1
|
Prashar N, Mohammed SB, Raja NS, Mohideen HS. Rerouting therapeutic peptides and unlocking their potential against SARS-CoV2. 3 Biotech 2025; 15:116. [PMID: 40191455 PMCID: PMC11971104 DOI: 10.1007/s13205-025-04270-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Accepted: 03/10/2025] [Indexed: 04/09/2025] Open
Abstract
The COVID-19 pandemic highlighted the potential of peptide-based therapies as an alternative to traditional pharmaceutical treatments for SARS-CoV-2 and its variants. Our review explores the role of therapeutic peptides in modulating immune responses, inhibiting viral entry, and disrupting replication. Despite challenges such as stability, bioavailability, and the rapid mutation of the virus, ongoing research and clinical trials show that peptide-based treatments are increasingly becoming integral to future viral outbreak responses. Advancements in computational modelling methods in combination with artificial intelligence will enable mass screening of therapeutic peptides and thereby, comprehending a peptide repurposing strategy similar to the small molecule repurposing. These findings suggest that peptide-based therapies play a critical and promising role in future pandemic preparedness and outbreak management.
Collapse
Affiliation(s)
- Namrata Prashar
- Bioinformatics and Entomoinformatics Lab, Department of Genetic Engineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu, Chennai, Tamil Nadu 603203 India
| | - Saharuddin Bin Mohammed
- Institute of Biological Sciences, Faculty of Science, Universiti Malaya, 50603 Kuala Lumpur, Malaysia
| | - N. S. Raja
- Deparmtent of Genetic Engineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu, Chennai, Tamil Nadu 603203 India
| | - Habeeb Shaik Mohideen
- Bioinformatics and Entomoinformatics Lab, Department of Genetic Engineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu, Chennai, Tamil Nadu 603203 India
| |
Collapse
|
2
|
Shah M, Moon SU, Shin J, Choi J, Kim D, Woo HG. Pan-Variant SARS-CoV-2 Vaccines Induce Protective Immunity by Targeting Conserved Epitopes. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2409919. [PMID: 40014015 PMCID: PMC12021035 DOI: 10.1002/advs.202409919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 01/27/2025] [Indexed: 02/28/2025]
Abstract
The development of a globally effective COVID-19 vaccine faces significant challenges, particularly in redirecting the B-cell response from immunodominant yet variable regions of viral proteins toward their conserved domains. To address this, an integrated strategy is implemented that combines classical B-cell epitope prediction with protein-antibody cluster docking and antibody titer analysis from 30 vaccinated and convalescent individuals. This approach yields stable immunodominant and immunoprevalent B-cell epitopes capable of eliciting robust antibody responses in BALB/c mice and effectively neutralizing pseudoviruses expressing the Spike protein of SARS-CoV-2 variants of concern, including Alpha, Beta, Gamma, Delta, and Omicron. To achieve a broader T-cell-based immune response, promiscuous T-cell epitopes are identified by integrating classical T-cell epitope predictions, differential scanning fluorimetry, and peptide-MHC structural analysis. Unique peptides with conserved MHC-anchoring residues are identified, enabling binding to a spectrum of MHC-I and MHC-II haplotypes. These peptides elicit strong interferon gamma responses in human peripheral blood mononuclear cells and demonstrate cross-species efficacy by activating both CD4+ and CD8+ T-cells in BALB/c mice. Collectively, these findings highlight the significance of innovative vaccine strategies targeting immunodominant/immunoprevalent B-cell and promiscuous T-cell epitopes to drive broad and robust humoral and cellular immune responses against a wide range of SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Masaud Shah
- Department of PhysiologyAjou University School of MedicineSuwon16499Republic of Korea
- Ajou Translational Omics Center (ATOC)Research Institute for Innovative MedicineAjou University Medical CenterSuwon16499Republic of Korea
| | - Sung Ung Moon
- Department of PhysiologyAjou University School of MedicineSuwon16499Republic of Korea
