51
|
Sharifi E, Yousefiasl S, Trovato M, Sartorius R, Esmaeili Y, Goodarzi H, Ghomi M, Bigham A, Moghaddam FD, Heidarifard M, Pourmotabed S, Nazarzadeh Zare E, Paiva-Santos AC, Rabiee N, Wang X, Tay FR. Nanostructures for prevention, diagnosis, and treatment of viral respiratory infections: from influenza virus to SARS-CoV-2 variants. J Nanobiotechnology 2023; 21:199. [PMID: 37344894 PMCID: PMC10283343 DOI: 10.1186/s12951-023-01938-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 05/24/2023] [Indexed: 06/23/2023] Open
Abstract
Viruses are a major cause of mortality and socio-economic downfall despite the plethora of biopharmaceuticals designed for their eradication. Conventional antiviral therapies are often ineffective. Live-attenuated vaccines can pose a safety risk due to the possibility of pathogen reversion, whereas inactivated viral vaccines and subunit vaccines do not generate robust and sustained immune responses. Recent studies have demonstrated the potential of strategies that combine nanotechnology concepts with the diagnosis, prevention, and treatment of viral infectious diseases. The present review provides a comprehensive introduction to the different strains of viruses involved in respiratory diseases and presents an overview of recent advances in the diagnosis and treatment of viral infections based on nanotechnology concepts and applications. Discussions in diagnostic/therapeutic nanotechnology-based approaches will be focused on H1N1 influenza, respiratory syncytial virus, human parainfluenza virus type 3 infections, as well as COVID-19 infections caused by the SARS-CoV-2 virus Delta variant and new emerging Omicron variant.
Collapse
Affiliation(s)
- Esmaeel Sharifi
- Department of Tissue Engineering and Biomaterials, School of Advanced Medical Sciences and Technologies, Hamadan University of Medical Sciences, Hamadan, 6517838736, Iran.
| | - Satar Yousefiasl
- Dental Research Center, Dentistry Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Maria Trovato
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council (CNR), 80131, Naples, Italy
| | - Rossella Sartorius
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council (CNR), 80131, Naples, Italy
| | - Yasaman Esmaeili
- School of Advanced Technologies in Medicine, Biosensor Research Center, Isfahan University of Medical Sciences, Isfahan, 8174673461, Iran
| | - Hamid Goodarzi
- Centre de recherche, Hôpital Maisonneuve-Rosemont, Montreal, QC, Canada
- Départment d'Ophtalmologie, Université de Montréal, Montreal, QC, Canada
| | - Matineh Ghomi
- School of Chemistry, Damghan University, Damghan, 36716-45667, Iran
| | - Ashkan Bigham
- Department of Tissue Engineering and Biomaterials, School of Advanced Medical Sciences and Technologies, Hamadan University of Medical Sciences, Hamadan, 6517838736, Iran
| | - Farnaz Dabbagh Moghaddam
- Institute for Photonics and Nanotechnologies, National Research Council, Via Fosso del Cavaliere, 100, 00133, Rome, Italy
| | - Maryam Heidarifard
- Centre de recherche, Hôpital Maisonneuve-Rosemont, Montreal, QC, Canada
- Départment d'Ophtalmologie, Université de Montréal, Montreal, QC, Canada
| | - Samiramis Pourmotabed
- Department of Emergency Medicine, School of Medicine, Hamadan University of Medical Sciences, Hamadan, 6517838736, Iran
| | | | - Ana Cláudia Paiva-Santos
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548, Coimbra, Portugal
- Group of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548, Coimbra, Portugal
| | - Navid Rabiee
- School of Engineering, Macquarie University, Sydney, NSW, 2109, Australia
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-ro, Nam-gu, Pohang, Gyeongbuk, 37673, Republic of Korea
| | - Xiangdong Wang
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University Shanghai Medical College, Shanghai, 200032, China
| | - Franklin R Tay
- The Graduate School, Augusta University, Augusta, GA, 30912, USA.
| |
Collapse
|
52
|
Hillebrandt N, Hubbuch J. Size-selective downstream processing of virus particles and non-enveloped virus-like particles. Front Bioeng Biotechnol 2023; 11:1192050. [PMID: 37304136 PMCID: PMC10248422 DOI: 10.3389/fbioe.2023.1192050] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 05/08/2023] [Indexed: 06/13/2023] Open
Abstract
Non-enveloped virus-like particles (VLPs) are versatile protein nanoparticles with great potential for biopharmaceutical applications. However, conventional protein downstream processing (DSP) and platform processes are often not easily applicable due to the large size of VLPs and virus particles (VPs) in general. The application of size-selective separation techniques offers to exploit the size difference between VPs and common host-cell impurities. Moreover, size-selective separation techniques offer the potential for wide applicability across different VPs. In this work, basic principles and applications of size-selective separation techniques are reviewed to highlight their potential in DSP of VPs. Finally, specific DSP steps for non-enveloped VLPs and their subunits are reviewed as well as the potential applications and benefits of size-selective separation techniques are shown.
Collapse
Affiliation(s)
| | - Jürgen Hubbuch
- Institute of Engineering in Life Sciences, Section IV: Biomolecular Separation Engineering, Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany
| |
Collapse
|
53
|
Yang JI, Sepúlveda D, Vardia I, Skov J, Goksøyr L, Sander AF, Lorenzen N. High immunogenicity of virus-like particles (VLPs) decorated with Aeromonas salmonicida VapA antigen in rainbow trout. Front Immunol 2023; 14:1139206. [PMID: 37283749 PMCID: PMC10239931 DOI: 10.3389/fimmu.2023.1139206] [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: 01/06/2023] [Accepted: 05/02/2023] [Indexed: 06/08/2023] Open
Abstract
The Gram-negative bacterium A. salmonicida is the causal agent of furunculosis and used to be one of the most loss-causing bacterial infections in the salmonid aquaculture industry with a mortality rate of about 90% until the 1990s, when an inactivated vaccine with mineral oil as adjuvant was successfully implemented to control the disease. However, the use of this vaccine is associated with inflammatory side effects in the peritoneal cavity as well as autoimmune reactions in Atlantic salmon, and incomplete protection has been reported in rainbow trout. We here aimed at developing and testing a recombinant alternative vaccine based on virus-like particles (VLPs) decorated with VapA, the key structural surface protein in the outer A-layer of A. salmonicida. The VLP carrier was based on either the capsid protein of a fish nodavirus, namely red grouper nervous necrotic virus (RGNNV) or the capsid protein of Acinetobacter phage AP205. The VapA and capsid proteins were expressed individually in E. coli and VapA was fused to auto-assembled VLPs using the SpyTag/SpyCatcher technology. Rainbow trout were vaccinated/immunized with the VapA-VLP vaccines by intraperitoneal injection and were challenged with A. salmonicida 7 weeks later. The VLP vaccines provided protection comparable to that of a bacterin-based vaccine and antibody response analysis demonstrated that vaccinated fish mounted a strong VapA-specific antibody response. To our knowledge, this is the first demonstration of the potential use of antigen-decorated VLPs for vaccination against a bacterial disease in salmonids.
Collapse
Affiliation(s)
- Jeong In Yang
- National Institute of Aquatic Resources, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Dagoberto Sepúlveda
- National Institute of Aquatic Resources, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Irina Vardia
- National Institute of Aquatic Resources, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Jakob Skov
- National Institute of Aquatic Resources, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Louise Goksøyr
- Centre for Medical Parasitology, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
- AdaptVac Aps, Copenhagen, Denmark
| | - Adam F. Sander
- Centre for Medical Parasitology, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
- AdaptVac Aps, Copenhagen, Denmark
| | - Niels Lorenzen
- National Institute of Aquatic Resources, Technical University of Denmark, Kongens Lyngby, Denmark
| |
Collapse
|
54
|
Ortiz R, Barajas A, Pons-Grífols A, Trinité B, Tarrés-Freixas F, Rovirosa C, Urrea V, Barreiro A, Gonzalez-Tendero A, Cardona M, Ferrer L, Clotet B, Carrillo J, Aguilar-Gurrieri C, Blanco J. Exploring FeLV-Gag-Based VLPs as a New Vaccine Platform-Analysis of Production and Immunogenicity. Int J Mol Sci 2023; 24:ijms24109025. [PMID: 37240371 DOI: 10.3390/ijms24109025] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/11/2023] [Accepted: 05/18/2023] [Indexed: 05/28/2023] Open
Abstract
Feline leukemia virus (FeLV) is one of the most prevalent infectious diseases in domestic cats. Although different commercial vaccines are available, none of them provides full protection. Thus, efforts to design a more efficient vaccine are needed. Our group has successfully engineered HIV-1 Gag-based VLPs that induce a potent and functional immune response against the HIV-1 transmembrane protein gp41. Here, we propose to use this concept to generate FeLV-Gag-based VLPs as a novel vaccine strategy against this retrovirus. By analogy to our HIV-1 platform, a fragment of the FeLV transmembrane p15E protein was exposed on FeLV-Gag-based VLPs. After optimization of Gag sequences, the immunogenicity of the selected candidates was evaluated in C57BL/6 and BALB/c mice, showing strong cellular and humoral responses to Gag but failing to generate anti-p15E antibodies. Altogether, this study not only tests the versatility of the enveloped VLP-based vaccine platform but also sheds light on FeLV vaccine research.
Collapse
Affiliation(s)
- Raquel Ortiz
- AIDS Research Institute, IrsiCaixa, Campus Can Ruti, 08916 Badalona, Spain
- Doctorate School, Microbiology Department, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Ana Barajas
- AIDS Research Institute, IrsiCaixa, Campus Can Ruti, 08916 Badalona, Spain
- Doctorate School, Medicine Department, University of Vic-Central University of Catalonia (UVic-UCC), 08500 Vic, Spain
| | - Anna Pons-Grífols
- AIDS Research Institute, IrsiCaixa, Campus Can Ruti, 08916 Badalona, Spain
- Doctorate School, Microbiology Department, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Benjamin Trinité
- AIDS Research Institute, IrsiCaixa, Campus Can Ruti, 08916 Badalona, Spain
| | | | - Carla Rovirosa
- AIDS Research Institute, IrsiCaixa, Campus Can Ruti, 08916 Badalona, Spain
| | - Victor Urrea
- AIDS Research Institute, IrsiCaixa, Campus Can Ruti, 08916 Badalona, Spain
| | | | | | | | | | - Bonaventura Clotet
- AIDS Research Institute, IrsiCaixa, Campus Can Ruti, 08916 Badalona, Spain
- Doctorate School, Medicine Department, University of Vic-Central University of Catalonia (UVic-UCC), 08500 Vic, Spain
- Infectious Diseases Department, Germans Trias I Pujol Hospital, 08916 Badalona, Spain
| | - Jorge Carrillo
- AIDS Research Institute, IrsiCaixa, Campus Can Ruti, 08916 Badalona, Spain
- CIBERINFEC, ISCIII, 28029 Madrid, Spain
| | | | - Julià Blanco
- AIDS Research Institute, IrsiCaixa, Campus Can Ruti, 08916 Badalona, Spain
- Doctorate School, Microbiology Department, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
- Doctorate School, Medicine Department, University of Vic-Central University of Catalonia (UVic-UCC), 08500 Vic, Spain
- CIBERINFEC, ISCIII, 28029 Madrid, Spain
- Germans Trias I Pujol Research Institute (IGTP), Campus Can Ruti, 08916 Badalona, Spain
| |
Collapse
|
55
|
Chang CC, Algaissi A, Lai CC, Chang CK, Lin JS, Wang YS, Chang BH, Chang YC, Chen WT, Fan YQ, Peng BH, Chao CY, Tzeng SR, Liang PH, Sung WC, Hu AYC, Chang SC, Chang MF. Subunit vaccines with a saponin-based adjuvant boost humoral and cellular immunity to MERS coronavirus. Vaccine 2023; 41:3337-3346. [PMID: 37085450 PMCID: PMC10083212 DOI: 10.1016/j.vaccine.2023.04.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 02/15/2023] [Accepted: 04/03/2023] [Indexed: 04/23/2023]
Abstract
Middle East respiratory syndrome coronavirus (MERS-CoV) outbreaks have constituted a public health issue with drastic mortality higher than 34%, necessitating the development of an effective vaccine. During MERS-CoV infection, the trimeric spike protein on the viral envelope is primarily responsible for attachment to host cellular receptor, dipeptidyl peptidase 4 (DPP4). With the goal of generating a protein-based prophylactic, we designed a subunit vaccine comprising the recombinant S1 protein with a trimerization motif (S1-Fd) and examined its immunogenicity and protective immune responses in combination with various adjuvants. We found that sera from immunized wild-type and human DPP4 transgenic mice contained S1-specific antibodies that can neutralize MERS-CoV infection in susceptible cells. Vaccination with S1-Fd protein in combination with a saponin-based QS-21 adjuvant provided long-term humoral as well as cellular immunity in mice. Our findings highlight the significance of the trimeric S1 protein in the development of MERS-CoV vaccines and offer a suitable adjuvant, QS-21, to induce robust and prolonged memory T cell response.
