1
|
Syed Altaf RR, Mohan A, Palani N, Mendonce KC, Monisha P, Rajadesingu S. A review of innovative design strategies: Artificial antigen presenting cells in cancer immunotherapy. Int J Pharm 2024; 669:125053. [PMID: 39667594 DOI: 10.1016/j.ijpharm.2024.125053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Revised: 11/07/2024] [Accepted: 12/05/2024] [Indexed: 12/14/2024]
Abstract
Developing nanocarriers that can carry medications directly to tumors is an exciting development in cancer nanomedicine. The efficacy of this intriguing therapeutic approach is, however, compromised by intricate and immunosuppressive circumstances that arise concurrently with the onset of cancer. The artificial antigen presenting cell (aAPC), a micro or nanoparticle based device that mimics an antigen presenting cell by providing crucial signal proteins to T lymphocytes to activate them against cancer, is one cutting-edge method for cancer immunotherapy. This review delves into the critical design considerations for aAPCs, particularly focusing on particle size, shape, and the non-uniform distribution of T cell activating proteins on their surfaces. Adequate surface contact between T cells and aAPCs is essential for activation, prompting engineers to develop nano-aAPCs with microscale contact areas through techniques such as shape modification and nanoparticle clustering. Additionally, we explore recommendations for future advancements in this field.
Collapse
Affiliation(s)
- Rabiya Riffath Syed Altaf
- Department of Physics and Nanotechnology, SRM Institute of Science and Technology, Kattankulathur 603203, Chengalpattu District, Tamil Nadu, India; Centre for Research in Environment, Sustainability Advocacy and Climate CHange (REACH), Directorate of Research, SRM Institute of Science and Technology, Kattankulathur 603203, Chengalpattu District, Tamil Nadu, India
| | - Agilandeswari Mohan
- Department of BioChemistry, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur 603203, Chengalpattu District, Tamil Nadu, India; Centre for Research in Environment, Sustainability Advocacy and Climate CHange (REACH), Directorate of Research, SRM Institute of Science and Technology, Kattankulathur 603203, Chengalpattu District, Tamil Nadu, India
| | - Naveen Palani
- Department of Physics and Nanotechnology, SRM Institute of Science and Technology, Kattankulathur 603203, Chengalpattu District, Tamil Nadu, India; Centre for Research in Environment, Sustainability Advocacy and Climate CHange (REACH), Directorate of Research, SRM Institute of Science and Technology, Kattankulathur 603203, Chengalpattu District, Tamil Nadu, India
| | - Keren Celestina Mendonce
- Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur 603203, Chengalpattu District, Tamil Nadu, India; Centre for Research in Environment, Sustainability Advocacy and Climate CHange (REACH), Directorate of Research, SRM Institute of Science and Technology, Kattankulathur 603203, Chengalpattu District, Tamil Nadu, India
| | - P Monisha
- PG & Research Department of Physics, Sri Sarada College for Women, Salem - 636016, Tamil Nadu, India
| | - Suriyaprakash Rajadesingu
- Centre for Research in Environment, Sustainability Advocacy and Climate CHange (REACH), Directorate of Research, SRM Institute of Science and Technology, Kattankulathur 603203, Chengalpattu District, Tamil Nadu, India.
| |
Collapse
|
2
|
Jha K, Jaishwal P, Yadav TP, Singh SP. Self-assembling of coiled-coil peptides into virus-like particles: Basic principles, properties, design, and applications with special focus on vaccine design and delivery. Biophys Chem 2024; 318:107375. [PMID: 39674128 DOI: 10.1016/j.bpc.2024.107375] [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: 10/01/2024] [Revised: 12/03/2024] [Accepted: 12/03/2024] [Indexed: 12/16/2024]
Abstract
Self-assembling peptide nanoparticles (SAPN) based delivery systems, including virus-like particles (VLP), have shown great potential for becoming prominent in next-generation vaccine and drug development. The VLP can mimic properties of natural viral capsid in terms of size (20-200 nm), geometry (i.e., icosahedral structures), and the ability to generate a robust immune response (with multivalent epitopes) through activation of innate and/or adaptive immune signals. In this regard, coiled-coil (CC) domains are suitable building blocks for designing VLP because of their programmable interaction specificity, affinity, and well-established sequence-to-structure relationships. Generally, two CC domains with different oligomeric states (trimer and pentamer) are fused to form a monomeric protein through a short, flexible spacer sequence. By using combinations of symmetry axes (2-, 3- and 5- folds) that are unique to the geometry of the desired protein cage, it is possible, in principle, to assemble well-defined protein cages like VLP. In this review, we have discussed the crystallographic rules and the basic principles involved in the design of CC-based VLP. It also explored the functions of numerous noncovalent interactions in generating stable VLP structures, which play a crucial role in improving the properties of vaccine immunogenicity, drug delivery, and 3D cell culturing.
Collapse
Affiliation(s)
- Kisalay Jha
- Department of Biotechnology, Mahatma Gandhi Central University, Motihari 845401, India
| | - Puja Jaishwal
- Department of Biotechnology, Mahatma Gandhi Central University, Motihari 845401, India
| | - Thakur Prasad Yadav
- Department of Physics, Faculty of Science, University of Allahabad, Prayagraj 211002, India.
| | | |
Collapse
|
3
|
Sanchez-Martinez ZV, Alpuche-Lazcano SP, Stuible M, Akache B, Renner TM, Deschatelets L, Dudani R, Harrison BA, McCluskie MJ, Hrapovic S, Blouin J, Wang X, Schuller M, Cui K, Cho JY, Durocher Y. SARS-CoV-2 spike-based virus-like particles incorporate influenza H1/N1 antigens and induce dual immunity in mice. Vaccine 2024; 42:126463. [PMID: 39481241 DOI: 10.1016/j.vaccine.2024.126463] [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: 08/16/2024] [Revised: 10/11/2024] [Accepted: 10/14/2024] [Indexed: 11/02/2024]
Abstract
A vaccine effective against both SARS-CoV-2 and influenza A (IAV) viruses could represent a cost-effective strategy to reduce their combined public health burden as well as potential complications arising from co-infection. Based on previous findings that full-length SARS-CoV-2 spike (S) expression can induce high-level, enveloped VLP (eVLP) production in CHO cells, we tested whether IAV H1N1 hemagglutinin (H1) and neuraminidase (N1) could also be displayed on these particles. We found that co-incorporation of the IAV surface antigens in spike VLPs (S-VLPs) was highly efficient: upon transient co-expression of S + H1 or S + H1 + N1 in CHO cells, the resulting VLPs contained similar amounts of the SARS-CoV-2 S and IAV antigens. The self-assembled bivalent (S/H1) and trivalent (S/H1/N1) VLPs released into the culture media were purified by single-step chromatography using a S-VLP affinity resin. Western blot analysis and immuno‑gold labeling transmission electron microscopy (TEM) of purified VLPs confirmed the coexistence of S, H1 and N1 antigens in the same particles. Finally, we demonstrated that two doses of adjuvanted bivalent and trivalent VLPs elicit specific functional antibodies and cellular immunity in a mouse model, suggesting potential for combined SARS-CoV-2/IAV vaccine development.
Collapse
MESH Headings
- Animals
- Spike Glycoprotein, Coronavirus/immunology
- Spike Glycoprotein, Coronavirus/genetics
- Mice
- Vaccines, Virus-Like Particle/immunology
- Vaccines, Virus-Like Particle/administration & dosage
- Influenza A Virus, H1N1 Subtype/immunology
- SARS-CoV-2/immunology
- Neuraminidase/immunology
- Antibodies, Viral/immunology
- Hemagglutinin Glycoproteins, Influenza Virus/immunology
- CHO Cells
- Cricetulus
- Influenza Vaccines/immunology
- Female
- Mice, Inbred BALB C
- COVID-19/prevention & control
- COVID-19/immunology
- COVID-19 Vaccines/immunology
- Orthomyxoviridae Infections/prevention & control
- Orthomyxoviridae Infections/immunology
- Humans
- Antigens, Viral/immunology
- Antigens, Viral/genetics
Collapse
Affiliation(s)
- Zalma V Sanchez-Martinez
- Human Health Therapeutics Research Centre, National Research Council of Canada, Montreal, QC H4P 2R2, Canada.; Department of Biochemistry and Molecular Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada
| | - Sergio P Alpuche-Lazcano
- Human Health Therapeutics Research Centre, National Research Council of Canada, Montreal, QC H4P 2R2, Canada.; Current address: Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), A.P. 510-3, Cuernavaca Morelos (CP 62250), Mexico
| | - Matthew Stuible
- Human Health Therapeutics Research Centre, National Research Council of Canada, Montreal, QC H4P 2R2, Canada
| | - Bassel Akache
- Human Health Therapeutics Research Centre, National Research Council Canada, 1200 Montreal Road, Ottawa, ON K1A 0R6, Canada
| | - Tyler M Renner
- Human Health Therapeutics Research Centre, National Research Council Canada, 1200 Montreal Road, Ottawa, ON K1A 0R6, Canada
| | - Lise Deschatelets
- Human Health Therapeutics Research Centre, National Research Council Canada, 1200 Montreal Road, Ottawa, ON K1A 0R6, Canada
| | - Renu Dudani
- Human Health Therapeutics Research Centre, National Research Council Canada, 1200 Montreal Road, Ottawa, ON K1A 0R6, Canada
| | - Blair A Harrison
- Human Health Therapeutics Research Centre, National Research Council Canada, 1200 Montreal Road, Ottawa, ON K1A 0R6, Canada
| | - Michael J McCluskie
- Human Health Therapeutics Research Centre, National Research Council Canada, 1200 Montreal Road, Ottawa, ON K1A 0R6, Canada
| | - Sabahudin Hrapovic
- Aquatic and Crop Resources Development Research Centre, National Research Council Canada, 6100 Royalmount Avenue, Montreal, QC H4P 2R2, Canada
| | - Julie Blouin
- Human Health Therapeutics Research Centre, National Research Council of Canada, Montreal, QC H4P 2R2, Canada
| | - Xinyu Wang
- Quantum and Nanotechnologies Research Centre, National Research Council Canada, 11421 Saskatchewan Drive, Edmonton, AB T6G 2M9, Canada
| | - Matthew Schuller
- Quantum and Nanotechnologies Research Centre, National Research Council Canada, 11421 Saskatchewan Drive, Edmonton, AB T6G 2M9, Canada
| | - Kai Cui
- Quantum and Nanotechnologies Research Centre, National Research Council Canada, 11421 Saskatchewan Drive, Edmonton, AB T6G 2M9, Canada
| | - Jae-Young Cho
- Quantum and Nanotechnologies Research Centre, National Research Council Canada, 11421 Saskatchewan Drive, Edmonton, AB T6G 2M9, Canada
| | - Yves Durocher
- Human Health Therapeutics Research Centre, National Research Council of Canada, Montreal, QC H4P 2R2, Canada.; Department of Biochemistry and Molecular Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada..
| |
Collapse
|
4
|
Zhang Y, Gao J, Xu W, Huo X, Wang J, Xu Y, Ding W, Guo Z, Liu R. Advances in protein subunit vaccines against H1N1/09 influenza. Front Immunol 2024; 15:1499754. [PMID: 39650643 PMCID: PMC11621219 DOI: 10.3389/fimmu.2024.1499754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Accepted: 11/05/2024] [Indexed: 12/11/2024] Open
Abstract
The A/H1N1pdm09 influenza virus, which caused the 2009 pandemic, has since become a recurring strain in seasonal influenza outbreaks. Given the ongoing threat of influenza, protein subunit vaccines have garnered significant attention for their safety and effectiveness. This review seeks to highlight the latest developments in protein subunit vaccines that specifically target the A/H1N1pdm09 virus. It will also examine the structure and replication cycle of influenza A viruses and compare different types of influenza vaccines. Additionally, the review will address key aspects of H1N1 protein subunit vaccine development, such as antigen selection, protein expression systems, and the use of adjuvants. The role of animal models in evaluating these vaccines will also be discussed. Despite challenges like antigenic variability and the complexities of vaccine production and distribution, protein subunit vaccines remain a promising option for future influenza prevention efforts.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Immunology, College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, China
- Department of Medical Imaging, School of Medicine, Zhoukou Vocational and Technical College, Zhoukou, China
| | - Jingyao Gao
- Department of Immunology, College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, China
| | - Wenqi Xu
- Department of Immunology, College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, China
| | - Xingyu Huo
- Department of Immunology, College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, China
| | - Jingyan Wang
- Department of Immunology, College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, China
| | - Yirui Xu
- Department of Immunology, College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, China
| | - Wenting Ding
- Department of Immunology, College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, China
| | - Zeliang Guo
- Department of Immunology, College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, China
| | - Rongzeng Liu
- Department of Immunology, College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, China
| |
Collapse
|
5
|
Stephens AD, Wilkinson T. Discovery of Therapeutic Antibodies Targeting Complex Multi-Spanning Membrane Proteins. BioDrugs 2024; 38:769-794. [PMID: 39453540 PMCID: PMC11530565 DOI: 10.1007/s40259-024-00682-1] [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: 09/04/2024] [Indexed: 10/26/2024]
Abstract
Complex integral membrane proteins, which are embedded in the cell surface lipid bilayer by multiple transmembrane spanning polypeptides, encompass families of proteins that are important target classes for drug discovery. These protein families include G protein-coupled receptors, ion channels, transporters, enzymes, and adhesion molecules. The high specificity of monoclonal antibodies and the ability to engineer their properties offers a significant opportunity to selectively bind these target proteins, allowing direct modulation of pharmacology or enabling other mechanisms of action such as cell killing. Isolation of antibodies that bind these types of membrane proteins and exhibit the desired pharmacological function has, however, remained challenging due to technical issues in preparing membrane protein antigens suitable for enabling and driving antibody drug discovery strategies. In this article, we review progress and emerging themes in defining discovery strategies for a generation of antibodies that target these complex membrane protein antigens. We also comment on how this field may develop with the emerging implementation of computational techniques, artificial intelligence, and machine learning.
Collapse
Affiliation(s)
- Amberley D Stephens
- Department of Biologics Engineering, Oncology R&D, The Discovery Centre, AstraZeneca, 1 Francis Crick Avenue, Cambridge, CB2 0AA, UK
| | - Trevor Wilkinson
- Department of Biologics Engineering, Oncology R&D, The Discovery Centre, AstraZeneca, 1 Francis Crick Avenue, Cambridge, CB2 0AA, UK.
| |
Collapse
|
6
|
Sharma A, Bhatia D. Programmable bionanomaterials for revolutionizing cancer immunotherapy. Biomater Sci 2024; 12:5415-5432. [PMID: 39291418 DOI: 10.1039/d4bm00815d] [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: 09/19/2024]
Abstract
Cancer immunotherapy involves a cutting-edge method that utilizes the immune system to detect and eliminate cancer cells. It has shown substantial effectiveness in treating different types of cancer. As a result, its growing importance is due to its distinct benefits and potential for sustained recovery. However, the general deployment of this treatment is hindered by ongoing issues in maintaining minimal toxicity, high specificity, and prolonged effectiveness. Nanotechnology offers promising solutions to these challenges due to its notable attributes, including expansive precise surface areas, accurate ability to deliver drugs and controlled surface chemistry. This review explores the current advancements in the application of nanomaterials in cancer immunotherapy, focusing on three primary areas: monoclonal antibodies, therapeutic cancer vaccines, and adoptive cell treatment. In adoptive cell therapy, nanomaterials enhance the expansion and targeting capabilities of immune cells, such as T cells, thereby improving their ability to locate and destroy cancer cells. For therapeutic cancer vaccines, nanoparticles serve as delivery vehicles that protect antigens from degradation and enhance their uptake by antigen-presenting cells, boosting the immune response against cancer. Monoclonal antibodies benefit from nanotechnology through improved delivery mechanisms and reduced off-target effects, which increase their specificity and effectiveness. By highlighting the intersection of nanotechnology and immunotherapy, we aim to underscore the transformative potential of nanomaterials in enhancing the effectiveness and safety of cancer immunotherapies. Nanoparticles' ability to deliver drugs and biomolecules precisely to tumor sites reduces systemic toxicity and enhances therapeutic outcomes.
Collapse
Affiliation(s)
- Ayushi Sharma
- Department of Biotechnology, Institute of Applied Sciences and Humanities, GLA University, Mathura, Uttar Pradesh-281406, India.
| | - Dhiraj Bhatia
- Department of Biological Sciences and Engineering, Indian Institute of Technology Gandhinagar, Palaj 382355, Gandhinagar, India
| |
Collapse
|
7
|
Vuitika L, Côrtes N, Malaquias VB, Silva JDQ, Lira A, Prates-Syed WA, Schimke LF, Luz D, Durães-Carvalho R, Balan A, Câmara NOS, Cabral-Marques O, Krieger JE, Hirata MH, Cabral-Miranda G. A self-adjuvanted VLPs-based Covid-19 vaccine proven versatile, safe, and highly protective. Sci Rep 2024; 14:24228. [PMID: 39414952 PMCID: PMC11484777 DOI: 10.1038/s41598-024-76163-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 10/10/2024] [Indexed: 10/18/2024] Open
Abstract
Vaccination has played a critical role in mitigating COVID-19. Despite the availability of licensed vaccines, there remains a pressing need for improved vaccine platforms that provide high protection, safety, and versatility, while also reducing vaccine costs. In response to these challenges, our aim is to create a self-adjuvanted vaccine against SARS-CoV-2, utilizing Virus-Like Particles (VLPs) as the foundation. To achieve this, we produced bacteriophage (Qβ) VLPs in a prokaryotic system and purified them using a rapid and cost-effective strategy involving organic solvents. This method aims to solubilize lipids and components of the cell membrane to eliminate endotoxins present in bacterial samples. For vaccine formulation, Receptor Binding Domain (RBD) antigens were conjugated using chemical crosslinkers, a process compatible with Good Manufacturing Practice (GMP) standards. Transmission Electron Microscopy (TEM) confirmed the expected folding and spatial configuration of the QβVLPs vaccine. Additionally, vaccine formulation assessment involved SDS-PAGE stained with Coomassie Brilliant Blue, Western blotting, and stereomicroscopic experiments. In vitro and in vivo evaluations of the vaccine formulation were conducted to assess its capacity to induce a protective immune response without causing side effects. Vaccine doses of 20 µg and 50 µg stimulated the production of neutralizing antibodies. In in vivo testing, the group of animals vaccinated with 50 µg of vaccine formulation provided complete protection against virus infection, maintaining stable body weight without showing signs of disease. In conclusion, the QβVLPs-RBD vaccine has proven to be effective and safe, eliminating the necessity for supplementary adjuvants and offering a financially feasible approach. Moreover, this vaccine platform demonstrates flexibility in targeting Variants of Concern (VOCs) via established conjugation protocols with VLPs.
