, China
| | - Hungyan Lau
- Guangdong Keguanda Pharmaceutical Technology Co., Ltd, Guangzhou, China
| | - Hudan Pan
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
- Chinese Medicine Guangdong Laboratory, Hengqin, China
| | - Xixin Chen
- Guangdong Keguanda Pharmaceutical Technology Co., Ltd, Guangzhou, China
| | - Runze Li
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
- Chinese Medicine Guangdong Laboratory, Hengqin, China
| | - Liang Liu
- Guangzhou National Laboratory, Guangzhou, China
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
- Chinese Medicine Guangdong Laboratory, Hengqin, China
| |
Collapse
|
3
|
Bergeron HC, Murray J, Arora A, Nuñez Castrejon AM, DuBois RM, Anderson LJ, Kauvar LM, Tripp RA. Immune Prophylaxis Targeting the Respiratory Syncytial Virus (RSV) G Protein. Viruses 2023; 15:1067. [PMID: 37243153 PMCID: PMC10221658 DOI: 10.3390/v15051067] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 04/21/2023] [Accepted: 04/26/2023] [Indexed: 05/28/2023] Open
Abstract
The respiratory syncytial virus (RSV) causes significant respiratory disease in young infants and the elderly. Immune prophylaxis in infants is currently limited to palivizumab, an anti-RSV fusion (F) protein monoclonal antibody (mAb). While anti-F protein mAbs neutralize RSV, they are unable to prevent aberrant pathogenic responses provoked by the RSV attachment (G) protein. Recently, the co-crystal structures of two high-affinity anti-G protein mAbs that bind the central conserved domain (CCD) at distinct non-overlapping epitopes were solved. mAbs 3D3 and 2D10 are broadly neutralizing and block G protein CX3C-mediated chemotaxis by binding antigenic sites γ1 and γ2, respectively, which is known to reduce RSV disease. Previous studies have established 3D3 as a potential immunoprophylactic and therapeutic; however, there has been no similar evaluation of 2D10 available. Here, we sought to determine the differences in neutralization and immunity to RSV Line19F infection which recapitulates human RSV infection in mouse models making it useful for therapeutic antibody studies. Prophylactic (24 h prior to infection) or therapeutic (72 h post-infection) treatment of mice with 3D3, 2D10, or palivizumab were compared to isotype control antibody treatment. The results show that 2D10 can neutralize RSV Line19F both prophylactically and therapeutically, and can reduce disease-causing immune responses in a prophylactic but not therapeutic context. In contrast, 3D3 was able to significantly (p < 0.05) reduce lung virus titers and IL-13 in a prophylactic and therapeutic regimen suggesting subtle but important differences in immune responses to RSV infection with mAbs that bind distinct epitopes.
Collapse
Affiliation(s)
- Harrison C. Bergeron
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA
| | - Jackelyn Murray
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA
| | - Aakash Arora
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA
| | - Ana M. Nuñez Castrejon
- Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, CA 95064, USA; (A.M.N.C.)
| | - Rebecca M. DuBois
- Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, CA 95064, USA; (A.M.N.C.)