Collapse
Affiliation(s)
- Chi-Chieh Chang
- Institute of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei 10051, Taiwan
| | - Abdullah Algaissi
- Department of Microbiology and Immunology, The University of Texas Medical Branch, Galveston, TX 77555, USA; Center for Biodefense and Emerging Disease, The University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Chia-Chun Lai
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli County 35053, Taiwan; College of Life Science, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Chun-Kai Chang
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei 100025, Taiwan
| | - Jr-Shiuan Lin
- Graduate Institute of Immunology, College of Medicine, National Taiwan University, Taipei 10051, Taiwan
| | - Yi-Shiang Wang
- Institute of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei 10051, Taiwan
| | - Bo-Hau Chang
- Institute of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei 10051, Taiwan
| | - Yu-Chiuan Chang
- Institute of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei 10051, Taiwan
| | - Wei-Ting Chen
- Department of Physics, National Taiwan University, Taipei 10617, Taiwan
| | - Yong-Qing Fan
- Institute of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei 10051, Taiwan
| | - Bi-Hung Peng
- Department of Neurosciences, The University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Chih-Yu Chao
- Department of Physics, National Taiwan University, Taipei 10617, Taiwan
| | - Shiou-Ru Tzeng
- Institute of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei 10051, Taiwan
| | - Pi-Hui Liang
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei 100025, Taiwan
| | - Wang-Chou Sung
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli County 35053, Taiwan
| | - Alan Yung-Chih Hu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli County 35053, Taiwan
| | - Shin C Chang
- Institute of Microbiology, College of Medicine, National Taiwan University, Taipei 10051, Taiwan
| | - Ming-Fu Chang
- Institute of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei 10051, Taiwan.
| |
Collapse
|
56
|
Ohba K, Sehara Y, Enoki T, Mineno J, Ozawa K, Mizukami H. Adeno-associated virus vector system controlling capsid expression improves viral quantity and quality. iScience 2023; 26:106487. [PMID: 37096037 PMCID: PMC10122016 DOI: 10.1016/j.isci.2023.106487] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 01/13/2023] [Accepted: 03/20/2023] [Indexed: 04/08/2023] Open
Abstract
Adeno-associated virus (AAV) vectors are promising tools for gene therapy. The current AAV vector system produces an abundance of empty capsids that are eliminated before clinical use, leading to increased costs for gene therapy. In the present study, we established an AAV production system that regulates the timing of capsid expression using a tetracycline-dependent promoter. Tetracycline-regulating capsid expression increased viral yield and reduced empty capsids in various serotypes without altering AAV vector infectivity in vitro and in vivo. The replicase expression pattern change observed in the developed AAV vector system improved viral quantity and quality, whereas timing control of capsid expression reduced empty capsids. These findings provide a new perspective on the development of AAV vector production systems in gene therapy.
Collapse
Affiliation(s)
- Kenji Ohba
- Division of Genetic Therapeutics, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Tochigi 329-0498, Japan
- Corresponding author
| | - Yoshihide Sehara
- Division of Genetic Therapeutics, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Tochigi 329-0498, Japan
| | - Tatsuji Enoki
- CDM Center, TAKARA Bio Inc., Kusatsu, Shiga 525-0058, Japan
| | - Junichi Mineno
- CDM Center, TAKARA Bio Inc., Kusatsu, Shiga 525-0058, Japan
| | - Keiya Ozawa
- Division of Genetic Therapeutics, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Tochigi 329-0498, Japan
- Department of Immuno-Gene & Cell Therapy (Takara Bio), Jichi Medical University, Shimotsuke, Tochigi 329-0498, Japan
| | - Hiroaki Mizukami
- Division of Genetic Therapeutics, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Tochigi 329-0498, Japan
| |
Collapse
|
57
|
Koopman G, Amacker M, Stegmann T, Verschoor EJ, Verstrepen BE, Bhoelan F, Bemelman D, Böszörményi KP, Fagrouch Z, Kiemenyi-Kayere G, Mortier D, Verel DE, Niphuis H, Acar RF, Kondova I, Kap YS, Bogers WMJM, Mooij P, Fleury S. A low dose of RBD and TLR7/8 agonist displayed on influenza virosome particles protects rhesus macaque against SARS-CoV-2 challenge. Sci Rep 2023; 13:5074. [PMID: 36977691 PMCID: PMC10044094 DOI: 10.1038/s41598-023-31818-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 03/17/2023] [Indexed: 03/30/2023] Open
Abstract
Influenza virosomes serve as antigen delivery vehicles and pre-existing immunity toward influenza improves the immune responses toward antigens. Here, vaccine efficacy was evaluated in non-human primates with a COVID-19 virosome-based vaccine containing a low dose of RBD protein (15 µg) and the adjuvant 3M-052 (1 µg), displayed together on virosomes. Vaccinated animals (n = 6) received two intramuscular administrations at week 0 and 4 and challenged with SARS-CoV-2 at week 8, together with unvaccinated control animals (n = 4). The vaccine was safe and well tolerated and serum RBD IgG antibodies were induced in all animals and in the nasal washes and bronchoalveolar lavages in the three youngest animals. All control animals became strongly sgRNA positive in BAL, while all vaccinated animals were protected, although the oldest vaccinated animal (V1) was transiently weakly positive. The three youngest animals had also no detectable sgRNA in nasal wash and throat. Cross-strain serum neutralizing antibodies toward Wuhan-like, Alpha, Beta, and Delta viruses were observed in animals with the highest serum titers. Pro-inflammatory cytokines IL-8, CXCL-10 and IL-6 were increased in BALs of infected control animals but not in vaccinated animals. Virosomes-RBD/3M-052 prevented severe SARS-CoV-2, as shown by a lower total lung inflammatory pathology score than control animals.
Collapse
Grants
- TRANSVAC2 2002-08-AVVAX-COVID-19, TRANSVAC2_TNA2002-08 European Commission
- TRANSVAC2 2002-08-AVVAX-COVID-19, TRANSVAC2_TNA2002-08 European Commission
- TRANSVAC2 2002-08-AVVAX-COVID-19, TRANSVAC2_TNA2002-08 European Commission
- TRANSVAC2 2002-08-AVVAX-COVID-19, TRANSVAC2_TNA2002-08 European Commission
- TRANSVAC2 2002-08-AVVAX-COVID-19, TRANSVAC2_TNA2002-08 European Commission
- TRANSVAC2 2002-08-AVVAX-COVID-19, TRANSVAC2_TNA2002-08 European Commission
- TRANSVAC2 2002-08-AVVAX-COVID-19, TRANSVAC2_TNA2002-08 European Commission
- TRANSVAC2 2002-08-AVVAX-COVID-19, TRANSVAC2_TNA2002-08 European Commission
- TRANSVAC2 2002-08-AVVAX-COVID-19, TRANSVAC2_TNA2002-08 European Commission
- TRANSVAC2 2002-08-AVVAX-COVID-19, TRANSVAC2_TNA2002-08 European Commission
- TRANSVAC2 2002-08-AVVAX-COVID-19, TRANSVAC2_TNA2002-08 European Commission
- TRANSVAC2 2002-08-AVVAX-COVID-19, TRANSVAC2_TNA2002-08 European Commission
- TRANSVAC2 2002-08-AVVAX-COVID-19, TRANSVAC2_TNA2002-08 European Commission
- TRANSVAC2 2002-08-AVVAX-COVID-19, TRANSVAC2_TNA2002-08 European Commission
- TRANSVAC2 2002-08-AVVAX-COVID-19, TRANSVAC2_TNA2002-08 European Commission
- TRANSVAC2 2002-08-AVVAX-COVID-19, TRANSVAC2_TNA2002-08 European Commission
Collapse
Affiliation(s)
- Gerrit Koopman
- Department of Virology, Biomedical Primate Research Centre (BPRC), Rijswijk, The Netherlands.
| | - Mario Amacker
- Mymetics SA, 4 Route de La Corniche, 1066, Epalinges, Switzerland
- Department for BioMedical Research DBMR, Department of Pulmonary Medicine, Inselspital, Bern University Hospital, University of Bern, 3008, Bern, Switzerland
| | - Toon Stegmann
- Mymetics BV, JH Oortweg 21, 2333 CH, Leiden, The Netherlands
| | - Ernst J Verschoor
- Department of Virology, Biomedical Primate Research Centre (BPRC), Rijswijk, The Netherlands
| | - Babs E Verstrepen
- Department of Virology, Biomedical Primate Research Centre (BPRC), Rijswijk, The Netherlands
| | - Farien Bhoelan
- Mymetics BV, JH Oortweg 21, 2333 CH, Leiden, The Netherlands
| | - Denzel Bemelman
- Mymetics BV, JH Oortweg 21, 2333 CH, Leiden, The Netherlands
| | - Kinga P Böszörményi
- Department of Virology, Biomedical Primate Research Centre (BPRC), Rijswijk, The Netherlands
| | - Zahra Fagrouch
- Department of Virology, Biomedical Primate Research Centre (BPRC), Rijswijk, The Netherlands
| | | | - Daniella Mortier
- Department of Virology, Biomedical Primate Research Centre (BPRC), Rijswijk, The Netherlands
| | - Dagmar E Verel
- Department of Virology, Biomedical Primate Research Centre (BPRC), Rijswijk, The Netherlands
| | - Henk Niphuis
- Department of Virology, Biomedical Primate Research Centre (BPRC), Rijswijk, The Netherlands
| | - Roja Fidel Acar
- Department of Virology, Biomedical Primate Research Centre (BPRC), Rijswijk, The Netherlands
| | - Ivanela Kondova
- Animal Science Department, Biomedical Primate Research Centre (BPRC), Rijswijk, The Netherlands
| | - Yolanda S Kap
- Department of Virology, Biomedical Primate Research Centre (BPRC), Rijswijk, The Netherlands
| | - Willy M J M Bogers
- Department of Virology, Biomedical Primate Research Centre (BPRC), Rijswijk, The Netherlands
| | - Petra Mooij
- Department of Virology, Biomedical Primate Research Centre (BPRC), Rijswijk, The Netherlands
| | - Sylvain Fleury
- Mymetics SA, 4 Route de La Corniche, 1066, Epalinges, Switzerland.
| |
Collapse
|
58
|
Brito C, Lourenço C, Magalhães J, Reis S, Borges M. Nanoparticles as a Delivery System of Antigens for the Development of an Effective Vaccine against Toxoplasma gondii. Vaccines (Basel) 2023; 11:vaccines11040733. [PMID: 37112645 PMCID: PMC10142924 DOI: 10.3390/vaccines11040733] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/10/2023] [Accepted: 03/21/2023] [Indexed: 03/29/2023] Open
Abstract
Nanoparticles include particles ranging in size from nanometers to micrometers, whose physicochemical characteristics are optimized to make them appropriate delivery vehicles for drugs or immunogens important in the fight and/or prevention of infectious diseases. There has been a rise in the use of nanoparticles in preventive vaccine formulations as immunostimulatory adjuvants, and as vehicles for immunogen delivery to target immune cells. Toxoplasma is important worldwide, and may cause human toxoplasmosis. In immunocompetent hosts, infection is usually asymptomatic, but in immunocompromised patients it can cause serious neurological and ocular consequences, such as encephalitis and retinochoroiditis. Primary infection during pregnancy may cause abortion or congenital toxoplasmosis. Currently, there is no effective human vaccine against this disease. Evidence has emerged from several experimental studies testing nanovaccines showing them to be promising tools in the prevention of experimental toxoplasmosis. For the present study, a literature review was carried out on articles published over the last 10 years through the PubMed database, pertaining to in vivo experimental models of T. gondii infection where nanovaccines were tested and protection and immune responses evaluated. This review aims to highlight the way forward in the search for an effective vaccine for toxoplasmosis.
Collapse
|
59
|
Li Y, Sun R, Li S, Tan Z, Li Z, Liu Y, Guo Y, Huang J. ASFV proteins presented at the surface of T7 phages induce strong antibody responses in mice. J Virol Methods 2023; 316:114725. [PMID: 36965632 DOI: 10.1016/j.jviromet.2023.114725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 03/21/2023] [Accepted: 03/22/2023] [Indexed: 03/27/2023]
Abstract
African swine fever virus (ASFV) infection causes substantial economic losses to the swine industry worldwide, and there are still no safe and effective vaccines or therapeutics available. The granulated virus antigen improves the antigen present process and elicits high antibody reaction than the subunit antigen. In this study, the SpyTag peptide-p10 fusion protein was altered and displayed on the surface of the T7 phage to construct an engineered phage (T7-ST). At the same time, ASFV antigen-Spycatcher C-terminal-fused protein (antigen-SC) was expressed and purified by an E. coli prokaryotic expression system. Five virus-like particles (VLPs) displaying the main ASFV antigenic proteins P30, P54, P72, CD2v, and K145R were reconstructed by the isopeptide bond between SpyTag and antigen-SC proteins. The stability of five ASFV VLPs in high temperature and extreme pH conditions was evaluated by transmission electron microscopy (TEM) and plaque analysis. All ASFV VLPs induced a high titer antigen-specific antibody response in mice. Our results showed that the granulated antigen displaying ASFV protein on the surface of the T7 phage provides a robust potential vaccine and diagnostic tool to address the challenge of the ASFV pandemic.
Collapse
Affiliation(s)
- Yuanfang Li
- School of Life Sciences, Tianjin University, Tianjin, China, 300072
| | - Ruiqi Sun
- School of Life Sciences, Tianjin University, Tianjin, China, 300072
| | - Shujun Li
- School of Life Sciences, Tianjin University, Tianjin, China, 300072
| | - Zheng Tan
- School of Life Sciences, Tianjin University, Tianjin, China, 300072
| | - Zexing Li
- School of Life Sciences, Tianjin University, Tianjin, China, 300072
| | - Yebin Liu
- China Institute of Veterinary Drug Control, Beijing, China, 100081
| | - Yanyu Guo
- School of Life Sciences, Tianjin University, Tianjin, China, 300072.
| | - Jinhai Huang
- School of Life Sciences, Tianjin University, Tianjin, China, 300072.
| |
Collapse
|
60
|
Radiom M, Keys T, Turgay Y, Ali A, Preet S, Chesnov S, Lutz-Bueno V, Slack E, Mezzenga R. Mechanical tuning of virus-like particles. J Colloid Interface Sci 2023; 634:963-971. [PMID: 36571858 DOI: 10.1016/j.jcis.2022.12.090] [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: 08/23/2022] [Revised: 12/18/2022] [Accepted: 12/18/2022] [Indexed: 12/24/2022]
Abstract
HYPOTHESIS Virus-like particles (VLPs) are promising scaffolds for developing mucosal vaccines. For their optimal performance, in addition to design parameters from an immunological perspective, biophysical properties may need to be considered. EXPERIMENTS We investigated the mechanical properties of VLPs scaffolded on the coat protein of Acinetobacter phage AP205 using atomic force microscopy and small angle X-ray scattering. FINDINGS Investigations showed that AP205 VLP is a tough nanoshell of stiffness 93 ± 23 pN/nm and elastic modulus 0.11 GPa. However, its mechanical properties are modulated by attaching muco-inert polyethylene glycol to 46 ± 10 pN/nm and 0.05 GPa. Addition of antigenic peptides derived from SARS-CoV2 spike protein by genetic fusion increased the stiffness to 146 ± 54 pN/nm although the elastic modulus remained unchanged. These results, which are interpreted in terms of shell thickness and coat protein net charge variations, demonstrate that surface conjugation can induce appreciable changes in the biophysical properties of VLP-scaffolded vaccines.