Collapse
Affiliation(s)
- Larissa Vuitika
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Nelson Côrtes
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- The Interunits Graduate Program in Biotechnology, University of São Paulo, the Butantan Institute and the Technological Research Institute of the State of São Paulo, São Paulo, Brazil
- Department of Infectious Diseases and Tropical Medicine, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| | - Vanessa B Malaquias
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, São Paulo, Brazil
- The Graduate Program in Pathophysiology and Toxicology, Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Jaqueline D Q Silva
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, São Paulo, Brazil
- The Graduate Program in Pathophysiology and Toxicology, Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
- Department of Infectious Diseases and Tropical Medicine, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| | - Aline Lira
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- The Interunits Graduate Program in Biotechnology, University of São Paulo, the Butantan Institute and the Technological Research Institute of the State of São Paulo, São Paulo, Brazil
- Department of Infectious Diseases and Tropical Medicine, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| | - Wasim A Prates-Syed
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- The Interunits Graduate Program in Biotechnology, University of São Paulo, the Butantan Institute and the Technological Research Institute of the State of São Paulo, São Paulo, Brazil
- Department of Infectious Diseases and Tropical Medicine, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| | - Lena F Schimke
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- Department of Medicine, Division of Molecular Medicine, Laboratory of Medical Investigation 29., University of São Paulo School of Medicine, São Paulo, Brazil
| | - Daniela Luz
- Laboratory of Bacteriology, Butantan Institute, São Paulo, Brazil
| | - Ricardo Durães-Carvalho
- São Paulo School of Medicine, Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo (UNIFESP), São Paulo, SP, Brazil
- Department of Morphology and Genetics, Federal University of São Paulo, São Paulo, Brazil
- Interunit Bioinformatics Graduate Program, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Andrea Balan
- Applied Structural Biology Laboratory, Institute of Biomedical Sciences, University of São Paulo, São Paulo, 05508-000, Brazil
| | - Niels O S Câmara
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- Nephrology Division, Department of Medicine, Federal University of São Paulo, São Paulo, Brazil
| | - Otavio Cabral-Marques
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- Department of Medicine, Division of Molecular Medicine, Laboratory of Medical Investigation 29., University of São Paulo School of Medicine, São Paulo, Brazil
- DO'R Institute for research, São Paulo, Brazil, IDOR, São Paulo, Brazil
| | - José E Krieger
- Heart Institute, Clinical Hospital, Faculty of Medicine, Laboratory of Genetics and Molecular Cardiology, Clinical Hospital, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| | - Mario H Hirata
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, São Paulo, Brazil
- The Graduate Program in Pathophysiology and Toxicology, Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Gustavo Cabral-Miranda
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil.
- The Interunits Graduate Program in Biotechnology, University of São Paulo, the Butantan Institute and the Technological Research Institute of the State of São Paulo, São Paulo, Brazil.
- The Graduate Program in Pathophysiology and Toxicology, Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil.
- Department of Infectious Diseases and Tropical Medicine, Faculty of Medicine, University of São Paulo, São Paulo, Brazil.
| |
Collapse
|
8
|
Liu Q, Huo X, Wang P, Zhao F, Yuan G, Yang C, Su J. Lactobacillus casei displaying MCP2α and FlaC delivered by PLA microspheres effectively enhances the immune protection of largemouth bass (Micropterus salmoides) against LMBV infection. FISH & SHELLFISH IMMUNOLOGY 2024; 153:109870. [PMID: 39218416 DOI: 10.1016/j.fsi.2024.109870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 08/29/2024] [Accepted: 08/29/2024] [Indexed: 09/04/2024]
Abstract
Largemouth bass ranavirus (LMBV) seriously affects the development of largemouth bass (Micropterus salmoides) industry and causes huge economic losses. Oral vaccine can be a promising method for viral disease precaution. In this study, MCP2α was identified as a valuable epitope region superior to MCP and MCP2 of LMBV by neutralizing antibody experiments. Then, recombinant Lactobacillus casei expressing the fusion protein MCP2αC (MCP2α as antigen, C represents flagellin C from Aeromonas hydrophila as adjuvant) on surface was constructed and verified. Further, PLA microsphere vaccine loading recombinant MCP2αC L. casei was prepared. The PLA microspheres vaccine were observed by scanning electron microscopy and showed a smooth, regular spherical surface with a particle size distribution between 100 and 200 μm. Furthermore, we evaluated the tolerance of PLA-MCP2αC vaccine in simulated gastric fluid and simulated intestinal fluid, and the results showed that PLA-MCP2αC can effectively resist the gastrointestinal environment. Moreover, the protective effect of PLA-MCP2αC against LMBV was evaluated after oral immunization and LMBV challenge. The results showed that PLA-MCP2αC effectively up-regulated the activity of serum biochemical enzymes (T-SOD, T-AOC, LZM, complement C3) and induced the mRNA expression of representative immune genes (IL-1β, TNF-α, IFN-γ, MHC-IIα, Mx, IgM) in spleen and head kidney tissues. The survival rate of largemouth bass vaccinated with PLA-MCP2αC increased from 24 % to 68 %. Meanwhile, PLA-MCP2αC inhibited the LMBV burden in spleen, head kidney and liver tissues and attenuated tissue damage in spleen. These results suggested that PLA-MCP2αC can be used as a candidate oral vaccine against LMBV infection in aquaculture.
Collapse
Affiliation(s)
- Qian Liu
- Hubei Hongshan Laboratory, College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, China; Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, 266237, China
| | - Xingchen Huo
- Hubei Hongshan Laboratory, College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, China
| | - Pengxu Wang
- Hubei Hongshan Laboratory, College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, China
| | - Fengxia Zhao
- Hubei Hongshan Laboratory, College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, China
| | - Gailing Yuan
- Hubei Hongshan Laboratory, College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, China
| | - Chunrong Yang
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Jianguo Su
- Hubei Hongshan Laboratory, College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, China; Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, 266237, China.
| |
Collapse
|
9
|
Ahmadivand S, Fux R, Palić D. Ferritin Vaccine Platform for Animal and Zoonotic Viruses. Vaccines (Basel) 2024; 12:1112. [PMID: 39460279 PMCID: PMC11511493 DOI: 10.3390/vaccines12101112] [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: 08/23/2024] [Revised: 09/23/2024] [Accepted: 09/25/2024] [Indexed: 10/28/2024] Open
Abstract
Viral infections in animals continue to pose a significant challenge, affecting livestock health, welfare, and food safety, and, in the case of zoonotic viruses, threatening global public health. The control of viral diseases currently relies on conventional approaches such as inactivated or attenuated vaccines produced via platforms with inherent limitations. Self-assembling ferritin nanocages represent a novel vaccine platform that has been utilized for several viruses, some of which are currently undergoing human clinical trials. Experimental evidence also supports the potential of this platform for developing commercial vaccines for veterinary viruses. In addition to improved stability and immunogenicity, ferritin-based vaccines are safe and DIVA-compatible, and can be rapidly deployed in response to emerging epidemics or pandemics. This review discusses the structural and functional properties of ferritin proteins, followed by an overview of the design and production of ferritin-based vaccines, the mechanisms of immune responses, and their applications in developing vaccines against animal and zoonotic viruses.
Collapse
Affiliation(s)
- Sohrab Ahmadivand
- Faculty of Veterinary Medicine, Ludwig-Maximilians-Universität München, 80539 Munich, Germany
| | - Robert Fux
- Institute for Infectious Diseases and Zoonoses, Ludwig-Maximilians-Universität München, 80539 Munich, Germany;
| | - Dušan Palić
- Faculty of Veterinary Medicine, Ludwig-Maximilians-Universität München, 80539 Munich, Germany
| |
Collapse
|
10
|
Wang H, Zhang S, Xue W, Zeng Y, Liu L, Cui L, Liu H, Zhang Y, Chen L, Nie M, Zhang R, Chen Z, Hong C, Zheng Q, Cheng T, Gu Y, Li T, Xia N, Li S. Glycoprotein E-Displaying Nanoparticles Induce Robust Neutralizing Antibodies and T-Cell Response against Varicella Zoster Virus. Int J Mol Sci 2024; 25:9872. [PMID: 39337359 PMCID: PMC11432701 DOI: 10.3390/ijms25189872] [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: 07/30/2024] [Revised: 09/05/2024] [Accepted: 09/09/2024] [Indexed: 09/30/2024] Open
Abstract
The Varicella zoster virus (VZV), responsible for both varicella (chickenpox) and herpes zoster (shingles), presents significant global health challenges. While primary VZV infection primarily affects children, leading to chickenpox, reactivation in later life can result in herpes zoster and associated post-herpetic neuralgia, among other complications. Vaccination remains the most effective strategy for VZV prevention, with current vaccines largely based on the attenuated vOka strains. Although these vaccines are generally effective, they can induce varicella-like rashes and have sparked concerns regarding cell virulence. As a safer alternative, subunit vaccines circumvent these issues. In this study, we developed a nanoparticle-based vaccine displaying the glycoprotein E (gE) on ferritin particles using the SpyCatcher/SpyTag system, termed FR-gE. This FR-gE nanoparticle antigen elicited substantial gE-specific binding and VZV-neutralizing antibody responses in BALB/c and C57BL/6 mice-responses that were up to 3.2-fold greater than those elicited by the subunit gE while formulated with FH002C, aluminum hydroxide, or a liposome-based XUA01 adjuvant. Antibody subclass analysis revealed that FR-gE produced comparable levels of IgG1 and significantly higher levels of IgG2a compared to subunit gE, indicating a Th1-biased immune response. Notably, XUA01-adjuvanted FR-gE induced a significant increase in neutralizing antibody response compared to the live attenuated varicella vaccine and recombinant vaccine, Shingrix. Furthermore, ELISPOT assays demonstrated that immunization with FR-gE/XUA01 generated IFN-γ and IL-2 levels comparable to those induced by Shingrix. These findings underscore the potential of FR-gE as a promising immunogen for the development of varicella and herpes zoster vaccines.
Collapse
Affiliation(s)
- Hong Wang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, School of Life Sciences, Xiamen University, Xiamen 361102, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, The Research Unit of Frontier Technology of Structural Vaccinology of Chinese Academy of Medical Sciences, Xiamen University, Xiamen 361102, China
| | - Sibo Zhang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, School of Life Sciences, Xiamen University, Xiamen 361102, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, The Research Unit of Frontier Technology of Structural Vaccinology of Chinese Academy of Medical Sciences, Xiamen University, Xiamen 361102, China
| | - Wenhui Xue
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, School of Life Sciences, Xiamen University, Xiamen 361102, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, The Research Unit of Frontier Technology of Structural Vaccinology of Chinese Academy of Medical Sciences, Xiamen University, Xiamen 361102, China
| | - Yarong Zeng
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, School of Life Sciences, Xiamen University, Xiamen 361102, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, The Research Unit of Frontier Technology of Structural Vaccinology of Chinese Academy of Medical Sciences, Xiamen University, Xiamen 361102, China
| | - Liqin Liu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, School of Life Sciences, Xiamen University, Xiamen 361102, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, The Research Unit of Frontier Technology of Structural Vaccinology of Chinese Academy of Medical Sciences, Xiamen University, Xiamen 361102, China
| | - Lingyan Cui
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, School of Life Sciences, Xiamen University, Xiamen 361102, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, The Research Unit of Frontier Technology of Structural Vaccinology of Chinese Academy of Medical Sciences, Xiamen University, Xiamen 361102, China
| | - Hongjing Liu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, School of Life Sciences, Xiamen University, Xiamen 361102, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, The Research Unit of Frontier Technology of Structural Vaccinology of Chinese Academy of Medical Sciences, Xiamen University, Xiamen 361102, China
| | - Yuyun Zhang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, School of Life Sciences, Xiamen University, Xiamen 361102, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, The Research Unit of Frontier Technology of Structural Vaccinology of Chinese Academy of Medical Sciences, Xiamen University, Xiamen 361102, China
| | - Lin Chen
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, School of Life Sciences, Xiamen University, Xiamen 361102, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, The Research Unit of Frontier Technology of Structural Vaccinology of Chinese Academy of Medical Sciences, Xiamen University, Xiamen 361102, China
| | - Meifeng Nie
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, School of Life Sciences, Xiamen University, Xiamen 361102, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, The Research Unit of Frontier Technology of Structural Vaccinology of Chinese Academy of Medical Sciences, Xiamen University, Xiamen 361102, China
| | - Rongwei Zhang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, School of Life Sciences, Xiamen University, Xiamen 361102, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, The Research Unit of Frontier Technology of Structural Vaccinology of Chinese Academy of Medical Sciences, Xiamen University, Xiamen 361102, China
| | - Zhenqin Chen
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, School of Life Sciences, Xiamen University, Xiamen 361102, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, The Research Unit of Frontier Technology of Structural Vaccinology of Chinese Academy of Medical Sciences, Xiamen University, Xiamen 361102, China
| | - Congming Hong
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, School of Life Sciences, Xiamen University, Xiamen 361102, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, The Research Unit of Frontier Technology of Structural Vaccinology of Chinese Academy of Medical Sciences, Xiamen University, Xiamen 361102, China
| | - Qingbing Zheng
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, School of Life Sciences, Xiamen University, Xiamen 361102, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, The Research Unit of Frontier Technology of Structural Vaccinology of Chinese Academy of Medical Sciences, Xiamen University, Xiamen 361102, China
| | - Tong Cheng
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, School of Life Sciences, Xiamen University, Xiamen 361102, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, The Research Unit of Frontier Technology of Structural Vaccinology of Chinese Academy of Medical Sciences, Xiamen University, Xiamen 361102, China
| | - Ying Gu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, School of Life Sciences, Xiamen University, Xiamen 361102, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, The Research Unit of Frontier Technology of Structural Vaccinology of Chinese Academy of Medical Sciences, Xiamen University, Xiamen 361102, China
| | - Tingting Li
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, School of Life Sciences, Xiamen University, Xiamen 361102, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, The Research Unit of Frontier Technology of Structural Vaccinology of Chinese Academy of Medical Sciences, Xiamen University, Xiamen 361102, China
| | - Ningshao Xia
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, School of Life Sciences, Xiamen University, Xiamen 361102, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, The Research Unit of Frontier Technology of Structural Vaccinology of Chinese Academy of Medical Sciences, Xiamen University, Xiamen 361102, China
| | - Shaowei Li
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, School of Life Sciences, Xiamen University, Xiamen 361102, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, The Research Unit of Frontier Technology of Structural Vaccinology of Chinese Academy of Medical Sciences, Xiamen University, Xiamen 361102, China
| |
Collapse
|
11
|
Vasyagin EA, Zykova AA, Mardanova ES, Nikitin NA, Shuklina MA, Ozhereleva OO, Stepanova LA, Tsybalova LM, Blokhina EA, Ravin NV. Influenza A Vaccine Candidates Based on Virus-like Particles Formed by Coat Proteins of Single-Stranded RNA Phages Beihai32 and PQ465. Vaccines (Basel) 2024; 12:1033. [PMID: 39340063 PMCID: PMC11435553 DOI: 10.3390/vaccines12091033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 08/30/2024] [Accepted: 09/06/2024] [Indexed: 09/30/2024] Open
Abstract
Efficient control of influenza A infection can potentially be achieved through the development of broad-spectrum recombinant vaccines based on conserved antigens. The extracellular domain of the transmembrane protein M2 of influenza A virus (M2e) is highly conserved but poorly immunogenic and needs to be fused to an adjuvant protein or carrier virus-like particles (VLPs) to increase immunogenicity and provide protection against infection. In this study, we obtained VLPs based on capsid proteins (CPs) of single-stranded RNA phages Beihai32 and PQ465 bearing the M2e peptides. Four copies of the M2e peptide were linked to the C-terminus of the CP of phage Beihai32 and to the N and C termini of the CP of phage PQ465. The hybrid proteins, being expressed in Escherichia coli, formed spherical VLPs of about 30 nm in size. Immunogold transmission electron microscopy showed that VLPs formed by the phage PQ465 CP with a C-terminal M2e fusion present the M2e peptide on the surface. Subcutaneous immunization of mice with VLPs formed by both CPs containing four copies of the M2e peptide at the C termini induced high levels of M2e-specific IgG antibodies in serum and provided mice with protection against lethal influenza A virus challenge. In the case of an N-terminal fusion of M2e with the phage PQ465 CP, the immune response against M2e was significantly lower. CPs of phages Beihai32 and PQ465, containing four copies of the M2e peptide at their C termini, can be used to develop recombinant influenza A vaccine.
Collapse
Affiliation(s)
- Egor A Vasyagin
- Institute of Bioengineering, Research Center of Biotechnology of the Russian Academy of Sciences, 119071 Moscow, Russia
| | - Anna A Zykova
- Institute of Bioengineering, Research Center of Biotechnology of the Russian Academy of Sciences, 119071 Moscow, Russia
| | - Eugenia S Mardanova
- Institute of Bioengineering, Research Center of Biotechnology of the Russian Academy of Sciences, 119071 Moscow, Russia
| | - Nikolai A Nikitin
- Department of Virology, Faculty of Biology, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Marina A Shuklina
- Smorodintsev Research Institute of Influenza, Russian Ministry of Health, 197376 St. Petersburg, Russia
| | - Olga O Ozhereleva
- Smorodintsev Research Institute of Influenza, Russian Ministry of Health, 197376 St. Petersburg, Russia
| | - Liudmila A Stepanova
- Smorodintsev Research Institute of Influenza, Russian Ministry of Health, 197376 St. Petersburg, Russia
| | - Liudmila M Tsybalova
- Smorodintsev Research Institute of Influenza, Russian Ministry of Health, 197376 St. Petersburg, Russia
| | - Elena A Blokhina
- Institute of Bioengineering, Research Center of Biotechnology of the Russian Academy of Sciences, 119071 Moscow, Russia
| | - Nikolai V Ravin
- Institute of Bioengineering, Research Center of Biotechnology of the Russian Academy of Sciences, 119071 Moscow, Russia
| |
Collapse
|
12
|
Raji AA, Dastjerdi PZ, Omar AR. Virus-like particles in poultry disease: an approach to effective and safe vaccination. Front Vet Sci 2024; 11:1405605. [PMID: 39315089 PMCID: PMC11417104 DOI: 10.3389/fvets.2024.1405605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Accepted: 08/19/2024] [Indexed: 09/25/2024] Open
Abstract
The poultry industry, a cornerstone of global food security, faces dynamic challenges exacerbated by viral diseases. This review traces the trajectory of poultry vaccination, evolving from traditional methods to the forefront of innovation Virus-Like Particle (VLP) vaccines. Vaccination has been pivotal in disease control, but traditional vaccines exhibit some limitations. This review examines the emergence of VLPs as a game-changer in poultry vaccination. VLPs, mimicking viruses without replication, offer a safer, targeted alternative with enhanced immunogenicity. The narrative encompasses VLP design principles, production methods, immunogenicity, and efficacy against major poultry viruses. Challenges and prospects are explored, presenting VLP vaccines as a transformative technique in poultry disease control. Understanding their potential empowers industry stakeholders to navigate poultry health management with precision, promising improved welfare, reduced economic losses, and heightened food safety.
