1
|
Gül A, Erkunt Alak S, Can H, Karakavuk M, Korukluoğlu G, Altaş AB, Gül C, Karakavuk T, Köseoğlu AE, Ülbeği Polat H, Yazıcı Malkoçoğlu H, Taş Ekiz A, Abacı İ, Aksoy Ö, Enül H, Adıay C, Uzar S, Saraç F, Ün C, Gürüz AY, Kantarcı AG, Akbaba H, Erel Akbaba G, Yılmaz H, Değirmenci Döşkaya A, Taşbakan M, Pullukçu H, Karasulu E, Tekin Ş, Döşkaya M. Immunogenicity and protection efficacy of a COVID-19 DNA vaccine encoding spike protein with D614G mutation and optimization of large-scale DNA vaccine production. Sci Rep 2024; 14:13865. [PMID: 38879684 PMCID: PMC11180131 DOI: 10.1038/s41598-024-64690-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 06/12/2024] [Indexed: 06/19/2024] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 had devastating consequences for human health. Despite the introduction of several vaccines, COVID-19 continues to pose a serious health risk due to emerging variants of concern. DNA vaccines gained importance during the pandemic due to their advantages such as induction of both arms of immune response, rapid development, stability, and safety profiles. Here, we report the immunogenicity and protective efficacy of a DNA vaccine encoding spike protein with D614G mutation (named pcoSpikeD614G) and define a large-scale production process. According to the in vitro studies, pcoSpikeD614G expressed abundant spike protein in HEK293T cells. After the administration of pcoSpikeD614G to BALB/c mice through intramuscular (IM) route and intradermal route using an electroporation device (ID + EP), it induced high level of anti-S1 IgG and neutralizing antibodies (P < 0.0001), strong Th1-biased immune response as shown by IgG2a polarization (P < 0.01), increase in IFN-γ levels (P < 0.01), and increment in the ratio of IFN-γ secreting CD4+ (3.78-10.19%) and CD8+ (5.24-12.51%) T cells. Challenging K18-hACE2 transgenic mice showed that pcoSpikeD614G administered through IM and ID + EP routes conferred 90-100% protection and there was no sign of pneumonia. Subsequently, pcoSpikeD614G was evaluated as a promising DNA vaccine candidate and scale-up studies were performed. Accordingly, a large-scale production process was described, including a 36 h fermentation process of E. coli DH5α cells containing pcoSpikeD614G resulting in a wet cell weight of 242 g/L and a three-step chromatography for purification of the pcoSpikeD614G DNA vaccine.
Collapse
MESH Headings
- Vaccines, DNA/immunology
- Vaccines, DNA/administration & dosage
- Vaccines, DNA/genetics
- Spike Glycoprotein, Coronavirus/immunology
- Spike Glycoprotein, Coronavirus/genetics
- Animals
- Humans
- SARS-CoV-2/immunology
- SARS-CoV-2/genetics
- Mice
- COVID-19/prevention & control
- COVID-19/immunology
- HEK293 Cells
- Mice, Inbred BALB C
- COVID-19 Vaccines/immunology
- COVID-19 Vaccines/administration & dosage
- Mutation
- Antibodies, Viral/immunology
- Antibodies, Viral/blood
- Antibodies, Neutralizing/immunology
- Antibodies, Neutralizing/blood
- Female
- Immunogenicity, Vaccine
- Immunoglobulin G/blood
- Immunoglobulin G/immunology
Collapse
Affiliation(s)
- Aytül Gül
- Vaccine Development Application and Research Center, Ege University, 35100, İzmir, Türkiye
- Department of Bioengineering, Faculty of Engineering, Ege University, İzmir, Türkiye
- Department of Bioengineering, Graduate School of Natural and Applied Sciences, Ege University, İzmir, Türkiye
| | - Sedef Erkunt Alak
- Vaccine Development Application and Research Center, Ege University, 35100, İzmir, Türkiye
- Department of Biology, Molecular Biology Section, Faculty of Science, Ege University, İzmir, Türkiye
| | - Hüseyin Can
- Vaccine Development Application and Research Center, Ege University, 35100, İzmir, Türkiye
- Department of Biology, Molecular Biology Section, Faculty of Science, Ege University, İzmir, Türkiye
- Department of Vaccine Studies, Institute of Health Sciences, Ege University, İzmir, Türkiye
| | - Muhammet Karakavuk
- Vaccine Development Application and Research Center, Ege University, 35100, İzmir, Türkiye
- Department of Vaccine Studies, Institute of Health Sciences, Ege University, İzmir, Türkiye
- Ödemiş Vocational School, Ege University, İzmir, Türkiye
| | - Gülay Korukluoğlu
- Republic of Türkiye, General Directorate of Public Health, Ministry of Health, National Virology Reference Central Laboratory, Ankara, Türkiye
- Department of Medical Microbiology, Ankara Bilkent City Hospital, University of Health Sciences, Ankara, Türkiye
| | - Ayşe Başak Altaş
- Republic of Türkiye, General Directorate of Public Health, Ministry of Health, National Virology Reference Central Laboratory, Ankara, Türkiye
| | - Ceren Gül
- Vaccine Development Application and Research Center, Ege University, 35100, İzmir, Türkiye
- Department of Biotechnology, Graduate School of Natural and Applied Sciences, Ege University, İzmir, Türkiye
| | - Tuğba Karakavuk
- Vaccine Development Application and Research Center, Ege University, 35100, İzmir, Türkiye
- Department of Biotechnology, Graduate School of Natural and Applied Sciences, Ege University, İzmir, Türkiye
| | - Ahmet Efe Köseoğlu
- Department of Biology, Molecular Biology Section, Faculty of Science, Ege University, İzmir, Türkiye
- Department of Environmental Microbiology and Biotechnology, Faculty of Chemistry, Duisburg-Essen University, Essen, Germany
| | - Hivda Ülbeği Polat
- TÜBİTAK Marmara Research Center, Vice Presidency of Life Sciences, Kocaeli, Türkiye
| | | | - Arzu Taş Ekiz
- TÜBİTAK Marmara Research Center, Vice Presidency of Life Sciences, Kocaeli, Türkiye
| | - İrem Abacı
- TÜBİTAK Marmara Research Center, Vice Presidency of Life Sciences, Kocaeli, Türkiye
| | - Özge Aksoy
- TÜBİTAK Marmara Research Center, Vice Presidency of Life Sciences, Kocaeli, Türkiye
| | - Hakan Enül
- Pendik Veterinary Control Institute, İstanbul, Türkiye
| | - Cumhur Adıay
- Pendik Veterinary Control Institute, İstanbul, Türkiye
| | - Serdar Uzar
- Pendik Veterinary Control Institute, İstanbul, Türkiye
| | - Fahriye Saraç
- Pendik Veterinary Control Institute, İstanbul, Türkiye
| | - Cemal Ün
- Vaccine Development Application and Research Center, Ege University, 35100, İzmir, Türkiye
- Department of Biology, Molecular Biology Section, Faculty of Science, Ege University, İzmir, Türkiye
- Department of Vaccine Studies, Institute of Health Sciences, Ege University, İzmir, Türkiye
| | - Adnan Yüksel Gürüz
- Vaccine Development Application and Research Center, Ege University, 35100, İzmir, Türkiye
- Department of Vaccine Studies, Institute of Health Sciences, Ege University, İzmir, Türkiye
- Department of Parasitology, Faculty of Medicine, Ege University, İzmir, Türkiye
| | - Ayşe Gülten Kantarcı
- Vaccine Development Application and Research Center, Ege University, 35100, İzmir, Türkiye
- Department of Vaccine Studies, Institute of Health Sciences, Ege University, İzmir, Türkiye
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Ege University, İzmir, Türkiye
| | - Hasan Akbaba
- Department of Vaccine Studies, Institute of Health Sciences, Ege University, İzmir, Türkiye
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Ege University, İzmir, Türkiye
| | - Gülşah Erel Akbaba
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, İzmir Katip Çelebi University, İzmir, Türkiye
| | - Habibe Yılmaz
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Trakya University, Edirne, Türkiye
| | - Aysu Değirmenci Döşkaya
- Vaccine Development Application and Research Center, Ege University, 35100, İzmir, Türkiye
- Department of Vaccine Studies, Institute of Health Sciences, Ege University, İzmir, Türkiye
- Department of Parasitology, Faculty of Medicine, Ege University, İzmir, Türkiye
| | - Meltem Taşbakan
- Department of Vaccine Studies, Institute of Health Sciences, Ege University, İzmir, Türkiye
- Department of Infectious Diseases, Faculty of Medicine, Ege University, İzmir, Türkiye
| | - Hüsnü Pullukçu
- Department of Vaccine Studies, Institute of Health Sciences, Ege University, İzmir, Türkiye
- Department of Infectious Diseases, Faculty of Medicine, Ege University, İzmir, Türkiye
| | - Ercüment Karasulu
- Ege University Research and Application Center of Drug Development and Pharmacokinetics, İzmir, Türkiye
| | - Şaban Tekin
- Department of Basic Medical Sciences, Medical Biology, Faculty of Medicine, University of Health Sciences, İstanbul, Türkiye
| | - Mert Döşkaya
- Vaccine Development Application and Research Center, Ege University, 35100, İzmir, Türkiye.
- Department of Vaccine Studies, Institute of Health Sciences, Ege University, İzmir, Türkiye.
- Department of Parasitology, Faculty of Medicine, Ege University, İzmir, Türkiye.
| |
Collapse
|
2
|
Leontieva G, Gupalova T, Desheva Y, Kramskaya T, Bormotova E, Koroleva I, Kopteva O, Suvorov A. Evaluation of Immune Response to Mucosal Immunization with an Oral Probiotic-Based Vaccine in Mice: Potential for Prime-Boost Immunization against SARS-CoV-2. Int J Mol Sci 2023; 25:215. [PMID: 38203387 PMCID: PMC10779021 DOI: 10.3390/ijms25010215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/06/2023] [Accepted: 12/18/2023] [Indexed: 01/12/2024] Open
Abstract
Following the conclusion of the COVID-19 pandemic, the persistent genetic variability in the virus and its ongoing circulation within the global population necessitate the enhancement of existing preventive vaccines and the development of novel ones. A while back, we engineered an orally administered probiotic-based vaccine, L3-SARS, by integrating a gene fragment that encodes the spike protein S of the SARS-CoV-2 virus into the genome of the probiotic strain E. faecium L3, inducing the expression of viral antigen on the surface of bacteria. Previous studies demonstrated the efficacy of this vaccine candidate in providing protection against the virus in Syrian hamsters. In this present study, utilizing laboratory mice, we assess the immune response subsequent to immunization via the gastrointestinal mucosa and discuss its potential as an initial phase in a two-stage vaccination strategy. Our findings indicate that the oral administration of L3-SARS elicits an adaptive immune response in mice. Pre-immunization with L3-SARS enhances and prolongs the humoral immune response following a single subcutaneous immunization with a recombinant S-protein analogous to the S-insert of the coronavirus in Enterococcus faecium L3.
Collapse
Affiliation(s)
| | | | - Yulia Desheva
- Scientific and Educational Center, Molecular Bases of Interaction of Microorganisms and Human of the World-Class Research Center, Center for Personalized Medicine, FSBSI, IEM, 197376 Saint Petersburg, Russia; (G.L.); (T.G.); (T.K.); (E.B.); (I.K.); (O.K.); (A.S.)
| | | | | | | | | | | |
Collapse
|
3
|
He Q, Liu S, Liang Z, Lu S, Cun W, Mao Q. Mouse study of combined DNA/protein COVID-19 vaccine to boost high levels of antibody and cell mediated immune responses. Emerg Microbes Infect 2023; 12:2152388. [PMID: 36426608 DOI: 10.1080/22221751.2022.2152388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Affiliation(s)
- Qian He
- National Institute for Food and Drug Control, Beijing, People's Republic of China
| | | | - Zhenglun Liang
- National Institute for Food and Drug Control, Beijing, People's Republic of China
| | - Shan Lu
- Laboratory of Nucleic Acid Vaccines, Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Wei Cun
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, People's Republic of China
| | - Qunyan Mao
- National Institute for Food and Drug Control, Beijing, People's Republic of China
| |
Collapse
|
4
|
Gislefoss E, Abdelrahim Gamil AA, Øvergård AC, Evensen Ø. Identification and characterization of two salmon louse heme peroxidases and their potential as vaccine antigens. iScience 2023; 26:107991. [PMID: 37854698 PMCID: PMC10579435 DOI: 10.1016/j.isci.2023.107991] [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: 05/07/2023] [Revised: 07/09/2023] [Accepted: 09/16/2023] [Indexed: 10/20/2023] Open
Abstract
Salmon louse, Lepeophtheirus salmonis, represents major challenge for salmon farming. Current treatments impose welfare issues and are costly, whereas prophylactic measures are unavailable. Two salmon louse heme peroxidases (LsPxtl-1 and LsPxtl-2) were tested for their importance for parasite development and as potential vaccine candidates. LsPxtl-1 possesses two heme peroxidase domains and is expressed in ovaries and gut, whereas LsPxtl-2 encodes one domain and contains N-terminal signal peptide and an Eph receptor ligand-binding domain. LsPxtl-1, but not LsPxtl-2, knockdown in nauplius II stage caused poor swimming and death, indicating its importance for parasite development. Immunizations using single DNA plasmid injection encoding the peroxidases or heterologous prime (DNA) and boost (recombinant LsPxtl-2 protein) gave non-significant reduction in lice numbers. Single injection gave low specific antibody levels compared with the prime-boost. The findings suggest LsPxtl-1 is important for parasite development but formulations and vaccination modalities used did not significantly reduce lice infestation.