Collapse
Affiliation(s)
- Milad Radiom
- Laboratory of Food Immunology, Institute of Food, Nutrition and Health, ETH Zürich, Zürich, Switzerland; Laboratory of Food and Soft Materials, Institute of Food, Nutrition and Health, ETH Zürich, Zürich, Switzerland.
| | - Tim Keys
- Laboratory of Food Immunology, Institute of Food, Nutrition and Health, ETH Zürich, Zürich, Switzerland
| | - Yagmur Turgay
- Laboratory of Food Immunology, Institute of Food, Nutrition and Health, ETH Zürich, Zürich, Switzerland
| | - Ahmed Ali
- Laboratory of Food Immunology, Institute of Food, Nutrition and Health, ETH Zürich, Zürich, Switzerland
| | - Swapan Preet
- Laboratory of Food Immunology, Institute of Food, Nutrition and Health, ETH Zürich, Zürich, Switzerland
| | - Serge Chesnov
- University of Zürich/ETH Zürich, Functional Genomics Centre Zürich, Zürich, Switzerland
| | | | - Emma Slack
- Laboratory of Food Immunology, Institute of Food, Nutrition and Health, ETH Zürich, Zürich, Switzerland.
| | - Raffaele Mezzenga
- Laboratory of Food and Soft Materials, Institute of Food, Nutrition and Health, ETH Zürich, Zürich, Switzerland.
| |
Collapse
|
61
|
Ali H, Akbar M, Iqbal B, Ali F, Kant Sharma N, Kumar N, Najmi A, Albratty M, Alhazmi HA, Madkhali OA, Zoghebi K, Shamsher Alam M. Virosome: An engineered virus for vaccine delivery. Saudi Pharm J 2023; 31:752-764. [PMID: 37181145 PMCID: PMC10172599 DOI: 10.1016/j.jsps.2023.03.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 03/22/2023] [Accepted: 03/24/2023] [Indexed: 04/03/2023] Open
Abstract
The purpose of immunization is the effective cellular and humoral immune response against antigens. Several studies on novel vaccine delivery approaches such as micro-particles, liposomes & nanoparticles, etc. against infectious diseases have been investigated so far. In contrast to the conventional approaches in vaccine development, a virosomes-based vaccine represents the next generation in the field of immunization because of its balance between efficacy and tolerability by virtue of its mechanism of immune instigation. The versatility of virosomes as a vaccine adjuvant, and delivery vehicle of molecules of different nature, such as peptides, nucleic acids, and proteins, as well as provide an insight into the prospect of drug targeting using virosomes. This article focuses on the basics of virosomes, structure, composition formulation and development, advantages, interplay with the immune system, current clinical status, different patents highlighting the applications of virosomes and their status, recent advances, and research associated with virosomes, the efficacy, safety, and tolerability of virosomes based vaccines and the future prospective.
Collapse
|
62
|
González-Davis O, Villagrana-Escareño MV, Trujillo MA, Gama P, Chauhan K, Vazquez-Duhalt R. Virus-like nanoparticles as enzyme carriers for Enzyme Replacement Therapy (ERT). Virology 2023; 580:73-87. [PMID: 36791560 DOI: 10.1016/j.virol.2023.01.017] [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: 08/31/2022] [Revised: 01/20/2023] [Accepted: 01/30/2023] [Indexed: 02/09/2023]
Abstract
Enzyme replacement therapy (ERT) has been used to treat a few of the many existing diseases which are originated from the lack of, or low enzymatic activity. Exogenous enzymes are administered to contend with the enzymatic activity deficiency. Enzymatic nanoreactors based on the enzyme encapsulation inside of virus-like particles (VLPs) appear as an interesting alternative for ERT. VLPs are excellent delivery vehicles for therapeutic enzymes as they are biodegradable, uniformly organized, and porous nanostructures that transport and could protect the biocatalyst from the external environment without much affecting the bioactivity. Consequently, significant efforts have been made in the production processes of virus-based enzymatic nanoreactors and their functionalization, which are critically reviewed. The use of virus-based enzymatic nanoreactors for the treatment of lysosomal storage diseases such as Gaucher, Fabry, and Pompe diseases, as well as potential therapies for galactosemia, and Hurler and Hunter syndromes are discussed.
Collapse
Affiliation(s)
- Oscar González-Davis
- Centro de Nanociencias y Nanotecnología, Universidad Nacional Autónoma de México, Km 107 carretera, Tijuana-Ensenada, Baja California, 22860, Mexico
| | - Maria V Villagrana-Escareño
- Centro de Nanociencias y Nanotecnología, Universidad Nacional Autónoma de México, Km 107 carretera, Tijuana-Ensenada, Baja California, 22860, Mexico
| | - Mario A Trujillo
- School of Medicine, Universidad Xochicalco, Ensenada, Baja California, Mexico
| | - Pedro Gama
- Centro de Nanociencias y Nanotecnología, Universidad Nacional Autónoma de México, Km 107 carretera, Tijuana-Ensenada, Baja California, 22860, Mexico
| | - Kanchan Chauhan
- Centro de Nanociencias y Nanotecnología, Universidad Nacional Autónoma de México, Km 107 carretera, Tijuana-Ensenada, Baja California, 22860, Mexico
| | - Rafael Vazquez-Duhalt
- Centro de Nanociencias y Nanotecnología, Universidad Nacional Autónoma de México, Km 107 carretera, Tijuana-Ensenada, Baja California, 22860, Mexico.
| |
Collapse
|
63
|
Assessing the Efficacy of VLP-Based Vaccine against Epstein-Barr Virus Using a Rabbit Model. Vaccines (Basel) 2023; 11:vaccines11030540. [PMID: 36992124 DOI: 10.3390/vaccines11030540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 02/02/2023] [Accepted: 02/03/2023] [Indexed: 03/02/2023] Open
Abstract
Epstein–Barr virus (EBV) is etiologically associated with a number of malignant and non-malignant conditions. Thus, a prophylactic vaccine against this virus could help to reduce the burden of many EBV-associated diseases. Previously, we reported that an EBV virus-like particle (VLP) vaccine was highly immunogenic and produced a strong humoral response in mice. However, since EBV does not infect mice, the efficacy of the VLP in preventing EBV infection could not be addressed. Here we examined, for the first time, the efficacy of the EBV-VLP vaccine using a novel rabbit model of EBV infection. Animals vaccinated with two doses of VLP elicited higher antibody responses to total EBV antigens compared to animals receiving one dose. Vaccinated animals also elicited both IgM and IgG to EBV-specific antigens, VCA and EBNA1. Analysis of peripheral blood and spleen for EBV copy number indicated that the viral load in both of these compartments was lower in animals receiving a 2-dose vaccine. However, the VLP vaccine was ineffective in preventing EBV infection. With several other EBV vaccine candidates currently at various stages of development and testing, we believe that the rabbit model of EBV infection could be a great platform for evaluating potential candidates.
Collapse
|
64
|
England C, TrejoMartinez J, PerezSanchez P, Karki U, Xu J. Plants as Biofactories for Therapeutic Proteins and Antiviral Compounds to Combat COVID-19. Life (Basel) 2023; 13:617. [PMID: 36983772 PMCID: PMC10054913 DOI: 10.3390/life13030617] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 02/14/2023] [Accepted: 02/20/2023] [Indexed: 02/25/2023] Open
Abstract
The outbreak of coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) had a profound impact on the world's health and economy. Although the end of the pandemic may come in 2023, it is generally believed that the virus will not be completely eradicated. Most likely, the disease will become an endemicity. The rapid development of vaccines of different types (mRNA, subunit protein, inactivated virus, etc.) and some other antiviral drugs (Remdesivir, Olumiant, Paxlovid, etc.) has provided effectiveness in reducing COVID-19's impact worldwide. However, the circulating SARS-CoV-2 virus has been constantly mutating with the emergence of multiple variants, which makes control of COVID-19 difficult. There is still a pressing need for developing more effective antiviral drugs to fight against the disease. Plants have provided a promising production platform for both bioactive chemical compounds (small molecules) and recombinant therapeutics (big molecules). Plants naturally produce a diverse range of bioactive compounds as secondary metabolites, such as alkaloids, terpenoids/terpenes and polyphenols, which are a rich source of countless antiviral compounds. Plants can also be genetically engineered to produce valuable recombinant therapeutics. This molecular farming in plants has an unprecedented opportunity for developing vaccines, antibodies, and other biologics for pandemic diseases because of its potential advantages, such as low cost, safety, and high production volume. This review summarizes the latest advancements in plant-derived drugs used to combat COVID-19 and discusses the prospects and challenges of the plant-based production platform for antiviral agents.
Collapse
Affiliation(s)
- Corbin England
- Arkansas Biosciences Institute, Arkansas State University, Jonesboro, AR 72401, USA
- Molecular Biosciences Program, Arkansas State University, Jonesboro, AR 72401, USA
| | | | - Paula PerezSanchez
- Department of Biological Sciences, Arkansas State University, Jonesboro, AR 72401, USA
| | - Uddhab Karki
- Arkansas Biosciences Institute, Arkansas State University, Jonesboro, AR 72401, USA
- Molecular Biosciences Program, Arkansas State University, Jonesboro, AR 72401, USA
| | - Jianfeng Xu
- Arkansas Biosciences Institute, Arkansas State University, Jonesboro, AR 72401, USA
- College of Agriculture, Arkansas State University, Jonesboro, AR 72401, USA
| |
Collapse
|
65
|
Lee J, Lee SK, Park JS, Lee KR. Plant-made pharmaceuticals: exploring studies for the production of recombinant protein in plants and assessing challenges ahead. PLANT BIOTECHNOLOGY REPORTS 2023; 17:53-65. [PMID: 36820221 PMCID: PMC9931573 DOI: 10.1007/s11816-023-00821-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 01/16/2023] [Accepted: 02/06/2023] [Indexed: 06/18/2023]
Abstract
The production of pharmaceutical compounds in plants is attracting increasing attention, as plant-based systems can be less expensive, safer, and more scalable than mammalian, yeast, bacterial, and insect cell expression systems. Here, we review the history and current status of plant-made pharmaceuticals. Producing pharmaceuticals in plants requires pairing the appropriate plant species with suitable transformation technology. Pharmaceuticals have been produced in tobacco, cereals, legumes, fruits, and vegetables via nuclear transformation, chloroplast transformation, transient expression, and transformation of suspension cell cultures. Despite this wide range of species and methods used, most such efforts have involved the nuclear transformation of tobacco. Tobacco readily generates large amounts of biomass, easily accepts foreign genes, and is amenable to stable gene expression via nuclear transformation. Although vaccines, antibodies, and therapeutic proteins have been produced in plants, such pharmaceuticals are not readily utilized by humans due to differences in glycosylation, and few such compounds have been approved due to a lack of clinical data. In addition, achieving an adequate immune response using plant-made pharmaceuticals can be difficult due to low rates of production compared to other expression systems. Various technologies have recently been developed to help overcome these limitations; however, plant systems are expected to increasingly become widely used expression systems for recombinant protein production.
Collapse
Affiliation(s)
- Juho Lee
- Department of Agricultural Biotechnology, National Institute of Agricultural Sciences, Rural Development Administration, Jeonju, 54874 Republic of Korea
| | - Seon-Kyeong Lee
- Department of Agricultural Biotechnology, National Institute of Agricultural Sciences, Rural Development Administration, Jeonju, 54874 Republic of Korea
| | - Jong-Sug Park
- Department of Agricultural Biotechnology, National Institute of Agricultural Sciences, Rural Development Administration, Jeonju, 54874 Republic of Korea
| | - Kyeong-Ryeol Lee
- Department of Agricultural Biotechnology, National Institute of Agricultural Sciences, Rural Development Administration, Jeonju, 54874 Republic of Korea
| |
Collapse
|
66
|
Gupta R, Arora K, Roy SS, Joseph A, Rastogi R, Arora NM, Kundu PK. Platforms, advances, and technical challenges in virus-like particles-based vaccines. Front Immunol 2023; 14:1123805. [PMID: 36845125 PMCID: PMC9947793 DOI: 10.3389/fimmu.2023.1123805] [Citation(s) in RCA: 56] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 01/30/2023] [Indexed: 02/11/2023] Open
Abstract
Viral infectious diseases threaten human health and global stability. Several vaccine platforms, such as DNA, mRNA, recombinant viral vectors, and virus-like particle-based vaccines have been developed to counter these viral infectious diseases. Virus-like particles (VLP) are considered real, present, licensed and successful vaccines against prevalent and emergent diseases due to their non-infectious nature, structural similarity with viruses, and high immunogenicity. However, only a few VLP-based vaccines have been commercialized, and the others are either in the clinical or preclinical phases. Notably, despite success in the preclinical phase, many vaccines are still struggling with small-scale fundamental research owing to technical difficulties. Successful production of VLP-based vaccines on a commercial scale requires a suitable platform and culture mode for large-scale production, optimization of transduction-related parameters, upstream and downstream processing, and monitoring of product quality at each step. In this review article, we focus on the advantages and disadvantages of various VLP-producing platforms, recent advances and technical challenges in VLP production, and the current status of VLP-based vaccine candidates at commercial, preclinical, and clinical levels.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Prabuddha K. Kundu
- Department of Research and Development, Premas Biotech Pvt Ltd., Sector IV, Industrial Model Township (IMT), Manesar, Gurgaon, India
| |
Collapse
|
67
|
Chu KB, Quan FS. Respiratory Viruses and Virus-like Particle Vaccine Development: How Far Have We Advanced? Viruses 2023; 15:v15020392. [PMID: 36851606 PMCID: PMC9965150 DOI: 10.3390/v15020392] [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: 12/12/2022] [Revised: 01/26/2023] [Accepted: 01/28/2023] [Indexed: 01/31/2023] Open
Abstract
With technological advancements enabling globalization, the intercontinental transmission of pathogens has become much easier. Respiratory viruses are one such group of pathogens that require constant monitoring since their outbreak leads to massive public health crises, as exemplified by the influenza virus, respiratory syncytial virus (RSV), and the recent coronavirus disease 2019 (COVID-19) outbreak caused by the SARS-CoV-2. To prevent the transmission of these highly contagious viruses, developing prophylactic tools, such as vaccines, is of considerable interest to the scientific community. Virus-like particles (VLPs) are highly sought after as vaccine platforms for their safety and immunogenicity profiles. Although several VLP-based vaccines against hepatitis B and human papillomavirus have been approved for clinical use by the United States Food and Drug Administration, VLP vaccines against the three aforementioned respiratory viruses are lacking. Here, we summarize the most recent progress in pre-clinical and clinical VLP vaccine development. We also outline various strategies that contributed to improving the efficacy of vaccines against each virus and briefly discuss the stability aspect of VLPs that makes it a highly desired vaccine platform.