Collapse
Affiliation(s)
- Abdullahi Abdullahi Raji
- Laboratory of Vaccine and Biomolecules, Institute of Bioscience, Universiti Putra Malaysia, Serdang, Malaysia
- Department of Veterinary Pathology, Faculty of Veterinary Medicine, Usmanu Danfodiyo University, Sokoto, Nigeria
| | - Paniz Zarghami Dastjerdi
- Laboratory of Vaccine and Biomolecules, Institute of Bioscience, Universiti Putra Malaysia, Serdang, Malaysia
| | - Abdul Rahman Omar
- Laboratory of Vaccine and Biomolecules, Institute of Bioscience, Universiti Putra Malaysia, Serdang, Malaysia
- Department of Veterinary Pathology and Microbiology, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang, Malaysia
| |
Collapse
|
13
|
Jung HG, Jeong S, Kang MJ, Hong I, Park YS, Ko E, Kim JO, Choi DY. Molecular Design of Encapsulin Protein Nanoparticles to Display Rotavirus Antigens for Enhancing Immunogenicity. Vaccines (Basel) 2024; 12:1020. [PMID: 39340050 PMCID: PMC11435836 DOI: 10.3390/vaccines12091020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 09/03/2024] [Accepted: 09/05/2024] [Indexed: 09/30/2024] Open
Abstract
Rotavirus considerably threatens global health, particularly for children <5 years. Current, licensed oral attenuated vaccine formulations have limitations including insufficient efficacy in children in low- and middle-income countries, warranting urgent development of novel vaccines with improved efficacy and safety profiles. Herein, we present a novel approach utilizing an encapsulin (ENC) nanoparticle (NP)-based non-replicating rotavirus vaccine. ENC, originating from bacteria, offers a self-assembling scaffold that displays rotavirus VP8* antigens on its surface. To enhance the correct folding and soluble expression of monomeric antigens and their subsequent assembly into NP, we adopted an RNA-interacting domain (RID) of mammalian transfer RNA synthetase as an expression tag fused to the N-terminus of the ENC-VP8* fusion protein. Using the RID-ENC-VP8* tripartite modular design, insertion of linkers of appropriate length and sequence and the universal T cell epitope P2 remarkably improved the production yield and immunogenicity. Cleavage of the RID rendered a homogenous assembly of ENC-P2-VP8* into protein NPs. Immunization with ENC-P2-VP8* induced markedly higher levels of VP8*-specific antibodies and virus neutralization titers in mice than those induced by P2-VP8* without ENC. Altogether, these results highlight the potential of the designed ENC NP-based rotavirus vaccine as an effective strategy against rotavirus disease to address global health challenges.
Collapse
Affiliation(s)
| | - Seonghun Jeong
- Molecular Immunology, Science Unit, International Vaccine Institute, Seoul 08826, Republic of Korea; (S.J.); (M.-J.K.); (I.H.); (Y.-S.P.); (E.K.)
| | - Min-Ji Kang
- Molecular Immunology, Science Unit, International Vaccine Institute, Seoul 08826, Republic of Korea; (S.J.); (M.-J.K.); (I.H.); (Y.-S.P.); (E.K.)
| | - Ingi Hong
- Molecular Immunology, Science Unit, International Vaccine Institute, Seoul 08826, Republic of Korea; (S.J.); (M.-J.K.); (I.H.); (Y.-S.P.); (E.K.)
| | - Young-Shin Park
- Molecular Immunology, Science Unit, International Vaccine Institute, Seoul 08826, Republic of Korea; (S.J.); (M.-J.K.); (I.H.); (Y.-S.P.); (E.K.)
| | - Eunbyeol Ko
- Molecular Immunology, Science Unit, International Vaccine Institute, Seoul 08826, Republic of Korea; (S.J.); (M.-J.K.); (I.H.); (Y.-S.P.); (E.K.)
| | - Jae-Ouk Kim
- Molecular Immunology, Science Unit, International Vaccine Institute, Seoul 08826, Republic of Korea; (S.J.); (M.-J.K.); (I.H.); (Y.-S.P.); (E.K.)
| | | |
Collapse
|
14
|
Atemin A, Ivanova A, Peppel W, Stamatov R, Gallegos R, Durden H, Uzunova S, Vershinin MD, Saffarian S, Stoynov SS. Kinetic Landscape of Single Virus-like Particles Highlights the Efficacy of SARS-CoV-2 Internalization. Viruses 2024; 16:1341. [PMID: 39205315 PMCID: PMC11359012 DOI: 10.3390/v16081341] [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: 06/26/2024] [Revised: 07/22/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024] Open
Abstract
The efficiency of virus internalization into target cells is a major determinant of infectivity. SARS-CoV-2 internalization occurs via S-protein-mediated cell binding followed either by direct fusion with the plasma membrane or endocytosis and subsequent fusion with the endosomal membrane. Despite the crucial role of virus internalization, the precise kinetics of the processes involved remains elusive. We developed a pipeline, which combines live-cell microscopy and advanced image analysis, for measuring the rates of multiple internalization-associated molecular events of single SARS-CoV-2-virus-like particles (VLPs), including endosome ingression and pH change. Our live-cell imaging experiments demonstrate that only a few minutes after binding to the plasma membrane, VLPs ingress into RAP5-negative endosomes via dynamin-dependent scission. Less than two minutes later, VLP speed increases in parallel with a pH drop below 5, yet these two events are not interrelated. By co-imaging fluorescently labeled nucleocapsid proteins, we show that nucleocapsid release occurs with similar kinetics to VLP acidification. Neither Omicron mutations nor abrogation of the S protein polybasic cleavage site affected the rate of VLP internalization, indicating that they do not confer any significant advantages or disadvantages during this process. Finally, we observe that VLP internalization occurs two to three times faster in VeroE6 than in A549 cells, which may contribute to the greater susceptibility of the former cell line to SARS-CoV-2 infection. Taken together, our precise measurements of the kinetics of VLP internalization-associated processes shed light on their contribution to the effectiveness of SARS-CoV-2 propagation in cells.
Collapse
Affiliation(s)
- Aleksandar Atemin
- Institute of Molecular Biology, Bulgarian Academy of Sciences, 21, G. Bontchev Str., 1113 Sofia, Bulgaria; (A.A.); (A.I.); (R.S.); (S.U.)
| | - Aneliya Ivanova
- Institute of Molecular Biology, Bulgarian Academy of Sciences, 21, G. Bontchev Str., 1113 Sofia, Bulgaria; (A.A.); (A.I.); (R.S.); (S.U.)
| | - Wiley Peppel
- Department of Physics and Astronomy, University of Utah, Salt Lake City, UT 84112, USA; (W.P.); (R.G.); (H.D.)
- Center for Cell and Genome Science, University of Utah, Salt Lake City, UT 84112, USA
| | - Rumen Stamatov
- Institute of Molecular Biology, Bulgarian Academy of Sciences, 21, G. Bontchev Str., 1113 Sofia, Bulgaria; (A.A.); (A.I.); (R.S.); (S.U.)
| | - Rodrigo Gallegos
- Department of Physics and Astronomy, University of Utah, Salt Lake City, UT 84112, USA; (W.P.); (R.G.); (H.D.)
- Center for Cell and Genome Science, University of Utah, Salt Lake City, UT 84112, USA
| | - Haley Durden
- Department of Physics and Astronomy, University of Utah, Salt Lake City, UT 84112, USA; (W.P.); (R.G.); (H.D.)
- Center for Cell and Genome Science, University of Utah, Salt Lake City, UT 84112, USA
| | - Sonya Uzunova
- Institute of Molecular Biology, Bulgarian Academy of Sciences, 21, G. Bontchev Str., 1113 Sofia, Bulgaria; (A.A.); (A.I.); (R.S.); (S.U.)
| | - Michael D. Vershinin
- Department of Physics and Astronomy, University of Utah, Salt Lake City, UT 84112, USA; (W.P.); (R.G.); (H.D.)
- Center for Cell and Genome Science, University of Utah, Salt Lake City, UT 84112, USA
- Department of Biology, University of Utah, Salt Lake City, UT 84112, USA
| | - Saveez Saffarian
- Department of Physics and Astronomy, University of Utah, Salt Lake City, UT 84112, USA; (W.P.); (R.G.); (H.D.)
- Center for Cell and Genome Science, University of Utah, Salt Lake City, UT 84112, USA
- Department of Biology, University of Utah, Salt Lake City, UT 84112, USA
| | - Stoyno S. Stoynov
- Institute of Molecular Biology, Bulgarian Academy of Sciences, 21, G. Bontchev Str., 1113 Sofia, Bulgaria; (A.A.); (A.I.); (R.S.); (S.U.)
| |
Collapse
|
15
|
Prosper P, Rodríguez Puertas R, Guérin DMA, Branda MM. Computational method for designing vaccines applied to virus-like particles (VLPs) as epitope carriers. Vaccine 2024; 42:3916-3929. [PMID: 38782665 DOI: 10.1016/j.vaccine.2024.05.025] [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: 09/21/2023] [Revised: 04/06/2024] [Accepted: 05/04/2024] [Indexed: 05/25/2024]
Abstract
Nonenveloped virus-like particles (VLPs) are self-assembled oligomeric structures composed of one or more proteins that originate from diverse viruses. Because these VLPs have similar antigenicity to the parental virus, they are successfully used as vaccines against cognate virus infection. Furthermore, after foreign antigenic sequences are inserted in their protein components (chimVLPs), some VLPs are also amenable to producing vaccines against pathogens other than the virus it originates from (these VLPs are named platform or epitope carrier). Designing chimVLP vaccines is challenging because the immunogenic response must be oriented against a given antigen without altering stimulant properties inherent to the VLP. An important step in this process is choosing the location of the sequence modifications because this must be performed without compromising the assembly and stability of the original VLP. Currently, many immunogenic data and computational tools can help guide the design of chimVLPs, thus reducing experimental costs and work. In this study, we analyze the structure of a novel VLP that originate from an insect virus and describe the putative regions of its three structural proteins amenable to insertion. For this purpose, we employed molecular dynamics (MD) simulations to assess chimVLP stability by comparing mutated and wild-type (WT) VLP protein trajectories. We applied this procedure to design a chimVLP that can serve as a prophylactic vaccine against the SARS-CoV-2 virus. The methodology described in this work is generally applicable for VLP-based vaccine development.
Collapse
Affiliation(s)
- Pascalita Prosper
- Instituto de Física Aplicada - INFAP, Universidad Nacional de San Luis/CONICET, Argentina, Av. Ejército de los Andes 950, 5700 San Luis, San Luis, Argentina
| | - Rafael Rodríguez Puertas
- Universidad del País Vasco (UPV/EHU), Dept. Farmacología, Facultad de Medicina, B° Sarriena S/N, 48940 Leioa, Vizcaya, Spain; Neurodegenerative Diseases, BioCruces Bizkaia Health Research Institute, Barakaldo, Spain
| | - Diego M A Guérin
- Universidad del País Vasco (UPV/EHU) and Instituto Biofisika (CSIC, UPV/EHU), B° Sarriena S/N, 48940 Leioa, Vizcaya, Spain
| | - María Marta Branda
- Instituto de Física Aplicada - INFAP, Universidad Nacional de San Luis/CONICET, Argentina, Av. Ejército de los Andes 950, 5700 San Luis, San Luis, Argentina.
| |
Collapse
|
16
|
Bayraktar C, Kayabolen A, Odabas A, Durgun A, Kok I, Sevinc K, Supramaniam A, Idris A, Bagci-Onder T. ACE2-Decorated Virus-Like Particles Effectively Block SARS-CoV-2 Infection. Int J Nanomedicine 2024; 19:6931-6943. [PMID: 39005960 PMCID: PMC11246629 DOI: 10.2147/ijn.s446093] [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: 10/23/2023] [Accepted: 05/02/2024] [Indexed: 07/16/2024] Open
Abstract
Purpose Over the past three years, extensive research has been dedicated to understanding and combating COVID-19. Targeting the interaction between the SARS-CoV-2 Spike protein and the ACE2 receptor has emerged as a promising therapeutic strategy against SARS-CoV-2. This study aimed to develop ACE2-coated virus-like particles (ACE2-VLPs), which can be utilized to prevent viral entry into host cells and efficiently neutralize the virus. Methods Virus-like particles were generated through the utilization of a packaging plasmid in conjunction with a plasmid containing the ACE2 envelope sequence. Subsequently, ACE2-VLPs and ACE2-EVs were purified via ultracentrifugation. The quantification of VLPs was validated through multiple methods, including Nanosight 3000, TEM imaging, and Western blot analysis. Various packaging systems were explored to optimize the ACE2-VLP configuration for enhanced neutralization capabilities. The evaluation of neutralization effectiveness was conducted using pseudoviruses bearing different spike protein variants. Furthermore, the study assessed the neutralization potential against the Omicron BA.1 variant in Vero E6 cells. Results ACE2-VLPs showed a high neutralization capacity even at low doses and demonstrated superior efficacy in in vitro pseudoviral assays compared to extracellular vesicles carrying ACE2. ACE2-VLPs remained stable under various environmental temperatures and effectively blocked all tested variants of concern in vitro. Notably, they exhibited significant neutralization against Omicron BA.1 variant in Vero E6 cells. Given their superior efficacy compared to extracellular vesicles and proven success against live virus, ACE2-VLPs stand out as crucial candidates for treating SARS-CoV-2 infections. Conclusion This novel therapeutic approach of coating VLPs with receptor particles provides a proof-of-concept for designing effective neutralization strategies for other viral diseases in the future.
Collapse
Affiliation(s)
- Canan Bayraktar
- Koç University Research Center for Translational Medicine (KUTTAM), Koç University, Istanbul, Turkey
| | - Alisan Kayabolen
- Koç University Research Center for Translational Medicine (KUTTAM), Koç University, Istanbul, Turkey
| | - Arda Odabas
- Koç University Research Center for Translational Medicine (KUTTAM), Koç University, Istanbul, Turkey
| | - Aysegul Durgun
- Koç University Research Center for Translational Medicine (KUTTAM), Koç University, Istanbul, Turkey
| | - Ipek Kok
- Koç University Research Center for Translational Medicine (KUTTAM), Koç University, Istanbul, Turkey
| | - Kenan Sevinc
- Koç University Research Center for Translational Medicine (KUTTAM), Koç University, Istanbul, Turkey
| | - Aroon Supramaniam
- Menzies Health Institute Queensland, School of Medical Science Griffith University, Gold Coast Campus, Brisbane, QLD, Australia
| | - Adi Idris
- Menzies Health Institute Queensland, School of Medical Science Griffith University, Gold Coast Campus, Brisbane, QLD, Australia
- Centre for Immunology and Infection Control, School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| | - Tugba Bagci-Onder
- Koç University Research Center for Translational Medicine (KUTTAM), Koç University, Istanbul, Turkey
| |
Collapse
|
17
|
Berreiros-Hortala H, Vilchez-Pinto G, Diaz-Perales A, Garrido-Arandia M, Tome-Amat J. Virus-like Particles as Vaccines for Allergen-Specific Therapy: An Overview of Current Developments. Int J Mol Sci 2024; 25:7429. [PMID: 39000536 PMCID: PMC11242184 DOI: 10.3390/ijms25137429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 07/02/2024] [Accepted: 07/04/2024] [Indexed: 07/16/2024] Open
Abstract
Immune engineering and modulation are the basis of a novel but powerful tool to treat immune diseases using virus-like particles (VLPs). VLPs are formed by the viral capsid without genetic material making them non-infective. However, they offer a wide variety of possibilities as antigen-presenting platforms, resulting in high immunogenicity and high efficacy in immune modulation, with low allergenicity. Both animal and plant viruses are being studied for use in the treatment of food allergies. These formulations are combined with adjuvants, T-stimulatory epitopes, TLR ligands, and other immune modulators to modulate or enhance the immune response toward the presented allergen. Here, the authors present an overview of VLP production systems, their immune modulation capabilities, and the applicability of actual VLP-based formulations targeting allergic diseases.
Collapse
Affiliation(s)
- Helena Berreiros-Hortala
- Centro de Biotecnología y Genómica de Plantas, Universidad Politécnica de Madrid (UPM)-Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA/CSIC), Campus de Montegancedo UPM, Pozuelo de Alarcón, 28223 Madrid, Spain; (H.B.-H.); (G.V.-P.); (A.D.-P.); (M.G.-A.)
- Departamento de Biotecnología-Biología Vegetal, Escuela Técnica Superior de Ingeniería Agronómica, Alimentaria y de Biosistemas, UPM, 28040 Madrid, Spain
| | - Gonzalo Vilchez-Pinto
- Centro de Biotecnología y Genómica de Plantas, Universidad Politécnica de Madrid (UPM)-Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA/CSIC), Campus de Montegancedo UPM, Pozuelo de Alarcón, 28223 Madrid, Spain; (H.B.-H.); (G.V.-P.); (A.D.-P.); (M.G.-A.)
- Departamento de Biotecnología-Biología Vegetal, Escuela Técnica Superior de Ingeniería Agronómica, Alimentaria y de Biosistemas, UPM, 28040 Madrid, Spain
| | - Araceli Diaz-Perales
- Centro de Biotecnología y Genómica de Plantas, Universidad Politécnica de Madrid (UPM)-Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA/CSIC), Campus de Montegancedo UPM, Pozuelo de Alarcón, 28223 Madrid, Spain; (H.B.-H.); (G.V.-P.); (A.D.-P.); (M.G.-A.)
- Departamento de Biotecnología-Biología Vegetal, Escuela Técnica Superior de Ingeniería Agronómica, Alimentaria y de Biosistemas, UPM, 28040 Madrid, Spain
| | - Maria Garrido-Arandia
- Centro de Biotecnología y Genómica de Plantas, Universidad Politécnica de Madrid (UPM)-Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA/CSIC), Campus de Montegancedo UPM, Pozuelo de Alarcón, 28223 Madrid, Spain; (H.B.-H.); (G.V.-P.); (A.D.-P.); (M.G.-A.)
- Departamento de Biotecnología-Biología Vegetal, Escuela Técnica Superior de Ingeniería Agronómica, Alimentaria y de Biosistemas, UPM, 28040 Madrid, Spain
| | - Jaime Tome-Amat
- Centro de Biotecnología y Genómica de Plantas, Universidad Politécnica de Madrid (UPM)-Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA/CSIC), Campus de Montegancedo UPM, Pozuelo de Alarcón, 28223 Madrid, Spain; (H.B.-H.); (G.V.-P.); (A.D.-P.); (M.G.-A.)
- Departamento de Biotecnología-Biología Vegetal, Escuela Técnica Superior de Ingeniería Agronómica, Alimentaria y de Biosistemas, UPM, 28040 Madrid, Spain
| |
Collapse
|
18
|
Wang Y, Li Q, Peng P, Zhang Q, Huang Y, Hu J, Hu Z, Liu X. Dual N-linked glycosylation at residues 133 and 158 in the hemagglutinin are essential for the efficacy of H7N9 avian influenza virus like particle vaccine in chickens and mice. Vet Microbiol 2024; 294:110108. [PMID: 38729093 DOI: 10.1016/j.vetmic.2024.110108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 04/25/2024] [Accepted: 05/04/2024] [Indexed: 05/12/2024]
Abstract
H7N9 subtype avian influenza virus (AIV) poses a great challenge to poultry industry. Virus-like particle (VLP) is a prospective alternative for the traditional egg-based influenza vaccines. N-linked glycosylation (NLG) regulates the efficacy of influenza vaccines, whereas the impact of NLG modifications on the efficacy of influenza VLP vaccines remains unclear. Here, H7N9 VLPs were assembled in insect cells through co-infection with the baculoviruses expressing the NLG-modified hemagglutinin (HA), neuraminidase and matrix proteins, and the VLP vaccines were assessed in chickens and mice. NLG modifications significantly enhanced hemagglutination-inhibition and virus neutralization antibody responses in mice, rather than in chickens, because different immunization strategies were used in these animal models. The presence of dual NLG at residues 133 and 158 significantly elevated HA-binding IgG titers in chickens and mice. The VLP vaccines conferred complete protection and significantly suppressed virus replication and lung pathology post challenge with H7N9 viruses in chickens and mice. VLP immunization activated T cell immunity-related cytokine response and inhibited inflammatory cytokine response in mouse lung. Of note, the presence of dual NLG at residues 133 and 158 optimized the capacity of the VLP vaccine to stimulate interleukin-4 expression, inhibit virus shedding or alleviate lung pathology in chickens or mice. Intriguingly, the VLP vaccine with NLG addition at residue 133 provided partial cross-protection against the H5Nx subtype AIVs in chickens and mice. In conclusion, dual NLG at residues 133 and 158 in HA can be potentially used to enhance the efficacy of H7N9 VLP vaccines in chickens and mammals.