Collapse
Affiliation(s)
- Elisabeth Gislefoss
- Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Ås, Norway
- Sea Lice Research Center, University of Bergen, Bergen, Norway
| | - Amr Ahmed Abdelrahim Gamil
- Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Ås, Norway
- Sea Lice Research Center, University of Bergen, Bergen, Norway
| | - Aina-Cathrine Øvergård
- Department of Biological Sciences, University of Bergen, Bergen, Norway
- Sea Lice Research Center, University of Bergen, Bergen, Norway
| | - Øystein Evensen
- Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Ås, Norway
- Sea Lice Research Center, University of Bergen, Bergen, Norway
| |
Collapse
|
5
|
Liao HC, Huang MS, Chiu FF, Chai KM, Liao CL, Wu SC, Chen HW, Liu SJ. Co-delivery of a trimeric spike DNA and protein vaccine with aluminum hydroxide enhanced Th1-dominant humoral and cellular immunity against SARS-CoV-2. J Med Virol 2023; 95:e29040. [PMID: 37635380 DOI: 10.1002/jmv.29040] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 07/18/2023] [Accepted: 08/07/2023] [Indexed: 08/29/2023]
Abstract
Protein subunit vaccines have been used as prophylactic vaccines for a long time. The well-established properties of these vaccines make them the first choice for the coronavirus disease 2019 (COVID-19) outbreak. However, it is not easy to develop a protein vaccine that induces cytotoxic T lymphocyte responses and requires a longer time for manufacturing, which limits the usage of this vaccine type. Here, we report the combination of a recombinant spike (S)-trimer protein with a DNA vaccine-encoded S protein as a novel COVID-19 vaccine. The recombinant S protein was formulated with different adjuvants and mixed with the DNA plasmid before injection. We found that the recombinant S protein formulated with the adjuvant aluminum hydroxide and mixed with the DNA plasmid could enhance antigen-specific antibody titers, neutralizing antibody titers. We further evaluated the IgG2a/IgG1 isotype and cytokine profiles of the specific boosted T-cell response, which indicated that the combined vaccine induced a T-helper 1 cell-biased immune response. Immunized hamsters were challenged with severe acute respiratory syndrome coronavirus 2, and the body weight of the hamsters that received the recombinant S protein with aluminum hydroxide and/or the DNA plasmid was not reduced. Alternatively, those that received control or only the DNA plasmid immunization were reduced. Interestingly, after the third day of the viral load in the lungs, the viral challenge could not be detected in hamsters immunized with the recombinant S protein in aluminum hydroxide mixed with DNA (tissue culture infectious dose < 10). The viral load in the lungs was 109 , 106 , and 107 for the phosphate-buffered saline, protein in aluminum hydroxide, and DNA-only immunizations, respectively. These results indicated that antiviral mechanisms neutralizing antibodies play important roles. Furthermore, we found that the combination of protein and DNA vaccination could induce relatively strong CD8+ T-cell responses. In summary, the protein subunit vaccine combined with a DNA vaccine could induce strong CD8+ T-cell responses to increase antiviral immunity for disease control.
Collapse
Affiliation(s)
- Hung-Chun Liao
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
- Institute of Biotechnology, College of Life Science and Medicine, National Tsing Hua University, Hsinchu, Taiwan
| | - Min-Syuan Huang
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
- Institute of Biotechnology, College of Life Science and Medicine, National Tsing Hua University, Hsinchu, Taiwan
| | - Fang-Feng Chiu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Kit Man Chai
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Ching-Len Liao
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Suh-Chin Wu
- Institute of Biotechnology, College of Life Science and Medicine, National Tsing Hua University, Hsinchu, Taiwan
| | - Hsin-Wei Chen
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Shih-Jen Liu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
6
|
An Y, Zhao G, Duan H, Zhang N, Duan M, Xu S, Liu X, Han Y, Zheng T, Li X, Hou J, Zhang Z, Bi Y, Zhao X, Xu K, Dai L, Wang B, Gao GF. Robust and protective immune responses induced by heterologous prime-boost vaccination with DNA-protein dimeric RBD vaccines for COVID-19. J Med Virol 2023; 95:e28948. [PMID: 37436839 DOI: 10.1002/jmv.28948] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 06/21/2023] [Accepted: 06/22/2023] [Indexed: 07/14/2023]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic posed great impacts on public health. To fight against the pandemic, robust immune responses induced by vaccination are indispensable. Previously, we developed a subunit vaccine adjuvanted by aluminum hydroxide, ZF2001, based on the dimeric tandem-repeat RBD immunogen, which has been approved for clinical use. This dimeric RBD design was also explored as an mRNA vaccine. Both showed potent immunogenicity. In this study, a DNA vaccine candidate encoding RBD-dimer was designed. The humoral and cellular immune responses induced by homologous and heterologous prime-boost approaches with DNA-RBD-dimer and ZF2001 were assessed in mice. Protection efficacy was studied by the SARS-CoV-2 challenge. We found that the DNA-RBD-dimer vaccine was robustly immunogenic. Priming with DNA-RBD-dimer followed by ZF2001 boosting induced higher levels of neutralizing antibodies than homologous vaccination with either DNA-RBD-dimer or ZF2001, elicited polyfunctional cellular immunity with a TH 1-biased polarization, and efficiently protected mice against SARS-CoV-2 infection in the lung. This study demonstrated the robust and protective immune responses induced by the DNA-RBD-dimer candidate and provided a heterologous prime-boost approach with DNA-RBD-dimer and ZF2001.
Collapse
Affiliation(s)
- Yaling An
- Savaid Medical School, University of Chinese Academy of Sciences (UCAS), Beijing, China
| | - Gan Zhao
- Advaccine Biopharmaceutics (Suzhou) Co. Ltd, Suzhou, China
| | - Huixin Duan
- Savaid Medical School, University of Chinese Academy of Sciences (UCAS), Beijing, China
| | - Ning Zhang
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Chinese Academy of Sciences (CAS), Beijing, China
| | - Minrun Duan
- School of Life Sciences, Yunnan University, Kunming, China
| | - Senyu Xu
- Savaid Medical School, University of Chinese Academy of Sciences (UCAS), Beijing, China
| | - Xueyuan Liu
- School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yuxuan Han
- Savaid Medical School, University of Chinese Academy of Sciences (UCAS), Beijing, China
| | - Tianyi Zheng
- Zhejiang University School of Medicine, Hangzhou, China
| | - Xin Li
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Chinese Academy of Sciences (CAS), Beijing, China
| | - Jiawang Hou
- Advaccine Biopharmaceutics (Suzhou) Co. Ltd, Suzhou, China
| | - Zhiyu Zhang
- Advaccine Biopharmaceutics (Suzhou) Co. Ltd, Suzhou, China
| | - Yuhai Bi
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Chinese Academy of Sciences (CAS), Beijing, China
- CAS Center for Influenza Research and Early-Warning (CASCIRE), CAS-TWAS Center of Excellence for Emerging Infectious Diseases (CEEID), Chinese Academy of Sciences, Beijing, China
| | - Xin Zhao
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Chinese Academy of Sciences (CAS), Beijing, China
| | - Kun Xu
- Research Network of Immunity and Health (RNIH), Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing, China
| | - Lianpan Dai
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Chinese Academy of Sciences (CAS), Beijing, China
| | - Bin Wang
- Advaccine Biopharmaceutics (Suzhou) Co. Ltd, Suzhou, China
| | - George F Gao
- Savaid Medical School, University of Chinese Academy of Sciences (UCAS), Beijing, China
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Chinese Academy of Sciences (CAS), Beijing, China
- Research Network of Immunity and Health (RNIH), Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
7
|
Pallavi P, Harini K, Elboughdiri N, Gowtham P, Girigoswami K, Girigoswami A. Infections associated with SARS-CoV-2 exploited via nanoformulated photodynamic therapy. ADMET AND DMPK 2023; 11:513-531. [PMID: 37937246 PMCID: PMC10626507 DOI: 10.5599/admet.1883] [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: 05/08/2023] [Revised: 06/30/2023] [Indexed: 11/09/2023] Open
Abstract
Background and purpose The pandemic of COVID-19 has highlighted the need for managing infectious diseases, which spreads by airborne transmission leading to serious health, social, and economic issues. SARS-CoV-2 is an enveloped virus with a 60-140 nm diameter and particle-like features, which majorly accounts for this disease. Expanding diagnostic capabilities, developing safe vaccinations with long-lasting immunity, and formulating effective medications are the strategies to be investigated. Experimental approach For the literature search, electronic databases such as Scopus, Google Scholar, MEDLINE, Embase, PubMed, and Web of Science were used as the source. Search terms like 'Nano-mediated PDT,' 'PDT for SARS-CoV-2', and 'Nanotechnology in treatment for SARS-CoV-2' were used. Out of 275 initially selected articles, 198 were chosen after the abstract screening. During the full-text screening, 80 papers were excluded, and 18 were eliminated during data extraction. Preference was given to articles published from 2018 onwards, but a few older references were cited for their valuable information. Key results Synthetic nanoparticles (NPs) have a close structural resemblance to viruses and interact greatly with their proteins due to their similarities in the configurations. NPs had previously been reported to be effective against a variety of viruses. In this way, with nanoparticles, photodynamic therapy (PDT) can be a viable alternative to antibiotics in fighting against microbial infections. The protocol of PDT includes the activation of photosensitizers using specific light to destroy microorganisms in the presence of oxygen, treating several respiratory diseases. Conclusion The use of PDT in treating COVID-19 requires intensive investigations, which has been reviewed in this manuscript, including a computational approach to formulating effective photosensitizers.
Collapse
Affiliation(s)
- Pragya Pallavi
- Medical Bionanotechnology, Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, Chennai, TN-603103, India
| | - Karthick Harini
- Medical Bionanotechnology, Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, Chennai, TN-603103, India
| | - Noureddine Elboughdiri
- Chemical Engineering Department, College of Engineering, University of Ha'il, P.O. Box 2440, Ha'il 81441, Saudi Arabia
- Chemical Engineering Process Department, National School of Engineers Gabes, University of Gabes, Gabes 6029, Tunisia
| | - Pemula Gowtham
- Medical Bionanotechnology, Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, Chennai, TN-603103, India
| | - Koyeli Girigoswami
- Medical Bionanotechnology, Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, Chennai, TN-603103, India
| | - Agnishwar Girigoswami
- Medical Bionanotechnology, Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, Chennai, TN-603103, India
| |
Collapse
|
8
|
Baghban R, Ghasemian A, Mahmoodi S. Nucleic acid-based vaccine platforms against the coronavirus disease 19 (COVID-19). Arch Microbiol 2023; 205:150. [PMID: 36995507 PMCID: PMC10062302 DOI: 10.1007/s00203-023-03480-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Revised: 03/11/2023] [Accepted: 03/11/2023] [Indexed: 03/31/2023]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic has infected 673,010,496 patients and caused the death of 6,854,959 cases globally until today. Enormous efforts have been made to develop fundamentally different COVID-19 vaccine platforms. Nucleic acid-based vaccines consisting of mRNA and DNA vaccines (third-generation vaccines) have been promising in terms of rapid and convenient production and efficient provocation of immune responses against the COVID-19. Several DNA-based (ZyCoV-D, INO-4800, AG0302-COVID19, and GX-19N) and mRNA-based (BNT162b2, mRNA-1273, and ARCoV) approved vaccine platforms have been utilized for the COVID-19 prevention. mRNA vaccines are at the forefront of all platforms for COVID-19 prevention. However, these vaccines have lower stability, while DNA vaccines are needed with higher doses to stimulate the immune responses. Intracellular delivery of nucleic acid-based vaccines and their adverse events needs further research. Considering re-emergence of the COVID-19 variants of concern, vaccine reassessment and the development of polyvalent vaccines, or pan-coronavirus strategies, is essential for effective infection prevention.
Collapse
Affiliation(s)
- Roghayyeh Baghban
- Poostchi Ophthalmology Research Center, Department of Ophthalmology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Abdolmajid Ghasemian
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| | - Shirin Mahmoodi
- Department of Medical Biotechnology, School of Medicine, Fasa University of Medical Sciences, Fasa, Iran.
| |
Collapse
|
9
|
You H, Jones MK, Gordon CA, Arganda AE, Cai P, Al-Wassiti H, Pouton CW, McManus DP. The mRNA Vaccine Technology Era and the Future Control of Parasitic Infections. Clin Microbiol Rev 2023; 36:e0024121. [PMID: 36625671 PMCID: PMC10035331 DOI: 10.1128/cmr.00241-21] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Despite intensive long-term efforts, with very few exceptions, the development of effective vaccines against parasitic infections has presented considerable challenges, given the complexity of parasite life cycles, the interplay between parasites and their hosts, and their capacity to escape the host immune system and to regulate host immune responses. For many parasitic diseases, conventional vaccine platforms have generally proven ill suited, considering the complex manufacturing processes involved and the costs they incur, the inability to posttranslationally modify cloned target antigens, and the absence of long-lasting protective immunity induced by these antigens. An effective antiparasite vaccine platform is required to assess the effectiveness of novel vaccine candidates at high throughput. By exploiting the approach that has recently been used successfully to produce highly protective COVID mRNA vaccines, we anticipate a new wave of research to advance the use of mRNA vaccines to prevent parasitic infections in the near future. This article considers the characteristics that are required to develop a potent antiparasite vaccine and provides a conceptual foundation to promote the development of parasite mRNA-based vaccines. We review the recent advances and challenges encountered in developing antiparasite vaccines and evaluate the potential of developing mRNA vaccines against parasites, including those causing diseases such as malaria and schistosomiasis, against which vaccines are currently suboptimal or not yet available.
Collapse
Affiliation(s)
- Hong You
- Department of Infection and Inflammation, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Malcolm K. Jones
- School of Veterinary Science, The University of Queensland, Brisbane, Australia
| | - Catherine A. Gordon
- Department of Infection and Inflammation, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Alexa E. Arganda
- Department of Infection and Inflammation, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Pengfei Cai
- Department of Infection and Inflammation, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Harry Al-Wassiti
- Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Australia
| | - Colin W. Pouton
- Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Australia
| | - Donald P. McManus
- Department of Infection and Inflammation, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| |
Collapse
|
10
|
He X, He C, Hong W, Yang J, Wei X. Research progress in spike mutations of SARS-CoV-2 variants and vaccine development. Med Res Rev 2023. [PMID: 36929527 DOI: 10.1002/med.21941] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 09/27/2022] [Accepted: 02/26/2023] [Indexed: 03/18/2023]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic can hardly end with the emergence of different variants over time. In the past 2 years, several variants of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), such as the Delta and Omicron variants, have emerged with higher transmissibility, immune evasion and drug resistance, leading to higher morbidity and mortality in the population. The prevalent variants of concern (VOCs) share several mutations on the spike that can affect virus characteristics, including transmissibility, antigenicity, and immune evasion. Increasing evidence has demonstrated that the neutralization capacity of sera from COVID-19 convalescent or vaccinated individuals is decreased against SARS-CoV-2 variants. Moreover, the vaccine effectiveness of current COVID-19 vaccines against SARS-CoV-2 VOCs is not as high as that against wild-type SARS-CoV-2. Therefore, more attention might be paid to how the mutations impact vaccine effectiveness. In this review, we summarized the current studies on the mutations of the SARS-CoV-2 spike, particularly of the receptor binding domain, to elaborate on how the mutations impact the infectivity, transmissibility and immune evasion of the virus. The effects of mutations in the SARS-CoV-2 spike on the current therapeutics were highlighted, and potential strategies for future vaccine development were suggested.