Collapse
Affiliation(s)
- Ki-Back Chu
- Medical Research Center for Bioreaction to Reactive Oxygen Species and Biomedical Science Institute, Core Research Institute (CRI), Kyung Hee University, Seoul 02447, Republic of Korea
| | - Fu-Shi Quan
- Medical Research Center for Bioreaction to Reactive Oxygen Species and Biomedical Science Institute, Core Research Institute (CRI), Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Medical Zoology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Correspondence:
| |
Collapse
|
68
|
Zhang J, Ge J, Li J, Li J, Zhang Y, Shi Y, Sun J, Wang Q, Zhang X, Zhao X. Expression of FMD virus-like particles in yeast Hansenula polymorpha and immunogenicity of combine with CpG and aluminum adjuvant. J Vet Sci 2023; 24:e15. [PMID: 36726280 PMCID: PMC9899949 DOI: 10.4142/jvs.22227] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 11/28/2022] [Accepted: 12/12/2022] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Inactivated vaccines are limited in preventing foot-and-mouth disease (FMD) due to safety problems. Recombinant virus-like particles (VLPs) are an excellent candidate for a novel vaccine for preventing FMD, given that VLPs have similar immunogenicity as natural viruses and are replication- and infection-incompetent. OBJECTIVES The 3C protease and P1 polyprotein of type O FMD virus (FDMV) was expressed in yeast Hansenula polymorpha to generate self-resembling VLPs, and the potential of recombinant VLPs as an FMD vaccine was evaluated. METHODS BALB/c mice were immunized with recombinant purified VLPs using CpG oligodeoxynucleotide and aluminum hydroxide gel as an adjuvant. Cytokines and lymphocytes from serum and spleen were analyzed by enzyme-linked immunosorbent assay, enzyme-linked immunospot assay, and flow cytometry. RESULTS The VLPs of FMD were purified successfully from yeast protein with a diameter of approximately 25 nm. The immunization of mice showed that animals produced high levels of FMDV antibodies and a higher level of antibodies for a longer time. In addition, higher levels of interferon-γ and CD4+ T cells were observed in mice immunized with VLPs. CONCLUSIONS The expression of VLPs of FMD in H. polymorpha provides a novel strategy for the generation of the FMDV vaccine.
Collapse
Affiliation(s)
- Jianhui Zhang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China
| | - Jun Ge
- Grand Theravac Life Sciences (Nanjing) Co., Ltd., Nanjing 210000, China
| | - Juyin Li
- Jiangsu Argi-animal Husbandry Vocational College, Taizhou 225300, China
| | - Jianqiang Li
- Grand Theravac Life Sciences (Nanjing) Co., Ltd., Nanjing 210000, China
| | - Yong Zhang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China
| | - Yinghui Shi
- Grand Theravac Life Sciences (Nanjing) Co., Ltd., Nanjing 210000, China
| | - Jiaojiao Sun
- Grand Theravac Life Sciences (Nanjing) Co., Ltd., Nanjing 210000, China
| | - Qiongjin Wang
- Grand Theravac Life Sciences (Nanjing) Co., Ltd., Nanjing 210000, China
| | - Xiaobo Zhang
- Department of Physiology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210000, China
| | - Xingxu Zhao
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China.
| |
Collapse
|
69
|
Kim KR, Lee AS, Kim SM, Heo HR, Kim CS. Virus-like nanoparticles as a theranostic platform for cancer. Front Bioeng Biotechnol 2023; 10:1106767. [PMID: 36714624 PMCID: PMC9878189 DOI: 10.3389/fbioe.2022.1106767] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 12/31/2022] [Indexed: 01/15/2023] Open
Abstract
Virus-like nanoparticles (VLPs) are natural polymer-based nanomaterials that mimic viral structures through the hierarchical assembly of viral coat proteins, while lacking viral genomes. VLPs have received enormous attention in a wide range of nanotechnology-based medical diagnostics and therapies, including cancer therapy, imaging, and theranostics. VLPs are biocompatible and biodegradable and have a uniform structure and controllable assembly. They can encapsulate a wide range of therapeutic and diagnostic agents, and can be genetically or chemically modified. These properties have led to sophisticated multifunctional theranostic platforms. This article reviews the current progress in developing and applying engineered VLPs for molecular imaging, drug delivery, and multifunctional theranostics in cancer research.
Collapse
Affiliation(s)
- Kyeong Rok Kim
- Graduate School of Biochemistry, Yeungnam University, Gyeongsan, South Korea
| | - Ae Sol Lee
- Graduate School of Biochemistry, Yeungnam University, Gyeongsan, South Korea
| | - Su Min Kim
- Graduate School of Biochemistry, Yeungnam University, Gyeongsan, South Korea
| | - Hye Ryoung Heo
- Senotherapy-Based Metabolic Disease Control Research Center, Yeungnam University, Gyeongsan, South Korea,*Correspondence: Chang Sup Kim, ; Hye Ryoung Heo,
| | - Chang Sup Kim
- Graduate School of Biochemistry, Yeungnam University, Gyeongsan, South Korea,School of Chemistry and Biochemistry, Yeungnam University, Gyeongsan, South Korea,*Correspondence: Chang Sup Kim, ; Hye Ryoung Heo,
| |
Collapse
|
70
|
Hossaini Alhashemi S, Ahmadi F, Dehshahri A. Lessons learned from COVID-19 pandemic: Vaccine platform is a key player. Process Biochem 2023; 124:269-279. [PMID: 36514356 PMCID: PMC9731819 DOI: 10.1016/j.procbio.2022.12.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 10/15/2022] [Accepted: 12/02/2022] [Indexed: 12/13/2022]
Abstract
The SARS-CoV-2 outbreak and emergence of COVID-19 resulted in the development of different vaccines based on various platforms to combat the disease. While the conventional platforms of inactivated/live attenuated, subunit proteins and virus-like particles (VLPs) have provided efficient and safe vaccines, novel platforms of viral vector- and nucleic acid-based vaccines opened up new horizons for vaccine development. The emergence of COVID-19 pandemic showed that the availability of platforms with high possibility of quick translation from bench to bedside is a prerequisite step in vaccine development in pandemics. Moreover, parallel development of different platforms as well as considering the shipping, storage condition, distribution infrastructure and route of administration are key players for successful and robust response. This review highlights the lessons learned from the current COVID-19 pandemic in terms of vaccine development to provide quick response to future outbreaks of infectious diseases and the importance of vaccine platform in its storage condition and shipping. Finally, the potential application of current COVID-19 vaccine platforms in the treatment of non-infectious diseases has been discussed.
Collapse
Affiliation(s)
| | - Fatemeh Ahmadi
- Department of Pharmaceutics, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran,Correspondence to: School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Dehshahri
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran,Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran,Correspondence to: School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
71
|
Zhao S, Han X, Lang Y, Xie Y, Yang Z, Zhao Q, Wen Y, Xia J, Wu R, Huang X, Huang Y, Cao S, Lan J, Luo L, Yan Q. Development and efficacy evaluation of remodeled canine parvovirus-like particles displaying major antigenic epitopes of a giant panda derived canine distemper virus. Front Microbiol 2023; 14:1117135. [PMID: 36922967 PMCID: PMC10008873 DOI: 10.3389/fmicb.2023.1117135] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 02/08/2023] [Indexed: 03/03/2023] Open
Abstract
Canine parvovirus (CPV) and Canine distemper virus (CDV) can cause fatal diseases in giant panda (Ailuropoda melanoleuca). The main capsid protein of CPV VP2 can be self-assembled to form virus-like particles (VLPs) in vitro, which is of great significance for potential vaccine development. In the present study, we remodeled the VP2 protein of a giant panda-derived CPV, where the major CDV F and N epitopes were incorporated in the N-terminal and loop2 region in two combinations to form chimeric VLPs. The reactivity ability and morphology of the recombinant proteins were confirmed by Western blot, hemagglutination (HA) test and electron microscopy. Subsequently, the immunogenicity of the VLPs was examined in vivo. Antigen-specific antibodies and neutralizing activity were measured by ELISA, hemagglutination inhibition (HI) test and serum neutralization test (SNT), respectively. In addition, antigen specific T cell activation were determined in splenic lymphocytes. The results indicated that the VLPs displayed good reaction with CDV/CPV antibodies, and the heterologous epitopes do not hamper solubility or activity. The VLPs showed decent HA activity, and resembled round-shaped particles with a diameter of 22-26 nm, which is identical to natural virions. VLPs could induce high levels of specific antibodies to CPV and CDV, shown by the indication of neutralizing antibodies in both VP2N and VP2L VLPs group. In addition, splenic lymphocytes of mice immunized with VLPs could proliferate rapidly after stimulation by specific antigen. Taken together, the CPV VP2 VLPs or chimeric VLPs are highly immunogenic, and henceforth could function as CPV/CDV vaccine candidates for giant pandas.
Collapse
Affiliation(s)
- Shan Zhao
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Diseases and Human Health of Sichuan Province, Chengdu, China
| | - Xinfeng Han
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Diseases and Human Health of Sichuan Province, Chengdu, China
| | - Yifei Lang
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Diseases and Human Health of Sichuan Province, Chengdu, China
| | - Yue Xie
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Diseases and Human Health of Sichuan Province, Chengdu, China
| | - Zhijie Yang
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Qin Zhao
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Diseases and Human Health of Sichuan Province, Chengdu, China
| | - Yiping Wen
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Diseases and Human Health of Sichuan Province, Chengdu, China
| | - Jing Xia
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Diseases and Human Health of Sichuan Province, Chengdu, China
| | - Rui Wu
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Diseases and Human Health of Sichuan Province, Chengdu, China
| | - Xiaobo Huang
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Diseases and Human Health of Sichuan Province, Chengdu, China
| | - Yong Huang
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Diseases and Human Health of Sichuan Province, Chengdu, China
| | - Sanjie Cao
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Diseases and Human Health of Sichuan Province, Chengdu, China
| | - Jingchao Lan
- Chengdu Research Base of Giant Panda Breeding, Chengdu, China
| | - Li Luo
- Chengdu Research Base of Giant Panda Breeding, Chengdu, China
| | - Qigui Yan
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Diseases and Human Health of Sichuan Province, Chengdu, China
| |
Collapse
|
72
|
Zhang L, Xu W, Ma X, Sun X, Fan J, Wang Y. Virus-like Particles as Antiviral Vaccine: Mechanism, Design, and Application. BIOTECHNOL BIOPROC E 2023; 28:1-16. [PMID: 36627930 PMCID: PMC9817464 DOI: 10.1007/s12257-022-0107-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 05/17/2022] [Accepted: 05/18/2022] [Indexed: 01/09/2023]
Abstract
Virus-like particles (VLPs) are viral structural protein that are noninfectious as they do not contain viral genetic materials. They are safe and effective immune stimulators and play important roles in vaccine development because of their intrinsic immunogenicity to induce cellular and humoral immune responses. In the design of antiviral vaccine, VLPs based vaccines are appealing multifunctional candidates with the advantages such as self-assembling nanoscaled structures, repetitive surface epitopes, ease of genetic and chemical modifications, versatility as antigen presenting platforms, intrinsic immunogenicity, higher safety profile in comparison with live-attenuated vaccines and inactivated vaccines. In this review, we discuss the mechanism of VLPs vaccine inducing cellular and humoral immune responses. We outline the impact of size, shape, surface charge, antigen presentation, genetic and chemical modification, and expression systems when constructing effective VLPs based vaccines. Recent applications of antiviral VLPs vaccines and their clinical trials are summarized.
Collapse
Affiliation(s)
- Lei Zhang
- Xi'an Key Laboratory of Pathogenic Microorganism and Tumor Immunity, Department of Basic Medicine, Xi'an Medical University, Xi'an, 710021, Shaanxi China
| | - Wen Xu
- Xi'an Key Laboratory of Pathogenic Microorganism and Tumor Immunity, Department of Basic Medicine, Xi'an Medical University, Xi'an, 710021, Shaanxi China
| | - Xi Ma
- Xi'an Key Laboratory of Pathogenic Microorganism and Tumor Immunity, Department of Basic Medicine, Xi'an Medical University, Xi'an, 710021, Shaanxi China
| | - XiaoJing Sun
- Xi'an Key Laboratory of Pathogenic Microorganism and Tumor Immunity, Department of Basic Medicine, Xi'an Medical University, Xi'an, 710021, Shaanxi China
| | - JinBo Fan
- Xi'an Key Laboratory of Pathogenic Microorganism and Tumor Immunity, Department of Basic Medicine, Xi'an Medical University, Xi'an, 710021, Shaanxi China
| | - Yang Wang
- Xi'an Key Laboratory of Pathogenic Microorganism and Tumor Immunity, Department of Basic Medicine, Xi'an Medical University, Xi'an, 710021, Shaanxi China
| |
Collapse
|
73
|
Singh VA, Kumar CS, Khare B, Kuhn RJ, Banerjee M, Tomar S. Surface decorated reporter-tagged chikungunya virus-like particles for clinical diagnostics and identification of virus entry inhibitors. Virology 2023; 578:92-102. [PMID: 36473281 DOI: 10.1016/j.virol.2022.11.012] [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: 10/29/2022] [Revised: 11/20/2022] [Accepted: 11/22/2022] [Indexed: 11/29/2022]
Abstract
The ever-evolving and versatile VLP technology is becoming an increasingly popular area of science. This study presents surface decorated reporter-tagged VLPs of CHIKV, an enveloped RNA virus of the genus alphavirus and its applications. Western blot, IFA and live-cell imaging confirm the expression of reporter-tagged CHIK-VLPs from transfected HEK293Ts. CryoEM micrographs reveal particle diameter as ∼67nm and 56-70 nm, respectively, for NLuc CHIK-VLPs and mCherry CHIK-VLPs. Our study demonstrates that by exploiting NLuc CHIK-VLPs as a detector probe, robust ratiometric luminescence signal in CHIKV-positive sera compared to healthy controls can be achieved swiftly. Moreover, the potential activity of the Suramin drug as a CHIKV entry inhibitor has been validated through the reporter-tagged CHIK-VLPs. The results reported in this study open new avenues in the eVLPs domain and offer potential for large-scale screening of clinical samples and antiviral agents targeting entry of CHIKV and other alphaviruses.