Collapse
Affiliation(s)
- Yufei Wang
- Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, Jiangsu, China
| | - Qun Li
- Yangzhou Uni-Bio Pharmaceutical Co., Ltd, Yangzhou, Jiangsu, China
| | - Peipei Peng
- Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, Jiangsu, China
| | - Qi Zhang
- Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, Jiangsu, China
| | - Yalan Huang
- Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, Jiangsu, China
| | - Jiao Hu
- Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, Jiangsu, China
| | - Zenglei Hu
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, Jiangsu, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, China.
| | - Xiufan Liu
- Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, Jiangsu, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, China
| |
Collapse
|
19
|
Lopes Chaves L, Dourado D, Prunache IB, Manuelle Marques da Silva P, Tacyana Dos Santos Lucena G, Cardoso de Souza Z, Muniz Mendes Freire de Moura P, Nunes Bordallo H, Rocha Formiga F, de Souza Rebouças J. Nanocarriers of antigen proteins for vaccine delivery. Int J Pharm 2024; 659:124162. [PMID: 38663646 DOI: 10.1016/j.ijpharm.2024.124162] [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: 12/07/2023] [Revised: 04/17/2024] [Accepted: 04/22/2024] [Indexed: 06/06/2024]
Abstract
Nanoformulations in vaccinology provide antigen stability and enhanced immunogenicity, in addition to providing targeted delivery and controlled release. In the last years, much research has been focused on vaccine development using virus-like particles, liposomes, emulsions, polymeric, lipid, and inorganic nanoparticles. Importantly, nanoparticle interactions with innate and adaptive immune systems must be clearly understood to guide the rational development of nanovaccines. This review provides a recap and updates on different aspects advocating nanoparticles as promising antigen carriers and immune cell activators for vaccination. Moreover, it offers a discussion of how the physicochemical properties of nanoparticles are modified to target specific cells and improve vaccine efficacy.
Collapse
Affiliation(s)
- Luíse Lopes Chaves
- Department of Immunology, Aggeu Magalhães Institute (IAM), Oswaldo Cruz Foundation (FIOCRUZ), 50670-420, Recife, PE, Brazil; Graduate Program in Applied Cellular and Molecular Biology (PGBCMA), Institute of Biological Sciences, University of Pernambuco (UPE), 50100-130, Recife, PE, Brazil
| | - Douglas Dourado
- Department of Immunology, Aggeu Magalhães Institute (IAM), Oswaldo Cruz Foundation (FIOCRUZ), 50670-420, Recife, PE, Brazil
| | - Ioana-Bianca Prunache
- Niels Bohr Institute, University of Copenhagen, Universitetsparken 5, Copenhagen 2100, Denmark
| | - Paloma Manuelle Marques da Silva
- Graduate Program in Applied Cellular and Molecular Biology (PGBCMA), Institute of Biological Sciences, University of Pernambuco (UPE), 50100-130, Recife, PE, Brazil
| | - Gislayne Tacyana Dos Santos Lucena
- Graduate Program in Applied Cellular and Molecular Biology (PGBCMA), Institute of Biological Sciences, University of Pernambuco (UPE), 50100-130, Recife, PE, Brazil
| | - Zilyane Cardoso de Souza
- Graduate Program in Applied Cellular and Molecular Biology (PGBCMA), Institute of Biological Sciences, University of Pernambuco (UPE), 50100-130, Recife, PE, Brazil
| | - Patrícia Muniz Mendes Freire de Moura
- Graduate Program in Applied Cellular and Molecular Biology (PGBCMA), Institute of Biological Sciences, University of Pernambuco (UPE), 50100-130, Recife, PE, Brazil
| | - Heloísa Nunes Bordallo
- Niels Bohr Institute, University of Copenhagen, Universitetsparken 5, Copenhagen 2100, Denmark
| | - Fabio Rocha Formiga
- Department of Immunology, Aggeu Magalhães Institute (IAM), Oswaldo Cruz Foundation (FIOCRUZ), 50670-420, Recife, PE, Brazil; Graduate Program in Applied Cellular and Molecular Biology (PGBCMA), Institute of Biological Sciences, University of Pernambuco (UPE), 50100-130, Recife, PE, Brazil.
| | - Juliana de Souza Rebouças
- Graduate Program in Applied Cellular and Molecular Biology (PGBCMA), Institute of Biological Sciences, University of Pernambuco (UPE), 50100-130, Recife, PE, Brazil
| |
Collapse
|
20
|
Laxmi B, Devi PUM, Thanjavur N, Buddolla V. The Applications of Artificial Intelligence (AI)-Driven Tools in Virus-Like Particles (VLPs) Research. Curr Microbiol 2024; 81:234. [PMID: 38904765 DOI: 10.1007/s00284-024-03750-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Accepted: 05/26/2024] [Indexed: 06/22/2024]
Abstract
Viral-like particles (VLPs) represent versatile nanoscale structures mimicking the morphology and antigenic characteristics of viruses, devoid of genetic material, making them promising candidates for various biomedical applications. The integration of artificial intelligence (AI) into VLP research has catalyzed significant advancements in understanding, production, and therapeutic applications of these nanostructures. This comprehensive review explores the collaborative utilization of AI tools, computational methodologies, and state-of-the-art technologies within the VLP domain. AI's involvement in bioinformatics facilitates sequencing and structure prediction, unraveling genetic intricacies and three-dimensional configurations of VLPs. Furthermore, AI-enabled drug discovery enables virtual screening, demonstrating promise in identifying compounds to inhibit VLP activity. In VLP production, AI optimizes processes by providing strategies for culture conditions, nutrient concentrations, and growth kinetics. AI's utilization in image analysis and electron microscopy expedites VLP recognition and quantification. Moreover, network analysis of protein-protein interactions through AI tools offers an understanding of VLP interactions. The integration of multi-omics data via AI analytics provides a comprehensive view of VLP behavior. Predictive modeling utilizing machine learning algorithms aids in forecasting VLP stability, guiding optimization efforts. Literature mining facilitated by text mining algorithms assists in summarizing information from the VLP knowledge corpus. Additionally, AI's role in laboratory automation enhances experimental efficiency. Addressing data security concerns, AI ensures the protection of sensitive information in the digital era of VLP research. This review serves as a roadmap, providing insights into AI's current and future applications in VLP research, thereby guiding innovative directions in medicine and beyond.
Collapse
Affiliation(s)
- Bugude Laxmi
- Department of Applied Microbiology, Sri Padmavati Mahila Visvavidyalayam, Padmavathi Nagar, Tirupati, Andhra Pradesh, 517502, India
| | - Palempalli Uma Maheswari Devi
- Department of Applied Microbiology, Sri Padmavati Mahila Visvavidyalayam, Padmavathi Nagar, Tirupati, Andhra Pradesh, 517502, India.
| | - Naveen Thanjavur
- Dr. Buddolla's Institute of Life Sciences (A Unit of Dr. Buddolla's Research and Educational Society), Tirupati, 517506, India
| | - Viswanath Buddolla
- Dr. Buddolla's Institute of Life Sciences (A Unit of Dr. Buddolla's Research and Educational Society), Tirupati, 517506, India.
| |
Collapse
|
21
|
Sobczak JM, Barkovska I, Balke I, Rothen DA, Mohsen MO, Skrastina D, Ogrina A, Martina B, Jansons J, Bogans J, Vogel M, Bachmann MF, Zeltins A. Identifying Key Drivers of Efficient B Cell Responses: On the Role of T Help, Antigen-Organization, and Toll-like Receptor Stimulation for Generating a Neutralizing Anti-Dengue Virus Response. Vaccines (Basel) 2024; 12:661. [PMID: 38932390 PMCID: PMC11209419 DOI: 10.3390/vaccines12060661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 05/14/2024] [Accepted: 05/23/2024] [Indexed: 06/28/2024] Open
Abstract
T help (Th), stimulation of toll-like receptors (pathogen-associated molecular patterns, PAMPs), and antigen organization and repetitiveness (pathogen-associated structural patterns, PASPs) were shown numerous times to be important in driving B-cell and antibody responses. In this study, we dissected the individual contributions of these parameters using newly developed "Immune-tag" technology. As model antigens, we used eGFP and the third domain of the dengue virus 1 envelope protein (DV1 EDIII), the major target of virus-neutralizing antibodies. The respective proteins were expressed alone or genetically fused to the N-terminal fragment of the cucumber mosaic virus (CMV) capsid protein-nCMV, rendering the antigens oligomeric. In a step-by-step manner, RNA was attached as a PAMP, and/or a universal Th-cell epitope was genetically added for additional Th. Finally, a PASP was added to the constructs by displaying the antigens highly organized and repetitively on the surface of CMV-derived virus-like particles (CuMV VLPs). Sera from immunized mice demonstrated that each component contributed stepwise to the immunogenicity of both proteins. All components combined in the CuMV VLP platform induced by far the highest antibody responses. In addition, the DV1 EDIII induced high levels of DENV-1-neutralizing antibodies only if displayed on VLPs. Thus, combining multiple cues typically associated with viruses results in optimal antibody responses.
Collapse
Affiliation(s)
- Jan M. Sobczak
- Department of Immunology, University Clinic of Rheumatology and Immunology, Inselspital, CH-3010 Bern, Switzerland; (D.A.R.); (M.O.M.); (M.V.); (M.F.B.)
- Department of BioMedical Research, University of Bern, CH-3008 Bern, Switzerland
| | - Irena Barkovska
- Latvian Biomedical Research and Study Centre, LV-1067 Riga, Latvia; (I.B.); (I.B.); (D.S.); (A.O.); (J.J.); (J.B.); (A.Z.)
| | - Ina Balke
- Latvian Biomedical Research and Study Centre, LV-1067 Riga, Latvia; (I.B.); (I.B.); (D.S.); (A.O.); (J.J.); (J.B.); (A.Z.)
| | - Dominik A. Rothen
- Department of Immunology, University Clinic of Rheumatology and Immunology, Inselspital, CH-3010 Bern, Switzerland; (D.A.R.); (M.O.M.); (M.V.); (M.F.B.)
- Department of BioMedical Research, University of Bern, CH-3008 Bern, Switzerland
| | - Mona O. Mohsen
- Department of Immunology, University Clinic of Rheumatology and Immunology, Inselspital, CH-3010 Bern, Switzerland; (D.A.R.); (M.O.M.); (M.V.); (M.F.B.)
- Department of BioMedical Research, University of Bern, CH-3008 Bern, Switzerland
| | - Dace Skrastina
- Latvian Biomedical Research and Study Centre, LV-1067 Riga, Latvia; (I.B.); (I.B.); (D.S.); (A.O.); (J.J.); (J.B.); (A.Z.)
| | - Anete Ogrina
- Latvian Biomedical Research and Study Centre, LV-1067 Riga, Latvia; (I.B.); (I.B.); (D.S.); (A.O.); (J.J.); (J.B.); (A.Z.)
| | - Byron Martina
- Artemis Bioservices, 2629 JD Delft, The Netherlands;
- Protinhi Therapeutics, 6534 AT Nijmegen, The Netherlands
| | - Juris Jansons
- Latvian Biomedical Research and Study Centre, LV-1067 Riga, Latvia; (I.B.); (I.B.); (D.S.); (A.O.); (J.J.); (J.B.); (A.Z.)
| | - Janis Bogans
- Latvian Biomedical Research and Study Centre, LV-1067 Riga, Latvia; (I.B.); (I.B.); (D.S.); (A.O.); (J.J.); (J.B.); (A.Z.)
| | - Monique Vogel
- Department of Immunology, University Clinic of Rheumatology and Immunology, Inselspital, CH-3010 Bern, Switzerland; (D.A.R.); (M.O.M.); (M.V.); (M.F.B.)
- Department of BioMedical Research, University of Bern, CH-3008 Bern, Switzerland
| | - Martin F. Bachmann
- Department of Immunology, University Clinic of Rheumatology and Immunology, Inselspital, CH-3010 Bern, Switzerland; (D.A.R.); (M.O.M.); (M.V.); (M.F.B.)
- Department of BioMedical Research, University of Bern, CH-3008 Bern, Switzerland
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford OX3 7BN, UK
| | - Andris Zeltins
- Latvian Biomedical Research and Study Centre, LV-1067 Riga, Latvia; (I.B.); (I.B.); (D.S.); (A.O.); (J.J.); (J.B.); (A.Z.)
| |
Collapse
|
22
|
BenDavid E, Ramezanian S, Lu Y, Rousseau J, Schroeder A, Lavertu M, Tremblay JP. Emerging Perspectives on Prime Editor Delivery to the Brain. Pharmaceuticals (Basel) 2024; 17:763. [PMID: 38931430 PMCID: PMC11206523 DOI: 10.3390/ph17060763] [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: 05/09/2024] [Revised: 06/02/2024] [Accepted: 06/03/2024] [Indexed: 06/28/2024] Open
Abstract
Prime editing shows potential as a precision genome editing technology, as well as the potential to advance the development of next-generation nanomedicine for addressing neurological disorders. However, turning in prime editors (PEs), which are macromolecular complexes composed of CRISPR/Cas9 nickase fused with a reverse transcriptase and a prime editing guide RNA (pegRNA), to the brain remains a considerable challenge due to physiological obstacles, including the blood-brain barrier (BBB). This review article offers an up-to-date overview and perspective on the latest technologies and strategies for the precision delivery of PEs to the brain and passage through blood barriers. Furthermore, it delves into the scientific significance and possible therapeutic applications of prime editing in conditions related to neurological diseases. It is targeted at clinicians and clinical researchers working on advancing precision nanomedicine for neuropathologies.
Collapse
Affiliation(s)
- Eli BenDavid
- Laboratory of Biomaterials and Tissue Engineering, Department of Chemical Engineering, Institute of Biomedical Engineering, Polytechnique Montréal, Montréal, QC H3C 3A7, Canada;
- Division of Human Genetics, Centre de Recherche du CHU de Québec—Université Laval, Québec, QC G1V 4G2, Canada
- Laboratory of Molecular Genetics and Gene Therapy, Department of Molecular Medicine, Faculty of Medicine, Laval University, Québec, QC G1V 0A6, Canada
- Laboratory of Nanopharmacology and Pharmaceutical Nanoscience, Faculty of Pharmacy, Laval University, Québec, QC G1V 4G2, Canada
- Rappaport Faculty of Medicine, Technion—Israel Institute of Technology, Haifa 3525433, Israel
| | - Sina Ramezanian
- Division of Human Genetics, Centre de Recherche du CHU de Québec—Université Laval, Québec, QC G1V 4G2, Canada
- Laboratory of Molecular Genetics and Gene Therapy, Department of Molecular Medicine, Faculty of Medicine, Laval University, Québec, QC G1V 0A6, Canada
| | - Yaoyao Lu
- Division of Human Genetics, Centre de Recherche du CHU de Québec—Université Laval, Québec, QC G1V 4G2, Canada
- Laboratory of Molecular Genetics and Gene Therapy, Department of Molecular Medicine, Faculty of Medicine, Laval University, Québec, QC G1V 0A6, Canada
| | - Joël Rousseau
- Division of Human Genetics, Centre de Recherche du CHU de Québec—Université Laval, Québec, QC G1V 4G2, Canada
| | - Avi Schroeder
- Laboratory for Targeted Drug Delivery and Personalized Medicine Technologies, Department of Chemical Engineering, Technion—Israel Institute of Technology, Haifa 3200003, Israel;
| | - Marc Lavertu
- Laboratory of Biomaterials and Tissue Engineering, Department of Chemical Engineering, Institute of Biomedical Engineering, Polytechnique Montréal, Montréal, QC H3C 3A7, Canada;
| | - Jacques P. Tremblay
- Division of Human Genetics, Centre de Recherche du CHU de Québec—Université Laval, Québec, QC G1V 4G2, Canada
- Laboratory of Molecular Genetics and Gene Therapy, Department of Molecular Medicine, Faculty of Medicine, Laval University, Québec, QC G1V 0A6, Canada
| |
Collapse
|
23
|
Karan S, Durán-Meza AL, Chapman A, Tanimoto C, Chan SK, Knobler CM, Gelbart WM, Steinmetz NF. In Vivo Delivery of Spherical and Cylindrical In Vitro Reconstituted Virus-like Particles Containing the Same Self-Amplifying mRNA. Mol Pharm 2024; 21:2727-2739. [PMID: 38709860 PMCID: PMC11250921 DOI: 10.1021/acs.molpharmaceut.3c01105] [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] [Indexed: 05/08/2024]
Abstract
The dramatic effectiveness of recent mRNA (mRNA)-based COVID vaccines delivered in lipid nanoparticles has highlighted the promise of mRNA therapeutics in general. In this report, we extend our earlier work on self-amplifying mRNAs delivered in spherical in vitro reconstituted virus-like particles (VLPs), and on drug delivery using cylindrical virus particles. In particular, we carry out separate in vitro assemblies of a self-amplifying mRNA gene in two different virus-like particles: one spherical, formed with the capsid protein of cowpea chlorotic mottle virus (CCMV), and the other cylindrical, formed from the capsid protein of tobacco mosaic virus (TMV). The mRNA gene is rendered self-amplifying by genetically fusing it to the RNA-dependent RNA polymerase (RdRp) of Nodamura virus, and the relative efficacies of cell uptake and downstream protein expression resulting from their CCMV- and TMV-packaged forms are compared directly. This comparison is carried out by their transfections into cells in culture: expressions of two self-amplifying genes, enhanced yellow fluorescent protein (EYFP) and Renilla luciferase (Luc), packaged alternately in CCMV and TMV VLPs, are quantified by fluorescence and chemiluminescence levels, respectively, and relative numbers of the delivered mRNAs are measured by quantitative real-time PCR. The cellular uptake of both forms of these VLPs is further confirmed by confocal microscopy of transfected cells. Finally, VLP-mediated delivery of the self-amplifying-mRNA in mice following footpad injection is shown by in vivo fluorescence imaging to result in robust expression of EYFP in the draining lymph nodes, suggesting the potential of these plant virus-like particles as a promising mRNA gene and vaccine delivery modality. These results establish that both CCMV and TMV VLPs can deliver their in vitro packaged mRNA genes to immune cells and that their self-amplifying forms significantly enhance in situ expression. Choice of one VLP (CCMV or TMV) over the other will depend on which geometry of nucleocapsid is self-assembled more efficiently for a given length and sequence of RNA, and suggests that these plant VLP gene delivery systems will prove useful in a wide variety of medical applications, both preventive and therapeutic.