| | - Ji‐Yon Shin
- Department of PhysiologyAjou University School of MedicineSuwon16499Republic of Korea
- AI‐Superconvergence KIURI Translational Research CenterAjou University School of MedicineSuwon16499Republic of Korea
| | - Ji‐Hye Choi
- Department of PhysiologyAjou University School of MedicineSuwon16499Republic of Korea
- Ajou Translational Omics Center (ATOC)Research Institute for Innovative MedicineAjou University Medical CenterSuwon16499Republic of Korea
| | - Doyoon Kim
- Ajou Translational Omics Center (ATOC)Research Institute for Innovative MedicineAjou University Medical CenterSuwon16499Republic of Korea
| | - Hyun Goo Woo
- Department of PhysiologyAjou University School of MedicineSuwon16499Republic of Korea
- Ajou Translational Omics Center (ATOC)Research Institute for Innovative MedicineAjou University Medical CenterSuwon16499Republic of Korea
- Department of Biomedical ScienceGraduate SchoolAjou UniversitySuwon16499Republic of Korea
| |
Collapse
|
3
|
Vega Rojas LJ, Ruíz-Manzano RA, Velasco-Elizondo MA, Carbajo-Mata MA, Hernández-Silva DJ, Rocha-Solache M, Hernández J, Pérez-Serrano RM, Zaldívar-Lelo de Larrea G, García-Gasca T, Mosqueda J. An Evaluation of the Cellular and Humoral Response of a Multi-Epitope Vaccine Candidate Against COVID-19 with Different Alum Adjuvants. Pathogens 2024; 13:1081. [PMID: 39770342 PMCID: PMC11728595 DOI: 10.3390/pathogens13121081] [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: 10/16/2024] [Revised: 12/03/2024] [Accepted: 12/04/2024] [Indexed: 01/16/2025] Open
Abstract
SARS-CoV-2 (Betacoronavirus pandemicum) is responsible for the disease identified by the World Health Organization (WHO) as COVID-19. We designed "CHIVAX 2.1", a multi-epitope vaccine, containing ten immunogenic peptides with conserved B-cell and T-cell epitopes in the receceptor binding domain (RBD) sequences of different SARS-CoV-2 variants of concern (VoCs). We evaluated the immune response of mice immunized with 20 or 60 µg of the chimeric protein with two different alum adjuvants (Alhydrogel® and Adju-Phos®), plus PHAD®, in a two-immunization regimen (0 and 21 days). Serum samples were collected on days 0, 21, 31, and 72 post first immunization, with antibody titers determined by indirect ELISA, while lymphoproliferation assays and cytokine production were evaluated by flow cytometry. The presence of neutralizing antibodies was assessed by surrogate neutralization assays. Higher titers of total IgG, IgG1, and IgG2a antibodies, as well as increased proliferation rates of specific CD4+ and CD8+ T cells, were observed in mice immunized with 60 μg of protein plus Adju-Phos®/PHAD®. This formulation also generated the highest levels of TNF-α and IFN-γ, in addition to the presence of neutralizing antibodies against Delta and Omicron VoC. These findings indicate the potential of this chimeric multi-epitope vaccine with combined adjuvants as a promising platform against viral infections, eliciting a TH1 or TH1:TH2 balanced cell response.
Collapse
MESH Headings
- Animals
- Mice
- COVID-19 Vaccines/immunology
- COVID-19/prevention & control
- COVID-19/immunology
- SARS-CoV-2/immunology
- Alum Compounds/pharmacology
- Alum Compounds/administration & dosage
- Adjuvants, Immunologic/pharmacology
- Adjuvants, Immunologic/administration & dosage
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- Immunity, Humoral/drug effects
- Immunity, Humoral/immunology
- Antibodies, Neutralizing/blood
- Antibodies, Neutralizing/immunology
- Female
- Epitopes, T-Lymphocyte/immunology
- Immunity, Cellular/drug effects
- Immunity, Cellular/immunology
- Mice, Inbred BALB C
- Adjuvants, Vaccine/pharmacology
- Epitopes, B-Lymphocyte/immunology
- Humans
- Immunoglobulin G/blood
- Immunoglobulin G/immunology
- CD8-Positive T-Lymphocytes/immunology
- Cytokines/metabolism
- Spike Glycoprotein, Coronavirus/immunology
Collapse
Affiliation(s)
- Lineth Juliana Vega Rojas
- Immunology and Vaccines Laboratory, Facultad de Ciencias Naturales, Universidad Autónoma de Querétaro, Campus Aeropuerto, Carretera a Chichimequillas, Ejido Bolaños, Querétaro 76140, Mexico; (L.J.V.R.); (R.A.R.-M.); (M.A.V.-E.); (D.J.H.-S.); (M.R.-S.)