Collapse
Affiliation(s)
- Xuemei He
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Cai He
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Weiqi Hong
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jingyun Yang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
11
|
Zhao G, Zhang Z, Ding Y, Hou J, Liu Y, Zhang M, Sui C, Wang L, Xu X, Gao X, Kou Z. A DNA Vaccine Encoding the Full-Length Spike Protein of Beta Variant (B.1.351) Elicited Broader Cross-Reactive Immune Responses against Other SARS-CoV-2 Variants. Vaccines (Basel) 2023; 11:513. [PMID: 36992097 PMCID: PMC10054764 DOI: 10.3390/vaccines11030513] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 02/16/2023] [Accepted: 02/21/2023] [Indexed: 02/25/2023] Open
Abstract
The SARS-CoV-2 pandemic remains an ongoing threat to global health with emerging variants, especially the Omicron variant and its sub-lineages. Although large-scale vaccination worldwide has delivered outstanding achievements for COVID-19 prevention, a declining effectiveness to a different extent in emerging SARS-CoV-2 variants was observed in the vaccinated population. Vaccines eliciting broader spectrum neutralizing antibodies and cellular immune responses are urgently needed and important. To achieve this goal, rational vaccine design, including antigen modeling, screening and combination, vaccine pipelines, and delivery, are keys to developing a next-generation COVID-19 vaccine. In this study, we designed several DNA constructs based on codon-optimized spike coding regions of several SARS-CoV-2 variants and analyzed their cross-reactive antibodies, including neutralizing antibodies, and cellular immune responses against several VOCs in C57BL/6 mice. The results revealed that different SARS-CoV-2 VOCs induced different cross-reactivity; pBeta, a DNA vaccine encoding the spike protein of the Beta variant, elicited broader cross-reactive neutralizing antibodies against other variants including the Omicron variants BA.1 and BA.4/5. This result demonstrates that the spike antigen from the Beta variant potentially serves as one of the antigens for multivalent vaccine design and development against variants of SARS-CoV-2.
Collapse
Affiliation(s)
- Gan Zhao
- Advaccine Biopharmaceutics (Suzhou) Co., Ltd., Suzhou 215000, China
| | | | | | | | | | | | | | | | | | | | - Zhihua Kou
- Advaccine Biopharmaceutics (Suzhou) Co., Ltd., Suzhou 215000, China
| |
Collapse
|
12
|
da Costa HHM, Orts DJB, Moura AD, Duarte-Neto AN, Cirqueira CS, Réssio RA, Kanamura CT, Miguita K, Ferreira JE, Santos RTM, Adriani PP, Cunha-Junior JP, Astray RM, Catarino RM, Lancelotti M, Prudencio CR. RBD and Spike DNA-Based Immunization in Rabbits Elicited IgG Avidity Maturation and High Neutralizing Antibody Responses against SARS-CoV-2. Viruses 2023; 15:555. [PMID: 36851769 PMCID: PMC9959588 DOI: 10.3390/v15020555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 01/23/2023] [Accepted: 02/11/2023] [Indexed: 02/19/2023] Open
Abstract
Neutralizing antibodies (nAbs) are a critical part of coronavirus disease 2019 (COVID-19) research as they are used to gain insight into the immune response to severe acute respiratory syndrome-related coronavirus 2 (SARS-CoV-2) infections. Among the technologies available for generating nAbs, DNA-based immunization methods are an alternative to conventional protocols. In this pilot study, we investigated whether DNA-based immunization by needle injection in rabbits was a viable approach to produce a functional antibody response. We demonstrated that three doses of DNA plasmid carrying the gene encoding the full-length spike protein (S) or the receptor binding domain (RBD) of SARS-CoV-2 induced a time-dependent increase in IgG antibody avidity maturation. Moreover, the IgG antibodies displayed high cross neutralization by live SARS-CoV-2 and pseudoviruses neutralization assays. Thus, we established a simple, low cost and feasible DNA-based immunization protocol in rabbits that elicited high IgG avidity maturation and nAbs production against SARS-CoV-2, highlighting the importance of DNA-based platforms for developing new immunization strategies against SARS-CoV-2 and future emerging epidemics.
Collapse
Affiliation(s)
- Hernan H. M. da Costa
- Immunology Center, Institute Adolfo Lutz, São Paulo 01246-902, Brazil
- Graduate Program Interunits in Biotechnology, University of São Paulo, São Paulo 05508-000, Brazil
| | - Diego J. B. Orts
- Immunology Center, Institute Adolfo Lutz, São Paulo 01246-902, Brazil
| | - Andrew D. Moura
- Immunology Center, Institute Adolfo Lutz, São Paulo 01246-902, Brazil
| | | | | | - Rodrigo A. Réssio
- Pathology Center, Institute Adolfo Lutz, São Paulo 01246-902, Brazil
| | | | - Karen Miguita
- Pathology Center, Institute Adolfo Lutz, São Paulo 01246-902, Brazil
| | | | | | - Patricia P. Adriani
- Skinzymes Biotechnology Ltd., São Paulo 05441-040, Brazil
- Laboratory of Nanopharmaceuticals and Delivery Systems, Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil
| | - Jair P. Cunha-Junior
- Laboratory of Immunochemistry and Immunotechnology, Department of Immunology, Federal University of Uberlândia, Uberlândia 38405-317, Brazil
| | - Renato M. Astray
- Multi-Purpose Laboratory, Butantan Institute, São Paulo 05503-900, Brazil
| | | | - Marcelo Lancelotti
- Faculty of Pharmaceutical Sciences, Campinas State University, Campinas 13083-871, Brazil
| | - Carlos R. Prudencio
- Immunology Center, Institute Adolfo Lutz, São Paulo 01246-902, Brazil
- Graduate Program Interunits in Biotechnology, University of São Paulo, São Paulo 05508-000, Brazil
| |
Collapse
|
13
|
Adenovirus Vaccine Containing Truncated SARS-CoV-2 Spike Protein S1 Subunit Leads to a Specific Immune Response in Mice. Vaccines (Basel) 2023; 11:vaccines11020429. [PMID: 36851306 PMCID: PMC9968167 DOI: 10.3390/vaccines11020429] [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: 01/04/2023] [Revised: 02/06/2023] [Accepted: 02/07/2023] [Indexed: 02/17/2023] Open
Abstract
The development of an efficient and safe coronavirus disease 2019 (COVID-19) vaccine is a crucial approach for managing the severe acute respiratory disease coronavirus 2 (SARS-CoV-2) pandemic in light of current conditions. In this study, we produced a shortened segment of the optimized SARS-CoV-2 spike gene (2043 bp, termed S1) that was able to encode a truncated S1 protein. The protein was tested to determine if it could elicit efficient immunization in mice against SARS-CoV-2. The presence of the S1 protein was confirmed with immunofluorescence and Western blotting. An adenovirus vaccine bearing the S1 gene fragment (Ad-S1) was administered intramuscularly to mice four times over 4 weeks. SARS-CoV-2 S1 protein humoral immunity was demonstrated in all immunized mice. The serum from immunized mice demonstrated excellent anti-infection activity in vitro. A robust humoral immune response against SARS-CoV-2 was observed in the mice after vaccination with Ad-S1, suggesting that the adenovirus vaccine may aid the development of vaccines against SARS-CoV-2 and other genetically distinct viruses.
Collapse
|
14
|
Zhang J, He Q, Yan X, Liu J, Bai Y, An C, Cui B, Gao F, Mao Q, Wang J, Xu M, Liang Z. Mixed formulation of mRNA and protein-based COVID-19 vaccines triggered superior neutralizing antibody responses. MedComm (Beijing) 2022; 3:e188. [PMID: 36474858 PMCID: PMC9717706 DOI: 10.1002/mco2.188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 11/02/2022] [Accepted: 11/03/2022] [Indexed: 12/03/2022] Open
Abstract
Integrating different types of vaccines into a singular immunization regimen is an effective and accessible approach to strengthen and broaden the immunogenicity of existing coronavirus disease 2019 (COVID-19) vaccine candidates. To optimize the immunization strategy of the novel mRNA-based vaccine and recombinant protein subunit vaccine that attracted much attention in COVID-19 vaccine development, we evaluated the immunogenicity of different combined regimens with the mRNA vaccine (RNA-RBD) and protein subunit vaccine (PS-RBD) in mice. Compared with homologous immunization of RNA-RBD or PS-RBD, heterologous prime-boost strategies for mRNA and protein subunit vaccines failed to simultaneously enhance neutralizing antibody (NAb) and Th1 cellular response in this study, showing modestly higher serum neutralizing activity and antibody-dependent cell-mediated cytotoxicity for "PS-RBD prime, RNA-RBD boost" and robust Th1 type cellular response for "RNA-RBD prime, PS-RBD boost". Interestingly, immunizing the mice with the mixed formulation of the two aforementioned vaccines in various proportions further significantly enhanced the NAb responses against ancestral, Delta, and Omicron strains and manifested increased Th1-type responses, suggesting that a mixed formulation of mRNA and protein vaccines might be a more prospective vaccination strategy. This study provides basic research data on the combined vaccination strategies of mRNA and protein-based COVID-19 vaccines.
Collapse
Affiliation(s)
- Jialu Zhang
- Division of Hepatitis and Enterovirus Vaccines, Institute of Biological Products, National Institutes for Food and Drug ControlNHC Key Laboratory of Research on Quality and Standardization of Biotech Products, NMPA Key Laboratory for Quality Research and Evaluation of Biological ProductsBeijingPeople's Republic of China
| | - Qian He
- Division of Hepatitis and Enterovirus Vaccines, Institute of Biological Products, National Institutes for Food and Drug ControlNHC Key Laboratory of Research on Quality and Standardization of Biotech Products, NMPA Key Laboratory for Quality Research and Evaluation of Biological ProductsBeijingPeople's Republic of China
| | - Xujia Yan
- Division of Hepatitis and Enterovirus Vaccines, Institute of Biological Products, National Institutes for Food and Drug ControlNHC Key Laboratory of Research on Quality and Standardization of Biotech Products, NMPA Key Laboratory for Quality Research and Evaluation of Biological ProductsBeijingPeople's Republic of China
| | - Jianyang Liu
- Division of Hepatitis and Enterovirus Vaccines, Institute of Biological Products, National Institutes for Food and Drug ControlNHC Key Laboratory of Research on Quality and Standardization of Biotech Products, NMPA Key Laboratory for Quality Research and Evaluation of Biological ProductsBeijingPeople's Republic of China
| | - Yu Bai
- Division of Hepatitis and Enterovirus Vaccines, Institute of Biological Products, National Institutes for Food and Drug ControlNHC Key Laboratory of Research on Quality and Standardization of Biotech Products, NMPA Key Laboratory for Quality Research and Evaluation of Biological ProductsBeijingPeople's Republic of China
| | - Chaoqiang An
- Division of Hepatitis and Enterovirus Vaccines, Institute of Biological Products, National Institutes for Food and Drug ControlNHC Key Laboratory of Research on Quality and Standardization of Biotech Products, NMPA Key Laboratory for Quality Research and Evaluation of Biological ProductsBeijingPeople's Republic of China
| | - Bopei Cui
- Division of Hepatitis and Enterovirus Vaccines, Institute of Biological Products, National Institutes for Food and Drug ControlNHC Key Laboratory of Research on Quality and Standardization of Biotech Products, NMPA Key Laboratory for Quality Research and Evaluation of Biological ProductsBeijingPeople's Republic of China
| | - Fan Gao
- Division of Hepatitis and Enterovirus Vaccines, Institute of Biological Products, National Institutes for Food and Drug ControlNHC Key Laboratory of Research on Quality and Standardization of Biotech Products, NMPA Key Laboratory for Quality Research and Evaluation of Biological ProductsBeijingPeople's Republic of China
| | - Qunying Mao
- Division of Hepatitis and Enterovirus Vaccines, Institute of Biological Products, National Institutes for Food and Drug ControlNHC Key Laboratory of Research on Quality and Standardization of Biotech Products, NMPA Key Laboratory for Quality Research and Evaluation of Biological ProductsBeijingPeople's Republic of China
| | - Junzhi Wang
- Division of Hepatitis and Enterovirus Vaccines, Institute of Biological Products, National Institutes for Food and Drug ControlNHC Key Laboratory of Research on Quality and Standardization of Biotech Products, NMPA Key Laboratory for Quality Research and Evaluation of Biological ProductsBeijingPeople's Republic of China
| | - Miao Xu
- Division of Hepatitis and Enterovirus Vaccines, Institute of Biological Products, National Institutes for Food and Drug ControlNHC Key Laboratory of Research on Quality and Standardization of Biotech Products, NMPA Key Laboratory for Quality Research and Evaluation of Biological ProductsBeijingPeople's Republic of China
| | - Zhenglun Liang
- Division of Hepatitis and Enterovirus Vaccines, Institute of Biological Products, National Institutes for Food and Drug ControlNHC Key Laboratory of Research on Quality and Standardization of Biotech Products, NMPA Key Laboratory for Quality Research and Evaluation of Biological ProductsBeijingPeople's Republic of China
| |
Collapse
|
15
|
Wang T, Miao F, Lv S, Li L, Wei F, Hou L, Sun R, Li W, Zhang J, Zhang C, Yang G, Xiang H, Meng K, Wan Z, Wang B, Feng G, Zhao Z, Luo D, Li N, Tu C, Wang H, Xue X, Liu Y, Gao Y. Proteomic and Metabolomic Characterization of SARS-CoV-2-Infected Cynomolgus Macaque at Early Stage. Front Immunol 2022; 13:954121. [PMID: 35903092 PMCID: PMC9315341 DOI: 10.3389/fimmu.2022.954121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 06/15/2022] [Indexed: 11/26/2022] Open
Abstract
Although tremendous effort has been exerted to elucidate the pathogenesis of severe COVID-19 cases, the detailed mechanism of moderate cases, which accounts for 90% of all patients, remains unclear yet, partly limited by lacking the biopsy tissues. Here, we established the COVID-19 infection model in cynomolgus macaques (CMs), monitored the clinical and pathological features, and analyzed underlying pathogenic mechanisms at early infection stage by performing proteomic and metabolomic profiling of lung tissues and sera samples from COVID-19 CMs models. Our data demonstrated that innate immune response, neutrophile and platelet activation were mainly dysregulated in COVID-19 CMs. The symptom of neutrophilia, lymphopenia and massive "cytokines storm", main features of severe COVID-19 patients, were greatly weakened in most of the challenged CMs, which are more semblable as moderate patients. Thus, COVID-19 model in CMs is rational to understand the pathogenesis of moderate COVID-19 and may be a candidate model to assess the safety and efficacy of therapeutics and vaccines against SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Tiecheng Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou, China
| | - Faming Miao
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou, China
| | - Shengnan Lv
- Department of Hepatobiliary and Pancreas Surgery, Jilin University First Hospital, Changchun, China
| | - Liang Li
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou, China
| | - Feng Wei
- Department of Hepatobiliary and Pancreas Surgery, Jilin University First Hospital, Changchun, China
| | - Lihua Hou
- Vaccine and Antibody Engineering Laboratory, Beijing Institute of Biotechnology, Beijing, China
| | - Renren Sun
- Key Laboratory of Organ Regeneration & Transplantation of Ministry of Education, and National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, The First Hospital of Jilin University, Changchun, China
| | - Wei Li
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Jian Zhang
- Department of Hepatobiliary and Pancreas Surgery, Jilin University First Hospital, Changchun, China
| | - Cheng Zhang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Guang Yang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Haiyang Xiang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Keyin Meng
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Zhonghai Wan
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Busen Wang
- Vaccine and Antibody Engineering Laboratory, Beijing Institute of Biotechnology, Beijing, China
| | - Guodong Feng
- Department of Neurology, Zhongshan Hospital Fudan University, Shanghai, China
| | - Zhongpeng Zhao
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Deyan Luo
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Nan Li
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Changchun Tu
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou, China
| | - Hui Wang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Xiaochang Xue
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi’an, China
| | - Yan Liu
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou, China
| | - Yuwei Gao
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou, China
| |
Collapse
|
16
|
Lin Q, Lu C, Hong Y, Li R, Chen J, Chen W, Chen J. Animal models for studying coronavirus infections and developing antiviral agents and vaccines. Antiviral Res 2022; 203:105345. [PMID: 35605699 PMCID: PMC9122840 DOI: 10.1016/j.antiviral.2022.105345] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 04/30/2022] [Accepted: 05/17/2022] [Indexed: 01/17/2023]
Abstract
In addition to severe acute respiratory syndrome coronavirus (SARS-CoV) and Middle East respiratory syndrome coronavirus (MERS-CoV), SARS-CoV-2 has become the third deadly coronavirus that infects humans and causes the new coronavirus disease (COVID-19). COVID-19 has already caused more than six million deaths worldwide and it is likely the biggest pandemic of this century faced by mankind. Although many studies on SARS-CoV-2 have been conducted, a detailed understanding of SARS-CoV-2 and COVID-19 is still lacking. Animal models are indispensable for studying its pathogenesis and developing vaccines and antivirals. In this review, we analyze animal models of coronavirus infections and explore their applications on antivirals and vaccines.