Collapse
Affiliation(s)
- Sweta Karan
- Department of NanoEngineering, University of California San Diego, La Jolla, California 92093, United States
- Center for Nano-ImmunoEngineering, University of California San Diego, La Jolla, California 92093, United States
- Shu and K. C. Chien and Peter Farrell Collaboratory, University of California San Diego, La Jolla, California 92093, United States
| | - Ana Luisa Durán-Meza
- Department of Chemistry and Biochemistry, University of California, Los Angeles, California 90095, United States
| | - Abigail Chapman
- Department of Chemistry and Biochemistry, University of California, Los Angeles, California 90095, United States
| | - Cheylene Tanimoto
- Department of Chemistry and Biochemistry, University of California, Los Angeles, California 90095, United States
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| | - Soo Khim Chan
- Department of NanoEngineering, University of California San Diego, La Jolla, California 92093, United States
- Center for Nano-ImmunoEngineering, University of California San Diego, La Jolla, California 92093, United States
| | - Charles M Knobler
- Department of Chemistry and Biochemistry, University of California, Los Angeles, California 90095, United States
| | - William M Gelbart
- Department of Chemistry and Biochemistry, University of California, Los Angeles, California 90095, United States
- UCLA Molecular Biology Institute, University of California, Los Angeles, California 90095, United States
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| | - Nicole F Steinmetz
- Department of NanoEngineering, University of California San Diego, La Jolla, California 92093, United States
- Center for Nano-ImmunoEngineering, University of California San Diego, La Jolla, California 92093, United States
- Institute for Materials Discovery and Design, University of California San Diego, La Jolla, California 92093, United States
- Department of Bioengineering, University of California San Diego, La Jolla, California 92093, United States
- Department of Radiology, University of California San Diego, La Jolla, California 92093, United States
- Moores Cancer Center, University of California San Diego, La Jolla, California 92093, United States
- Center for Engineering in Cancer, Institute for Engineering in Medicine, University of California, San Diego, La Jolla, California 92093, United States
- Shu and K. C. Chien and Peter Farrell Collaboratory, University of California San Diego, La Jolla, California 92093, United States
| |
Collapse
|
24
|
Karaaslan C, Wirz O, Tan G, Globinska A, Boonpiyathad T, Hedman K, Vaselek S, Venermo MS, Jartti T, Akdis M, Akdis CA. B cell immune response to human bocaviruses. Clin Exp Allergy 2024; 54:388-401. [PMID: 38321724 DOI: 10.1111/cea.14453] [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: 05/24/2023] [Revised: 01/03/2024] [Accepted: 01/15/2024] [Indexed: 02/08/2024]
Abstract
BACKGROUND Human bocaviruses (HBoVs) have been demonstrated in respiratory and gastrointestinal infections; however, the immune response to them has not been studied in detail. In this study, we investigated the B cell immune responses to HBoV1 and HBoV2, representing two different species of bocaviruses in humans. METHODS We analyzed the effects of stimulations with HBoV1 and 2 virus-like particles (VLPs) and of co-stimulation with HBoV1-rhinovirus (RV) on cells of the immune system by flow cytometry, transcriptomics, and luminometric immune assays. RESULTS Human B cells, and particularly B regulatory cells (Breg cells), showed an increased immune response to HBoV1-VLPs stimulation. These immune responses were also supported by increased IL-1RA and PDL1 expressions in IL-10+ B cells from peripheral blood mononuclear cells (PBMCs) stimulated with HBoV1-VLPs. In addition, increased levels of IL-10 and IL-1RA were determined in the supernatants of PBMCs following HBoV1-VLPs stimulation. HBoV1-VLPs and RV co-stimulation increased the IL-10+ B cell population. Transcriptome analysis by next-generation RNA sequencing showed an increased expression of IL-10 signalling and Breg cell markers in PBMCs stimulated with HBoV1-VLPs. Furthermore, TGF-β and chemoattractants MIP-1α, MIP-1β and IP10 protein levels were high in the supernatants of PBMCs stimulated with HBoV1-VLPs. CONCLUSIONS The findings demonstrate that in Breg cells, IL-10 signalling pathways, and anti-inflammatory activity are induced by HBoV1, which can explain the often mild nature of the disease. In addition, the immune regulatory response induced by HBoV1-VLPs may indicate a potential immunomodulatory role of HBoV1 on the immune system and may represent an immune regulatory strategy.
Collapse
Affiliation(s)
- Cagatay Karaaslan
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- Molecular Biology Section, Biology Department, Faculty of Science, Hacettepe University, Ankara, Turkey
| | - Oliver Wirz
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Ge Tan
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Anna Globinska
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Tadech Boonpiyathad
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Klaus Hedman
- Department of Virology, University of Helsinki, Helsinki, Finland
- Helsinki University Hospital Diagnostics Center, Helsinki, Finland
| | - Slavica Vaselek
- Molecular Biology Section, Biology Department, Faculty of Science, Hacettepe University, Ankara, Turkey
| | | | - Tuomas Jartti
- Research Unit of Clinical Medicine, University of Oulu, Oulu, Finland
- Department of Pediatrics and Adolescent Medicine, Oulu University Hospital, Oulu, Finland
- Department of Pediatrics and Adolescent Medicine, Turku University Hospital and University of Turku, Turku, Finland
| | - Mubeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Cezmi A Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| |
Collapse
|
25
|
Puarattana-aroonkorn S, Tharakaraman K, Suriyawipada D, Ruchirawat M, Fuangthong M, Sasisekharan R, Artpradit C. Rapid and Scalable Production of Functional SARS-CoV-2 Virus-like Particles (VLPs) by a Stable HEK293 Cell Pool. Vaccines (Basel) 2024; 12:561. [PMID: 38932290 PMCID: PMC11209123 DOI: 10.3390/vaccines12060561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 05/06/2024] [Accepted: 05/09/2024] [Indexed: 06/28/2024] Open
Abstract
At times of pandemics, such as the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, the situation demands rapid development and production timelines of safe and effective vaccines for delivering life-saving medications quickly to patients. Typical biologics production relies on using the lengthy and arduous approach of stable single-cell clones. Here, we used an alternative approach, a stable cell pool that takes only weeks to generate compared to a stable single-cell clone that needs several months to complete. We employed the membrane, envelope, and highly immunogenic spike proteins of SARS-CoV-2 to produce virus-like particles (VLPs) using the HEK293-F cell line as a host system with an economical transfection reagent. The cell pool showed the stability of protein expression for more than one month. We demonstrated that the production of SARS-CoV-2 VLPs using this cell pool was scalable up to a stirred-tank 2 L bioreactor in fed-batch mode. The purified VLPs were properly assembled, and their size was consistent with the authentic virus. Our particles were functional as they specifically entered the cell that naturally expresses ACE-2. Notably, this work reports a practical and cost-effective manufacturing platform for scalable SARS-CoV-2 VLPs production and chromatographic purification.
Collapse
Affiliation(s)
| | - Kannan Tharakaraman
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Disapan Suriyawipada
- Translational Research Unit, Chulabhorn Research Institute, Bangkok 10210, Thailand (M.F.)
| | - Mathuros Ruchirawat
- Translational Research Unit, Chulabhorn Research Institute, Bangkok 10210, Thailand (M.F.)
- Center of Excellence on Environmental Health and Toxicology (EHT), OPS, MHESI, Bangkok 10400, Thailand
| | - Mayuree Fuangthong
- Translational Research Unit, Chulabhorn Research Institute, Bangkok 10210, Thailand (M.F.)
- Center of Excellence on Environmental Health and Toxicology (EHT), OPS, MHESI, Bangkok 10400, Thailand
- Program in Applied Biological Sciences, Chulabhorn Graduate Institute, Bangkok 10210, Thailand
| | - Ram Sasisekharan
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Charlermchai Artpradit
- Translational Research Unit, Chulabhorn Research Institute, Bangkok 10210, Thailand (M.F.)
| |
Collapse
|
26
|
Sanchez-Martinez ZV, Alpuche-Lazcano SP, Stuible M, Durocher Y. CHO cells for virus-like particle and subunit vaccine manufacturing. Vaccine 2024; 42:2530-2542. [PMID: 38503664 DOI: 10.1016/j.vaccine.2024.03.034] [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: 01/14/2024] [Revised: 02/27/2024] [Accepted: 03/13/2024] [Indexed: 03/21/2024]
Abstract
Chinese Hamster Ovary (CHO) cells, employed primarily for manufacturing monoclonal antibodies and other recombinant protein (r-protein) therapeutics, are emerging as a promising host for vaccine antigen production. This is exemplified by the recently approved CHO cell-derived subunit vaccines (SUV) against respiratory syncytial virus (RSV) and varicella-zoster virus (VZV), as well as the enveloped virus-like particle (eVLP) vaccine against hepatitis B virus (HBV). Here, we summarize the design, production, and immunogenicity features of these vaccine and review the most recent progress of other CHO-derived vaccines in pre-clinical and clinical development. We also discuss the challenges associated with vaccine production in CHO cells, with a focus on ensuring viral clearance for eVLP products.
Collapse
Affiliation(s)
- Zalma V Sanchez-Martinez
- Human Health Therapeutics Research Centre, National Research Council of Canada, Montreal, QC H4P 2R2, Canada; Department of Biochemistry and Molecular Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada
| | - Sergio P Alpuche-Lazcano
- Human Health Therapeutics Research Centre, National Research Council of Canada, Montreal, QC H4P 2R2, Canada
| | - Matthew Stuible
- Human Health Therapeutics Research Centre, National Research Council of Canada, Montreal, QC H4P 2R2, Canada
| | - Yves Durocher
- Human Health Therapeutics Research Centre, National Research Council of Canada, Montreal, QC H4P 2R2, Canada; Department of Biochemistry and Molecular Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada; PROTEO: The Quebec Network for Research on Protein Function, Structure, and Engineering, Université du Québec à Montréal, 201 Avenue du Président Kennedy, Montréal, QC H2X 3Y7, Canada.
| |
Collapse
|
27
|
Grigoryan L, Feng Y, Bellusci L, Lai L, Wali B, Ellis M, Yuan M, Arunachalam PS, Hu M, Kowli S, Gupta S, Maysel-Auslender S, Maecker HT, Samaha H, Rouphael N, Wilson IA, Moreno AC, Suthar MS, Khurana S, Pillet S, Charland N, Ward BJ, Pulendran B. AS03 adjuvant enhances the magnitude, persistence, and clonal breadth of memory B cell responses to a plant-based COVID-19 vaccine in humans. Sci Immunol 2024; 9:eadi8039. [PMID: 38579013 DOI: 10.1126/sciimmunol.adi8039] [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: 05/18/2023] [Accepted: 03/11/2024] [Indexed: 04/07/2024]
Abstract
Vaccine adjuvants increase the breadth of serum antibody responses, but whether this is due to the generation of antigen-specific B cell clones with distinct specificities or the maturation of memory B cell clones that produce broadly cross-reactive antibodies is unknown. Here, we longitudinally analyzed immune responses in healthy adults after two-dose vaccination with either a virus-like particle COVID-19 vaccine (CoVLP), CoVLP adjuvanted with AS03 (CoVLP+AS03), or a messenger RNA vaccination (mRNA-1273). CoVLP+AS03 enhanced the magnitude and durability of circulating antibodies and antigen-specific CD4+ T cell and memory B cell responses. Antigen-specific CD4+ T cells in the CoVLP+AS03 group at day 42 correlated with antigen-specific memory B cells at 6 months. CoVLP+AS03 induced memory B cell responses, which accumulated somatic hypermutations over 6 months, resulting in enhanced neutralization breadth of monoclonal antibodies. Furthermore, the fraction of broadly neutralizing antibodies encoded by memory B cells increased between day 42 and 6 months. These results indicate that AS03 enhances the antigenic breadth of B cell memory at the clonal level and induces progressive maturation of the B cell response.
Collapse
Affiliation(s)
- Lilit Grigoryan
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Yupeng Feng
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA 94305, USA
| | | | - Lilin Lai
- Department of Pediatrics and Department of Microbiology and Immunology, Emory Vaccine Center, Emory National Primate Research Center, Emory School of Medicine, Atlanta, GA 30329, USA
| | - Bushra Wali
- Department of Pediatrics and Department of Microbiology and Immunology, Emory Vaccine Center, Emory National Primate Research Center, Emory School of Medicine, Atlanta, GA 30329, USA
| | - Madison Ellis
- Department of Pediatrics and Department of Microbiology and Immunology, Emory Vaccine Center, Emory National Primate Research Center, Emory School of Medicine, Atlanta, GA 30329, USA
| | - Meng Yuan
- Department of Integrative Structural and Computational Biology, Scripps Research Institute, La Jolla, CA 92037, USA
| | - Prabhu S Arunachalam
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Mengyun Hu
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Sangeeta Kowli
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Sheena Gupta
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Sofia Maysel-Auslender
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Holden T Maecker
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Hady Samaha
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Nadine Rouphael
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
- Hope Clinic of Emory Vaccine Center, Emory University, Decatur, GA 30030, USA
| | - Ian A Wilson
- Department of Integrative Structural and Computational Biology, Scripps Research Institute, La Jolla, CA 92037, USA
| | - Alberto C Moreno
- Department of Medicine, Emory Vaccine Center, Emory National Primate Research Center, Emory University School of Medicine, Atlanta, GA 30329, USA
| | - Mehul S Suthar
- Department of Pediatrics and Department of Microbiology and Immunology, Emory Vaccine Center, Emory National Primate Research Center, Emory School of Medicine, Atlanta, GA 30329, USA
| | | | - Stéphane Pillet
- Medicago Inc., Québec, QC G1V 3V9, Canada
- Research Institute of the McGill University Health Center, 1001 Decarie St., Montréal, QC H4A 3J1, Canada
| | | | - Brian J Ward
- Medicago Inc., Québec, QC G1V 3V9, Canada
- Research Institute of the McGill University Health Center, 1001 Decarie St., Montréal, QC H4A 3J1, Canada
| | - Bali Pulendran
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
28
|
Rani NV, Baig MS, Pathak B, Kapoor N, Krishnan A. Mutation of conserved histidine residues of dengue virus envelope protein impairs viral like particle maturation and secretion. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119682. [PMID: 38301907 DOI: 10.1016/j.bbamcr.2024.119682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 01/23/2024] [Accepted: 01/23/2024] [Indexed: 02/03/2024]
Abstract
Dengue virus (DENV) envelope protein plays crucial role in virus entry and maturation of virus during infection. Maturation of DENV occurs in the trans Golgi network at slightly acidic pH which is close to pKa of histidine. When exposed to the acidic environment of the late secretory pathway, dengue virus particles go through a significant conformational change, whereby interactions of structural proteins envelope (E) and prM proteins are reorganised and enable furin protease to cleave prM resulting in mature virus. In order to study the role of histidine of E protein in DENV maturation, we mutated 7 conserved histidine residues of envelope protein and assessed the percent of budding using viral like particle (VLP) system. Histidine mutants; H144A, H244A, H261A and H282A severely disrupted VLP formation without any significant change in expression in cell and its oligomerization ability. Treatment with acidotropic amine reversed the defect for all 4 mutants suggesting that these histidines could be involved in maturation and release. Over expression of capsid protein slightly enhanced VLP release of H244A and H261A. Similarly, furin over expression increased VLP release of these mutants. Co-immunoprecipitation studies revealed that prM and E interaction is lost for H244A, H261A and H282A mutants at acidic pH but not at neutral pH indicating that they could be involved in histidine switch during maturation at acidic pH. Detailed analysis of the mutants could provide novel insights on the interplay of envelop protein during maturation and aid in target for drug development.
Collapse
Affiliation(s)
- N Veena Rani
- School of Sciences, IGNOU, New Delhi 110068, India
| | - Mirza Sarwar Baig
- Centre for Virology, Jamia Hamdard, New Delhi 110062, India; Department of Molecular Medicine, School of Interdisciplinary Sciences and Technology, Jamia Hamdard, New Delhi 110062, India
| | - Bharti Pathak
- Department of Molecular Medicine, School of Interdisciplinary Sciences and Technology, Jamia Hamdard, New Delhi 110062, India
| | - Neera Kapoor
- School of Sciences, IGNOU, New Delhi 110068, India
| | - Anuja Krishnan
- Department of Molecular Medicine, School of Interdisciplinary Sciences and Technology, Jamia Hamdard, New Delhi 110062, India.
| |
Collapse
|
29
|
Tatarūnas V, Čiapienė I, Giedraitienė A. Precise Therapy Using the Selective Endogenous Encapsidation for Cellular Delivery Vector System. Pharmaceutics 2024; 16:292. [PMID: 38399346 PMCID: PMC10893373 DOI: 10.3390/pharmaceutics16020292] [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: 12/13/2023] [Revised: 02/13/2024] [Accepted: 02/16/2024] [Indexed: 02/25/2024] Open
Abstract
Interindividual variability in drug response is a major problem in the prescription of pharmacological treatments. The therapeutic effect of drugs can be influenced by human genes. Pharmacogenomic guidelines for individualization of treatment have been validated and used for conventional dosage forms. However, drugs can often target non-specific areas and produce both desired and undesired pharmacological effects. The use of nanoparticles, liposomes, or other available forms for drug formulation could help to overcome the latter problem. Virus-like particles based on retroviruses could be a potential envelope for safe and efficient drug formulations. Human endogenous retroviruses would make it possible to overcome the host immune response and deliver drugs to the desired target. PEG10 is a promising candidate that can bind to mRNA because it is secreted like an enveloped virus-like extracellular vesicle. PEG10 is a retrotransposon-derived gene that has been domesticated. Therefore, formulations with PEG10 may have a lower immunogenicity. The use of existing knowledge can lead to the development of suitable drug formulations for the precise treatment of individual diseases.
Collapse
Affiliation(s)
- Vacis Tatarūnas
- Institute of Cardiology, Lithuanian University of Health Sciences, Sukileliu 15, LT 50103 Kaunas, Lithuania; (V.T.); (I.Č.)
| | - Ieva Čiapienė
- Institute of Cardiology, Lithuanian University of Health Sciences, Sukileliu 15, LT 50103 Kaunas, Lithuania; (V.T.); (I.Č.)
| | - Agnė Giedraitienė
- Institute of Microbiology and Virology, Lithuanian University of Health Sciences, Eiveniu 4, LT 50161 Kaunas, Lithuania
| |
Collapse
|
30
|
Park G, Na W, Lim JW, Park C, Lee S, Yeom M, Ga E, Hwang J, Moon S, Jeong DG, Jeong HH, Song D, Haam S. Self-Assembled Nanostructures Presenting Repetitive Arrays of Subunit Antigens for Enhanced Immune Response. ACS NANO 2024; 18:4847-4861. [PMID: 38189789 DOI: 10.1021/acsnano.3c09672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Infectious diseases pose persistent threats to public health, demanding advanced vaccine technologies. Nanomaterial-based delivery systems offer promising solutions to enhance immunogenicity while minimizing reactogenicity. We introduce a self-assembled vaccine (SAV) platform employing antigen-polymer conjugates designed to facilitate robust immune responses. The SAVs exhibit efficient cellular uptake by dendritic cells (DCs) and macrophages, which are crucial players in the innate immune system. The high-density antigen presentation of this SAV platform enhances the affinity for DCs through multivalent recognition, significantly augmenting humoral immunity. SAV induced high levels of immunoglobulin G (IgG), IgG1, and IgG2a, suggesting that mature DCs efficiently induced B cell activation through multivalent antigen recognition. Universality was confirmed by applying it to respiratory viruses, showcasing its potential as a versatile vaccine platform. Furthermore, we have also demonstrated strong protection against influenza A virus infection with SAV containing hemagglutinin, which is used in influenza A virus subunit vaccines. The efficacy and adaptability of this nanostructured vaccine present potential utility in combating infectious diseases.