Collapse
Affiliation(s)
- Vedita Anand Singh
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, 247667, Uttarakhand, India; Department of Biological Sciences, Purdue University, West Lafayette, IN, 47907, USA
| | - Chandra Shekhar Kumar
- Kusuma School of Biological Sciences, Indian Institute of Technology - Delhi, Hauz Khas, New Delhi, 110016, India
| | - Baldeep Khare
- Department of Biological Sciences, Purdue University, West Lafayette, IN, 47907, USA
| | - Richard J Kuhn
- Department of Biological Sciences, Purdue University, West Lafayette, IN, 47907, USA
| | - Manidipa Banerjee
- Kusuma School of Biological Sciences, Indian Institute of Technology - Delhi, Hauz Khas, New Delhi, 110016, India
| | - Shailly Tomar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, 247667, Uttarakhand, India.
| |
Collapse
|
74
|
High-Yield Production of Chimeric Hepatitis E Virus-Like Particles Bearing the M2e Influenza Epitope and Receptor Binding Domain of SARS-CoV-2 in Plants Using Viral Vectors. Int J Mol Sci 2022; 23:ijms232415684. [PMID: 36555326 PMCID: PMC9779006 DOI: 10.3390/ijms232415684] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 12/02/2022] [Accepted: 12/08/2022] [Indexed: 12/14/2022] Open
Abstract
Capsid protein of Hepatitis E virus (HEV) is capable of self-assembly into virus-like particles (VLPs) when expressed in Nicotiana benthamiana plants. Such VLPs could be used as carriers of antigens for vaccine development. In this study, we obtained VLPs based on truncated coat protein of HEV bearing the M2e peptide of Influenza A virus or receptor-binding domain of SARS-CoV-2 spike glycoprotein (RBD). We optimized the immunogenic epitopes' presentation by inserting them into the protruding domain of HEV ORF2 at position Tyr485. The fusion proteins were expressed in Nicotiana benthamiana plants using self-replicating potato virus X (PVX)-based vector. The fusion protein HEV/M2, targeted to the cytosol, was expressed at the level of about 300-400 μg per gram of fresh leaf tissue and appeared to be soluble. The fusion protein was purified using metal affinity chromatography under native conditions with the final yield about 200 μg per gram of fresh leaf tissue. The fusion protein HEV/RBD, targeted to the endoplasmic reticulum, was expressed at about 80-100 μg per gram of fresh leaf tissue; the yield after purification was up to 20 μg per gram of fresh leaf tissue. The recombinant proteins HEV/M2 and HEV/RBD formed nanosized virus-like particles that could be recognized by antibodies against inserted epitopes. The ELISA assay showed that antibodies of COVID-19 patients can bind plant-produced HEV/RBD virus-like particles. This study shows that HEV capsid protein is a promising carrier for presentation of foreign antigen.
Collapse
|
75
|
Castro KM, Scheck A, Xiao S, Correia BE. Computational design of vaccine immunogens. Curr Opin Biotechnol 2022; 78:102821. [PMID: 36279815 DOI: 10.1016/j.copbio.2022.102821] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 09/13/2022] [Accepted: 09/19/2022] [Indexed: 12/14/2022]
Abstract
Computational protein engineering has enabled the rational design of customized proteins, which has propelled both sequence-based and structure-based immunogen engineering and delivery. By discerning antigenic determinants of viral pathogens, computational methods have been implemented to successfully engineer representative viral strains able to elicit broadly neutralizing responses or present antigenic sites of viruses for focused immune responses. Combined with improvements in customizable nanoparticle design, immunogens are multivalently displayed to enhance immune responses. These rationally designed immunogens offer unique and powerful approaches to engineer vaccines for pathogens, which have eluded traditional approaches.
Collapse
Affiliation(s)
- Karla M Castro
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland; Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland
| | - Andreas Scheck
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland; Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland
| | - Shuhao Xiao
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland; Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland
| | - Bruno E Correia
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland; Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland.
| |
Collapse
|
76
|
Hansoongnern P, Phecharat N, Wasanasuk K, Tommeurd W, Chankeeree P, Lekcharoensuk C, Semkum P, Pinitkiatisakul S, Lekcharoensuk P. Encapsidated-CpG ODN enhances immunogenicity of porcine circovirus type 2 virus-like particles. Vet Microbiol 2022; 275:109583. [DOI: 10.1016/j.vetmic.2022.109583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 10/14/2022] [Accepted: 10/15/2022] [Indexed: 11/27/2022]
|
77
|
Babaeimarzangou SS, Zaker H, Soleimannezhadbari E, Gamchi NS, Kazeminia M, Tarighi S, Seyedian H, Tsatsakis A, Spandidos DA, Margina D. Vaccine development for zoonotic viral diseases caused by positive‑sense single‑stranded RNA viruses belonging to the Coronaviridae and Togaviridae families (Review). Exp Ther Med 2022; 25:42. [PMID: 36569444 PMCID: PMC9768462 DOI: 10.3892/etm.2022.11741] [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: 08/30/2022] [Accepted: 11/10/2022] [Indexed: 12/02/2022] Open
Abstract
Outbreaks of zoonotic viral diseases pose a severe threat to public health and economies worldwide, with this currently being more prominent than it previously was human history. These emergency zoonotic diseases that originated and transmitted from vertebrates to humans have been estimated to account for approximately one billion cases of illness and have caused millions of deaths worldwide annually. The recent emergence of severe acute respiratory syndrome coronavirus-2 (coronavirus disease 2019) is an excellent example of the unpredictable public health threat causing a pandemic. The present review summarizes the literature data regarding the main vaccine developments in human clinical phase I, II and III trials against the zoonotic positive-sense single-stranded RNA viruses belonging to the Coronavirus and Alphavirus genera, including severe acute respiratory syndrome, Middle east respiratory syndrome, Venezuelan equine encephalitis virus, Semliki Forest virus, Ross River virus, Chikungunya virus and O'nyong-nyong virus. That there are neither vaccines nor effective antiviral drugs available against most of these viruses is undeniable. Therefore, new explosive outbreaks of these zoonotic viruses may surely be expected. The present comprehensive review provides an update on the status of vaccine development in different clinical trials against these viruses, as well as an overview of the present results of these trials.
Collapse
Affiliation(s)
- Seyed Sajjad Babaeimarzangou
- Division of Poultry Health and Diseases, Department of Clinical Sciences, Faculty of Veterinary Medicine, Urmia University, Urmia 5756151818, Iran
| | - Himasadat Zaker
- Histology and Microscopic Analysis Division, RASTA Specialized Research Institute (RSRI), West Azerbaijan Science and Technology Park (WASTP), Urmia 5756115322, Iran
| | | | - Naeimeh Shamsi Gamchi
- Histology and Microscopic Analysis Division, RASTA Specialized Research Institute (RSRI), West Azerbaijan Science and Technology Park (WASTP), Urmia 5756115322, Iran
| | - Masoud Kazeminia
- Department of Food Hygiene and Quality Control, Faculty of Veterinary Medicine, University of Tehran, Tehran 1417935840, Iran
| | - Shima Tarighi
- Veterinary Office of West Azerbaijan Province, Urmia 5717617695, Iran
| | - Homayon Seyedian
- Faculty of Veterinary Medicine, Urmia University, Urmia 5756151818, Iran
| | - Aristidis Tsatsakis
- Laboratory of Toxicology, Department of Medicine, University of Crete, 71307 Heraklion, Greece,Correspondence to: Professor Denisa Margina, Department of Biochemistry, Faculty of Pharmacy, ‘Carol Davila’ University of Medicine and Pharmacy, 6 Traian Vuia Street, 020956 Bucharest, Romania
| | - Demetrios A. Spandidos
- Laboratory of Clinical Virology, School of Medicine, University of Crete, 71003 Heraklion, Greece
| | - Denisa Margina
- Department of Biochemistry, Faculty of Pharmacy, ‘Carol Davila’ University of Medicine and Pharmacy, 020956 Bucharest, Romania,Correspondence to: Professor Denisa Margina, Department of Biochemistry, Faculty of Pharmacy, ‘Carol Davila’ University of Medicine and Pharmacy, 6 Traian Vuia Street, 020956 Bucharest, Romania
| |
Collapse
|
78
|
Olshefsky A, Richardson C, Pun SH, King NP. Engineering Self-Assembling Protein Nanoparticles for Therapeutic Delivery. Bioconjug Chem 2022; 33:2018-2034. [PMID: 35487503 PMCID: PMC9673152 DOI: 10.1021/acs.bioconjchem.2c00030] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Despite remarkable advances over the past several decades, many therapeutic nanomaterials fail to overcome major in vivo delivery barriers. Controlling immunogenicity, optimizing biodistribution, and engineering environmental responsiveness are key outstanding delivery problems for most nanotherapeutics. However, notable exceptions exist including some lipid and polymeric nanoparticles, some virus-based nanoparticles, and nanoparticle vaccines where immunogenicity is desired. Self-assembling protein nanoparticles offer a powerful blend of modularity and precise designability to the field, and have the potential to solve many of the major barriers to delivery. In this review, we provide a brief overview of key designable features of protein nanoparticles and their implications for therapeutic delivery applications. We anticipate that protein nanoparticles will rapidly grow in their prevalence and impact as clinically relevant delivery platforms.
Collapse
Affiliation(s)
- Audrey Olshefsky
- Department
of Bioengineering, University of Washington, Seattle, Washington 98195, United States
- Institute
for Protein Design, University of Washington, Seattle, Washington 98195, United States
| | - Christian Richardson
- Department
of Bioengineering, University of Washington, Seattle, Washington 98195, United States
- Institute
for Protein Design, University of Washington, Seattle, Washington 98195, United States
| | - Suzie H. Pun
- Department
of Bioengineering, University of Washington, Seattle, Washington 98195, United States
- Molecular
Engineering and Sciences Institute, University
of Washington, Seattle, Washington 98195, United States
| | - Neil P. King
- Institute
for Protein Design, University of Washington, Seattle, Washington 98195, United States
- Department
of Biochemistry, University of Washington, Seattle, Washington 98195, United States
| |
Collapse
|
79
|
Rabies Vaccine: Recent Update and Comprehensive Review of in vitro and in vivo Studies. Process Biochem 2022. [DOI: 10.1016/j.procbio.2022.11.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
80
|
Hu K, Palmieri E, Samnuan K, Ricchetti B, Oldrini D, McKay PF, Wu G, Thorne L, Fooks AR, McElhinney LM, Goharriz H, Golding M, Shattock RJ, Micoli F. Generalized Modules for Membrane Antigens (GMMA), an outer membrane vesicle-based vaccine platform, for efficient viral antigen delivery. J Extracell Vesicles 2022; 11:e12247. [PMID: 36377074 PMCID: PMC9663859 DOI: 10.1002/jev2.12247] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 04/19/2022] [Accepted: 04/30/2022] [Indexed: 11/17/2022] Open
Abstract
Vaccine platforms enable fast development, testing, and manufacture of more affordable vaccines. Here, we evaluated Generalized Modules for Membrane Antigens (GMMA), outer membrane vesicles (OMVs) generated by genetically modified Gram-negative bacteria, as a vaccine platform for viral pathogens. Influenza A virus hemagglutinin (HA), either physically mixed with GMMA (HA+STmGMMA mix), or covalently linked to GMMA surface (HA-STmGMMA conjugate), significantly increased antigen-specific humoral and cellular responses, with HA-STmGMMA conjugate inducing further enhancement than HA+STmGMMA mix. HA-STmGMMA conjugate protected mice from lethal challenge. The versatility for this platform was confirmed by conjugation of rabies glycoprotein (RABVG) onto GMMA through the same method. RABVG+STmGMMA mix and RABVG-STmGMMA conjugate exhibited similar humoral and cellular response patterns and protection efficacy as the HA formulations, indicating relatively consistent responses for different vaccines based on the GMMA platform. Comparing to soluble protein, GMMA was more efficiently taken up in vivo and exhibited a B-cell preferential uptake in the draining lymph nodes (LNs). Together, GMMA enhances immunity against viral antigens, and the platform works well with different antigens while retaining similar immunomodulatory patterns. The findings of our study imply the great potential of GMMA-based vaccine platform also against viral infectious diseases.
Collapse
Affiliation(s)
- Kai Hu
- Department of Infectious Diseases, Imperial College London, London, UK
| | - Elena Palmieri
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., Siena, Italy
| | - Karnyart Samnuan
- Department of Infectious Diseases, Imperial College London, London, UK
| | | | - Davide Oldrini
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., Siena, Italy
| | - Paul F McKay
- Department of Infectious Diseases, Imperial College London, London, UK
| | - Guanghui Wu
- Animal and Plant Health Agency (APHA), OIE Rabies Reference Laboratory, New Haw, Addlestone, Surrey, UK
| | - Leigh Thorne
- Animal and Plant Health Agency (APHA), OIE Rabies Reference Laboratory, New Haw, Addlestone, Surrey, UK
| | - Anthony R Fooks
- Animal and Plant Health Agency (APHA), OIE Rabies Reference Laboratory, New Haw, Addlestone, Surrey, UK
| | - Lorraine M McElhinney
- Animal and Plant Health Agency (APHA), OIE Rabies Reference Laboratory, New Haw, Addlestone, Surrey, UK
| | - Hooman Goharriz
- Animal and Plant Health Agency (APHA), OIE Rabies Reference Laboratory, New Haw, Addlestone, Surrey, UK
| | - Megan Golding
- Animal and Plant Health Agency (APHA), OIE Rabies Reference Laboratory, New Haw, Addlestone, Surrey, UK
| | - Robin J Shattock
- Department of Infectious Diseases, Imperial College London, London, UK
| | - Francesca Micoli
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., Siena, Italy
| |
Collapse
|
81
|
Lu Z, Yu S, Wang W, Chen W, Wang X, Wu K, Li X, Fan S, Ding H, Yi L, Chen J. Development of Foot-and-Mouth Disease Vaccines in Recent Years. Vaccines (Basel) 2022; 10:1817. [PMID: 36366327 PMCID: PMC9693445 DOI: 10.3390/vaccines10111817] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 10/24/2022] [Accepted: 10/26/2022] [Indexed: 12/20/2023] Open
Abstract
Foot-and-mouth disease (FMD) is a serious disease affecting the global graziery industry. Once an epidemic occurs, it can lead to economic and trade stagnation. In recent decades, FMD has been effectively controlled and even successfully eradicated in some countries or regions through mandatory vaccination with inactivated foot-and-mouth disease vaccines. Nevertheless, FMD still occurs in some parts of Africa and Asia. The transmission efficiency of foot-and-mouth disease is high. Both disease countries and disease-free countries should always be prepared to deal with outbreaks of FMD. The development of vaccines has played a key role in this regard. This paper summarizes the development of several promising vaccines including progress and design ideas. It also provides ways to develop a new generation of vaccines for FMDV and other major diseases.