Collapse
Affiliation(s)
- Qisheng Lin
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Chunni Lu
- Centre for Inflammatory Diseases, Monash University Department of Medicine, Monash Medical Centre, Monash University, Clayton, Victoria 3168, Australia
| | - Yuqi Hong
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Runfeng Li
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| | - Jinding Chen
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Weisan Chen
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, 3086, Australia.
| | - Jianxin Chen
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China.
| |
Collapse
|
17
|
Sapkota B, Saud B, Shrestha R, Al-Fahad D, Sah R, Shrestha S, Rodriguez-Morales AJ. Heterologous prime-boost strategies for COVID-19 vaccines. J Travel Med 2022; 29:taab191. [PMID: 34918097 PMCID: PMC8754745 DOI: 10.1093/jtm/taab191] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 09/10/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND/OBJECTIVE Heterologous prime-boost doses of COVID-19 vaccines ('mix-and-match' approach) are being studied to test for the effectiveness of Oxford (AZD1222), Pfizer (BNT162b2), Moderna (mRNA-1273) and Novavax (NVX-CoV2373) vaccines for COVID in 'Com-Cov2 trial' in UK, and that of Oxford and Pfizer vaccines in 'CombivacS trial' in Spain. Later, other heterologous combinations of CoronaVac (DB15806), Janssen (JNJ-78436735), CanSino (AD5-nCOV) and other were also being trialled to explore their effectiveness. Previously, such a strategy was deployed for HIV, Ebola virus, malaria, tuberculosis, influenza and hepatitis B to develop the artificial acquired active immunity. The present review explores the science behind such an approach for candidate COVID-19 vaccines developed using 11 different platforms approved by the World Health Organization. METHODS The candidate vaccines' pharmaceutical parameters (e.g. platforms, number needed to vaccinate and intervals, adjuvanted status, excipients and preservatives added, efficacy and effectiveness, vaccine adverse events, and boosters), and clinical aspects must be analysed for the mix-and-match approach. Results prime-boost trials showed safety, effectiveness, higher systemic reactogenicity, well tolerability with improved immunogenicity, and flexibility profiles for future vaccinations, especially during acute and global shortages, compared to the homologous counterparts. CONCLUSION Still, large controlled trials are warranted to address challenging variants of concerns including Omicron and other, and to generalize the effectiveness of the approach in regular as well as emergency use during vaccine scarcity.
Collapse
Affiliation(s)
- Binaya Sapkota
- Nobel College Faculty of Health Sciences, Department of Pharmaceutical Sciences, Kathmandu, Nepal
| | - Bhuvan Saud
- Department of Medical Laboratory Technology, Janamaitri Foundation Institute of Health Sciences, Lalitpur, Nepal
- Central Department of Biotechnology, Institute of Science and Technology, Tribhuvan University, Kirtipur, Nepal
| | - Ranish Shrestha
- Infection Control Unit, Outbreak Investigation and Response Sub-committee, Nepal Cancer Hospital and Research Center, Lalitpur, Nepal
- Nepal Health Research and Innovation Foundation, Lalitpur, Nepal
| | - Dhurgham Al-Fahad
- Department of Pathological Analysis, College of Science, University of Thi-Qar, Thi-Qar, Iraq
| | - Ranjit Sah
- Tribhuvan University Teaching Hospital, Institute of Medicine, Kathmandu, Nepal
| | - Sunil Shrestha
- School of Pharmacy, Monash University Malaysia, Selangor, Malaysia
| | - Alfonso J Rodriguez-Morales
- Grupo de Investigación Biomedicina, Faculty of Medicine, Fundacion Universitaria Autonoma de las Americas, Pereira, Colombia
- Master of Clinical Epidemiology and Biostatistics, Faculty of Health Sciences, Universidad Cientifica del Sur, Lima, Peru
| |
Collapse
|
18
|
Banihashemi SR, Es-haghi A, Fallah Mehrabadi MH, Nofeli M, Mokarram AR, Ranjbar A, Salman M, Hajimoradi M, Razaz SH, Taghdiri M, Bagheri M, Dadar M, Hassan ZM, Eslampanah M, Salehi Najafabadi Z, Lotfi M, Khorasani A, Rahmani F. Safety and Efficacy of Combined Intramuscular/Intranasal RAZI-COV PARS Vaccine Candidate Against SARS-CoV-2: A Preclinical Study in Several Animal Models. Front Immunol 2022; 13:836745. [PMID: 35693788 PMCID: PMC9179012 DOI: 10.3389/fimmu.2022.836745] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 04/25/2022] [Indexed: 12/23/2022] Open
Abstract
Several vaccine candidates for COVID-19 have been developed, and few vaccines received emergency approval with an acceptable level of efficacy and safety. We herein report the development of the first recombinant protein-based vaccine in Iran based on the recombinant SARS-CoV-2 spike protein in its monomeric (encompassing amino acid 1-674 for S1 and 685-1211 for S2 subunits) and trimer form (S-Trimer) formulated in the oil-in-water adjuvant system RAS-01 (Razi Adjuvant System-01). The safety and immunity of the candidate vaccine, referred to as RAZI-COV PARS, were evaluated in Syrian hamster, BALB/c mice, Pirbright guinea pig, and New Zeeland white (NZW) rabbit. All vaccinated animals received two intramuscular (IM) and one intranasal (IN) candidate vaccine at 3-week intervals (days 0, 21, and 51). The challenge study was performed intranasally with 5×106 pfu of SARS-CoV-2 35 days post-vaccination. None of the vaccinated mice, hamsters, guinea pigs, or rabbits showed any changes in general clinical observations; body weight and food intake, clinical indicators, hematology examination, blood chemistry, and pathological examination of vital organs. Safety of vaccine after the administration of single and repeated dose was also established. Three different doses of candidate vaccine stimulated remarkable titers of neutralizing antibodies, S1, Receptor-Binding Domain (RBD), and N-terminal domain (NTD) specific IgG antibodies as well as IgA antibodies compared to placebo and control groups (P<0.01). Middle and high doses of RAZI-COV PARS vaccine significantly induced a robust and quick immune response from the third-week post-immunization. Histopathological studies on vaccinated hamsters showed that the challenge with SARS-CoV-2 did not induce any modifications in the lungs. The protection of the hamster was documented by the absence of lung pathology, the decreased virus load in the lung, rapid clearance of the virus from the lung, and strong humoral and cellular immune response. These findings confirm the immunogenicity and efficacy of the RAZI-COV PARS vaccine. Of the three tested vaccine regimens, the middle dose of the vaccine showed the best protective immune parameters. This vaccine with heterologous prime-boost vaccination method can be a good candidate to control the viral infection and its spread by stimulating central and mucosal immunity.
Collapse
Affiliation(s)
- Seyed Reza Banihashemi
- Department of immunology, Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| | - Ali Es-haghi
- Department of Physico Chemistry, Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| | - Mohammad Hossein Fallah Mehrabadi
- Department of Epidemiology, Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| | - Mojtaba Nofeli
- Department of Research and Development, Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| | - Ali Rezaei Mokarram
- Department of Quality Assurance, Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| | - Alireza Ranjbar
- Clinic of Pediatrics, Institute of Interventional Allergology and Immunology, Bonn, Germany
| | - Mo Salman
- Animal Population Health Institute of College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| | - Monireh Hajimoradi
- Department of immunology, Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| | - Seyad Hossein Razaz
- Department of immunology, Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| | - Maryam Taghdiri
- Department of immunology, Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| | - Mohsen Bagheri
- Department of Physico Chemistry, Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| | - Maryam Dadar
- Department of Research and Development, Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| | - Zuhair Mohammad Hassan
- Department of Immunology, School of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mohammad Eslampanah
- Department of Pathology, Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| | - Zahra Salehi Najafabadi
- Department of Research and Development, Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| | - Mohsen Lotfi
- Department of Quality Control, Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| | - Akbar Khorasani
- Department of Research and Development, Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| | - Fereidoon Rahmani
- Department of Physico Chemistry, Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| |
Collapse
|
19
|
Willcox AC, Sung K, Garrett ME, Galloway JG, Erasmus JH, Logue JK, Hawman DW, Chu HY, Hasenkrug KJ, Fuller DH, Matsen IV FA, Overbaugh J. Detailed analysis of antibody responses to SARS-CoV-2 vaccination and infection in macaques. PLoS Pathog 2022; 18:e1010155. [PMID: 35404959 PMCID: PMC9022802 DOI: 10.1371/journal.ppat.1010155] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 04/21/2022] [Accepted: 03/21/2022] [Indexed: 02/02/2023] Open
Abstract
Macaques are a commonly used model for studying immunity to human viruses, including for studies of SARS-CoV-2 infection and vaccination. However, it is unknown whether macaque antibody responses resemble the response in humans. To answer this question, we employed a phage-based deep mutational scanning approach (Phage-DMS) to compare which linear epitopes are targeted on the SARS-CoV-2 Spike protein in convalescent humans, convalescent (re-infected) rhesus macaques, mRNA-vaccinated humans, and repRNA-vaccinated pigtail macaques. We also used Phage-DMS to determine antibody escape pathways within each epitope, enabling a granular comparison of antibody binding specificities at the locus level. Overall, we identified some common epitope targets in both macaques and humans, including in the fusion peptide (FP) and stem helix-heptad repeat 2 (SH-H) regions. Differences between groups included a response to epitopes in the N-terminal domain (NTD) and C-terminal domain (CTD) in vaccinated humans but not vaccinated macaques, as well as recognition of a CTD epitope and epitopes flanking the FP in convalescent macaques but not convalescent humans. There was also considerable variability in the escape pathways among individuals within each group. Sera from convalescent macaques showed the least variability in escape overall and converged on a common response with vaccinated humans in the SH-H epitope region, suggesting highly similar antibodies were elicited. Collectively, these findings suggest that the antibody response to SARS-CoV-2 in macaques shares many features with humans, but with substantial differences in the recognition of certain epitopes and considerable individual variability in antibody escape profiles, suggesting a diverse repertoire of antibodies that can respond to major epitopes in both humans and macaques. Differences in macaque species and exposure type may also contribute to these findings.
Collapse
Affiliation(s)
- Alexandra C. Willcox
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- Medical Scientist Training Program, University of Washington, Seattle, Washington, United States of America
- Molecular and Cellular Biology Program, University of Washington, Seattle, Washington, United States of America
| | - Kevin Sung
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Meghan E. Garrett
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- Molecular and Cellular Biology Program, University of Washington, Seattle, Washington, United States of America
| | - Jared G. Galloway
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Jesse H. Erasmus
- Department of Microbiology, University of Washington, Seattle, Washington, United States of America
- HDT Bio, Seattle, Washington, United States of America
| | - Jennifer K. Logue
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - David W. Hawman
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America
| | - Helen Y. Chu
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Kim J. Hasenkrug
- Laboratory of Persistent Viral Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America
| | - Deborah H. Fuller
- Department of Microbiology, University of Washington, Seattle, Washington, United States of America
- Infectious Diseases and Translational Medicine, Washington National Primate Research Center, Seattle, Washington, United States of America
- Center for Innate Immunity and Immune Disease, University of Washington, Seattle, Washington, United States of America
| | - Frederick A. Matsen IV
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Julie Overbaugh
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| |
Collapse
|
20
|
Borgoyakova MB, Karpenko LI, Rudometov AP, Volosnikova EA, Merkuleva IA, Starostina EV, Zadorozhny AM, Isaeva AA, Nesmeyanova VS, Shanshin DV, Baranov KO, Volkova NV, Zaitsev BN, Orlova LA, Zaykovskaya AV, Pyankov OV, Danilenko ED, Bazhan SI, Shcherbakov DN, Taranin AV, Ilyichev AA. Self-Assembled Particles Combining SARS-CoV-2 RBD Protein and RBD DNA Vaccine Induce Synergistic Enhancement of the Humoral Response in Mice. Int J Mol Sci 2022; 23:2188. [PMID: 35216301 PMCID: PMC8876144 DOI: 10.3390/ijms23042188] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 02/13/2022] [Accepted: 02/13/2022] [Indexed: 12/23/2022] Open
Abstract
Despite the fact that a range of vaccines against COVID-19 have already been created and are used for mass vaccination, the development of effective, safe, technological, and affordable vaccines continues. We have designed a vaccine that combines the recombinant protein and DNA vaccine approaches in a self-assembled particle. The receptor-binding domain (RBD) of the spike protein of SARS-CoV-2 was conjugated to polyglucin:spermidine and mixed with DNA vaccine (pVAXrbd), which led to the formation of particles of combined coronavirus vaccine (CCV-RBD) that contain the DNA vaccine inside and RBD protein on the surface. CCV-RBD particles were characterized with gel filtration, electron microscopy, and biolayer interferometry. To investigate the immunogenicity of the combined vaccine and its components, mice were immunized with the DNA vaccine pVAXrbd or RBD protein as well as CCV-RBD particles. The highest antigen-specific IgG and neutralizing activity were induced by CCV-RBD, and the level of antibodies induced by DNA or RBD alone was significantly lower. The cellular immune response was detected only in the case of DNA or CCV-RBD vaccination. These results demonstrate that a combination of DNA vaccine and RBD protein in one construct synergistically increases the humoral response to RBD protein in mice.