Collapse
Affiliation(s)
- Geunseon Park
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul 03722, Republic of Korea
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Woonsung Na
- College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Jong-Woo Lim
- Department of Virology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul 08826, Republic of Korea
| | - Chaewon Park
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul 03722, Republic of Korea
| | - Sojeong Lee
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul 03722, Republic of Korea
| | - Minjoo Yeom
- Department of Virology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul 08826, Republic of Korea
| | - Eulhae Ga
- College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Jaehyun Hwang
- College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Suyun Moon
- College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Dae Gwin Jeong
- Bionanotechnology Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea
- Department of Proteome Structural Biology, KRIBB School of Bioscience, University of Science and Technology, Daejeon 34141, Republic of Korea
| | | | - Daesub Song
- Department of Virology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul 08826, Republic of Korea
| | - Seungjoo Haam
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul 03722, Republic of Korea
| |
Collapse
|
31
|
Li J, Ding J, Chen K, Xu X, Shao Y, Zhang D, Yu X, Guo C, Qian J, Ding Z. Protective effects of a novel chimeric virus-like particle vaccine against virulent NDV and IBDV challenge. Vaccine 2024; 42:332-338. [PMID: 38065771 DOI: 10.1016/j.vaccine.2023.11.062] [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: 08/03/2023] [Revised: 11/26/2023] [Accepted: 11/29/2023] [Indexed: 01/01/2024]
Abstract
Newcastle disease (ND) and infectious bursal disease (IBD) pose significant threats to the chicken industry, causing substantial economic losses. Currently, immunization through vaccination is the most effective strategy to prevent ND and IBD but currently used traditional vaccines, including inactivated or attenuated vaccines, face challenges in achieving a balance between immunogenicity and safety. To develop a green and efficient novel vaccine for ND and IBD, we developed a bivalent chimeric virus-like particle vaccine (ND-IBD cVLPs) displaying the ND virus (NDV) HN protein and the IBD virus (IBDV) VP2 protein based on the ND VLPs carrier platform and insect baculovirus expression system. This study aimed to evaluate the immunogenicity and protective efficacy of ND-IBD cVLPs in specific pathogen-free chickens. Chickens were immunized with 50 µg of purified ND-IBD cVLPs at 7 days old, boosted at 21 days old, and challenged at 42 days old. The results demonstrated that ND-IBD cVLPs stimulated highly effective hemagglutination inhibition antibody levels against NDV HN protein and enzyme-linked immunosorbent assay antibody levels against the IBDV VP2 protein. Furthermore, ND-IBD cVLPs provided complete protection against virulent NDV and IBDV challenges and mitigated pathological damage to the lung caused by NDV infection and the bursa of Fabricius caused by IBDV infection. These findings suggest that ND-IBD cVLPs hold promise as a safe and efficient novel vaccine candidate for the effective prevention of ND and IBD, extending the development of a foreign protein delivery platform of ND VLPs.
Collapse
Affiliation(s)
- Jindou Li
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Jiaxin Ding
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Kainan Chen
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Xiaohong Xu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Yanan Shao
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Di Zhang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Xibing Yu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Chunhong Guo
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Jing Qian
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China.
| | - Zhuang Ding
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, College of Veterinary Medicine, Jilin University, Changchun 130062, China.
| |
Collapse
|
32
|
Gashti AB, Agbayani G, Hrapovic S, Nassoury N, Coulombe N, Dudani R, Harrison BA, Akache B, Gilbert R, Chahal PS. Production, purification and immunogenicity of Gag virus-like particles carrying SARS-CoV-2 components. Vaccine 2024; 42:40-52. [PMID: 38042697 DOI: 10.1016/j.vaccine.2023.11.048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 10/28/2023] [Accepted: 11/23/2023] [Indexed: 12/04/2023]
Abstract
The virus-like particle (VLP) platform is a robust inducer of humoral and cellular immune responses; hence, it has been used in vaccine development for several infectious diseases. In the current work, VLPs carrying SARS-CoV-2 Spike (S) protein (Wuhan strain) with an HIV-1 Gag core were produced using suspension HEK 293SF-3F6 cells by transient transfection. The Gag was fused with green fluorescent protein (GFP) for rapid quantification of the VLPs. Five different versions of Gag-Spike VLPs (Gag-S-VLPs) consisting of Gag-S alone or combined with other SARS-CoV-2 components, namely Gag-S-Nucleocapsid (N), Gag-S-Matrix (M), Gag-S-Envelope (E), Gag-S-MEN, along with Gag alone were produced and processed by clarification, nuclease treatment, concentration by tangential flow filtration (TFF) and diafiltration. A pilot mouse study was performed to evaluate the immunogenicity of the Gag-S-VLPs through the measurement of the humoral and/or cellular responses against all the mentioned SARS-CoV-2 components. Antibody response to Spike was observed in all variants. The highest number of Spike-specific IFN-γ + T cells was detected with Gag-S-VLPs. No induction of antigen-specific cellular responses to M, N or E proteins were detected with any of the Gag-S, M, E/or N VLPs tested. Therefore, the Gag-S-VLP, by reason of consistently eliciting strong antigen-specific cellular and antibody responses, was selected for further evaluation. The purification process was improved by replacing the conventional centrifugation by serial microfiltration in the clarification step, followed by Spike-affinity chromatography to get concentrated VLPs with higher purity. Three different doses of Gag-S-VLP in conjunction with two adjuvants (Quil-A or AddaVax) were used to assess the dose-dependent antigen-specific cellular and antibody responses in mice. The Gag-S-VLP adjuvanted with Quil-A resulted in a stronger Spike-specific cellular response compared to that adjuvanted with AddaVax. A strong spike neutralisation activity was observed for all doses, independent of the adjuvant combination.
Collapse
Affiliation(s)
| | - Gerard Agbayani
- Human Health Therapeutics, National Research Council Canada Ottawa, ON, Canada
| | - Sabahudin Hrapovic
- Aquatic and Crop Resource Development Research Centre, National Research Council Canada, Montréal, Canada
| | - Nasha Nassoury
- Human Health Therapeutics, National Research Council Canada, Montreal, QC, Canada
| | - Nathalie Coulombe
- Human Health Therapeutics, National Research Council Canada, Montreal, QC, Canada
| | - Renu Dudani
- Human Health Therapeutics, National Research Council Canada Ottawa, ON, Canada
| | - Blair A Harrison
- Human Health Therapeutics, National Research Council Canada Ottawa, ON, Canada
| | - Bassel Akache
- Human Health Therapeutics, National Research Council Canada Ottawa, ON, Canada
| | - Rénald Gilbert
- Human Health Therapeutics, National Research Council Canada, Montreal, QC, Canada; Department of Bioengineering, McGill University, Montreal, QC, Canada.
| | | |
Collapse
|
33
|
Taghizadeh MS, Niazi A, Afsharifar A. Virus-like particles (VLPs): A promising platform for combating against Newcastle disease virus. Vaccine X 2024; 16:100440. [PMID: 38283623 PMCID: PMC10811427 DOI: 10.1016/j.jvacx.2024.100440] [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: 10/12/2023] [Revised: 12/11/2023] [Accepted: 01/13/2024] [Indexed: 01/30/2024] Open
Abstract
The global poultry industry plays a pivotal role in providing eggs and meat for human consumption. However, outbreaks of viral disease, especially Newcastle virus disease (NDV), within poultry farms have detrimental effects on various zootechnical parameters, such as body weight gain, feed intake, feed conversion ratio, as well as the quality of egg and meat production. Cases of vaccine failure have been reported in regions where highly pathogenic strains of NDV are prevalent. To tackle this challenge, virus-like particles (VLPs) have emerged as a potential solution. VLPs closely resemble natural viruses, offering biocompatibility and immune-stimulating properties that make them highly promising for therapeutic applications against NDV. Hence, this review emphasizes the significance of NDV and the need for effective treatments. The manuscript will contain several key aspects, starting with an exploration of the structure and properties of NDV. Subsequently, the paper will delve into the characteristics and benefits of VLPs compared to conventional drug delivery systems. A comprehensive analysis of VLPs as potential vaccine candidates targeting NDV will be presented, along with a discussion on strategies for loading cargo into these NDV-targeting VLPs. The review will also examine various expression systems utilized in the production of NDV-targeting VLPs. Additionally, the manuscript will address future prospects and challenges in the field, concluding with recommendations for further research.
Collapse
Affiliation(s)
| | - Ali Niazi
- Institute of Biotechnology, Shiraz University, Shiraz, Iran
| | - Alireza Afsharifar
- Plant Virus Research Center, School of Agriculture, Shiraz University, Shiraz, Iran
| |
Collapse
|
34
|
Bárcena J, Zamora-Ceballos M, Blanco E. Design of Novel Vaccines Based on Virus-Like Particles. Subcell Biochem 2024; 105:785-821. [PMID: 39738963 DOI: 10.1007/978-3-031-65187-8_21] [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: 01/02/2025]
Abstract
Virus-like particles (VLPs) are formed by viral proteins that, when overexpressed, spontaneously self-assemble into particles that structurally are similar to infectious virus or subviral particles (e.g. the viral capsid). VLPs are appealing as vaccine candidates because their inherent properties (i.e. virus-sized, multimeric antigens, highly organised and repetitive structure, not infectious) are suitable for the induction of safe and efficient humoral and cellular immune responses. VLP-based vaccines have already been licensed for human and veterinary use, and many more vaccine candidates are currently in late stages of evaluation. Moreover, the development of VLPs as platforms for foreign antigen display has further broadened their potential applicability both as prophylactic and therapeutic vaccines. This chapter provides an overview on the design and use of VLPs for the development of new-generation vaccines.
Collapse
Affiliation(s)
- Juan Bárcena
- Centro de Investigación en Sanidad Animal (CISA-INIA/CSIC), Madrid, Spain.
| | | | - Esther Blanco
- Centro de Investigación en Sanidad Animal (CISA-INIA/CSIC), Madrid, Spain
| |
Collapse
|
35
|
Liu S, Hu M, Liu X, Liu X, Chen T, Zhu Y, Liang T, Xiao S, Li P, Ma X. Nanoparticles and Antiviral Vaccines. Vaccines (Basel) 2023; 12:30. [PMID: 38250843 PMCID: PMC10819235 DOI: 10.3390/vaccines12010030] [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: 11/22/2023] [Revised: 12/19/2023] [Accepted: 12/22/2023] [Indexed: 01/23/2024] Open
Abstract
Viruses have threatened human lives for decades, causing both chronic and acute infections accompanied by mild to severe symptoms. During the long journey of confrontation, humans have developed intricate immune systems to combat viral infections. In parallel, vaccines are invented and administrated to induce strong protective immunity while generating few adverse effects. With advancements in biochemistry and biophysics, different kinds of vaccines in versatile forms have been utilized to prevent virus infections, although the safety and effectiveness of these vaccines are diverse from each other. In this review, we first listed and described major pathogenic viruses and their pandemics that emerged in the past two centuries. Furthermore, we summarized the distinctive characteristics of different antiviral vaccines and adjuvants. Subsequently, in the main body, we reviewed recent advances of nanoparticles in the development of next-generation vaccines against influenza viruses, coronaviruses, HIV, hepatitis viruses, and many others. Specifically, we described applications of self-assembling protein polymers, virus-like particles, nano-carriers, and nano-adjuvants in antiviral vaccines. We also discussed the therapeutic potential of nanoparticles in developing safe and effective mucosal vaccines. Nanoparticle techniques could be promising platforms for developing broad-spectrum, preventive, or therapeutic antiviral vaccines.
Collapse
Affiliation(s)
- Sen Liu
- Guangzhou National Laboratory, Guangzhou International Bio-Island, Guangzhou 510005, China; (S.L.); (M.H.); (X.L.); (X.L.); (T.C.); (Y.Z.); (T.L.); (S.X.); (P.L.)
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Meilin Hu
- Guangzhou National Laboratory, Guangzhou International Bio-Island, Guangzhou 510005, China; (S.L.); (M.H.); (X.L.); (X.L.); (T.C.); (Y.Z.); (T.L.); (S.X.); (P.L.)
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou 511400, China
| | - Xiaoqing Liu
- Guangzhou National Laboratory, Guangzhou International Bio-Island, Guangzhou 510005, China; (S.L.); (M.H.); (X.L.); (X.L.); (T.C.); (Y.Z.); (T.L.); (S.X.); (P.L.)
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - Xingyu Liu
- Guangzhou National Laboratory, Guangzhou International Bio-Island, Guangzhou 510005, China; (S.L.); (M.H.); (X.L.); (X.L.); (T.C.); (Y.Z.); (T.L.); (S.X.); (P.L.)
| | - Tao Chen
- Guangzhou National Laboratory, Guangzhou International Bio-Island, Guangzhou 510005, China; (S.L.); (M.H.); (X.L.); (X.L.); (T.C.); (Y.Z.); (T.L.); (S.X.); (P.L.)
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou 511400, China
| | - Yiqiang Zhu
- Guangzhou National Laboratory, Guangzhou International Bio-Island, Guangzhou 510005, China; (S.L.); (M.H.); (X.L.); (X.L.); (T.C.); (Y.Z.); (T.L.); (S.X.); (P.L.)
| | - Taizhen Liang
- Guangzhou National Laboratory, Guangzhou International Bio-Island, Guangzhou 510005, China; (S.L.); (M.H.); (X.L.); (X.L.); (T.C.); (Y.Z.); (T.L.); (S.X.); (P.L.)
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou 511400, China
| | - Shiqi Xiao
- Guangzhou National Laboratory, Guangzhou International Bio-Island, Guangzhou 510005, China; (S.L.); (M.H.); (X.L.); (X.L.); (T.C.); (Y.Z.); (T.L.); (S.X.); (P.L.)
| | - Peiwen Li
- Guangzhou National Laboratory, Guangzhou International Bio-Island, Guangzhou 510005, China; (S.L.); (M.H.); (X.L.); (X.L.); (T.C.); (Y.Z.); (T.L.); (S.X.); (P.L.)
| | - Xiancai Ma
- Guangzhou National Laboratory, Guangzhou International Bio-Island, Guangzhou 510005, China; (S.L.); (M.H.); (X.L.); (X.L.); (T.C.); (Y.Z.); (T.L.); (S.X.); (P.L.)
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou 511400, China
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| |
Collapse
|
36
|
Luo H, Ma Y, Ren Y, Li Z, Sheng Y, Wang Y, Su Z, Bi J, Zhang S. Study of self-assembling properties of HBc-VLP derivatives aided by molecular dynamic simulations from a thermodynamic perspective. J Biomol Struct Dyn 2023; 42:12822-12835. [PMID: 37908124 DOI: 10.1080/07391102.2023.2273438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 10/14/2023] [Indexed: 11/02/2023]
Abstract
Self-assembling protein nanoparticles showed promise for vaccine design due to efficient antigen presentations and safety. However, the unpredictable formations of epitopes-fused protein assemblies remain challenging in the upstream design. This study suggests employing molecular dynamic (MD) simulations to investigate the assembly properties of Hepatitis B core protein (HBc) from thermodynamic perspectives. Eight HBc derivatives were expressed in E. coli, with their self-assembly properties characterised by high-performance liquid chromatography and transmission electron microscopy. MD simulations on the dimers, based on AlphaFold-predicted 3D structures, analysed the derivative at the atomic level. Results revealed that HBc derivatives can form dissociative polymers or large multi-subunit structures due to assembly failures. The instability of the dimer in aqueous solvents or inappropriate intradimer distances could cause major assembly failures. Polar solvation energies played a vital role too in forming assemble-incompetent dimers. Importantly, our study demonstrated that MD simulations on dimers can provide preliminary predictions on the assembly properties of HBc derivatives, thus aiding vaccine design by lowering the risk of self-assembling failures in engineered proteins.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Hong Luo
- School of Chemical Engineering, Faculty of Sciences, Engineering and Technology, University of Adelaide, Adelaide, Australia
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, PR China
- Institute of Pharmaceutical and Food Engineering, Shanxi University of Chinese Medicine, Jinzhong, PR China
| | - Yanyan Ma
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, PR China
| | - Ying Ren
- State Key Laboratory of Multiphase Complex Systems, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, PR China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, PR China
| | - Zhengjun Li
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, PR China
| | - Yanan Sheng
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, PR China
| | - Yingli Wang
- Institute of Pharmaceutical and Food Engineering, Shanxi University of Chinese Medicine, Jinzhong, PR China
| | - Zhiguo Su
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, PR China
| | - Jingxiu Bi
- School of Chemical Engineering, Faculty of Sciences, Engineering and Technology, University of Adelaide, Adelaide, Australia
| | - Songping Zhang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, PR China
| |
Collapse
|
37
|
Boix-Besora A, Gòdia F, Cervera L. Gag Virus-like Particles Functionalized with SARS-CoV-2 Variants: Generation, Characterization and Recognition by COVID-19 Convalescent Patients' Sera. Vaccines (Basel) 2023; 11:1641. [PMID: 38005972 PMCID: PMC10675557 DOI: 10.3390/vaccines11111641] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 10/16/2023] [Accepted: 10/21/2023] [Indexed: 11/26/2023] Open
Abstract
The robustness, safety, versatility, and high immunogenicity of virus-like particles (VLPs) make them a promising approach for the generation of vaccines against a broad range of pathogens. VLPs are recombinant macromolecular structures that closely mimic the native conformation of viruses without carrying viral genetic material. Particularly, HIV-1 Gag-based VLPs are a suitable platform for the presentation of the SARS-CoV-2 Spike (S) protein on their surface. In this context, this work studies the effect of different rationally engineered mutations of the S protein to improve some of its characteristics. The studied variants harbored mutations such as proline substitutions for S stabilization, D614G from the early dominant pandemic form, the elimination of the S1/S2 furin cleavage site to improve S homogeneity, the suppression of a retention motif to favor its membrane localization, and cysteine substitutions to increase its immunogenicity and avoid potential undesired antibody-dependent enhancement (ADE) effects. The influence of the mutations on VLP expression was studied, as well as their immunogenic potential, by testing the recognition of the generated VLP variants by COVID-19 convalescent patients' sera. The results of this work are conceived to give insights on the selection of S protein candidates for their use as immunogens and to showcase the potential of VLPs as carriers for antigen presentation.