- Consejo Nacional de Humanidades, Ciencias y Tecnologías (CONAHCYT), Av. Insurgentes Sur 1582, Alcaldía Benito Juárez, Crédito Constructor, Ciudad de México 03940, Mexico
| | - Rocío Alejandra Ruíz-Manzano
- Immunology and Vaccines Laboratory, Facultad de Ciencias Naturales, Universidad Autónoma de Querétaro, Campus Aeropuerto, Carretera a Chichimequillas, Ejido Bolaños, Querétaro 76140, Mexico; (L.J.V.R.); (R.A.R.-M.); (M.A.V.-E.); (D.J.H.-S.); (M.R.-S.)
| | - Miguel Andrés Velasco-Elizondo
- Immunology and Vaccines Laboratory, Facultad de Ciencias Naturales, Universidad Autónoma de Querétaro, Campus Aeropuerto, Carretera a Chichimequillas, Ejido Bolaños, Querétaro 76140, Mexico; (L.J.V.R.); (R.A.R.-M.); (M.A.V.-E.); (D.J.H.-S.); (M.R.-S.)
| | - María Antonieta Carbajo-Mata
- Instituto de Neurobiología UNAM, Laboratorio Universitario del Bioterio, Universidad Nacional Autónoma de México, Querétaro 76230, Mexico
| | - Diego Josimar Hernández-Silva
- Immunology and Vaccines Laboratory, Facultad de Ciencias Naturales, Universidad Autónoma de Querétaro, Campus Aeropuerto, Carretera a Chichimequillas, Ejido Bolaños, Querétaro 76140, Mexico; (L.J.V.R.); (R.A.R.-M.); (M.A.V.-E.); (D.J.H.-S.); (M.R.-S.)
| | - Mariana Rocha-Solache
- Immunology and Vaccines Laboratory, Facultad de Ciencias Naturales, Universidad Autónoma de Querétaro, Campus Aeropuerto, Carretera a Chichimequillas, Ejido Bolaños, Querétaro 76140, Mexico; (L.J.V.R.); (R.A.R.-M.); (M.A.V.-E.); (D.J.H.-S.); (M.R.-S.)
| | - Jesús Hernández
- Laboratorio de Inmunología, Centro de Investigación en Alimentación y Desarrollo, A.C, Hermosillo 83304, Mexico;
| | - Rosa Martha Pérez-Serrano
- Advanced Biomedical Research Center, School of Medicine, Universidad Autónoma de Querétaro, Querétaro 76176, Mexico; (R.M.P.-S.); (G.Z.-L.d.L.)
| | - Guadalupe Zaldívar-Lelo de Larrea
- Advanced Biomedical Research Center, School of Medicine, Universidad Autónoma de Querétaro, Querétaro 76176, Mexico; (R.M.P.-S.); (G.Z.-L.d.L.)
| | - Teresa García-Gasca
- Facultad de Ciencias Naturales, Universidad Autónoma de Querétaro, Av. de las Ciencias s/n, Juriquilla, Querétaro 76230, Mexico
| | - Juan Mosqueda
- Immunology and Vaccines Laboratory, Facultad de Ciencias Naturales, Universidad Autónoma de Querétaro, Campus Aeropuerto, Carretera a Chichimequillas, Ejido Bolaños, Querétaro 76140, Mexico; (L.J.V.R.); (R.A.R.-M.); (M.A.V.-E.); (D.J.H.-S.); (M.R.-S.)
| |
Collapse
|
4
|
Zornikova K, Dianov D, Ivanova N, Davydova V, Nenasheva T, Fefelova E, Bogolyubova A. Features of Highly Homologous T-Cell Receptor Repertoire in the Immune Response to Mutations in Immunogenic Epitopes. Int J Mol Sci 2024; 25:12591. [PMID: 39684303 DOI: 10.3390/ijms252312591] [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: 10/18/2024] [Revised: 11/21/2024] [Accepted: 11/21/2024] [Indexed: 12/18/2024] Open
Abstract
CD8+ T-cell immunity, mediated through interactions between human leukocyte antigen (HLA) and the T-cell receptor (TCR), plays a pivotal role in conferring immune memory and protection against viral infections. The emergence of SARS-CoV-2 variants presents a significant challenge to the existing population immunity. While numerous SARS-CoV-2 mutations have been associated with immune evasion from CD8+ T cells, the molecular effects of most mutations on epitope-specific TCR recognition remain largely unexplored, particularly for epitope-specific repertoires characterized by common TCRs. In this study, we investigated an HLA-A*24-restricted NYN epitope (Spike448-456) that elicits broad and highly homologous CD8+ T cell responses in COVID-19 patients. Eleven naturally occurring mutations in the NYN epitope, all of which retained cell surface presentation by HLA, were tested against four transgenic Jurkat reporter cell lines. Our findings demonstrate that, with the exception of L452R and the combined mutation L452Q + Y453F, these mutations have minimal impact on the avidity of recognition by NYN peptide-specific TCRs. Additionally, we observed that a similar TCR responded differently to mutant epitopes and demonstrated cross-reactivity to the unrelated VYF epitope (ORF3a112-120). The results contradict the idea that immune responses with limited receptor diversity are insufficient to provide protection against emerging variants.