Collapse
Affiliation(s)
- Zhimin Lu
- College of Veterinary Medicine, South China Agricultural University, No. 483 Wushan Road, Tianhe District, Guangzhou 510642, China
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
| | - Shu Yu
- College of Veterinary Medicine, South China Agricultural University, No. 483 Wushan Road, Tianhe District, Guangzhou 510642, China
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
| | - Weijun Wang
- College of Veterinary Medicine, South China Agricultural University, No. 483 Wushan Road, Tianhe District, Guangzhou 510642, China
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
| | - Wenxian Chen
- College of Veterinary Medicine, South China Agricultural University, No. 483 Wushan Road, Tianhe District, Guangzhou 510642, China
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
| | - Xinyan Wang
- College of Veterinary Medicine, South China Agricultural University, No. 483 Wushan Road, Tianhe District, Guangzhou 510642, China
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
| | - Keke Wu
- College of Veterinary Medicine, South China Agricultural University, No. 483 Wushan Road, Tianhe District, Guangzhou 510642, China
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
| | - Xiaowen Li
- College of Veterinary Medicine, South China Agricultural University, No. 483 Wushan Road, Tianhe District, Guangzhou 510642, China
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
| | - Shuangqi Fan
- College of Veterinary Medicine, South China Agricultural University, No. 483 Wushan Road, Tianhe District, Guangzhou 510642, China
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
| | - Hongxing Ding
- College of Veterinary Medicine, South China Agricultural University, No. 483 Wushan Road, Tianhe District, Guangzhou 510642, China
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
| | - Lin Yi
- College of Veterinary Medicine, South China Agricultural University, No. 483 Wushan Road, Tianhe District, Guangzhou 510642, China
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
| | - Jingding Chen
- College of Veterinary Medicine, South China Agricultural University, No. 483 Wushan Road, Tianhe District, Guangzhou 510642, China
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
| |
Collapse
|
82
|
Transient Expression of Flavivirus Structural Proteins in Nicotiana benthamiana. Vaccines (Basel) 2022; 10:vaccines10101667. [PMID: 36298532 PMCID: PMC9610170 DOI: 10.3390/vaccines10101667] [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: 08/29/2022] [Revised: 09/27/2022] [Accepted: 10/04/2022] [Indexed: 11/05/2022] Open
Abstract
Flaviviruses are a threat to public health and can cause major disease outbreaks. Tick-borne encephalitis (TBE) is caused by a flavivirus, and it is one of the most important causes of viral encephalitis in Europe and is on the rise in Sweden. As there is no antiviral treatment available, vaccination remains the best protective measure against TBE. Currently available TBE vaccines are based on formalin-inactivated virus produced in cell culture. These vaccines must be delivered by intramuscular injection, have a burdensome immunization schedule, and may exhibit vaccine failure in certain populations. This project aimed to develop an edible TBE vaccine to trigger a stronger immune response through oral delivery of viral antigens to mucosal surfaces. We demonstrated successful expression and post-translational processing of flavivirus structural proteins which then self-assembled to form virus-like particles in Nicotiana benthamiana. We performed oral toxicity tests in mice using various plant species as potential bioreactors and evaluated the immunogenicity of the resulting edible vaccine candidate. Mice immunized with the edible vaccine candidate did not survive challenge with TBE virus. Interestingly, immunization of female mice with a commercial TBE vaccine can protect their offspring against TBE virus infection.
Collapse
|
83
|
Gao X, Feng Q, Wang J, Zhao X. Bacterial outer membrane vesicle-based cancer nanovaccines. Cancer Biol Med 2022; 19:j.issn.2095-3941.2022.0452. [PMID: 36172794 PMCID: PMC9500226 DOI: 10.20892/j.issn.2095-3941.2022.0452] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 08/24/2022] [Indexed: 11/05/2022] Open
Abstract
Tumor vaccines, a type of personalized tumor immunotherapy, have developed rapidly in recent decades. These vaccines evoke tumor antigen-specific T cells to achieve immune recognition and killing of tumor cells. Because the immunogenicity of tumor antigens alone is insufficient, immune adjuvants and nanocarriers are often required to enhance anti-tumor immune responses. At present, vaccine carrier development often integrates nanocarriers and immune adjuvants. Among them, outer membrane vesicles (OMVs) are receiving increasing attention as a delivery platform for tumor vaccines. OMVs are natural nanovesicles derived from Gram-negative bacteria, which have adjuvant function because they contain pathogen associated molecular patterns. Importantly, OMVs can be functionally modified by genetic engineering of bacteria, thus laying a foundation for applications as a delivery platform for tumor nanovaccines. This review summarizes 5 aspects of recent progress in, and future development of, OMV-based tumor nanovaccines: strain selection, heterogeneity, tumor antigen loading, immunogenicity and safety, and mass production of OMVs.
Collapse
Affiliation(s)
- Xiaoyu Gao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qingqing Feng
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China
| | - Jing Wang
- Center of Drug Evaluation, National Medical Products Administration, Beijing 100022, China
| | - Xiao Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- IGDB-NCNST Joint Research Center, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| |
Collapse
|
84
|
Chu KB, Lee SH, Kim MJ, Kim AR, Moon EK, Quan FS. Virus-like particles coexpressing the PreF and Gt antigens of respiratory syncytial virus confer protection in mice. Nanomedicine (Lond) 2022; 17:1159-1171. [DOI: 10.2217/nnm-2022-0082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aims: The purpose of this study was to assess the protective efficacy of virus-like particles (VLPs) co-expressing the pre-fusogenic (PreF) and G protein with tandem repeats (Gt) antigens of respiratory syncytial virus (RSV) in mice. Materials & methods: VLP constructs expressing PreF, Gt or both were used to immunize mice, and the protective efficacies were evaluated using antibody responses, neutralizing antibody titers, T-cell responses, histopathological assessment and plaque assay. Results: PreF+Gt VLP immunization elicited strong RSV-specific antibody responses and pulmonary T-cell responses that contributed to lessening virus titer and inflammation. Conclusion: Our findings suggest that coexpressing PreF and Gt antigens elicits better protection than either one alone. This combinatorial approach could assist in future RSV vaccine development.
Collapse
Affiliation(s)
- Ki-Back Chu
- Medical Research Center for Bioreaction to Reactive Oxygen Species & Biomedical Science Institute, School of Medicine, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Su-Hwa Lee
- Department of Medical Zoology, School of Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Min-Ju Kim
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Ah-Ra Kim
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Eun-Kyung Moon
- Department of Medical Zoology, School of Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Fu-Shi Quan
- Medical Research Center for Bioreaction to Reactive Oxygen Species & Biomedical Science Institute, School of Medicine, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea
- Department of Medical Zoology, School of Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
| |
Collapse
|
85
|
Morales-Hernández S, Ugidos-Damboriena N, López-Sagaseta J. Self-Assembling Protein Nanoparticles in the Design of Vaccines: 2022 Update. Vaccines (Basel) 2022; 10:1447. [PMID: 36146525 PMCID: PMC9505534 DOI: 10.3390/vaccines10091447] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 08/25/2022] [Accepted: 08/26/2022] [Indexed: 11/16/2022] Open
Abstract
Vaccines constitute a pillar in the prevention of infectious diseases. The unprecedented emergence of novel immunization strategies due to the COVID-19 pandemic has again positioned vaccination as a pivotal measure to protect humankind and reduce the clinical impact and socioeconomic burden worldwide. Vaccination pursues the ultimate goal of eliciting a protective response in immunized individuals. To achieve this, immunogens must be efficiently delivered to prime the immune system and produce robust protection. Given their safety, immunogenicity, and flexibility to display varied and native epitopes, self-assembling protein nanoparticles represent one of the most promising immunogen delivery platforms. Currently marketed vaccines against the human papillomavirus, for instance, illustrate the potential of these nanoassemblies. This review is intended to provide novelties, since 2015, on the ground of vaccine design and self-assembling protein nanoparticles, as well as a comparison with the current emergence of mRNA-based vaccines.
Collapse
Affiliation(s)
- Sergio Morales-Hernández
- Unit of Protein Crystallography and Structural Immunology, Navarrabiomed-Public University of Navarra (UPNA), 31008 Pamplona, Spain
- Navarra University Hospital, 31008 Pamplona, Spain
| | - Nerea Ugidos-Damboriena
- Unit of Protein Crystallography and Structural Immunology, Navarrabiomed-Public University of Navarra (UPNA), 31008 Pamplona, Spain
- Navarra University Hospital, 31008 Pamplona, Spain
| | - Jacinto López-Sagaseta
- Unit of Protein Crystallography and Structural Immunology, Navarrabiomed-Public University of Navarra (UPNA), 31008 Pamplona, Spain
- Navarra University Hospital, 31008 Pamplona, Spain
| |
Collapse
|
86
|
Park JE, Kim JH, Park JY, Jun SH, Shin HJ. A chimeric MERS-CoV virus-like particle vaccine protects mice against MERS-CoV challenge. Virol J 2022; 19:112. [PMID: 35761402 PMCID: PMC9235161 DOI: 10.1186/s12985-022-01844-9] [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: 02/24/2022] [Accepted: 06/20/2022] [Indexed: 11/30/2022] Open
Abstract
Background Middle East respiratory syndrome coronavirus (MERS-CoV) causes severe respiratory disease in humans, with a case fatality rate of approximately 35%, thus posing a considerable threat to public health. The lack of approved vaccines or antivirals currently constitutes a barrier in controlling disease outbreaks and spread. Methods In this study, using a mammalian expression system, which is advantageous for maintaining correct protein glycosylation patterns, we constructed chimeric MERS-CoV virus-like particles (VLPs) and determined their immunogenicity and protective efficacy in mice. Results Western blot and cryo-electron microscopy analyses demonstrated that MERS-CoV VLPs were efficiently produced in cells co-transfected with MERS-CoV spike (S), envelope, membrane and murine hepatitis virus nucleocapsid genes. We examined their ability as a vaccine in a human dipeptidyl peptidase 4 knock-in C57BL/6 congenic mouse model. Mice immunized with MERS VLPs produced S-specific antibodies with virus neutralization activity. Furthermore, MERS-CoV VLP immunization provided complete protection against a lethal challenge with mouse-adapted MERS-CoV and improved virus clearance in the lung. Conclusions Overall, these data demonstrate that MERS-CoV VLPs have excellent immunogenicity and represent a promising vaccine candidate.
Collapse
Affiliation(s)
- Jung-Eun Park
- Laboratory of Veterinary Public Health, College of Veterinary Medicine, Chungnam National University, Daejeon, 34134, Republic of Korea. .,Research Institute of Veterinary Research, Chungnam National University, Daejeon, 34134, Republic of Korea.
| | - Ji-Hee Kim
- Laboratory of Veterinary Public Health, College of Veterinary Medicine, Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Jae-Yeon Park
- Laboratory of Veterinary Infectious Diseases, College of Veterinary Medicine, Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Sung-Hoon Jun
- Electron Microscopy & Spectroscopy Team, Korea Basic Science Institute, Cheongju, Chungcheongbukdo, 28119, Republic of Korea
| | - Hyun-Jin Shin
- Laboratory of Veterinary Infectious Diseases, College of Veterinary Medicine, Chungnam National University, Daejeon, 34134, Republic of Korea. .,Research Institute of Veterinary Research, Chungnam National University, Daejeon, 34134, Republic of Korea.
| |
Collapse
|
87
|
Facciolà A, Visalli G, Laganà A, Di Pietro A. An Overview of Vaccine Adjuvants: Current Evidence and Future Perspectives. Vaccines (Basel) 2022; 10:vaccines10050819. [PMID: 35632575 PMCID: PMC9147349 DOI: 10.3390/vaccines10050819] [Citation(s) in RCA: 115] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 05/18/2022] [Accepted: 05/20/2022] [Indexed: 01/27/2023] Open
Abstract
Vaccinations are one of the most important preventive tools against infectious diseases. Over time, many different types of vaccines have been developed concerning the antigen component. Adjuvants are essential elements that increase the efficacy of vaccination practises through many different actions, especially acting as carriers, depots, and stimulators of immune responses. For many years, few adjuvants have been included in vaccines, with aluminium salts being the most commonly used adjuvant. However, recent research has focused its attention on many different new compounds with effective adjuvant properties and improved safety. Modern technologies such as nanotechnologies and molecular biology have forcefully entered the production processes of both antigen and adjuvant components, thereby improving vaccine efficacy. Microparticles, emulsions, and immune stimulators are currently in the spotlight for their huge potential in vaccine production. Although studies have reported some potential side effects of vaccine adjuvants such as the recently recognised ASIA syndrome, the huge worth of vaccines remains unquestionable. Indeed, the recent COVID-19 pandemic has highlighted the importance of vaccines, especially in regard to managing future potential pandemics. In this field, research into adjuvants could play a leading role in the production of increasingly effective vaccines.
Collapse
Affiliation(s)
- Alessio Facciolà
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, 98125 Messina, Italy; (G.V.); (A.L.); (A.D.P.)
- Correspondence:
| | - Giuseppa Visalli
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, 98125 Messina, Italy; (G.V.); (A.L.); (A.D.P.)
| | - Antonio Laganà
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, 98125 Messina, Italy; (G.V.); (A.L.); (A.D.P.)