Collapse
Affiliation(s)
- Arnau Boix-Besora
- Grup d’Enginyeria de Bioprocessos i Biocatàlisi Aplicada ENG4BIO, Escola d’Enginyeria, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, 08193 Barcelona, Spain
| | | | | |
Collapse
|
38
|
Ramirez A, Felgner J, Jain A, Jan S, Albin TJ, Badten AJ, Gregory AE, Nakajima R, Jasinskas A, Felgner PL, Burkhardt AM, Davies DH, Wang SW. Engineering Protein Nanoparticles Functionalized with an Immunodominant Coxiella burnetii Antigen to Generate a Q Fever Vaccine. Bioconjug Chem 2023; 34:1653-1666. [PMID: 37682243 PMCID: PMC10515490 DOI: 10.1021/acs.bioconjchem.3c00317] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 08/25/2023] [Indexed: 09/09/2023]
Abstract
Coxiella burnetii is the causative agent of Q fever, for which there is yet to be an FDA-approved vaccine. This bacterial pathogen has both extra- and intracellular stages in its life cycle, and therefore both a cell-mediated (i.e., T lymphocyte) and humoral (i.e., antibody) immune response are necessary for effective eradication of this pathogen. However, most proposed vaccines elicit strong responses to only one mechanism of adaptive immunity, and some can either cause reactogenicity or lack sufficient immunogenicity. In this work, we aim to apply a nanoparticle-based platform toward producing both antibody and T cell immune responses against C. burnetii. We investigated three approaches for conjugation of the immunodominant outer membrane protein antigen (CBU1910) to the E2 nanoparticle to obtain a consistent antigen orientation: direct genetic fusion, high affinity tris-NTA-Ni conjugation to polyhistidine-tagged CBU1910, and the SpyTag/SpyCatcher (ST/SC) system. Overall, we found that the ST/SC approach yielded nanoparticles loaded with the highest number of antigens while maintaining stability, enabling formulations that could simultaneously co-deliver the protein antigen (CBU1910) and adjuvant (CpG1826) on one nanoparticle (CBU1910-CpG-E2). Using protein microarray analyses, we found that after immunization, antigen-bound nanoparticle formulations elicited significantly higher antigen-specific IgG responses than soluble CBU1910 alone and produced more balanced IgG1/IgG2c ratios. Although T cell recall assays from these protein antigen formulations did not show significant increases in antigen-specific IFN-γ production compared to soluble CBU1910 alone, nanoparticles conjugated with a CD4 peptide epitope from CBU1910 generated elevated T cell responses in mice to both the CBU1910 peptide epitope and whole CBU1910 protein. These investigations highlight the feasibility of conjugating antigens to nanoparticles for tuning and improving both humoral- and cell-mediated adaptive immunity against C. burnetii.
Collapse
Affiliation(s)
- Aaron Ramirez
- Department
of Chemical and Biomolecular Engineering, Vaccine Research and Development
Center, Department of Physiology and Biophysics, Department of Chemistry, Department of Biomedical
Engineering, Chao Family Comprehensive Cancer Center, and Institute for Immunology, University of California, Irvine, California 92697, United States
| | - Jiin Felgner
- Department
of Chemical and Biomolecular Engineering, Vaccine Research and Development
Center, Department of Physiology and Biophysics, Department of Chemistry, Department of Biomedical
Engineering, Chao Family Comprehensive Cancer Center, and Institute for Immunology, University of California, Irvine, California 92697, United States
| | - Aarti Jain
- Department
of Chemical and Biomolecular Engineering, Vaccine Research and Development
Center, Department of Physiology and Biophysics, Department of Chemistry, Department of Biomedical
Engineering, Chao Family Comprehensive Cancer Center, and Institute for Immunology, University of California, Irvine, California 92697, United States
| | - Sharon Jan
- Department
of Chemical and Biomolecular Engineering, Vaccine Research and Development
Center, Department of Physiology and Biophysics, Department of Chemistry, Department of Biomedical
Engineering, Chao Family Comprehensive Cancer Center, and Institute for Immunology, University of California, Irvine, California 92697, United States
| | - Tyler J. Albin
- Department
of Chemical and Biomolecular Engineering, Vaccine Research and Development
Center, Department of Physiology and Biophysics, Department of Chemistry, Department of Biomedical
Engineering, Chao Family Comprehensive Cancer Center, and Institute for Immunology, University of California, Irvine, California 92697, United States
| | - Alexander J. Badten
- Department
of Chemical and Biomolecular Engineering, Vaccine Research and Development
Center, Department of Physiology and Biophysics, Department of Chemistry, Department of Biomedical
Engineering, Chao Family Comprehensive Cancer Center, and Institute for Immunology, University of California, Irvine, California 92697, United States
| | - Anthony E. Gregory
- Department
of Chemical and Biomolecular Engineering, Vaccine Research and Development
Center, Department of Physiology and Biophysics, Department of Chemistry, Department of Biomedical
Engineering, Chao Family Comprehensive Cancer Center, and Institute for Immunology, University of California, Irvine, California 92697, United States
| | - Rie Nakajima
- Department
of Chemical and Biomolecular Engineering, Vaccine Research and Development
Center, Department of Physiology and Biophysics, Department of Chemistry, Department of Biomedical
Engineering, Chao Family Comprehensive Cancer Center, and Institute for Immunology, University of California, Irvine, California 92697, United States
| | - Algimantas Jasinskas
- Department
of Chemical and Biomolecular Engineering, Vaccine Research and Development
Center, Department of Physiology and Biophysics, Department of Chemistry, Department of Biomedical
Engineering, Chao Family Comprehensive Cancer Center, and Institute for Immunology, University of California, Irvine, California 92697, United States
| | - Philip L. Felgner
- Department
of Chemical and Biomolecular Engineering, Vaccine Research and Development
Center, Department of Physiology and Biophysics, Department of Chemistry, Department of Biomedical
Engineering, Chao Family Comprehensive Cancer Center, and Institute for Immunology, University of California, Irvine, California 92697, United States
| | - Amanda M. Burkhardt
- Department
of Clinical Pharmacy, School of Pharmacy, University of Southern California, Los Angeles, California 90089, United States
| | - D. Huw Davies
- Department
of Chemical and Biomolecular Engineering, Vaccine Research and Development
Center, Department of Physiology and Biophysics, Department of Chemistry, Department of Biomedical
Engineering, Chao Family Comprehensive Cancer Center, and Institute for Immunology, University of California, Irvine, California 92697, United States
| | - Szu-Wen Wang
- Department
of Chemical and Biomolecular Engineering, Vaccine Research and Development
Center, Department of Physiology and Biophysics, Department of Chemistry, Department of Biomedical
Engineering, Chao Family Comprehensive Cancer Center, and Institute for Immunology, University of California, Irvine, California 92697, United States
| |
Collapse
|
39
|
Xue Q, Swevers L, Taning CNT. Plant and insect virus-like particles: emerging nanoparticles for agricultural pest management. PEST MANAGEMENT SCIENCE 2023; 79:2975-2991. [PMID: 37103223 DOI: 10.1002/ps.7514] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 04/20/2023] [Accepted: 04/27/2023] [Indexed: 06/05/2023]
Abstract
Virus-like particles (VLPs) represent a biodegradable, biocompatible nanomaterial made from viral coat proteins that can improve the delivery of antigens, drugs, nucleic acids, and other substances, with most applications in human and veterinary medicine. Regarding agricultural viruses, many insect and plant virus coat proteins have been shown to assemble into VLPs accurately. In addition, some plant virus-based VLPs have been used in medical studies. However, to our knowledge, the potential application of plant/insect virus-based VLPs in agriculture remains largely underexplored. This review focuses on why and how to engineer coat proteins of plant/insect viruses as functionalized VLPs, and how to exploit VLPs in agricultural pest control. The first part of the review describes four different engineering strategies for loading cargo at the inner or the outer surface of VLPs depending on the type of cargo and purpose. Second, the literature on plant and insect viruses the coat proteins of which have been confirmed to self-assemble into VLPs is reviewed. These VLPs are good candidates for developing VLP-based agricultural pest control strategies. Lastly, the concepts of plant/insect virus-based VLPs for delivering insecticidal and antiviral components (e.g., double-stranded RNA, peptides, and chemicals) are discussed, which provides future prospects of VLP application in agricultural pest control. In addition, some concerns are raised about VLP production on a large scale and the short-term resistance of hosts to VLP uptake. Overall, this review is expected to stimulate interest and research exploring plant/insect virus-based VLP applications in agricultural pest management. © 2023 Society of Chemical Industry.
Collapse
Affiliation(s)
- Qi Xue
- Department of Plants and Crops, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Luc Swevers
- Insect Molecular Genetics and Biotechnology, Institute of Biosciences and Applications, National Centre for Scientific Research "Demokritos", Athens, Greece
| | - Clauvis Nji Tizi Taning
- Department of Plants and Crops, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| |
Collapse
|
40
|
Mo J, Spackman E, Swayne DE. Prediction of highly pathogenic avian influenza vaccine efficacy in chickens by comparison of in vitro and in vivo data: A meta-analysis and systematic review. Vaccine 2023; 41:5507-5517. [PMID: 37537093 DOI: 10.1016/j.vaccine.2023.07.076] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 07/29/2023] [Accepted: 07/30/2023] [Indexed: 08/05/2023]
Abstract
Vaccines for avian influenza (AI) can protect poultry against disease, mortality, and virus transmission. Numerous factors, including: vaccine platform, immunogenicity, and relatedness to the field strain, are known to be important to achieving optimal AI vaccine efficacy. To better understand how these factors contribute to vaccine protection, a systematic meta-analysis was conducted to evaluate efficacy data for vaccines in chickens challenged with highly pathogenic (HP) AI. Data from a total of 120 individual trials from 25 publications were selected and evaluated. Two vaccine criteria were evaluated for their effects on two metrics of protection. The vaccine criteria were: 1) the relatedness of the vaccine antigen and challenge strain in the hemagglutinin 1 domain (HA1) protein sequence; 2) vaccine-induced antibody titers to the challenge virus (VIAC). The metrics of protection were: A) survival of vaccinated chickens vs unvaccinated controls; and B) reduction in oral virus-shedding by vaccinated vs unvaccinated controls 2-4 days post challenge. Three vaccine platforms were evaluated: oil-adjuvanted inactivated whole AI virus, recombinant herpes virus of turkeys (rHVT) vectored, and a non-replicating alpha-virus vectored RNA particle (RP) vaccine. Higher VIAC correlated with greater reduction of virus-shed and vaccine efficacy by all vaccine platforms. Both higher HA1 relatedness and higher VIAC using challenge virus as antigen correlated with better survival by inactivated vaccines and rHVT-vectored vaccines. However, rHVT-vectored and RP based vaccines were more tolerant of variation in the HA1; the relatedness of the HA1 of the vaccine and challenge virus did not significantly correlate with survival with rHVT-vectored vaccines. Protection was achieved with the lowest aa similarity for which there was data, 90-93 % for rHVT vaccines and 88 % for the RP vaccine.
Collapse
Affiliation(s)
- Jongseo Mo
- Southeast Poultry Research Laboratory, U.S. National Poultry Research Center, U.S. Department of Agriculture-Agricultural Research Service, 934 College Station Road, Athens, GA 30605, USA
| | - Erica Spackman
- Southeast Poultry Research Laboratory, U.S. National Poultry Research Center, U.S. Department of Agriculture-Agricultural Research Service, 934 College Station Road, Athens, GA 30605, USA.
| | - David E Swayne
- Southeast Poultry Research Laboratory, U.S. National Poultry Research Center, U.S. Department of Agriculture-Agricultural Research Service, 934 College Station Road, Athens, GA 30605, USA
| |
Collapse
|
41
|
Alpuche-Lazcano SP, Stuible M, Akache B, Tran A, Kelly J, Hrapovic S, Robotham A, Haqqani A, Star A, Renner TM, Blouin J, Maltais JS, Cass B, Cui K, Cho JY, Wang X, Zoubchenok D, Dudani R, Duque D, McCluskie MJ, Durocher Y. Preclinical evaluation of manufacturable SARS-CoV-2 spike virus-like particles produced in Chinese Hamster Ovary cells. COMMUNICATIONS MEDICINE 2023; 3:116. [PMID: 37612423 PMCID: PMC10447459 DOI: 10.1038/s43856-023-00340-7] [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: 03/16/2023] [Accepted: 07/25/2023] [Indexed: 08/25/2023] Open
Abstract
BACKGROUND As the COVID-19 pandemic continues to evolve, novel vaccines need to be developed that are readily manufacturable and provide clinical efficacy against emerging SARS-CoV-2 variants. Virus-like particles (VLPs) presenting the spike antigen at their surface offer remarkable benefits over other vaccine antigen formats; however, current SARS-CoV-2 VLP vaccines candidates in clinical development suffer from challenges including low volumetric productivity, poor spike antigen density, expression platform-driven divergent protein glycosylation and complex upstream/downstream processing requirements. Despite their extensive use for therapeutic protein manufacturing and proven ability to produce enveloped VLPs, Chinese Hamster Ovary (CHO) cells are rarely used for the commercial production of VLP-based vaccines. METHODS Using CHO cells, we aimed to produce VLPs displaying the full-length SARS-CoV-2 spike. Affinity chromatography was used to capture VLPs released in the culture medium from engineered CHO cells expressing spike. The structure, protein content, and glycosylation of spikes in VLPs were characterized by several biochemical and biophysical methods. In vivo, the generation of neutralizing antibodies and protection against SARS-CoV-2 infection was tested in mouse and hamster models. RESULTS We demonstrate that spike overexpression in CHO cells is sufficient by itself to generate high VLP titers. These VLPs are evocative of the native virus but with at least three-fold higher spike density. In vivo, purified VLPs elicit strong humoral and cellular immunity at nanogram dose levels which grant protection against SARS-CoV-2 infection. CONCLUSIONS Our results show that CHO cells are amenable to efficient manufacturing of high titers of a potently immunogenic spike protein-based VLP vaccine antigen.
Collapse
Affiliation(s)
- Sergio P Alpuche-Lazcano
- Human Health Therapeutics Research Centre, National Research Council Canada, 6100 Royalmount Avenue, Montreal, QC, H4P 2R2, Canada
| | - Matthew Stuible
- Human Health Therapeutics Research Centre, National Research Council Canada, 6100 Royalmount Avenue, Montreal, QC, H4P 2R2, Canada
| | - Bassel Akache
- Human Health Therapeutics Research Centre, National Research Council Canada, 1200 Montreal Road, Ottawa, ON, K1A 0R6, Canada
| | - Anh Tran
- Human Health Therapeutics Research Centre, National Research Council Canada, 1200 Montreal Road, Ottawa, ON, K1A 0R6, Canada
| | - John Kelly
- Human Health Therapeutics Research Centre, National Research Council Canada, 100 Sussex Dr, Ottawa, ON, K1A 0R6, Canada
| | - Sabahudin Hrapovic
- Aquatic and Crop Resources Development Research Centre, National Research Council Canada, 6100 Royalmount Avenue, Montreal, QC, H4P 2R2, Canada
| | - Anna Robotham
- Human Health Therapeutics Research Centre, National Research Council Canada, 100 Sussex Dr, Ottawa, ON, K1A 0R6, Canada
| | - Arsalan Haqqani
- Human Health Therapeutics Research Centre, National Research Council Canada, 100 Sussex Dr, Ottawa, ON, K1A 0R6, Canada
| | - Alexandra Star
- Human Health Therapeutics Research Centre, National Research Council Canada, 100 Sussex Dr, Ottawa, ON, K1A 0R6, Canada
| | - Tyler M Renner
- Human Health Therapeutics Research Centre, National Research Council Canada, 1200 Montreal Road, Ottawa, ON, K1A 0R6, Canada
| | - Julie Blouin
- Human Health Therapeutics Research Centre, National Research Council Canada, 6100 Royalmount Avenue, Montreal, QC, H4P 2R2, Canada
| | - Jean-Sébastien Maltais
- Human Health Therapeutics Research Centre, National Research Council Canada, 6100 Royalmount Avenue, Montreal, QC, H4P 2R2, Canada
| | - Brian Cass
- Human Health Therapeutics Research Centre, National Research Council Canada, 6100 Royalmount Avenue, Montreal, QC, H4P 2R2, Canada
| | - Kai Cui
- Nanotechnology Research Centre, National Research Council Canada, 11421 Saskatchewan Drive, Edmonton, AB, T6G 2M9, Canada
| | - Jae-Young Cho
- Nanotechnology Research Centre, National Research Council Canada, 11421 Saskatchewan Drive, Edmonton, AB, T6G 2M9, Canada
| | - Xinyu Wang
- Nanotechnology Research Centre, National Research Council Canada, 11421 Saskatchewan Drive, Edmonton, AB, T6G 2M9, Canada
| | - Daria Zoubchenok
- Human Health Therapeutics Research Centre, National Research Council Canada, 6100 Royalmount Avenue, Montreal, QC, H4P 2R2, Canada
| | - Renu Dudani
- Human Health Therapeutics Research Centre, National Research Council Canada, 1200 Montreal Road, Ottawa, ON, K1A 0R6, Canada
| | - Diana Duque
- Human Health Therapeutics Research Centre, National Research Council Canada, 1200 Montreal Road, Ottawa, ON, K1A 0R6, Canada
| | - Michael J McCluskie
- Human Health Therapeutics Research Centre, National Research Council Canada, 1200 Montreal Road, Ottawa, ON, K1A 0R6, Canada
| | - Yves Durocher
- Human Health Therapeutics Research Centre, National Research Council Canada, 6100 Royalmount Avenue, Montreal, QC, H4P 2R2, Canada.
| |
Collapse
|
42
|
Ruzzi F, Semprini MS, Scalambra L, Palladini A, Angelicola S, Cappello C, Pittino OM, Nanni P, Lollini PL. Virus-like Particle (VLP) Vaccines for Cancer Immunotherapy. Int J Mol Sci 2023; 24:12963. [PMID: 37629147 PMCID: PMC10454695 DOI: 10.3390/ijms241612963] [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: 07/31/2023] [Revised: 08/14/2023] [Accepted: 08/17/2023] [Indexed: 08/27/2023] Open
Abstract
Cancer vaccines are increasingly being studied as a possible strategy to prevent and treat cancers. While several prophylactic vaccines for virus-caused cancers are approved and efficiently used worldwide, the development of therapeutic cancer vaccines needs to be further implemented. Virus-like particles (VLPs) are self-assembled protein structures that mimic native viruses or bacteriophages but lack the replicative material. VLP platforms are designed to display single or multiple antigens with a high-density pattern, which can trigger both cellular and humoral responses. The aim of this review is to provide a comprehensive overview of preventive VLP-based vaccines currently approved worldwide against HBV and HPV infections or under evaluation to prevent virus-caused cancers. Furthermore, preclinical and early clinical data on prophylactic and therapeutic VLP-based cancer vaccines were summarized with a focus on HER-2-positive breast cancer.