Collapse
Affiliation(s)
- Ksenia Zornikova
- National Medical Research Center for Hematology, Moscow 125167, Russia
| | - Dmitry Dianov
- National Medical Research Center for Hematology, Moscow 125167, Russia
| | - Natalia Ivanova
- National Medical Research Center for Hematology, Moscow 125167, Russia
| | - Vassa Davydova
- National Medical Research Center for Hematology, Moscow 125167, Russia
| | - Tatiana Nenasheva
- National Medical Research Center for Hematology, Moscow 125167, Russia
| | | | | |
Collapse
|
5
|
Chang TY, Li CJ, Chao TL, Chang SY, Chang SC. Design of the conserved epitope peptide of SARS-CoV-2 spike protein as the broad-spectrum COVID-19 vaccine. Appl Microbiol Biotechnol 2024; 108:486. [PMID: 39412657 PMCID: PMC11485143 DOI: 10.1007/s00253-024-13331-y] [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: 08/29/2024] [Revised: 10/02/2024] [Accepted: 10/10/2024] [Indexed: 10/19/2024]
Abstract
Our previous study has found that monoclonal antibodies targeting a conserved epitope peptide spanning from residues 1144 to 1156 of SARS-CoV-2 spike (S) protein, namely S(1144-1156), can broadly neutralize all of the prevalent SARS-CoV-2 strains, including the wild type, Alpha, Epsilon, Delta, and Gamma variants. In the study, S(1144-1156) was conjugated with bovine serum albumin (BSA) and formulated with Montanide ISA 51 adjuvant for inoculation in BALB/c mice to study its potential as a vaccine candidate. Results showed that the titers of S protein-specific IgGs and the neutralizing antibodies in mouse sera against various SARS-CoV-2 variants, including the Omicron sublineages, were largely induced along with three doses of immunization. The significant release of IFN-γ and IL-2 was also observed by ELISpot assays through stimulating vaccinated mouse splenocytes with the S(1144-1156) peptide. Furthermore, the vaccination of the S(1143-1157)- and S(1142-1158)-EGFP fusion proteins can elicit more SARS-CoV-2 neutralizing antibodies in mouse sera than the S(1144-1156)-EGFP fusion protein. Interestingly, the antisera collected from mice inoculated with the S(1144-1156) peptide vaccine exhibited better efficacy for neutralizing Omicron BA.2.86 and JN.1 subvariants than Omicron BA.1, BA.2, and XBB subvariants. Since the amino acid sequences of the S(1144-1156) are highly conserved among various SARS-CoV-2 variants, the immunogen containing the S(1144-1156) core epitope can be designed as a broadly effective COVID-19 vaccine. KEY POINTS: • Inoculation of mice with the S(1144-1156) peptide vaccine can induce bnAbs against various SARS-CoV-2 variants. • The S(1144-1156) peptide stimulated significant release of IFN-γ and IL-2 in vaccinated mouse splenocytes. • The S(1143-1157) and S(1142-1158) peptide vaccines can elicit more SARS-CoV-2 nAbs in mice.
Collapse
Affiliation(s)
- Ting-Yu Chang
- Department of Biochemical Science and Technology, College of Life Science, National Taiwan University, Taipei, 106, Taiwan
| | - Chia-Jung Li
- Department of Biochemical Science and Technology, College of Life Science, National Taiwan University, Taipei, 106, Taiwan
| | - Tai-Ling Chao
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei, 100, Taiwan
| | - Sui-Yuan Chang
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei, 100, Taiwan.
- Department of Laboratory Medicine, College of Medicine, National Taiwan University Hospital, National Taiwan University, Taipei, 100, Taiwan.
| | - Shih-Chung Chang
- Department of Biochemical Science and Technology, College of Life Science, National Taiwan University, Taipei, 106, Taiwan.