Collapse
Affiliation(s)
- Mariya B. Borgoyakova
- State Research Center of Virology and Biotechnology “Vector”, 630559 Koltsovo, Novosibirsk Region, Russia; (M.B.B.); (A.P.R.); (E.A.V.); (I.A.M.); (E.V.S.); (A.M.Z.); (A.A.I.); (V.S.N.); (D.V.S.); (N.V.V.); (B.N.Z.); (L.A.O.); (A.V.Z.); (O.V.P.); (E.D.D.); (S.I.B.); (D.N.S.); (A.A.I.)
| | - Larisa I. Karpenko
- State Research Center of Virology and Biotechnology “Vector”, 630559 Koltsovo, Novosibirsk Region, Russia; (M.B.B.); (A.P.R.); (E.A.V.); (I.A.M.); (E.V.S.); (A.M.Z.); (A.A.I.); (V.S.N.); (D.V.S.); (N.V.V.); (B.N.Z.); (L.A.O.); (A.V.Z.); (O.V.P.); (E.D.D.); (S.I.B.); (D.N.S.); (A.A.I.)
| | - Andrey P. Rudometov
- State Research Center of Virology and Biotechnology “Vector”, 630559 Koltsovo, Novosibirsk Region, Russia; (M.B.B.); (A.P.R.); (E.A.V.); (I.A.M.); (E.V.S.); (A.M.Z.); (A.A.I.); (V.S.N.); (D.V.S.); (N.V.V.); (B.N.Z.); (L.A.O.); (A.V.Z.); (O.V.P.); (E.D.D.); (S.I.B.); (D.N.S.); (A.A.I.)
| | - Ekaterina A. Volosnikova
- State Research Center of Virology and Biotechnology “Vector”, 630559 Koltsovo, Novosibirsk Region, Russia; (M.B.B.); (A.P.R.); (E.A.V.); (I.A.M.); (E.V.S.); (A.M.Z.); (A.A.I.); (V.S.N.); (D.V.S.); (N.V.V.); (B.N.Z.); (L.A.O.); (A.V.Z.); (O.V.P.); (E.D.D.); (S.I.B.); (D.N.S.); (A.A.I.)
| | - Iuliia A. Merkuleva
- State Research Center of Virology and Biotechnology “Vector”, 630559 Koltsovo, Novosibirsk Region, Russia; (M.B.B.); (A.P.R.); (E.A.V.); (I.A.M.); (E.V.S.); (A.M.Z.); (A.A.I.); (V.S.N.); (D.V.S.); (N.V.V.); (B.N.Z.); (L.A.O.); (A.V.Z.); (O.V.P.); (E.D.D.); (S.I.B.); (D.N.S.); (A.A.I.)
| | - Ekaterina V. Starostina
- State Research Center of Virology and Biotechnology “Vector”, 630559 Koltsovo, Novosibirsk Region, Russia; (M.B.B.); (A.P.R.); (E.A.V.); (I.A.M.); (E.V.S.); (A.M.Z.); (A.A.I.); (V.S.N.); (D.V.S.); (N.V.V.); (B.N.Z.); (L.A.O.); (A.V.Z.); (O.V.P.); (E.D.D.); (S.I.B.); (D.N.S.); (A.A.I.)
| | - Alexey M. Zadorozhny
- State Research Center of Virology and Biotechnology “Vector”, 630559 Koltsovo, Novosibirsk Region, Russia; (M.B.B.); (A.P.R.); (E.A.V.); (I.A.M.); (E.V.S.); (A.M.Z.); (A.A.I.); (V.S.N.); (D.V.S.); (N.V.V.); (B.N.Z.); (L.A.O.); (A.V.Z.); (O.V.P.); (E.D.D.); (S.I.B.); (D.N.S.); (A.A.I.)
| | - Anastasiya A. Isaeva
- State Research Center of Virology and Biotechnology “Vector”, 630559 Koltsovo, Novosibirsk Region, Russia; (M.B.B.); (A.P.R.); (E.A.V.); (I.A.M.); (E.V.S.); (A.M.Z.); (A.A.I.); (V.S.N.); (D.V.S.); (N.V.V.); (B.N.Z.); (L.A.O.); (A.V.Z.); (O.V.P.); (E.D.D.); (S.I.B.); (D.N.S.); (A.A.I.)
| | - Valentina S. Nesmeyanova
- State Research Center of Virology and Biotechnology “Vector”, 630559 Koltsovo, Novosibirsk Region, Russia; (M.B.B.); (A.P.R.); (E.A.V.); (I.A.M.); (E.V.S.); (A.M.Z.); (A.A.I.); (V.S.N.); (D.V.S.); (N.V.V.); (B.N.Z.); (L.A.O.); (A.V.Z.); (O.V.P.); (E.D.D.); (S.I.B.); (D.N.S.); (A.A.I.)
| | - Daniil V. Shanshin
- State Research Center of Virology and Biotechnology “Vector”, 630559 Koltsovo, Novosibirsk Region, Russia; (M.B.B.); (A.P.R.); (E.A.V.); (I.A.M.); (E.V.S.); (A.M.Z.); (A.A.I.); (V.S.N.); (D.V.S.); (N.V.V.); (B.N.Z.); (L.A.O.); (A.V.Z.); (O.V.P.); (E.D.D.); (S.I.B.); (D.N.S.); (A.A.I.)
| | - Konstantin O. Baranov
- Institute of Molecular and Cellular Biology, Siberian Branch of the Russian Academy of Science, 630090 Novosibirsk, Russia; (K.O.B.); (A.V.T.)
| | - Natalya V. Volkova
- State Research Center of Virology and Biotechnology “Vector”, 630559 Koltsovo, Novosibirsk Region, Russia; (M.B.B.); (A.P.R.); (E.A.V.); (I.A.M.); (E.V.S.); (A.M.Z.); (A.A.I.); (V.S.N.); (D.V.S.); (N.V.V.); (B.N.Z.); (L.A.O.); (A.V.Z.); (O.V.P.); (E.D.D.); (S.I.B.); (D.N.S.); (A.A.I.)
| | - Boris N. Zaitsev
- State Research Center of Virology and Biotechnology “Vector”, 630559 Koltsovo, Novosibirsk Region, Russia; (M.B.B.); (A.P.R.); (E.A.V.); (I.A.M.); (E.V.S.); (A.M.Z.); (A.A.I.); (V.S.N.); (D.V.S.); (N.V.V.); (B.N.Z.); (L.A.O.); (A.V.Z.); (O.V.P.); (E.D.D.); (S.I.B.); (D.N.S.); (A.A.I.)
| | - Lyubov A. Orlova
- State Research Center of Virology and Biotechnology “Vector”, 630559 Koltsovo, Novosibirsk Region, Russia; (M.B.B.); (A.P.R.); (E.A.V.); (I.A.M.); (E.V.S.); (A.M.Z.); (A.A.I.); (V.S.N.); (D.V.S.); (N.V.V.); (B.N.Z.); (L.A.O.); (A.V.Z.); (O.V.P.); (E.D.D.); (S.I.B.); (D.N.S.); (A.A.I.)
| | - Anna V. Zaykovskaya
- State Research Center of Virology and Biotechnology “Vector”, 630559 Koltsovo, Novosibirsk Region, Russia; (M.B.B.); (A.P.R.); (E.A.V.); (I.A.M.); (E.V.S.); (A.M.Z.); (A.A.I.); (V.S.N.); (D.V.S.); (N.V.V.); (B.N.Z.); (L.A.O.); (A.V.Z.); (O.V.P.); (E.D.D.); (S.I.B.); (D.N.S.); (A.A.I.)
| | - Oleg V. Pyankov
- State Research Center of Virology and Biotechnology “Vector”, 630559 Koltsovo, Novosibirsk Region, Russia; (M.B.B.); (A.P.R.); (E.A.V.); (I.A.M.); (E.V.S.); (A.M.Z.); (A.A.I.); (V.S.N.); (D.V.S.); (N.V.V.); (B.N.Z.); (L.A.O.); (A.V.Z.); (O.V.P.); (E.D.D.); (S.I.B.); (D.N.S.); (A.A.I.)
| | - Elena D. Danilenko
- State Research Center of Virology and Biotechnology “Vector”, 630559 Koltsovo, Novosibirsk Region, Russia; (M.B.B.); (A.P.R.); (E.A.V.); (I.A.M.); (E.V.S.); (A.M.Z.); (A.A.I.); (V.S.N.); (D.V.S.); (N.V.V.); (B.N.Z.); (L.A.O.); (A.V.Z.); (O.V.P.); (E.D.D.); (S.I.B.); (D.N.S.); (A.A.I.)
| | - Sergei I. Bazhan
- State Research Center of Virology and Biotechnology “Vector”, 630559 Koltsovo, Novosibirsk Region, Russia; (M.B.B.); (A.P.R.); (E.A.V.); (I.A.M.); (E.V.S.); (A.M.Z.); (A.A.I.); (V.S.N.); (D.V.S.); (N.V.V.); (B.N.Z.); (L.A.O.); (A.V.Z.); (O.V.P.); (E.D.D.); (S.I.B.); (D.N.S.); (A.A.I.)
| | - Dmitry N. Shcherbakov
- State Research Center of Virology and Biotechnology “Vector”, 630559 Koltsovo, Novosibirsk Region, Russia; (M.B.B.); (A.P.R.); (E.A.V.); (I.A.M.); (E.V.S.); (A.M.Z.); (A.A.I.); (V.S.N.); (D.V.S.); (N.V.V.); (B.N.Z.); (L.A.O.); (A.V.Z.); (O.V.P.); (E.D.D.); (S.I.B.); (D.N.S.); (A.A.I.)
| | - Alexander V. Taranin
- Institute of Molecular and Cellular Biology, Siberian Branch of the Russian Academy of Science, 630090 Novosibirsk, Russia; (K.O.B.); (A.V.T.)
| | - Alexander A. Ilyichev
- State Research Center of Virology and Biotechnology “Vector”, 630559 Koltsovo, Novosibirsk Region, Russia; (M.B.B.); (A.P.R.); (E.A.V.); (I.A.M.); (E.V.S.); (A.M.Z.); (A.A.I.); (V.S.N.); (D.V.S.); (N.V.V.); (B.N.Z.); (L.A.O.); (A.V.Z.); (O.V.P.); (E.D.D.); (S.I.B.); (D.N.S.); (A.A.I.)
| |
Collapse
|
21
|
Antibody Response of Combination of BNT162b2 and CoronaVac Platforms of COVID-19 Vaccines against Omicron Variant. Vaccines (Basel) 2022; 10:vaccines10020160. [PMID: 35214619 PMCID: PMC8877145 DOI: 10.3390/vaccines10020160] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/16/2022] [Accepted: 01/17/2022] [Indexed: 02/04/2023] Open
Abstract
By vaccinating SARS-CoV-2 naïve individuals who have already received two doses of COVID-19 vaccines, we aimed to investigate whether a heterologous prime-boost strategy, using vaccines of different platforms as the booster dose, can enhance the immune response against SARS-CoV-2 virus variants. Participants were assigned into four groups, each receiving different combination of vaccinations: two doses of BNT162b2 followed by one dose of BNT162b2 booster (B-B-B); Combination of BNT162b2 (first dose) and CoronaVac (second dose) followed by one dose of BNT162b2 booster (B-C-B); two doses of CoronaVac followed by one dose of CoronaVac booster (C-C-C); two doses of CoronaVac followed by one dose of BNT162b2 booster (C-C-B). The neutralizing antibody in sera against the virus was determined with live virus microneutralization assay (vMN). The B-B-B group and C-C-B group demonstrated significantly higher immunogenicity against SARS-CoV-2 Wild type (WT), Beta variant (BV) and Delta variant (DV). In addition, the B-B-B group and C-C-B group showed reduced but existing protection against Omicron variant (OV). Moreover, A persistent rise in vMN titre against OV was observed 3 days after booster dose. Regarding safety, a heterologous prime-boost vaccine strategy is well tolerated. In this study, it was demonstrated that using vaccines of different platforms as booster dose can enhance protection against SARS-CoV-2 variants, offering potent neutralizing activity against wild-type virus (WT), Beta variant (BV), Delta variant (DV) and some protection against the Omicron variant (OV). In addition, a booster mRNA vaccine results in a more potent immune response than inactivated vaccine regardless of which platform was used for prime doses.
Collapse
|
22
|
Negi N, Maurya SP, Singh R, Das BK. An update on host immunity correlates and prospects of re-infection in COVID-19. Int Rev Immunol 2021; 41:367-392. [PMID: 34961403 PMCID: PMC8787841 DOI: 10.1080/08830185.2021.2019727] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 10/18/2021] [Accepted: 12/08/2021] [Indexed: 01/08/2023]
Abstract
Reinfection with SARS-CoV-2 is not frequent yet the incidence rate of it is increasing globally owing to the slow emergence of drift variants that pose a perpetual threat to vaccination strategies and have a greater propensity for disease reoccurrence. Long-term protection against SARS-CoV-2 reinfection relies on the induction of the innate as well as the adaptive immune response endowed with immune memory. However, a multitude of factors including the selection pressure, the waning immunity against SARS-CoV-2 over the first year after infection possibly favors evolution of more infectious immune escape variants, amplifying the risk of reinfection. Additionally, the correlates of immune protection, the novel SARS-CoV-2 variants of concern (VOC), the durability of the adaptive and mucosal immunity remain major challenges for the development of therapeutic and prophylactic interventions. Interestingly, a recent body of evidence indicated that the gastrointestinal (GI) tract is another important target organ for SARS-CoV-2 besides the respiratory system, potentially increasing the likelihood of reinfection by impacting the microbiome and the immune response via the gut-lung axis. In this review, we summarized the latest development in SARS-CoV-2 reinfection, and explored the untapped potential of trained immunity. We also highlighted the immune memory kinetics of the humoral and cell-mediated immune response, genetic drift of the emerging viral variants, and discussed the current challenges in vaccine development. Understanding the dynamics and the quality of immune response by unlocking the power of the innate, humoral and cell-mediated immunity during SARS-CoV-2 reinfection would open newer avenues for drug discovery and vaccine designs.