Collapse
Affiliation(s)
- Francesca Ruzzi
- Department of Medical and Surgical Sciences (DIMEC) and Alma Mater Institute on Healthy Planet, University of Bologna, 40126 Bologna, Italy; (F.R.); (M.S.S.); (L.S.); (S.A.); (C.C.); (O.M.P.); (P.N.)
| | - Maria Sofia Semprini
- Department of Medical and Surgical Sciences (DIMEC) and Alma Mater Institute on Healthy Planet, University of Bologna, 40126 Bologna, Italy; (F.R.); (M.S.S.); (L.S.); (S.A.); (C.C.); (O.M.P.); (P.N.)
| | - Laura Scalambra
- Department of Medical and Surgical Sciences (DIMEC) and Alma Mater Institute on Healthy Planet, University of Bologna, 40126 Bologna, Italy; (F.R.); (M.S.S.); (L.S.); (S.A.); (C.C.); (O.M.P.); (P.N.)
| | - Arianna Palladini
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy;
| | - Stefania Angelicola
- Department of Medical and Surgical Sciences (DIMEC) and Alma Mater Institute on Healthy Planet, University of Bologna, 40126 Bologna, Italy; (F.R.); (M.S.S.); (L.S.); (S.A.); (C.C.); (O.M.P.); (P.N.)
| | - Chiara Cappello
- Department of Medical and Surgical Sciences (DIMEC) and Alma Mater Institute on Healthy Planet, University of Bologna, 40126 Bologna, Italy; (F.R.); (M.S.S.); (L.S.); (S.A.); (C.C.); (O.M.P.); (P.N.)
| | - Olga Maria Pittino
- Department of Medical and Surgical Sciences (DIMEC) and Alma Mater Institute on Healthy Planet, University of Bologna, 40126 Bologna, Italy; (F.R.); (M.S.S.); (L.S.); (S.A.); (C.C.); (O.M.P.); (P.N.)
| | - Patrizia Nanni
- Department of Medical and Surgical Sciences (DIMEC) and Alma Mater Institute on Healthy Planet, University of Bologna, 40126 Bologna, Italy; (F.R.); (M.S.S.); (L.S.); (S.A.); (C.C.); (O.M.P.); (P.N.)
| | - Pier-Luigi Lollini
- Department of Medical and Surgical Sciences (DIMEC) and Alma Mater Institute on Healthy Planet, University of Bologna, 40126 Bologna, Italy; (F.R.); (M.S.S.); (L.S.); (S.A.); (C.C.); (O.M.P.); (P.N.)
| |
Collapse
|
43
|
Ren H, Jia W, Xie Y, Yu M, Chen Y. Adjuvant physiochemistry and advanced nanotechnology for vaccine development. Chem Soc Rev 2023; 52:5172-5254. [PMID: 37462107 DOI: 10.1039/d2cs00848c] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/01/2023]
Abstract
Vaccines comprising innovative adjuvants are rapidly reaching advanced translational stages, such as the authorized nanotechnology adjuvants in mRNA vaccines against COVID-19 worldwide, offering new strategies to effectively combat diseases threatening human health. Adjuvants are vital ingredients in vaccines, which can augment the degree, extensiveness, and longevity of antigen specific immune response. The advances in the modulation of physicochemical properties of nanoplatforms elevate the capability of adjuvants in initiating the innate immune system and adaptive immunity, offering immense potential for developing vaccines against hard-to-target infectious diseases and cancer. In this review, we provide an essential introduction of the basic principles of prophylactic and therapeutic vaccination, key roles of adjuvants in augmenting and shaping immunity to achieve desired outcomes and effectiveness, and the physiochemical properties and action mechanisms of clinically approved adjuvants for humans. We particularly focus on the preclinical and clinical progress of highly immunogenic emerging nanotechnology adjuvants formulated in vaccines for cancer treatment or infectious disease prevention. We deliberate on how the immune system can sense and respond to the physicochemical cues (e.g., chirality, deformability, solubility, topology, and chemical structures) of nanotechnology adjuvants incorporated in the vaccines. Finally, we propose possible strategies to accelerate the clinical implementation of nanotechnology adjuvanted vaccines, such as in-depth elucidation of nano-immuno interactions, antigen identification and optimization by the deployment of high-dimensional multiomics analysis approaches, encouraging close collaborations among scientists from different scientific disciplines and aggressive exploration of novel nanotechnologies.
Collapse
Affiliation(s)
- Hongze Ren
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
- School of Medicine, Shanghai University, Shanghai, 200444, P. R. China
| | - Wencong Jia
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
- School of Medicine, Shanghai University, Shanghai, 200444, P. R. China
| | - Yujie Xie
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
- School of Medicine, Shanghai University, Shanghai, 200444, P. R. China
| | - Meihua Yu
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
| | - Yu Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
- School of Medicine, Shanghai University, Shanghai, 200444, P. R. China
| |
Collapse
|
44
|
Sarrami Z, Sedghi M, Mohammadi I, Bedford M, Miranzadeh H, Ghasemi R. Effects of bacteriophage on Salmonella Enteritidis infection in broilers. Sci Rep 2023; 13:12198. [PMID: 37500690 PMCID: PMC10374914 DOI: 10.1038/s41598-023-38791-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 07/14/2023] [Indexed: 07/29/2023] Open
Abstract
Bacteriophages (BP) are viruses that can infect bacteria. The present study evaluated the effect of BP on Salmonella infected broilers. A number of 150 day-old broilers were used in a completely randomized design with five treatments that included: (1) basal diet from day 0 to 28; (2) basal diet + 0.3 g/kg of colistin from day 0 to 28; (3) basal diet from day 1 to 13, and basal diet + 0.4 g/kg of colistin from day 14 to 28; (4) basal diet + 1 g/kg of BP from day 0 to 28; (5) basal diet + 1.5 g/kg of BP from day 0 to 28. On day 13, 15 chickens from each treatment were challenged by Salmonella Enteritidis (SE), while fifteen from each treatment were not; instead, they were kept in the same cage with the challenged chickens (exposed chickens). At 7 and 14 days post-challenge, the number of SE and coliform bacteria in the cecum and liver of colistin and BP-fed birds was lower than the control treatment. In exposed and challenged chickens, the height and surface area of villus were greater in the BP and colistin-supplemented groups. Serum concentrations of aspartate aminotransferase and alanine transaminase were greater, while serum albumin and triglycerides concentrations were lower in the control treatment. The liver of the challenged chickens had more pathological lesions than exposed birds. BP significantly decreased PPARγ gene expression in exposed chickens. In the challenged and exposed chickens, TLR4 gene expression was lower in BP and colistin-treated birds as compared to the control. In conclusion, adding BP to the diet from the day of age prevents the spread of Salmonella.
Collapse
Affiliation(s)
- Zahra Sarrami
- Department of Animal Sciences, College of Agriculture, Isfahan University of Technology, Isfahan, 84156-83111, Iran
| | - Mohammad Sedghi
- Department of Animal Sciences, College of Agriculture, Isfahan University of Technology, Isfahan, 84156-83111, Iran.
| | - Ishmael Mohammadi
- Department of Animal Sciences, College of Agriculture, Isfahan University of Technology, Isfahan, 84156-83111, Iran
| | | | - Hadi Miranzadeh
- Department of Animal Sciences, College of Agriculture, Isfahan University of Technology, Isfahan, 84156-83111, Iran
| | - Razie Ghasemi
- Department of Animal Sciences, College of Agriculture, Isfahan University of Technology, Isfahan, 84156-83111, Iran
| |
Collapse
|
45
|
Nowak I, Madej M, Secemska J, Sarna R, Strzalka-Mrozik B. Virus-Based Biological Systems as Next-Generation Carriers for the Therapy of Central Nervous System Diseases. Pharmaceutics 2023; 15:1931. [PMID: 37514117 PMCID: PMC10384784 DOI: 10.3390/pharmaceutics15071931] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 06/29/2023] [Accepted: 07/10/2023] [Indexed: 07/30/2023] Open
Abstract
Central nervous system (CNS) diseases are currently a major challenge in medicine. One reason is the presence of the blood-brain barrier, which is a significant limitation for currently used medicinal substances that are characterized by a high molecular weight and a short half-life. Despite the application of nanotechnology, there is still the problem of targeting and the occurrence of systemic toxicity. Viral vectors and virus-like particles (VLPs) may provide a promising solution to these challenges. Their small size, biocompatibility, ability to carry medicinal substances, and specific targeting of neural cells make them useful in research when formulating a new generation of biological carriers. Additionally, the possibility of genetic modification has the potential for gene therapy. Among the most promising viral vectors are adeno-associated viruses, adenoviruses, and retroviruses. This is due to their natural tropism to neural cells, as well as the possibility of genetic and surface modification. Moreover, VLPs that are devoid of infectious genetic material in favor of increasing capacity are also leading the way for research on new drug delivery systems. The aim of this study is to review the most recent reports on the use of viral vectors and VLPs in the treatment of selected CNS diseases.
Collapse
Affiliation(s)
- Ilona Nowak
- Department of Molecular Biology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 40-055 Katowice, Poland
| | - Marcel Madej
- Department of Molecular Biology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 40-055 Katowice, Poland
| | - Julia Secemska
- Department of Molecular Biology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 40-055 Katowice, Poland
| | - Robert Sarna
- Department of Molecular Biology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 40-055 Katowice, Poland
| | - Barbara Strzalka-Mrozik
- Department of Molecular Biology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 40-055 Katowice, Poland
| |
Collapse
|
46
|
Filipić B, Pantelić I, Nikolić I, Majhen D, Stojić-Vukanić Z, Savić S, Krajišnik D. Nanoparticle-Based Adjuvants and Delivery Systems for Modern Vaccines. Vaccines (Basel) 2023; 11:1172. [PMID: 37514991 PMCID: PMC10385383 DOI: 10.3390/vaccines11071172] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 05/31/2023] [Accepted: 06/15/2023] [Indexed: 07/30/2023] Open
Abstract
Ever since the development of the first vaccine, vaccination has had the great impact on global health, leading to the decrease in the burden of numerous infectious diseases. However, there is a constant need to improve existing vaccines and develop new vaccination strategies and vaccine platforms that induce a broader immune response compared to traditional vaccines. Modern vaccines tend to rely on certain nanotechnology platforms but are still expected to be readily available and easy for large-scale manufacturing and to induce a durable immune response. In this review, we present an overview of the most promising nanoadjuvants and nanoparticulate delivery systems and discuss their benefits from tehchnological and immunological standpoints as well as their objective drawbacks and possible side effects. The presented nano alums, silica and clay nanoparticles, nanoemulsions, adenoviral-vectored systems, adeno-associated viral vectors, vesicular stomatitis viral vectors, lentiviral vectors, virus-like particles (including bacteriophage-based ones) and virosomes indicate that vaccine developers can now choose different adjuvants and/or delivery systems as per the requirement, specific to combatting different infectious diseases.
Collapse
Affiliation(s)
- Brankica Filipić
- Department of Microbiology and Immunology, University of Belgrade-Faculty of Pharmacy, 11000 Belgrade, Serbia
| | - Ivana Pantelić
- Department of Pharmaceutical Technology and Cosmetology, University of Belgrade-Faculty of Pharmacy, 11000 Belgrade, Serbia
| | - Ines Nikolić
- Department of Pharmaceutical Technology and Cosmetology, University of Belgrade-Faculty of Pharmacy, 11000 Belgrade, Serbia
- Section of Pharmaceutical Sciences, University of Geneva, 1206 Geneva, Switzerland
| | - Dragomira Majhen
- Division of Molecular Biology, Ruđer Bošković Institute, 10000 Zagreb, Croatia
| | - Zorica Stojić-Vukanić
- Department of Microbiology and Immunology, University of Belgrade-Faculty of Pharmacy, 11000 Belgrade, Serbia
| | - Snežana Savić
- Department of Pharmaceutical Technology and Cosmetology, University of Belgrade-Faculty of Pharmacy, 11000 Belgrade, Serbia
| | - Danina Krajišnik
- Department of Pharmaceutical Technology and Cosmetology, University of Belgrade-Faculty of Pharmacy, 11000 Belgrade, Serbia
| |
Collapse
|
47
|
Wang Z, Li Z, Shi W, Zhu D, Hu S, Dinh PUC, Cheng K. A SARS-CoV-2 and influenza double hit vaccine based on RBD-conjugated inactivated influenza A virus. SCIENCE ADVANCES 2023; 9:eabo4100. [PMID: 37352360 PMCID: PMC10289656 DOI: 10.1126/sciadv.abo4100] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 05/19/2023] [Indexed: 06/25/2023]
Abstract
The circulating flu viruses merging with the ongoing COVID-19 pandemic raises a more severe threat that promotes the infectivity of SARS-CoV-2 associated with higher mortality rates. Here, we conjugated recombinant receptor binding domain (RBD) of SARS-CoV-2 spike protein onto inactivated influenza A virus (Flu) to develop a SARS-CoV-2 virus-like particle (VLP) vaccine with two-hit protection. This double-hit vaccine (Flu-RBD) not only induced protective immunities against SARS-CoV-2 but also remained functional as a flu vaccine. The Flu core improved the retention and distribution of Flu-RBD vaccine in the draining lymph nodes, with enhanced immunogenicity. In a hamster model of live SARS-CoV-2 infection, two doses of Flu-RBD efficiently protected animals against viral infection. Furthermore, Flu-RBD VLP elicited a strong neutralization activity against both SARS-CoV-2 Delta pseudovirus and wild-type influenza A H1N1 inactivated virus in mice. Overall, the Flu-RBD VLP vaccine is a promising candidate for combating COVID-19, influenza A, and coinfection.
Collapse
Affiliation(s)
- Zhenzhen Wang
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC 27607, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, and North Carolina State University, Raleigh, NC 27606, USA
| | - Zhenhua Li
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC 27607, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, and North Carolina State University, Raleigh, NC 27606, USA
| | - Weiwei Shi
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC 27607, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, and North Carolina State University, Raleigh, NC 27606, USA
| | - Dashuai Zhu
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC 27607, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, and North Carolina State University, Raleigh, NC 27606, USA
| | - Shiqi Hu
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC 27607, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, and North Carolina State University, Raleigh, NC 27606, USA
| | - Phuong-Uyen C. Dinh
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC 27607, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, and North Carolina State University, Raleigh, NC 27606, USA
| | - Ke Cheng
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC 27607, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, and North Carolina State University, Raleigh, NC 27606, USA
- Division of Pharmacoengineering and Molecular Pharmaceutics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
48
|
Nowill AE, Caruso M, de Campos-Lima PO. T-cell immunity to SARS-CoV-2: what if the known best is not the optimal course for the long run? Adapting to evolving targets. Front Immunol 2023; 14:1133225. [PMID: 37388738 PMCID: PMC10303130 DOI: 10.3389/fimmu.2023.1133225] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 05/11/2023] [Indexed: 07/01/2023] Open
Abstract
Humanity did surprisingly well so far, considering how unprepared it was to respond to the coronavirus disease 2019 (COVID-19) threat. By blending old and ingenious new technology in the context of the accumulated knowledge on other human coronaviruses, several vaccine candidates were produced and tested in clinical trials in record time. Today, five vaccines account for the bulk of the more than 13 billion doses administered worldwide. The ability to elicit biding and neutralizing antibodies most often against the spike protein is a major component of the protection conferred by immunization but alone it is not enough to limit virus transmission. Thus, the surge in numbers of infected individuals by newer variants of concern (VOCs) was not accompanied by a proportional increase in severe disease and death rate. This is likely due to antiviral T-cell responses, whose evasion is more difficult to achieve. The present review helps navigating the very large literature on T cell immunity induced by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and vaccination. We examine the successes and shortcomings of the vaccinal protection in the light of the emergence of VOCs with breakthrough potential. SARS-CoV-2 and human beings will likely coexist for a long while: it will be necessary to update existing vaccines to improve T-cell responses and attain better protection against COVID-19.
Collapse
Affiliation(s)
- Alexandre E. Nowill
- Integrated Center for Pediatric OncoHaematological Research, State University of Campinas, Campinas, SP, Brazil
| | - Manuel Caruso
- CHU de Québec-Université Laval Research Center (Oncology Division), Université Laval Cancer Research Center, Québec, QC, Canada
| | - Pedro O. de Campos-Lima
- Boldrini Children’s Center, Campinas, SP, Brazil
- Molecular and Morphofunctional Biology Graduate Program, Institute of Biology, State University of Campinas, Campinas, SP, Brazil
| |
Collapse
|
49
|
Kumar K, Tan WS, Arshad SS, Ho KL. Virus-like Particles of Nodavirus Displaying the Receptor Binding Domain of SARS-CoV-2 Spike Protein: A Potential VLP-Based COVID-19 Vaccine. Int J Mol Sci 2023; 24:ijms24054398. [PMID: 36901827 PMCID: PMC10001971 DOI: 10.3390/ijms24054398] [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: 12/03/2022] [Revised: 02/06/2023] [Accepted: 02/19/2023] [Indexed: 02/25/2023] Open
Abstract
Since the outbreak of the coronavirus disease 2019 (COVID-19), various vaccines have been developed for emergency use. The efficacy of the initial vaccines based on the ancestral strain of severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2) has become a point of contention due to the emergence of new variants of concern (VOCs). Therefore, continuous innovation of new vaccines is required to target upcoming VOCs. The receptor binding domain (RBD) of the virus spike (S) glycoprotein has been extensively used in vaccine development due to its role in host cell attachment and penetration. In this study, the RBDs of the Beta (β) and Delta (δ) variants were fused to the truncated Macrobrachium rosenbergii nodavirus capsid protein without the protruding domain (CΔ116-MrNV-CP). Immunization of BALB/c mice with the virus-like particles (VLPs) self-assembled from the recombinant CP showed that, with AddaVax as an adjuvant, a significantly high level of humoral response was elicited. Specifically, mice injected with equimolar of adjuvanted CΔ116-MrNV-CP fused with the RBD of the β- and δ-variants increased T helper (Th) cell production with a CD8+/CD4+ ratio of 0.42. This formulation also induced proliferation of macrophages and lymphocytes. Overall, this study demonstrated that the nodavirus truncated CP fused with the SARS-CoV-2 RBD has potential to be developed as a VLP-based COVID-19 vaccine.
Collapse
Affiliation(s)
- Kiven Kumar
- Department of Pathology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, UPM, Serdang 43400, Selangor, Malaysia
| | - Wen Siang Tan
- Department of Microbiology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, UPM, Serdang 43400, Selangor, Malaysia
| | - Siti Suri Arshad
- Department of Veterinary Pathology and Microbiology, Faculty of Veterinary Medicine, Universiti Putra Malaysia, UPM, Serdang 43400, Selangor, Malaysia
| | - Kok Lian Ho
- Department of Pathology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, UPM, Serdang 43400, Selangor, Malaysia
- Correspondence: ; Tel.: +603-9769-2729
| |
Collapse
|
50
|
Kaplonek P, Cizmeci D, Lee JSL, Shin SA, Fischinger S, Gobeil P, Pillet S, Charland N, Ward BJ, Alter G. Robust induction of functional humoral response by a plant-derived Coronavirus-like particle vaccine candidate for COVID-19. NPJ Vaccines 2023; 8:13. [PMID: 36781879 PMCID: PMC9924894 DOI: 10.1038/s41541-023-00612-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Accepted: 01/27/2023] [Indexed: 02/15/2023] Open
Abstract
Despite the success of existing COVID-19 vaccine platforms, the persistent limitations in global deployment of vaccines and waning immunity exhibited by many of the currently deployed vaccine platforms have led to perpetual outbreaks of SARS-CoV-2 variants of concern. Thus, there is an urgent need to develop new durable vaccine candidates, to expand the global vaccine pipeline, and provide safe and effective solutions for every country worldwide. Here we deeply profiled the functional humoral response induced by two doses of AS03-adjuvanted and non-adjuvanted plant-derived Coronavirus-like particle (CoVLP) vaccine candidate from the phase 1 clinical trial, at peak immunogenicity and six months post-vaccination. AS03-adjuvanted CoVLP induced robust and durable SARS-CoV-2 specific humoral immunity, marked by strong IgG1antibody responses, potent FcγR binding, and antibody effector function. Contrary to a decline in neutralizing antibody titers, the FcγR2A-receptor binding capacity and antibody-mediated effector functions, such as opsonophagocytosis, remained readily detectable for at least six months.
Collapse
Affiliation(s)
| | - Deniz Cizmeci
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | | | - Sally A Shin
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | | | | | | | | | - Brian J Ward
- Medicago Inc., Quebec City, QC, Canada.
- Research Institute of the McGill University Health Centre, Montréal, QC, Canada.
| | - Galit Alter
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA.
| |
Collapse
|