- Multi-Specialist Clinical Institute for Orthopaedic Trauma Care (COT), 98124 Messina, Italy
| | - Angela Di Pietro
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, 98125 Messina, Italy; (G.V.); (A.L.); (A.D.P.)
| |
Collapse
|
88
|
Zhao Y, Li Z, Voyer J, Li Y, Chen X. Flagellin/Virus-like Particle Hybrid Platform with High Immunogenicity, Safety, and Versatility for Vaccine Development. ACS APPLIED MATERIALS & INTERFACES 2022; 14:21872-21885. [PMID: 35467839 PMCID: PMC9121874 DOI: 10.1021/acsami.2c01028] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 04/13/2022] [Indexed: 05/07/2023]
Abstract
Hepatitis B core (HBc) virus-like particles (VLPs) and flagellin are highly immunogenic and widely explored vaccine delivery platforms. Yet, HBc VLPs mainly allow the insertion of relatively short antigenic epitopes into the immunodominant c/e1 loop without affecting VLP assembly, and flagellin-based vaccines carry the risk of inducing systemic adverse reactions. This study explored a hybrid flagellin/HBc VLP (FH VLP) platform to present heterologous antigens by replacing the surface-exposed D3 domain of flagellin. FH VLPs were prepared by the insertion of flagellin gene into the c/e1 loop of HBc, followed by E. coli expression, purification, and self-assembly into VLPs. Using the ectodomain of influenza matrix protein 2 (M2e) and ovalbumin (OVA) as models, we found that the D3 domain of flagellin could be replaced with four tandem copies of M2e or the cytotoxic T lymphocyte (CTL) epitope of OVA without interfering with the FH VLP assembly, while the insertion of four tandem copies of M2e into the c/e1 loop of HBc disrupted the VLP assembly. FH VLP-based M2e vaccine elicited potent anti-M2e antibody responses and conferred significant protection against multiple influenza A viral strains, while FljB- or HBc-based M2e vaccine failed to elicit significant protection. FH VLP-based OVA peptide vaccine elicited more potent CTL responses and protection against OVA-expressing lymphoma or melanoma challenges than FljB- or HBc-based OVA peptide vaccine. FH VLP-based vaccines showed a good systemic safety, while flagellin-based vaccines significantly increased serum interleukin 6 and tumor necrosis factor α levels and also rectal temperature at increased doses. We further found that the incorporation of a clinical CpG 1018 adjuvant could enhance the efficacy of FH VLP-based vaccines. Our data support FH VLPs to be a highly immunogenic, safe, and versatile platform for vaccine development to elicit potent humoral and cellular immune responses.
Collapse
Affiliation(s)
- Yiwen Zhao
- Biomedical & Pharmaceutical
Sciences, College of Pharmacy, University
of Rhode Island, 7 Greenhouse Road, Avedisian Hall, Room 480, Kingston, Rhode Island 02881, United States
| | - Zhuofan Li
- Biomedical & Pharmaceutical
Sciences, College of Pharmacy, University
of Rhode Island, 7 Greenhouse Road, Avedisian Hall, Room 480, Kingston, Rhode Island 02881, United States
| | - Jewel Voyer
- Biomedical & Pharmaceutical
Sciences, College of Pharmacy, University
of Rhode Island, 7 Greenhouse Road, Avedisian Hall, Room 480, Kingston, Rhode Island 02881, United States
| | - Yibo Li
- Biomedical & Pharmaceutical
Sciences, College of Pharmacy, University
of Rhode Island, 7 Greenhouse Road, Avedisian Hall, Room 480, Kingston, Rhode Island 02881, United States
| | - Xinyuan Chen
- Biomedical & Pharmaceutical
Sciences, College of Pharmacy, University
of Rhode Island, 7 Greenhouse Road, Avedisian Hall, Room 480, Kingston, Rhode Island 02881, United States
| |
Collapse
|
89
|
Li Y, Bao Q, Yang S, Yang M, Mao C. Bionanoparticles in cancer imaging, diagnosis, and treatment. VIEW 2022. [DOI: 10.1002/viw.20200027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Affiliation(s)
- Yan Li
- Institute of Applied Bioresource Research College of Animal Science Zhejiang University Hangzhou Zhejiang China
| | - Qing Bao
- School of Materials Science and Engineering Zhejiang University Hangzhou Zhejiang China
| | - Shuxu Yang
- Department of Neurosurgery Sir Run Run Shaw Hospital School of Medicine Zhejiang University Hangzhou Zhejiang China
| | - Mingying Yang
- Institute of Applied Bioresource Research College of Animal Science Zhejiang University Hangzhou Zhejiang China
| | - Chuanbin Mao
- School of Materials Science and Engineering Zhejiang University Hangzhou Zhejiang China
- Department of Chemistry and Biochemistry Stephenson Life Science Research Center University of Oklahoma Norman Oklahoma USA
| |
Collapse
|
90
|
Lu W, Zhao Z, Huang YW, Wang B. Review: A systematic review of virus-like particles of coronavirus: Assembly, generation, chimerism and their application in basic research and in the clinic. Int J Biol Macromol 2022; 200:487-497. [PMID: 35065135 PMCID: PMC8769907 DOI: 10.1016/j.ijbiomac.2022.01.108] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 01/14/2022] [Accepted: 01/17/2022] [Indexed: 12/27/2022]
Abstract
Virus-like particles (VLPs) are nano-scale particles that are morphologically similar to a live virus but which lack a genetic component. Since the pandemic spread of COVID-19, much focus has been placed on coronavirus (CoV)-related VLPs. CoVs contain four structural proteins, though the minimum requirement for VLP formation differs among virus species. CoV VLPs are commonly produced in mammalian and insect cell systems, sometimes in the form of chimeric VLPs that enable surface display of CoV epitopes. VLPs are an ideal model for virological research and have been applied as vaccines and diagnostic reagents to aid in clinical disease control. This review summarizes and updates the research progress on the characteristics of VLPs from different known CoVs, mainly focusing on assembly, in vitro expression systems for VLP generation, VLP chimerism, protein-based nanoparticles and their applications in basic research and clinical settings, which may aid in development of novel VLP vaccines against emerging coronavirus diseases such as SARS-CoV-2.
Collapse
Affiliation(s)
- Wan Lu
- Department of Veterinary Medicine, Zhejiang University, Hangzhou 310058, China
| | - Zhuangzhuang Zhao
- Department of Veterinary Medicine, Zhejiang University, Hangzhou 310058, China
| | - Yao-Wei Huang
- Department of Veterinary Medicine, Zhejiang University, Hangzhou 310058, China; Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China.
| | - Bin Wang
- Department of Veterinary Medicine, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
91
|
Martín F, Carreño A, Mendoza R, Caruana P, Rodríguez F, Bravo M, Benito A, Ferrer-Miralles N, Céspedes MV, Corchero JL. All-in-one biofabrication and loading of recombinant vaults in human cells. Biofabrication 2022; 14. [PMID: 35203066 DOI: 10.1088/1758-5090/ac584d] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 02/24/2022] [Indexed: 11/12/2022]
Abstract
One of the most promising approaches in the drug delivery field is the use of naturally occurring self-assembling protein nanoparticles, such as virus-like particles, bacterial microcompartments or vault ribonucleoprotein particles as drug delivery systems (DDS). Among them, eukaryotic vaults show a promising future due to their structural features, in vitro stability and non-immunogenicity. Recombinant vaults are routinely produced in insect cells and purified through several ultracentrifugations, both tedious and time-consuming processes. As an alternative, this work proposes a new approach and protocols for the production of recombinant vaults in human cells by transient gene expression of a His-tagged version of the Major Vault Protein (MVP-H6), the development of new affinity-based purification processes for such recombinant vaults, and the all-in-one biofabrication and encapsulation of a cargo recombinant protein within such vaults by their co-expression in human cells. Protocols proposed here allow the easy and straightforward biofabrication and purification of engineered vaults loaded with virtually any INT-tagged cargo protein, in very short times, paving the way to faster and easier engineering and production of better and more efficient DDS.
Collapse
Affiliation(s)
- Fernando Martín
- Universitat Autonoma de Barcelona, Institut de Biotecnologia i de Biomedicina, Campus Universitari Bellaterra, Bellaterra, Bellaterra, Catalunya, 08193, SPAIN
| | - Aida Carreño
- Universitat Autonoma de Barcelona, Institut de Biotecnologia i de Biomedicina, Campus Universitari Bellaterra, Bellaterra, Bellaterra, Catalunya, 08193, SPAIN
| | - Rosa Mendoza
- CIBER-BBN, Institut de Biotecnologia i de Biomedicina, Campus Universitari Bellaterra, Bellaterra, Bellaterra, 08193, SPAIN
| | - Pablo Caruana
- Hospital de la Santa Creu i Sant Pau, Sant Pau Biomedical Research Institute (IIB Sant Pau) Carrer Sant Quintí, 77-79, Barcelona, Catalunya, 08041, SPAIN
| | - Francisco Rodríguez
- Hospital de la Santa Creu i Sant Pau, Sant Pau Biomedical Research Institute (IIB Sant Pau) Carrer Sant Quintí, 77-79 08041. Barcelona, Spain, Barcelona, Catalunya, 08041, SPAIN
| | - Marlon Bravo
- Universitat de Girona, Laboratori Enginyeria Proteines, Dept biologia, Universitat de Girona, Girona, Catalunya, 17003, SPAIN
| | - Antoni Benito
- Universitat de Girona, Facultat de Ciències, Universitat de Girona, Campus de Montilivi, Carrer Maria Aurèlia Capmany, 40,, Girona, Catalunya, 17003, SPAIN
| | - Neus Ferrer-Miralles
- Universitat Autonoma de Barcelona, Institut de Biotecnologia i de Biomedicina, Campus Universitari Bellaterra, Bellaterra, Bellaterra, Catalunya, 08193, SPAIN
| | - Mª Virtudes Céspedes
- Hospital de la Santa Creu i Sant Pau, Sant Pau Biomedical Research Institute (IIB Sant Pau) Carrer Sant Quintí, 77-79, Barcelona, Catalunya, 08041, SPAIN
| | - Jose Luis Corchero
- CIBER-BBN, Institut de Biotecnologia i de Biomedicina, Campus Universitari Bellaterra, Bellaterra, 08193, SPAIN
| |
Collapse
|
92
|
Abstract
This review discusses peptide epitopes used as antigens in the development of vaccines in clinical trials as well as future vaccine candidates. It covers peptides used in potential immunotherapies for infectious diseases including SARS-CoV-2, influenza, hepatitis B and C, HIV, malaria, and others. In addition, peptides for cancer vaccines that target examples of overexpressed proteins are summarized, including human epidermal growth factor receptor 2 (HER-2), mucin 1 (MUC1), folate receptor, and others. The uses of peptides to target cancers caused by infective agents, for example, cervical cancer caused by human papilloma virus (HPV), are also discussed. This review also provides an overview of model peptide epitopes used to stimulate non-specific immune responses, and of self-adjuvanting peptides, as well as the influence of other adjuvants on peptide formulations. As highlighted in this review, several peptide immunotherapies are in advanced clinical trials as vaccines, and there is great potential for future therapies due the specificity of the response that can be achieved using peptide epitopes.
Collapse
Affiliation(s)
- Ian W Hamley
- Department of Chemistry, University of Reading, Whiteknights, Reading RG6 6AD, U.K
| |
Collapse
|
93
|
Firdaus FZ, Skwarczynski M, Toth I. Developments in Vaccine Adjuvants. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2022; 2412:145-178. [PMID: 34918245 DOI: 10.1007/978-1-0716-1892-9_8] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Vaccines, including subunit, recombinant, and conjugate vaccines, require the use of an immunostimulator/adjuvant for maximum efficacy. Adjuvants not only enhance the strength and longevity of immune responses but may also influence the type of response. In this chapter, we review the adjuvants that are available for use in human vaccines, such as alum, MF59, AS03, and AS01. We extensively discuss their composition, characteristics, mechanism of action, and effects on the immune system. Additionally, we summarize recent trends in adjuvant discovery, providing a brief overview of saponins, TLRs agonists, polysaccharides, nanoparticles, cytokines, and mucosal adjuvants.
Collapse
Affiliation(s)
- Farrhana Ziana Firdaus
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, Australia
| | - Mariusz Skwarczynski
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, Australia
| | - Istvan Toth
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, Australia. .,Institute of Molecular Biosciences, The University of Queensland, St Lucia, QLD, Australia. .,School of Pharmacy, The University of Queensland, Woolloongabba, QLD, Australia.
| |
Collapse
|
94
|
Tariq H, Batool S, Asif S, Ali M, Abbasi BH. Virus-Like Particles: Revolutionary Platforms for Developing Vaccines Against Emerging Infectious Diseases. Front Microbiol 2022; 12:790121. [PMID: 35046918 PMCID: PMC8761975 DOI: 10.3389/fmicb.2021.790121] [Citation(s) in RCA: 115] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 12/10/2021] [Indexed: 02/06/2023] Open
Abstract
Virus-like particles (VLPs) are nanostructures that possess diverse applications in therapeutics, immunization, and diagnostics. With the recent advancements in biomedical engineering technologies, commercially available VLP-based vaccines are being extensively used to combat infectious diseases, whereas many more are in different stages of development in clinical studies. Because of their desired characteristics in terms of efficacy, safety, and diversity, VLP-based approaches might become more recurrent in the years to come. However, some production and fabrication challenges must be addressed before VLP-based approaches can be widely used in therapeutics. This review offers insight into the recent VLP-based vaccines development, with an emphasis on their characteristics, expression systems, and potential applicability as ideal candidates to combat emerging virulent pathogens. Finally, the potential of VLP-based vaccine as viable and efficient immunizing agents to induce immunity against virulent infectious agents, including, SARS-CoV-2 and protein nanoparticle-based vaccines has been elaborated. Thus, VLP vaccines may serve as an effective alternative to conventional vaccine strategies in combating emerging infectious diseases.
Collapse
Affiliation(s)
- Hasnat Tariq
- Department of Biotechnology, Quaid-i-Azam University, Islamabad, Pakistan
| | - Sannia Batool
- Department of Biotechnology, Quaid-i-Azam University, Islamabad, Pakistan
| | - Saaim Asif
- Department of Biosciences, COMSATS University, Islamabad, Pakistan
| | - Mohammad Ali
- Center for Biotechnology and Microbiology, University of Swat, Swat, Pakistan
| | | |
Collapse
|
95
|
Aertgeerts K, Ho TT, Yan YG. Optimization of Recombinant GPCR Proteins for Biophysical and Structural Studies Using Virus-like Particles. Methods Mol Biol 2022; 2507:327-336. [PMID: 35773590 DOI: 10.1007/978-1-0716-2368-8_17] [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: 06/15/2023]
Abstract
Overexpression of biologically functional GPCRs and homogeneous purified protein solutions are required to enable structural studies and protein-based biophysical assay development. Iterative and time-consuming optimization cycles of protein engineering, expression, and purification are often needed to achieve the desired protein quantity and quality. Here, we describe the reconstitution of GPCRs in virus-like particles (VLPs) and their use in biophysical assays to characterize protein yield, stability, and small molecule ligand binding. This approach prevents the need for time-consuming detergent solubilization and protein purification during recombinant GPCR protein optimization.