Collapse
Affiliation(s)
- Neema Negi
- Department of Chemical Sciences, University of Limerick, Limerick, Ireland
- Bernal Institute, University of Limerick,Limerick, Ireland
| | - Shesh Prakash Maurya
- Department of Microbiology, All India Institute of Medical Sciences, New Delhi, India
| | - Ravinder Singh
- Department of Microbiology, All India Institute of Medical Sciences, New Delhi, India
| | - Bimal Kumar Das
- Department of Microbiology, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
23
|
Long-term stability and protection efficacy of the RBD-targeting COVID-19 mRNA vaccine in nonhuman primates. Signal Transduct Target Ther 2021; 6:438. [PMID: 34952914 PMCID: PMC8703211 DOI: 10.1038/s41392-021-00861-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 10/28/2021] [Accepted: 12/07/2021] [Indexed: 01/08/2023] Open
Abstract
Messenger RNA (mRNA) vaccine technology has shown its power in preventing the ongoing COVID-19 pandemic. Two mRNA vaccines targeting the full-length S protein of SARS-CoV-2 have been authorized for emergency use. Recently, we have developed a lipid nanoparticle-encapsulated mRNA (mRNA-LNP) encoding the receptor-binding domain (RBD) of SARS-CoV-2 (termed ARCoV), which confers complete protection in mouse model. Herein, we further characterized the protection efficacy of ARCoV in nonhuman primates and the long-term stability under normal refrigerator temperature. Intramuscular immunization of two doses of ARCoV elicited robust neutralizing antibodies as well as cellular response against SARS-CoV-2 in cynomolgus macaques. More importantly, ARCoV vaccination in macaques significantly protected animals from acute lung lesions caused by SARS-CoV-2, and viral replication in lungs and secretion in nasal swabs were completely cleared in all animals immunized with low or high doses of ARCoV. No evidence of antibody-dependent enhancement of infection was observed throughout the study. Finally, extensive stability assays showed that ARCoV can be stored at 2-8 °C for at least 6 months without decrease of immunogenicity. All these promising results strongly support the ongoing clinical trial.
Collapse
|
24
|
Fan S, Xiao K, Li D, Zhao H, Zhang J, Yu L, Chang P, Zhu S, Xu X, Liao Y, Ji T, Jiang G, Yan D, Zeng F, Duan S, Xia B, Wang L, Yang F, He Z, Song Y, Cui P, Li X, Zhang Y, Zheng B, Zhang Y, Xu W, Li Q. Preclinical immunological evaluation of an intradermal heterologous vaccine against SARS-CoV-2 variants. Emerg Microbes Infect 2021; 11:212-226. [PMID: 34931939 PMCID: PMC8745378 DOI: 10.1080/22221751.2021.2021807] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The recent emergence of COVID-19 variants has necessitated the development of new vaccines that stimulate the formation of high levels of neutralizing antibodies against S antigen variants. A new strategy involves the intradermal administration of heterologous vaccines composed of one or two doses of inactivated vaccine and a booster dose with the mutated S1 protein (K-S). Such vaccines improve the immune efficacy by increasing the neutralizing antibody titers and promoting specific T cell responses against five variants of the RBD protein. A viral challenge test with the B.1.617.2 (Delta) variant confirmed that both administration schedules (i.e. “1 + 1” and “2 + 1”) ensured protection against this strain. These results suggest that the aforementioned strategy is effective for protecting against new variants and enhances the anamnestic immune response in the immunized population.
Collapse
Affiliation(s)
- Shengtao Fan
- Institute of Medical Biology, Chinese Academy of Medicine Sciences & Peking Union Medical College, Yunnan Key Laboratory of Vaccine Research and Development for Severe Infectious Diseases, Kunming, 650118, China
| | - Kang Xiao
- National Institute for Viral Disease Control and Prevention, China CDC, Beijing, 102206, China
| | - Dandan Li
- Institute of Medical Biology, Chinese Academy of Medicine Sciences & Peking Union Medical College, Yunnan Key Laboratory of Vaccine Research and Development for Severe Infectious Diseases, Kunming, 650118, China
| | - Heng Zhao
- Institute of Medical Biology, Chinese Academy of Medicine Sciences & Peking Union Medical College, Yunnan Key Laboratory of Vaccine Research and Development for Severe Infectious Diseases, Kunming, 650118, China
| | - Jingjing Zhang
- Institute of Medical Biology, Chinese Academy of Medicine Sciences & Peking Union Medical College, Yunnan Key Laboratory of Vaccine Research and Development for Severe Infectious Diseases, Kunming, 650118, China
| | - Li Yu
- Institute of Medical Biology, Chinese Academy of Medicine Sciences & Peking Union Medical College, Yunnan Key Laboratory of Vaccine Research and Development for Severe Infectious Diseases, Kunming, 650118, China
| | - Penglan Chang
- Institute of Medical Biology, Chinese Academy of Medicine Sciences & Peking Union Medical College, Yunnan Key Laboratory of Vaccine Research and Development for Severe Infectious Diseases, Kunming, 650118, China
| | - Shuangli Zhu
- National Institute for Viral Disease Control and Prevention, China CDC, Beijing, 102206, China
| | - Xingli Xu
- Institute of Medical Biology, Chinese Academy of Medicine Sciences & Peking Union Medical College, Yunnan Key Laboratory of Vaccine Research and Development for Severe Infectious Diseases, Kunming, 650118, China
| | - Yun Liao
- Institute of Medical Biology, Chinese Academy of Medicine Sciences & Peking Union Medical College, Yunnan Key Laboratory of Vaccine Research and Development for Severe Infectious Diseases, Kunming, 650118, China
| | - Tianjiao Ji
- National Institute for Viral Disease Control and Prevention, China CDC, Beijing, 102206, China
| | - Guorun Jiang
- Institute of Medical Biology, Chinese Academy of Medicine Sciences & Peking Union Medical College, Yunnan Key Laboratory of Vaccine Research and Development for Severe Infectious Diseases, Kunming, 650118, China
| | - Dongmei Yan
- National Institute for Viral Disease Control and Prevention, China CDC, Beijing, 102206, China
| | - Fengyuan Zeng
- Institute of Medical Biology, Chinese Academy of Medicine Sciences & Peking Union Medical College, Yunnan Key Laboratory of Vaccine Research and Development for Severe Infectious Diseases, Kunming, 650118, China
| | - Suqin Duan
- Institute of Medical Biology, Chinese Academy of Medicine Sciences & Peking Union Medical College, Yunnan Key Laboratory of Vaccine Research and Development for Severe Infectious Diseases, Kunming, 650118, China
| | - Baicheng Xia
- National Institute for Viral Disease Control and Prevention, China CDC, Beijing, 102206, China
| | - Lichun Wang
- Institute of Medical Biology, Chinese Academy of Medicine Sciences & Peking Union Medical College, Yunnan Key Laboratory of Vaccine Research and Development for Severe Infectious Diseases, Kunming, 650118, China
| | - Fengmei Yang
- Institute of Medical Biology, Chinese Academy of Medicine Sciences & Peking Union Medical College, Yunnan Key Laboratory of Vaccine Research and Development for Severe Infectious Diseases, Kunming, 650118, China
| | - Zhanlong He
- Institute of Medical Biology, Chinese Academy of Medicine Sciences & Peking Union Medical College, Yunnan Key Laboratory of Vaccine Research and Development for Severe Infectious Diseases, Kunming, 650118, China
| | - Yang Song
- National Institute for Viral Disease Control and Prevention, China CDC, Beijing, 102206, China
| | - Pingfang Cui
- Institute of Medical Biology, Chinese Academy of Medicine Sciences & Peking Union Medical College, Yunnan Key Laboratory of Vaccine Research and Development for Severe Infectious Diseases, Kunming, 650118, China
| | - Xiaolei Li
- National Institute for Viral Disease Control and Prevention, China CDC, Beijing, 102206, China
| | - Yaxing Zhang
- Institute of Medical Biology, Chinese Academy of Medicine Sciences & Peking Union Medical College, Yunnan Key Laboratory of Vaccine Research and Development for Severe Infectious Diseases, Kunming, 650118, China
| | - Bangyi Zheng
- Institute of Medical Biology, Chinese Academy of Medicine Sciences & Peking Union Medical College, Yunnan Key Laboratory of Vaccine Research and Development for Severe Infectious Diseases, Kunming, 650118, China
| | - Ying Zhang
- Institute of Medical Biology, Chinese Academy of Medicine Sciences & Peking Union Medical College, Yunnan Key Laboratory of Vaccine Research and Development for Severe Infectious Diseases, Kunming, 650118, China
| | - Wenbo Xu
- National Institute for Viral Disease Control and Prevention, China CDC, Beijing, 102206, China
| | - Qihan Li
- Institute of Medical Biology, Chinese Academy of Medicine Sciences & Peking Union Medical College, Yunnan Key Laboratory of Vaccine Research and Development for Severe Infectious Diseases, Kunming, 650118, China
| |
Collapse
|
25
|
Willcox AC, Sung K, Garrett ME, Galloway JG, O’Connor MA, Erasmus JH, Logue JK, Hawman DW, Chu HY, Hasenkrug KJ, Fuller DH, Matsen FA, Overbaugh J. Macaque-human differences in SARS-CoV-2 Spike antibody response elicited by vaccination or infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021:2021.12.01.470697. [PMID: 34909774 PMCID: PMC8669841 DOI: 10.1101/2021.12.01.470697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Macaques are a commonly used model for studying immunity to human viruses, including for studies of SARS-CoV-2 infection and vaccination. However, it is unknown whether macaque antibody responses recapitulate, and thus appropriately model, the response in humans. To answer this question, we employed a phage-based deep mutational scanning approach (Phage-DMS) to compare which linear epitopes are targeted on the SARS-CoV-2 Spike protein in humans and macaques following either vaccination or infection. We also used Phage-DMS to determine antibody escape pathways within each epitope, enabling a granular comparison of antibody binding specificities at the locus level. Overall, we identified some common epitope targets in both macaques and humans, including in the fusion peptide (FP) and stem helix-heptad repeat 2 (SH-H) regions. Differences between groups included a response to epitopes in the N-terminal domain (NTD) and C-terminal domain (CTD) in vaccinated humans but not vaccinated macaques, as well as recognition of a CTD epitope and epitopes flanking the FP in convalescent macaques but not convalescent humans. There was also considerable variability in the escape pathways among individuals within each group. Sera from convalescent macaques showed the least variability in escape overall and converged on a common response with vaccinated humans in the SH-H epitope region, suggesting highly similar antibodies were elicited. Collectively, these findings suggest that the antibody response to SARS-CoV-2 in macaques shares many features with humans, but with substantial differences in the recognition of certain epitopes and considerable individual variability in antibody escape profiles, suggesting a diverse repertoire of antibodies that can respond to major epitopes in both humans and macaques.
Collapse
Affiliation(s)
- Alexandra C. Willcox
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Medical Scientist Training Program, University of Washington, Seattle, WA, USA
- Molecular and Cellular Biology Program, University of Washington, Seattle, WA, USA
| | - Kevin Sung
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Meghan E. Garrett
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Molecular and Cellular Biology Program, University of Washington, Seattle, WA, USA
| | - Jared G. Galloway
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Megan A. O’Connor
- Department of Microbiology, University of Washington, Seattle, WA, USA
- Infectious Diseases and Translational Medicine, Washington National Primate Research Center, Seattle, WA, USA
| | - Jesse H. Erasmus
- Department of Microbiology, University of Washington, Seattle, WA, USA
- HDT Bio, Seattle, WA, USA
| | | | - David W. Hawman
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Helen Y. Chu
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Kim J. Hasenkrug
- Laboratory of Persistent Viral Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Deborah H. Fuller
- Department of Microbiology, University of Washington, Seattle, WA, USA
- Infectious Diseases and Translational Medicine, Washington National Primate Research Center, Seattle, WA, USA
- Center for Innate Immunity and Immune Disease, University of Washington, Seattle, WA, USA
| | - Frederick A. Matsen
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Julie Overbaugh
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| |
Collapse
|
26
|
Pérez P, Martín-Acebes MA, Poderoso T, Lázaro-Frías A, Saiz JC, Sorzano CÓS, Esteban M, García-Arriaza J. The combined vaccination protocol of DNA/MVA expressing Zika virus structural proteins as efficient inducer of T and B cell immune responses. Emerg Microbes Infect 2021; 10:1441-1456. [PMID: 34213405 PMCID: PMC8284158 DOI: 10.1080/22221751.2021.1951624] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Zika virus (ZIKV) is a mosquito-borne pathogen with public health importance due to the high risk of its mosquito vector dissemination and the severe neurological and teratogenic sequelae associated with infection. Vaccines with broad immune specificity and control against this re-emerging virus are needed. Here, we described that mice immunized with a priming dose of a DNA plasmid mammalian expression vector encoding ZIKV prM-E antigens (DNA-ZIKV) followed by a booster dose of a modified vaccinia virus Ankara (MVA) vector expressing the same prM-E ZIKV antigens (MVA-ZIKV) induced broad, polyfunctional and long-lasting ZIKV-specific CD4+ and CD8+ T-cell immune responses, with high levels of CD4+ T follicular helper cells, together with the induction of neutralizing antibodies. All those immune parameters were significantly stronger in the heterologous DNA-ZIKV/MVA-ZIKV immunization group compared to the homologous prime/boost immunizations regimens. Collectively, these results provided an optimized immunization protocol able to induce high levels of ZIKV-specific T-cell responses, as well as neutralizing antibodies and reinforce the combined use of DNA-based vectors and MVA-ZIKV as promising prophylactic vaccination schedule against ZIKV.
Collapse
Affiliation(s)
- Patricia Pérez
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Miguel A. Martín-Acebes
- Department of Biotechnology, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Teresa Poderoso
- Molecular Virology Group, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Adrián Lázaro-Frías
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Juan-Carlos Saiz
- Department of Biotechnology, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Carlos Óscar S. Sorzano
- Biocomputing Unit, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Mariano Esteban
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain, Mariano Esteban
| | - Juan García-Arriaza
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain,Juan García-Arriaza
| |
Collapse
|
27
|
He X, He C, Hong W, Zhang K, Wei X. The challenges of COVID-19 Delta variant: Prevention and vaccine development. MedComm (Beijing) 2021; 2:846-854. [PMID: 34909755 PMCID: PMC8661803 DOI: 10.1002/mco2.95] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 09/22/2021] [Accepted: 09/23/2021] [Indexed: 02/05/2023] Open
Abstract
Several SARS-CoV-2 variants have emerged since the pandemic, bringing about a renewed threat to the public. Delta variant (B.1.617.2) was first detected in October 2020 in India and was characterized as variants of concern (VOC) by WHO on May 11, 2021. Delta variant rapidly outcompeted other variants to become the dominant circulating lineages due to its clear competitive advantage. There is emerging evidence of enhanced transmissibility and reduced vaccine effectiveness (VE) against Delta variant. Therefore, it is crucial to understand the features and phenotypic effects of this variant. Herein, we comprehensively described the evaluation and features of Delta variant, summarized the effects of mutations in spike on the infectivity, transmission ability, immune evasion, and provided a perspective on efficient approaches for preventing and overcoming COVID-19.