| | - Larry J. Anderson
- Division of Pediatric Infectious Disease, Emory University School of Medicine, Atlanta, GA 30322, USA;
| | | | - Ralph A. Tripp
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA
| |
Collapse
|
4
|
Lu Z, Zhang A, Dai Y. CX3CL1 deficiency ameliorates inflammation, apoptosis and accelerates osteogenic differentiation, mineralization in LPS-treated MC3T3-E1 cells via its receptor CX3CR1. Ann Anat 2023; 246:152036. [PMID: 36436718 DOI: 10.1016/j.aanat.2022.152036] [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: 08/24/2022] [Revised: 10/24/2022] [Accepted: 11/16/2022] [Indexed: 11/27/2022]
Abstract
BACKGROUND Osteoporosis is a devastating skeletal disease responsible for bone fragility and fracture. CX3C chemokine ligand 1 (CX3CL1) is an inflammatory chemokine which has been identified to possess increased expression in the serum of postmenopausal osteoporotic patients. This paper was to illuminate the impacts of CX3CL1 on inflammation, apoptosis and osteogenic differentiation, mineralization in LPS-treated osteoblasts and investigate the regulatory mechanism. METHODS The viability of MC3T3-E1 cells exposed to elevating doses of LPS was detected by CCK-8 assay. CX3CL1 and C-X3-C motif chemokine receptor 1 (CX3CR1) expression were detected by RT-qPCR and western blot. CX3CR1 expression was examined again following CX3CL1 depletion. The binding of CX3CL1 with CX3CR1 was testified through Co-IP assay. In MC3T3-E1 cells co-transduced with CX3CL1 interference and CX3CR1 overexpression plasmids following LPS exposure, cell activity and inflammation were separately estimated via CCK-8 assay and RT-qPCR. Apoptosis was measured by TUNEL assay and western blot. Osteoblast differentiation was evaluated by ALP activity assay, RT-qPCR and western blot. Osteoblast mineralization was assessed by ARS staining, RT-qPCR and western blot. Results The experimental data presented that LPS attenuated the viability and enhanced CX3CL1 and CX3CR1 expression in MC3T3-E1 cells in a dose-dependent manner. CX3CR1 interacted with CX3CL1 and was positively modulated by CX3CL1. The suppressive role of CX3CL1 absence in LPS-evoked viability decrease, inflammation and apoptosis in MC3T3-E1 cells was reversed by CX3CR1 elevation. Besides, CX3CR1 reversed the promoted osteoblast differentiation and mineralization imposed by CX3CL1 interference. CONCLUSIONS CX3CL1 knockdown eased inflammation, apoptosis and promoted osteogenic differentiation, mineralization in MC3T3-E1 cells upon LPS exposure through down-regulating CX3CR1.
Collapse
Affiliation(s)
- Zhihua Lu
- Medical school, Yangzhou Polytechnic College, Yangzhou, Jiangsu 225009, China
| | - Aihua Zhang
- Department of Rehabilitation, Northern Jiangsu People's Hospital, Yangzhou, Jiangsu 225001, China; Clinical Medical College of Yangzhou University, Yangzhou, Jiangsu 225009, China
| | - Yan Dai
- Medical research center, Northern Jiangsu People's Hospital, Yangzhou, Jiangsu 225001, China; Clinical Medical College of Yangzhou University, Yangzhou, Jiangsu 225009, China.
| |
Collapse
|
5
|
Influenza Virus-like Particle (VLP) Vaccines Expressing the SARS-CoV-2 S Glycoprotein, S1, or S2 Domains. Vaccines (Basel) 2021; 9:vaccines9080920. [PMID: 34452044 PMCID: PMC8402567 DOI: 10.3390/vaccines9080920] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 08/02/2021] [Accepted: 08/12/2021] [Indexed: 02/08/2023] Open
Abstract
The ongoing severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) pandemic had brought disastrous consequences throughout the entire world. While several manufactured vaccines have been approved for emergency use, continuous efforts to generate novel vaccines are needed. In this study, we developed SARS-CoV-2 virus-like particles (VLPs) containing the full length of spike (S) glycoprotein (S full), S1, or S2 together with the influenza matrix protein 1 (M1) as a core protein. Successfully constructed VLPs expressing the S full, S1, and S2 via Sf9 cell transfections were confirmed and characterized by Western blot and transmission electron microscopy (TEM). VLP immunization in mice induced higher levels of spike protein-specific IgG and its subclasses compared to naïve control, with IgG2a being the most predominant subclass. S full and S1 immune sera elicited virus-neutralizing activities, but these were not strong enough to fully inhibit receptor–ligand binding of the SARS-CoV-2. Neutralizing activities were not observed from the S2 VLP immune sera. Overall, our findings revealed that S full or S1 containing VLPs can be developed into effective vaccines.
Collapse
|