Collapse
Affiliation(s)
| | - Thao T Ho
- Biology Department, Vertex Pharmaceuticals, San Diego, CA, USA
| | - Yingzhou G Yan
- Neuroscience TRC, Bristol Myers Squibb, San Diego, CA, USA
| |
Collapse
|
96
|
Chiozzini C, Ridolfi B, Federico M. Extracellular Vesicles and Their Use as Vehicles of Immunogens. Methods Mol Biol 2022; 2504:177-198. [PMID: 35467287 DOI: 10.1007/978-1-0716-2341-1_13] [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: 06/14/2023]
Abstract
Healthy cells constitutively release lipid bilayered vesicles of different sizes and recognizing different biogenesis, collectively referred to as extracellular vesicles (EVs). EVs can be distinguished in exosomes and microvesicles. Biological and biomedical research on EVs is an emerging field that is rapidly growing. Many EV features including biogenesis, cell uptake, and functions still require unambiguous elucidation. Nevertheless, it has been well established that EVs are involved in communication among cells, tissues, and organs under both healthy and disease conditions by virtue of their ability to deliver macromolecules to target cells. Here, we summarize most recent findings regarding biogenesis, structure, and functions of both exosomes and microvesicles. In addition, the use of EVs as delivery tools to induce CD8+ T cell immunity is addressed compared to current designs exploiting enveloped viral vectors and virus-like particles. Finally, we describe a both safe and original approach conceived for the induction of strong CTL immunity against antigens uploaded in EVs constitutively released by muscle cells.
Collapse
Affiliation(s)
- Chiara Chiozzini
- National Center for Global Health, Istituto Superiore di Sanità (ISS), Rome, Italy.
| | - Barbara Ridolfi
- National Center for Global Health, Istituto Superiore di Sanità (ISS), Rome, Italy
| | - Maurizio Federico
- National Center for Global Health, Istituto Superiore di Sanità (ISS), Rome, Italy
| |
Collapse
|
97
|
Heinimäki S, Lampinen V, Tamminen K, Hankaniemi MM, Malm M, Hytönen VP, Blazevic V. Antigenicity and immunogenicity of HA2 and M2e influenza virus antigens conjugated to norovirus-like, VP1 capsid-based particles by the SpyTag/SpyCatcher technology. Virology 2021; 566:89-97. [PMID: 34894525 DOI: 10.1016/j.virol.2021.12.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 11/30/2021] [Accepted: 12/01/2021] [Indexed: 11/30/2022]
Abstract
Virus-like particles (VLPs) modified through different molecular technologies are employed as delivery vehicles or platforms for heterologous antigen display. We have recently created a norovirus (NoV) VLP platform, where two influenza antigens, the extracellular domain of matrix protein M2 (M2e) or the stem domain of the major envelope glycoprotein hemagglutinin (HA2) are displayed on the surface of the NoV VLPs by SpyTag/SpyCatcher conjugation. To demonstrate the feasibility of the platform to deliver foreign antigens, this study examined potential interference of the conjugation with induction of antibodies against conjugated M2e peptide, HA2, and NoV VLP carrier. High antibody response was induced by HA2 but not M2e decorated VLPs. Furthermore, HA2-elicited antibodies did not neutralize the homologous influenza virus in vitro. Conjugated NoV VLPs retained intact receptor binding capacity and self-immunogenicity. The results demonstrate that NoV VLPs could be simultaneously used as a platform to deliver foreign antigens and a NoV vaccine.
Collapse
Affiliation(s)
- Suvi Heinimäki
- Vaccine Development and Immunology/Vaccine Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.
| | - Vili Lampinen
- Protein Dynamics Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Kirsi Tamminen
- Vaccine Development and Immunology/Vaccine Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Minna M Hankaniemi
- Protein Dynamics Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Maria Malm
- Vaccine Development and Immunology/Vaccine Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Vesa P Hytönen
- Protein Dynamics Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland; Fimlab Laboratories, Tampere, Finland
| | - Vesna Blazevic
- Vaccine Development and Immunology/Vaccine Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| |
Collapse
|
98
|
Yang D, Zhang L, Duan J, Huang Q, Yu Y, Zhou J, Lu H. A Single Vaccination of IBDV Subviral Particles Generated by Kluyveromyces marxianus Efficiently Protects Chickens against Novel Variant and Classical IBDV Strains. Vaccines (Basel) 2021; 9:vaccines9121443. [PMID: 34960188 PMCID: PMC8706917 DOI: 10.3390/vaccines9121443] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 12/03/2021] [Accepted: 12/04/2021] [Indexed: 01/26/2023] Open
Abstract
Infectious bursal disease (IBD), caused by the infectious bursal disease virus (IBDV), is a highly contagious and immunosuppressive disease in chickens worldwide. The novel variant IBDV (nvIBDV) has been emerging in Chinese chicken farms since 2017, but there are no available vaccines that can provide effective protection. Herein, the capsid protein VP2 from nvIBDV strain FJ-18 was expressed in Kluyveromyces marxianus with the aim to produce nvIBDV subviral particles (SVPs). Two recombinant strains constructed for expression of nvIBDV VP2 (nvVP2) and His-tagged VP2 (nvHVP2) formed two types of nvIBDV subviral particles (SVPs), namely nvVP2-SVPs and nvHVP2-SVPs. TEM scans showed that both SVPs were about 25 nm in diameter, but there was a large portion of nvVP2-SVPs showing non-spherical particles. Molecular dynamics simulations indicate that an N-terminal His tag strengthened the interaction of the nvHVP2 monomer and contributed to the assembly of SVPs. Vaccination of chicks with the nvHVP2-SVPs provided 100% protection against novel variant IBDV infection when challenged with the FJ-18 strain, as well as the classical strain BC6/85. By contrast, vaccination with the nvVP2-SVPs only provided 60% protection against their parent FJ-18 strain, suggesting that the stable conformation of subviral particles posed a great impact on their protective efficacy. Our results showed that the nvHVP2-SVPs produced by the recombinant K. marxianus strain is an ideal vaccine candidate for IBDV eradication.
Collapse
Affiliation(s)
- Deqiang Yang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, 2005 Songhu Road, Shanghai 200438, China; (D.Y.); (Q.H.); (Y.Y.)
- Shanghai Engineering Research Center of Industrial Microorganisms, 2005 Songhu Road, Shanghai 200438, China
| | - Lixia Zhang
- Tianjin Ruipu Biotechnology Co. Ltd., Tianjin 300350, China; (L.Z.); (J.D.)
| | - Jinkun Duan
- Tianjin Ruipu Biotechnology Co. Ltd., Tianjin 300350, China; (L.Z.); (J.D.)
| | - Qiang Huang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, 2005 Songhu Road, Shanghai 200438, China; (D.Y.); (Q.H.); (Y.Y.)
- Shanghai Engineering Research Center of Industrial Microorganisms, 2005 Songhu Road, Shanghai 200438, China
| | - Yao Yu
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, 2005 Songhu Road, Shanghai 200438, China; (D.Y.); (Q.H.); (Y.Y.)
- Shanghai Engineering Research Center of Industrial Microorganisms, 2005 Songhu Road, Shanghai 200438, China
| | - Jungang Zhou
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, 2005 Songhu Road, Shanghai 200438, China; (D.Y.); (Q.H.); (Y.Y.)
- Shanghai Engineering Research Center of Industrial Microorganisms, 2005 Songhu Road, Shanghai 200438, China
- Correspondence: (J.Z.); (H.L.); Tel.: +86-021-31246579 (H.L.)
| | - Hong Lu
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, 2005 Songhu Road, Shanghai 200438, China; (D.Y.); (Q.H.); (Y.Y.)
- Shanghai Engineering Research Center of Industrial Microorganisms, 2005 Songhu Road, Shanghai 200438, China
- Shanghai Collaborative Innovation Center for Biomanufacturing (SCICB), East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
- Correspondence: (J.Z.); (H.L.); Tel.: +86-021-31246579 (H.L.)
| |
Collapse
|
99
|
Du X, Tan D, Gong Y, Zhang Y, Han J, Lv W, Xie T, He P, Hou Z, Xu K, Tan J, Zhu B. A new poly(I:C)-decorated PLGA-PEG nanoparticle promotes Mycobacterium tuberculosis fusion protein to induce comprehensive immune responses in mice intranasally. Microb Pathog 2021; 162:105335. [PMID: 34861347 DOI: 10.1016/j.micpath.2021.105335] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 11/03/2021] [Accepted: 11/27/2021] [Indexed: 11/25/2022]
Abstract
Protein-based subunit vaccine against tuberculosis (TB) is regarded as safer but with lower immunogenicity. To investigate effective adjuvant to improve the immunogenicity of TB subunit vaccine, we modified ploy(I:C) onto PLGA-PEG copolymer nanoparticle with polydopamine to produce a new nanoparticle adjuvant named "PLGA-PEG-poly(I:C)" (NP). M. tuberculosis fusion proteins Mtb10.4-HspX and ESAT-6-Rv2626c (M4) were encapsulated in the nanoparticles to produce the NP/M4 subunit vaccine. The PLGA-PEG/M4 nanoparticle was 200.21 ± 1.07 nm in diameter, and the polydispersity index (PDI) was 0.127 ± 0.02. Following modification with poly(I:C) by polydopamine, the NP/M4 was administered to C57BL/6 female mice intranasally and the immune responses were evaluated. The NP/M4 significantly induced antigen-specific CD4+ T cells proliferation, IL-2 and IFN-γ production. In addition, the NP/M4 could promote the production of antigen-specific IgG, IgG1, IgG2c in serum, and sIgA in lung washings. Overall, our results indicated that the NP would be a potential TB subunit vaccine adjuvant with the ability to induce strong Th1-type cell-mediated immunity and humoral immune responses.
Collapse
Affiliation(s)
- Xiufen Du
- Gansu Provincial Key Laboratory of Evidence Based Medicine and Clinical Translation and Lanzhou Center for Tuberculosis Research, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China; Department of Immunology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Daquan Tan
- Gansu Provincial Key Laboratory of Evidence Based Medicine and Clinical Translation and Lanzhou Center for Tuberculosis Research, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China; Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Yang Gong
- Gansu Provincial Key Laboratory of Evidence Based Medicine and Clinical Translation and Lanzhou Center for Tuberculosis Research, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China; Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Yifan Zhang
- Gansu Provincial Key Laboratory of Evidence Based Medicine and Clinical Translation and Lanzhou Center for Tuberculosis Research, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China; Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Jiangyuan Han
- Gansu Provincial Key Laboratory of Evidence Based Medicine and Clinical Translation and Lanzhou Center for Tuberculosis Research, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China; Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Wei Lv
- Gansu Provincial Key Laboratory of Evidence Based Medicine and Clinical Translation and Lanzhou Center for Tuberculosis Research, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China; Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Tao Xie
- Gansu Provincial Key Laboratory of Evidence Based Medicine and Clinical Translation and Lanzhou Center for Tuberculosis Research, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China; Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Pu He
- Gansu Provincial Key Laboratory of Evidence Based Medicine and Clinical Translation and Lanzhou Center for Tuberculosis Research, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China; Department of Immunology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Zongjie Hou
- Department of Immunology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Kun Xu
- Gansu Provincial Key Laboratory of Evidence Based Medicine and Clinical Translation and Lanzhou Center for Tuberculosis Research, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China; Department of Immunology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Jiying Tan
- Gansu Provincial Key Laboratory of Evidence Based Medicine and Clinical Translation and Lanzhou Center for Tuberculosis Research, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China; Department of Immunology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China.
| | - Bingdong Zhu
- Gansu Provincial Key Laboratory of Evidence Based Medicine and Clinical Translation and Lanzhou Center for Tuberculosis Research, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China; Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China.
| |
Collapse
|
100
|
Hemmati F, Hemmati-Dinarvand M, Karimzade M, Rutkowska D, Eskandari MH, Khanizadeh S, Afsharifar A. Plant-derived VLP: a worthy platform to produce vaccine against SARS-CoV-2. Biotechnol Lett 2021; 44:45-57. [PMID: 34837582 PMCID: PMC8626723 DOI: 10.1007/s10529-021-03211-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 11/10/2021] [Indexed: 02/06/2023]
Abstract
After its emergence in late 2019 SARS-CoV-2 was declared a pandemic by the World Health Organization on 11 March 2020 and has claimed more than 2.8 million lives. There has been a massive global effort to develop vaccines against SARS-CoV-2 and the rapid and low cost production of large quantities of vaccine is urgently needed to ensure adequate supply to both developed and developing countries. Virus-like particles (VLPs) are composed of viral antigens that self-assemble into structures that mimic the structure of native viruses but lack the viral genome. Thus they are not only a safer alternative to attenuated or inactivated vaccines but are also able to induce potent cellular and humoral immune responses and can be manufactured recombinantly in expression systems that do not require viral replication. VLPs have successfully been produced in bacteria, yeast, insect and mammalian cell cultures, each production platform with its own advantages and limitations. Plants offer a number of advantages in one production platform, including proper eukaryotic protein modification and assembly, increased safety, low cost, high scalability as well as rapid production speed, a critical factor needed to control outbreaks of potential pandemics. Plant-based VLP-based viral vaccines currently in clinical trials include, amongst others, Hepatitis B virus, Influenza virus and SARS-CoV-2 vaccines. Here we discuss the importance of plants as a next generation expression system for the fast, scalable and low cost production of VLP-based vaccines.
Collapse
Affiliation(s)
- Farshad Hemmati
- Plant Virology Research Center, College of Agriculture, Shiraz University, Shiraz, Iran.
| | - Mohsen Hemmati-Dinarvand
- Department of Clinical Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Marziye Karimzade
- Plant Pathology Department, Faculty of Agriculture, Tarbiat Modares University, Tehran, Iran
| | - Daria Rutkowska
- CSIR Next Generation Health, PO Box 395, Pretoria, 0001, South Africa
| | - Mohammad Hadi Eskandari
- Department of Food Science and Technology, College of Agriculture, Shiraz University, Shiraz, Iran
| | - Sayyad Khanizadeh
- Hepatitis Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Alireza Afsharifar
- Plant Virology Research Center, College of Agriculture, Shiraz University, Shiraz, Iran.
| |
Collapse
|