Collapse
Affiliation(s)
- Xuemei He
- Laboratory of Aging Research and Cancer Drug TargetState Key Laboratory of Biotherapy and Cancer CenterNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengduSichuanP. R. China
| | - Cai He
- Laboratory of Aging Research and Cancer Drug TargetState Key Laboratory of Biotherapy and Cancer CenterNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengduSichuanP. R. China
| | - Weiqi Hong
- Laboratory of Aging Research and Cancer Drug TargetState Key Laboratory of Biotherapy and Cancer CenterNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengduSichuanP. R. China
| | - Kang Zhang
- Center for Biomedicine and InnovationsFaculty of MedicineMacau University of Science and TechnologyTaipaMacauP. R. China
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug TargetState Key Laboratory of Biotherapy and Cancer CenterNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengduSichuanP. R. China
| |
Collapse
|
28
|
Bi Z, Hong W, Yang J, Lu S, Peng X. Animal models for SARS-CoV-2 infection and pathology. MedComm (Beijing) 2021; 2:548-568. [PMID: 34909757 PMCID: PMC8662225 DOI: 10.1002/mco2.98] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Revised: 10/10/2021] [Accepted: 10/13/2021] [Indexed: 02/05/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the etiology of coronavirus disease 2019 (COVID-19) pandemic. Current variants including Alpha, Beta, Gamma, Delta, and Lambda increase the capacity of infection and transmission of SARS-CoV-2, which might disable the in-used therapies and vaccines. The COVID-19 has now put an enormous strain on health care system all over the world. Therefore, the development of animal models that can capture characteristics and immune responses observed in COVID-19 patients is urgently needed. Appropriate models could accelerate the testing of therapeutic drugs and vaccines against SARS-CoV-2. In this review, we aim to summarize the current animal models for SARS-CoV-2 infection, including mice, hamsters, nonhuman primates, and ferrets, and discuss the details of transmission, pathology, and immunology induced by SARS-CoV-2 in these animal models. We hope this could throw light to the increased usefulness in fundamental studies of COVID-19 and the preclinical analysis of vaccines and therapeutic agents.
Collapse
Affiliation(s)
- Zhenfei Bi
- Laboratory of Aging Research and Cancer Drug TargetState Key Laboratory of BiotherapyNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengduSichuanChina
| | - Weiqi Hong
- Laboratory of Aging Research and Cancer Drug TargetState Key Laboratory of BiotherapyNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengduSichuanChina
| | - Jingyun Yang
- Laboratory of Aging Research and Cancer Drug TargetState Key Laboratory of BiotherapyNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengduSichuanChina
| | - Shuaiyao Lu
- National Kunming High‐level Biosafety Primate Research CenterInstitute of Medical BiologyChinese Academy of Medical Sciences and Peking Union Medical CollegeYunnanChina
| | - Xiaozhong Peng
- National Kunming High‐level Biosafety Primate Research CenterInstitute of Medical BiologyChinese Academy of Medical Sciences and Peking Union Medical CollegeYunnanChina
| |
Collapse
|
29
|
Yuan L, Tang Q, Zhu H, Guan Y, Cheng T, Xia N. SARS-CoV-2 infection and disease outcomes in non-human primate models: advances and implications. Emerg Microbes Infect 2021; 10:1881-1889. [PMID: 34490832 PMCID: PMC8451603 DOI: 10.1080/22221751.2021.1976598] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 08/17/2021] [Accepted: 08/31/2021] [Indexed: 01/07/2023]
Abstract
SARS-CoV-2 has been the causative pathogen of the pandemic of COVID-19, resulting in catastrophic health issues globally. It is important to develop human-like animal models for investigating the mechanisms that SARS-CoV-2 uses to infect humans and cause COVID-19. Several studies demonstrated that the non-human primate (NHP) is permissive for SARS-CoV-2 infection to cause typical clinical symptoms including fever, cough, breathing difficulty, and other diagnostic abnormalities such as immunopathogenesis and hyperplastic lesions in the lung. These NHP models have been used for investigating the potential infection route and host immune response to SARS-CoV-2, as well as testing vaccines and drugs. This review aims to summarize the benefits and caveats of NHP models currently available for SARS-CoV-2, and to discuss key topics including model optimization, extended application, and clinical translation.
Collapse
Affiliation(s)
- Lunzhi Yuan
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, People’s Republic of China
| | - Qiyi Tang
- Department of Microbiology, Howard University College of Medicine, Washington, DC, USA
| | - Huachen Zhu
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong, People’s Republic of China
- Joint Institute of Virology (Shantou University and The University of Hong Kong), Guangdong-Hongkong Joint Laboratory of Emerging Infectious Diseases, Shantou University, Shantou, People’s Republic of China
| | - Yi Guan
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong, People’s Republic of China
- Joint Institute of Virology (Shantou University and The University of Hong Kong), Guangdong-Hongkong Joint Laboratory of Emerging Infectious Diseases, Shantou University, Shantou, People’s Republic of China
| | - Tong Cheng
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, People’s Republic of China
| | - Ningshao Xia
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, People’s Republic of China
- Research Unit of Frontier Technology of Structural Vaccinology, Chinese Academy of Medical Sciences, Xiamen, People’s Republic of China
| |
Collapse
|
30
|
Shafaati M, Saidijam M, Soleimani M, Hazrati F, Mirzaei R, Amirheidari B, Tanzadehpanah H, Karampoor S, Kazemi S, Yavari B, Mahaki H, Safaei M, Rahbarizadeh F, Samadi P, Ahmadyousefi Y. A brief review on DNA vaccines in the era of COVID-19. Future Virol 2021; 17:10.2217/fvl-2021-0170. [PMID: 34858516 PMCID: PMC8629371 DOI: 10.2217/fvl-2021-0170] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Accepted: 11/05/2021] [Indexed: 02/08/2023]
Abstract
This article provides a brief overview of DNA vaccines. First, the basic DNA vaccine design strategies are described, then specific issues related to the industrial production of DNA vaccines are discussed, including the production and purification of DNA products such as plasmid DNA, minicircle DNA, minimalistic, immunologically defined gene expression (MIDGE) and Doggybone™. The use of adjuvants to enhance the immunogenicity of DNA vaccines is then discussed. In addition, different delivery routes and several physical and chemical methods to increase the efficacy of DNA delivery into cells are explained. Recent preclinical and clinical trials of DNA vaccines for COVID-19 are then summarized. Lastly, the advantages and obstacles of DNA vaccines are discussed.
Collapse
Affiliation(s)
- Maryam Shafaati
- Department of Microbiology, Faculty of Sciences, Jahrom Branch, Islamic Azad University, Jahrom, Iran
| | - Massoud Saidijam
- Department of Medical Biotechnology, School of Advanced Medical Sciences & Technologies, Hamadan University of Medical Sciences, Hamadan, Iran
- Research Center for Molecular Medicine, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Meysam Soleimani
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Fereshte Hazrati
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Rasoul Mirzaei
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Bagher Amirheidari
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran
- Extremophile and Productive Microorganisms Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Hamid Tanzadehpanah
- Department of Medical Biotechnology, School of Advanced Medical Sciences & Technologies, Hamadan University of Medical Sciences, Hamadan, Iran
- Research Center for Molecular Medicine, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Sajad Karampoor
- Department of Virology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Sima Kazemi
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Bahram Yavari
- Department of Medical Biotechnology, School of Advanced Medical Sciences & Technologies, Hamadan University of Medical Sciences, Hamadan, Iran
- Research Center for Molecular Medicine, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Hanie Mahaki
- Vascular & Endovascular Surgery Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohsen Safaei
- Department of Medical Biotechnology, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Fatemeh Rahbarizadeh
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Pouria Samadi
- Department of Medical Biotechnology, School of Advanced Medical Sciences & Technologies, Hamadan University of Medical Sciences, Hamadan, Iran
- Research Center for Molecular Medicine, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Yaghoub Ahmadyousefi
- Department of Medical Biotechnology, School of Advanced Medical Sciences & Technologies, Hamadan University of Medical Sciences, Hamadan, Iran
- Research Center for Molecular Medicine, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
31
|
Alleva DG, Delpero AR, Scully MM, Murikipudi S, Ragupathy R, Greaves EK, Sathiyaseelan T, Haworth JR, Shah NJ, Rao V, Nagre S, Lancaster TM, Webb SS, Jasa AI, Ronca SE, Green FM, Elyard HA, Yee J, Klein J, Karnes L, Sollie F, Zion TC. Development of an IgG-Fc fusion COVID-19 subunit vaccine, AKS-452. Vaccine 2021; 39:6601-6613. [PMID: 34642088 PMCID: PMC8491978 DOI: 10.1016/j.vaccine.2021.09.077] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 09/29/2021] [Accepted: 09/30/2021] [Indexed: 12/12/2022]
Abstract
AKS-452 is a biologically-engineered vaccine comprising an Fc fusion protein of the SARS-CoV-2 viral spike protein receptor binding domain antigen (Ag) and human IgG1 Fc (SP/RBD-Fc) in clinical development for the induction and augmentation of neutralizing IgG titers against SARS-CoV-2 viral infection to address the COVID-19 pandemic. The Fc moiety is designed to enhance immunogenicity by increasing uptake via Fc-receptors (FcγR) on Ag-presenting cells (APCs) and prolonging exposure due to neonatal Fc receptor (FcRn) recycling. AKS-452 induced approximately 20-fold greater neutralizing IgG titers in mice relative to those induced by SP/RBD without the Fc moiety and induced comparable long-term neutralizing titers with a single dose vs. two doses. To further enhance immunogenicity, AKS-452 was evaluated in formulations containing a panel of adjuvants in which the water-in-oil adjuvant, Montanide™ ISA 720, enhanced neutralizing IgG titers by approximately 7-fold after one and two doses in mice, including the neutralization of live SARS-CoV-2 virus infection of VERO-E6 cells. Furthermore, ISA 720-adjuvanted AKS-452 was immunogenic in rabbits and non-human primates (NHPs) and protected from infection and clinical symptoms with live SARS-CoV-2 virus in NHPs (USA-WA1/2020 viral strain) and the K18 human ACE2-trangenic (K18-huACE2-Tg) mouse (South African B.1.351 viral variant). These preclinical studies support the initiation of Phase I clinical studies with adjuvanted AKS-452 with the expectation that this room-temperature stable, Fc-fusion subunit vaccine can be rapidly and inexpensively manufactured to provide billions of doses per year especially in regions where the cold-chain is difficult to maintain.
Collapse
Affiliation(s)
- David G Alleva
- Akston Biosciences Corporation., 100 Cummings Center, Suite 454C, Beverly, MA 01915, United States
| | - Andrea R Delpero
- Akston Biosciences Corporation., 100 Cummings Center, Suite 454C, Beverly, MA 01915, United States
| | - Melanie M Scully
- Akston Biosciences Corporation., 100 Cummings Center, Suite 454C, Beverly, MA 01915, United States
| | - Sylaja Murikipudi
- Akston Biosciences Corporation., 100 Cummings Center, Suite 454C, Beverly, MA 01915, United States
| | - Ramya Ragupathy
- Akston Biosciences Corporation., 100 Cummings Center, Suite 454C, Beverly, MA 01915, United States
| | - Emma K Greaves
- Akston Biosciences Corporation., 100 Cummings Center, Suite 454C, Beverly, MA 01915, United States
| | | | - Jeffrey R Haworth
- Akston Biosciences Corporation., 100 Cummings Center, Suite 454C, Beverly, MA 01915, United States
| | - Nishit J Shah
- Akston Biosciences Corporation., 100 Cummings Center, Suite 454C, Beverly, MA 01915, United States
| | - Vidhya Rao
- Akston Biosciences Corporation., 100 Cummings Center, Suite 454C, Beverly, MA 01915, United States
| | - Shashikant Nagre
- Akston Biosciences Corporation., 100 Cummings Center, Suite 454C, Beverly, MA 01915, United States
| | - Thomas M Lancaster
- Akston Biosciences Corporation., 100 Cummings Center, Suite 454C, Beverly, MA 01915, United States
| | - Sarah S Webb
- Biomere Biomedical Research Models, 57 Union St., Worcester, MA 01608, United States
| | - Allison I Jasa
- Biomere Biomedical Research Models, 57 Union St., Worcester, MA 01608, United States
| | - Shannon E Ronca
- Feigin ABSL-3 Facility, Baylor, College of Medicine, 1102 Bates Ave, 300.15, Houston, TX 77030, United States
| | - Freedom M Green
- Feigin ABSL-3 Facility, Baylor, College of Medicine, 1102 Bates Ave, 300.15, Houston, TX 77030, United States
| | - Hanne Andersen Elyard
- BIOQUAL, Inc., 9600 Medical Center Drive, Suite 101, Rockville, MD 20850-3336, United States
| | - JoAnn Yee
- Primate Assay Laboratory, CA National Primate Research Center, University of California, Davis, CA 95616, United States
| | - Jeffrey Klein
- Sinclair Research Center, 562 State Road DD, Auxvasse, MO 65231, United States
| | - Larry Karnes
- Sinclair Research Center, 562 State Road DD, Auxvasse, MO 65231, United States
| | - Frans Sollie
- Pharmaceutical Research Associates Group B.V., Amerikaweg 18, 9407 TK Assen, Netherlands
| | - Todd C Zion
- Akston Biosciences Corporation., 100 Cummings Center, Suite 454C, Beverly, MA 01915, United States.
| |
Collapse
|
32
|
Wang S, Li L, Yan F, Gao Y, Yang S, Xia X. COVID-19 Animal Models and Vaccines: Current Landscape and Future Prospects. Vaccines (Basel) 2021; 9:1082. [PMID: 34696190 PMCID: PMC8537799 DOI: 10.3390/vaccines9101082] [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: 08/15/2021] [Revised: 09/17/2021] [Accepted: 09/23/2021] [Indexed: 12/23/2022] Open
Abstract
The worldwide pandemic of coronavirus disease 2019 (COVID-19) has become an unprecedented challenge to global public health. With the intensification of the COVID-19 epidemic, the development of vaccines and therapeutic drugs against the etiological agent severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is also widespread. To prove the effectiveness and safety of these preventive vaccines and therapeutic drugs, available animal models that faithfully recapitulate clinical hallmarks of COVID-19 are urgently needed. Currently, animal models including mice, golden hamsters, ferrets, nonhuman primates, and other susceptible animals have been involved in the study of COVID-19. Moreover, 117 vaccine candidates have entered clinical trials after the primary evaluation in animal models, of which inactivated vaccines, subunit vaccines, virus-vectored vaccines, and messenger ribonucleic acid (mRNA) vaccines are promising vaccine candidates. In this review, we summarize the landscape of animal models for COVID-19 vaccine evaluation and advanced vaccines with an efficacy range from about 50% to more than 95%. In addition, we point out future directions for COVID-19 animal models and vaccine development, aiming at providing valuable information and accelerating the breakthroughs confronting SARS-CoV-2.
Collapse
Affiliation(s)
- Shen Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130122, China; (S.W.); (X.X.)
| | - Ling Li
- National Research Center for Exotic Animal Diseases, China Animal Health and Epidemiology Center, Qingdao 266000, China;
| | - Feihu Yan
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130122, China; (S.W.); (X.X.)
| | - Yuwei Gao
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130122, China; (S.W.); (X.X.)
| | - Songtao Yang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130122, China; (S.W.); (X.X.)
| | - Xianzhu Xia
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130122, China; (S.W.); (X.X.)
| |
Collapse
|
33
|
Rosati M, Agarwal M, Hu X, Devasundaram S, Stellas D, Chowdhury B, Bear J, Burns R, Donohue D, Pessaint L, Andersen H, Lewis MG, Terpos E, Dimopoulos MA, Wlodawer A, Mullins JI, Venzon DJ, Pavlakis GN, Felber BK. Control of SARS-CoV-2 infection after Spike DNA or Spike DNA+Protein co-immunization in rhesus macaques. PLoS Pathog 2021; 17:e1009701. [PMID: 34551020 PMCID: PMC8489704 DOI: 10.1371/journal.ppat.1009701] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 10/04/2021] [Accepted: 09/07/2021] [Indexed: 12/28/2022] Open
Abstract
The speed of development, versatility and efficacy of mRNA-based vaccines have been amply demonstrated in the case of SARS-CoV-2. DNA vaccines represent an important alternative since they induce both humoral and cellular immune responses in animal models and in human trials. We tested the immunogenicity and protective efficacy of DNA-based vaccine regimens expressing different prefusion-stabilized Wuhan-Hu-1 SARS-CoV-2 Spike antigens upon intramuscular injection followed by electroporation in rhesus macaques. Different Spike DNA vaccine regimens induced antibodies that potently neutralized SARS-CoV-2 in vitro and elicited robust T cell responses. The antibodies recognized and potently neutralized a panel of different Spike variants including Alpha, Delta, Epsilon, Eta and A.23.1, but to a lesser extent Beta and Gamma. The DNA-only vaccine regimens were compared to a regimen that included co-immunization of Spike DNA and protein in the same anatomical site, the latter of which showed significant higher antibody responses. All vaccine regimens led to control of SARS-CoV-2 intranasal/intratracheal challenge and absence of virus dissemination to the lower respiratory tract. Vaccine-induced binding and neutralizing antibody titers and antibody-dependent cellular phagocytosis inversely correlated with transient virus levels in the nasal mucosa. Importantly, the Spike DNA+Protein co-immunization regimen induced the highest binding and neutralizing antibodies and showed the strongest control against SARS-CoV-2 challenge in rhesus macaques. Anti-Spike neutralizing antibodies provide strong protection against SARS-CoV-2 infection in animal models, and correlate with protection in humans, supporting the notion that induction of strong humoral immunity is key to protection. We show induction of robust antibody and T cell responses by different Spike DNA-based vaccine regimens able to effectively mediate protection and to control SARS-CoV-2 infection in the rhesus macaque model. This study provides the opportunity to compare vaccines able to induce different humoral and cellular immune responses in an effort to develop durable immunity against the SARS-CoV-2. A vaccine regimen comprising simultaneous co-immunization of DNA and Protein at the same anatomical site showed best neutralizing abilities and was more effective than DNA alone in inducing protective immune responses and controlling SARS-CoV-2 infection. Thus, an expansion of the DNA vaccine regimen to include co-immunization with Spike protein may be of advantage also for SARS-CoV-2.
Collapse
Affiliation(s)
- Margherita Rosati
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, Maryland, United States of America
| | - Mahesh Agarwal
- Human Retrovirus Pathogenesis Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, Maryland, United States of America
| | - Xintao Hu
- Human Retrovirus Pathogenesis Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, Maryland, United States of America
| | - Santhi Devasundaram
- Human Retrovirus Pathogenesis Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, Maryland, United States of America
| | - Dimitris Stellas
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, Maryland, United States of America
| | - Bhabadeb Chowdhury
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, Maryland, United States of America
| | - Jenifer Bear
- Human Retrovirus Pathogenesis Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, Maryland, United States of America
| | - Robert Burns
- Human Retrovirus Pathogenesis Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, Maryland, United States of America
| | - Duncan Donohue
- MS Applied Information and Management Sciences, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | | | - Hanne Andersen
- BIOQUAL, Inc.; Rockville, Maryland, United States of America
| | - Mark G. Lewis
- BIOQUAL, Inc.; Rockville, Maryland, United States of America
| | - Evangelos Terpos
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | | | - Alexander Wlodawer
- Center for Structural Biology, National Cancer Institute, Frederick, Maryland, United States of America
| | - James I. Mullins
- Department of Microbiology, University of Washington, Seattle, Washington, United States of America
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
- Department of Global Health, University of Washington, Seattle, Washington, United States of America
| | - David J. Venzon
- Biostatistics and Data Management Section, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - George N. Pavlakis
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, Maryland, United States of America
| | - Barbara K. Felber
- Human Retrovirus Pathogenesis Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, Maryland, United States of America
- * E-mail:
| |
Collapse
|
34
|
Zhang J, He Q, An C, Mao Q, Gao F, Bian L, Wu X, Wang Q, Liu P, Song L, Huo Y, Liu S, Yan X, Yang J, Cui B, Li C, Wang J, Liang Z, Xu M. Boosting with heterologous vaccines effectively improves protective immune responses of the inactivated SARS-CoV-2 vaccine. Emerg Microbes Infect 2021; 10:1598-1608. [PMID: 34278956 PMCID: PMC8381941 DOI: 10.1080/22221751.2021.1957401] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Since the outbreak of COVID-19, a variety of vaccine platforms have been developed. Amongst these, inactivated vaccines have been authorized for emergency use or conditional marketing in many countries. To further enhance the protective immune responses in populations that have completed vaccination regimen, we investigated the immunogenic characteristics of different vaccine platforms and tried homologous or heterologous boost strategy post two doses of inactivated vaccines in a mouse model. Our results showed that the humoral and cellular immune responses induced by different vaccines when administered individually differ significantly. In particular, inactivated vaccines showed relatively lower level of neutralizing antibody and T cell responses, but a higher IgG2a/IgG1 ratio compared with other vaccines. Boosting with either recombinant subunit, adenovirus vectored or mRNA vaccine after two-doses of inactivated vaccine further improved both neutralizing antibody and Spike-specific Th1-type T cell responses compared to boosting with a third dose of inactivated vaccine. Our results provide new ideas for prophylactic inoculation strategy of SARS-CoV-2 vaccines.
Collapse
Affiliation(s)
- Jialu Zhang
- National Institutes for Food and Drug Control, Beijing, People's Republic of China
| | - Qian He
- National Institutes for Food and Drug Control, Beijing, People's Republic of China
| | - Chaoqiang An
- National Institutes for Food and Drug Control, Beijing, People's Republic of China
| | - Qunying Mao
- National Institutes for Food and Drug Control, Beijing, People's Republic of China
| | - Fan Gao
- National Institutes for Food and Drug Control, Beijing, People's Republic of China
| | - Lianlian Bian
- National Institutes for Food and Drug Control, Beijing, People's Republic of China
| | - Xing Wu
- National Institutes for Food and Drug Control, Beijing, People's Republic of China
| | - Qian Wang
- National Institutes for Food and Drug Control, Beijing, People's Republic of China
| | - Pei Liu
- National Institutes for Food and Drug Control, Beijing, People's Republic of China
| | - Lifang Song
- National Institutes for Food and Drug Control, Beijing, People's Republic of China
| | - Yaqian Huo
- National Institutes for Food and Drug Control, Beijing, People's Republic of China
| | - Siyuan Liu
- National Institutes for Food and Drug Control, Beijing, People's Republic of China
| | - Xujia Yan
- National Institutes for Food and Drug Control, Beijing, People's Republic of China
| | - Jinghuan Yang
- National Institutes for Food and Drug Control, Beijing, People's Republic of China
| | - Bopei Cui
- National Institutes for Food and Drug Control, Beijing, People's Republic of China
| | - Changgui Li
- National Institutes for Food and Drug Control, Beijing, People's Republic of China
| | - Junzhi Wang
- National Institutes for Food and Drug Control, Beijing, People's Republic of China
| | - Zhenglun Liang
- National Institutes for Food and Drug Control, Beijing, People's Republic of China
| | - Miao Xu
- National Institutes for Food and Drug Control, Beijing, People's Republic of China
| |
Collapse
|
35
|
Wang Z, Zhao X, Wang Y, Sun C, Sun M, Gao X, Jia F, Shan C, Yang G, Wang J, Huang H, Shi C, Yang W, Qian A, Wang C, Jiang Y. In Vivo Production of HN Protein Increases the Protection Rates of a Minicircle DNA Vaccine against Genotype VII Newcastle Disease Virus. Vaccines (Basel) 2021; 9:vaccines9070723. [PMID: 34358140 PMCID: PMC8310180 DOI: 10.3390/vaccines9070723] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 06/26/2021] [Accepted: 06/26/2021] [Indexed: 01/09/2023] Open
Abstract
The Cre-recombinase mediated in vivo minicircle DNA vaccine platform (CRIM) provided a novel option to replace a traditional DNA vaccine. To further improve the immune response of our CRIM vaccine, we designed a dual promoter expression plasmid named pYL87 which could synthesize short HN protein under a prokaryotic in vivo promoter PpagC and full length HN protein of genotype VII Newcastle disease virus (NDV) under the previous eukaryotic CMV promoter at the same time. Making use of the self-lysed Salmonella strain as a delivery vesicle, chickens immunized with the pYL87 construction showed an increased serum haemagglutination inhibition antibody response, as well as an increased cell proliferation level and cellular IL-4 and IL-18 cytokines, compared with the previous CRIM vector pYL47. After the virus challenge, the pYL87 vector could provide 80% protection compared to 50% protection against genotype VII NDV in pYL47 immunized chickens, indicating a promising dual promoter strategy used in vaccine design.
Collapse
|
36
|
Huang C, Jiang Y, Yan J. Comparative analyses of ACE2 and TMPRSS2 gene: Implications for the risk to which vertebrate animals are susceptible to SARS-CoV-2. J Med Virol 2021; 93:5487-5504. [PMID: 33974296 PMCID: PMC8242802 DOI: 10.1002/jmv.27073] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 05/01/2021] [Accepted: 05/03/2021] [Indexed: 12/30/2022]
Abstract
Along with the control and prevention of coronavirus disease 2019 transmission, infected animals might have potential to carry the virus to spark new outbreaks. However, very few studies explore the susceptibility of animals to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Viral attachment as a crucial step for cross-species infection requires angiotensin-converting enzyme 2 (ACE2) as a receptor and depends on TMPRSS2 protease activity. Here, we searched the genomes of metazoans from different classes using an extensive BLASTP survey and found ACE2 and TMPRSS2 occur in vertebrates, but some vertebrates lack Tmprss2. We identified 6 amino acids among 25 known human ACE2 residues are highly associated with the binding of ACE2 to SARS-CoV-2 (p value < .01) by Fisher exact test, and following this, calculated the probability of viral attachment within each species by the randomForest function from R randomForest library. Furthermore, we observed that Ace2 selected from seven animals based on the above analysis lack the hydrophobic contacts identified on human ACE2, indicating less affinity of SARS-CoV-2 to Ace2 in animals than humans. Finally, the alignment of 3D structure between human ACE2 and other animals by I-TASSER and TM-align displayed a reasonable structure for viral attachment within these species. Taken together, our data may shed light on the human-to-animal transmission of SARS-CoV-2.
Collapse
Affiliation(s)
- Chen Huang
- Department of Veterinary Science, University of Kentucky, Lexington, Kentucky, USA
| | - Yu Jiang
- Division of Epidemiology, Biostatistics, and Environmental Health, School of Public Health, University of Memphis, Memphis, Tennessee, USA
| | - Jie Yan
- Department of Diagnosis, School of Medicine, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| |
Collapse
|
37
|
Li H, Wang S, Hu G, Zhang L, Liu S, Lu S. DNA priming immunization is more effective than recombinant protein vaccine in eliciting antigen-specific B cell responses. Emerg Microbes Infect 2021; 10:833-841. [PMID: 33853515 PMCID: PMC8812797 DOI: 10.1080/22221751.2021.1918026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
While DNA prime-protein boost vaccination approach has been widely used in preclinical and clinical studies especially in the field of HIV vaccine development, the exact role of DNA immunization has not been fully identified. Our previous work demonstrated that DNA immunization was able to elicit T follicular helper (Tfh) cell responses and germinal center (GC) B cell development in a mouse model. In the current report, a mouse immunogenicity study was conducted to further ask whether DNA immunization is able to elicit antigen-specific B cell responses. Using HIV-1 Env as model antigen delivered in the form of DNA prime-protein boost, our data demonstrated that DNA prime was able to enhance the antigen-specific B cell responses for both Env-specific antibody secreting cells (ASC) and memory B cells. Furthermore, the DNA priming can greatly reduce the need of including an adjuvant as part of the recombinant protein vaccine boost formulation. Our findings revealed one mechanism that supports the value of DNA priming in assisting the inductin of high affinity and long lasting antigen specific antibody responses.
Collapse
Affiliation(s)
- Haiying Li
- Laboratory of Nucleic Acid Vaccines, Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Shixia Wang
- Laboratory of Nucleic Acid Vaccines, Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Guangnan Hu
- Laboratory of Nucleic Acid Vaccines, Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Lu Zhang
- Department of General Surgery, The First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | | | - Shan Lu
- Laboratory of Nucleic Acid Vaccines, Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| |
Collapse
|
38
|
Ulmer JB, Liu MA. Path to Success and Future Impact of Nucleic Acid Vaccines: DNA and mRNA. MOLECULAR FRONTIERS JOURNAL 2021. [DOI: 10.1142/s2529732521400022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The rapid development of mRNA vaccines for COVID-19 has both astonished the world and raised concerns about their safety, perhaps because many people do not realize the decades’ long efforts for nucleic acid vaccines, both mRNA and DNA vaccines, including the licensure of several veterinary DNA vaccines. This manuscript traces the milestones for nucleic acid vaccine research and development (R&D), with a focus on the immune and safety issues they both raised and answered. The characteristics of the two entities are compared, demonstrating the similarities and differences between them, the advantages and disadvantages, which might lead toward using one or the other technology for different indications. In addition, as the SARS-CoV-2 pandemic has once again highlighted the importance of One Health, that is, the interactions between animal and human pathogens, focus will also be given to how DNA vaccine utilization and studies both in large domestic animals and in wildlife pave the way for more integrated approaches for vaccines to respond quickly to, and prevent, the global impacts of emerging diseases.
Collapse
|