- Center of Biotechnology, National Taiwan University, Taipei, 106, Taiwan.
| |
Collapse
|
6
|
Zhang X, Luo F, Zhang H, Guo H, Zhou J, Li T, Chen S, Song S, Shen M, Wu Y, Gao Y, Han X, Wang Y, Hu C, Zhao X, Guo H, Zhang D, Lu Y, Wang W, Wang K, Tang N, Jin T, Ding M, Luo S, Lin C, Lu T, Lu B, Tian Y, Yang C, Cheng G, Yang H, Jin A, Ji X, Gong R, Chiu S, Huang A. Prophylactic efficacy of an intranasal spray with 2 synergetic antibodies neutralizing Omicron. JCI Insight 2024; 9:e171034. [PMID: 38587080 PMCID: PMC11128199 DOI: 10.1172/jci.insight.171034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 02/27/2024] [Indexed: 04/09/2024] Open
Abstract
BACKGROUNDAs Omicron is prompted to replicate in the upper airway, neutralizing antibodies (NAbs) delivered through inhalation might inhibit early-stage infection in the respiratory tract. Thus, elucidating the prophylactic efficacy of NAbs via nasal spray addresses an important clinical need.METHODSThe applicable potential of a nasal spray cocktail containing 2 NAbs was characterized by testing its neutralizing potency, synergetic neutralizing mechanism, emergency protective and therapeutic efficacy in a hamster model, and pharmacokinetics/pharmacodynamic (PK/PD) in human nasal cavity.RESULTSThe 2 NAbs displayed broad neutralizing efficacy against Omicron, and they could structurally compensate each other in blocking the Spike-ACE2 interaction. When administrated through the intranasal mucosal route, this cocktail demonstrated profound efficacy in the emergency prevention in hamsters challenged with authentic Omicron BA.1. The investigator-initiated trial in healthy volunteers confirmed the safety and the PK/PD of the NAb cocktail delivered via nasal spray. Nasal samples from the participants receiving 4 administrations over a course of 16 hours demonstrated potent neutralization against Omicron BA.5 in an ex vivo pseudovirus neutralization assay.CONCLUSIONThese results demonstrate that the NAb cocktail nasal spray provides a good basis for clinical prophylactic efficacy against Omicron infections.TRIAL REGISTRATIONwww.chictr.org.cn, ChiCTR2200066525.FUNDINGThe National Science and Technology Major Project (2017ZX10202203), the National Key Research and Development Program of China (2018YFA0507100), Guangzhou National Laboratory (SRPG22-015), Lingang Laboratory (LG202101-01-07), Science and Technology Commission of Shanghai Municipality (YDZX20213100001556), and the Emergency Project from the Science & Technology Commission of Chongqing (cstc2021jscx-fyzxX0001).
Collapse
Affiliation(s)
- Xinghai Zhang
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, China
| | - Feiyang Luo
- Department of Immunology, College of Basic Medicine, and
- Chongqing Key Laboratory of Basic and Translational Research of Tumor Immunology, Chongqing Medical University, Chongqing, China
| | - Huajun Zhang
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, China
| | - Hangtian Guo
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Institute of Viruses and Infectious Diseases, Chemistry and Biomedicine Innovation Center (ChemBIC), Institute of Artificial Intelligence Biomedicine, Nanjing University, Nanjing, China
- Shanghai Institute for Advanced Immunochemical Studies and School of Life Science and Technology, Shanghai Tech University, Shanghai, China
| | - Junhui Zhou
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Tingting Li
- Department of Immunology, College of Basic Medicine, and
- Chongqing Key Laboratory of Basic and Translational Research of Tumor Immunology, Chongqing Medical University, Chongqing, China
| | - Shaohong Chen
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Shuyi Song
- Department of Immunology, College of Basic Medicine, and
- Chongqing Key Laboratory of Basic and Translational Research of Tumor Immunology, Chongqing Medical University, Chongqing, China
| | - Meiying Shen
- Department of Endocrine Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yan Wu
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, China
| | - Yan Gao
- Shanghai Institute for Advanced Immunochemical Studies and School of Life Science and Technology, Shanghai Tech University, Shanghai, China
- Shanghai Clinical Research and Trial Center, Shanghai, China
| | - Xiaojian Han
- Department of Immunology, College of Basic Medicine, and
- Chongqing Key Laboratory of Basic and Translational Research of Tumor Immunology, Chongqing Medical University, Chongqing, China
| | - Yingming Wang
- Department of Immunology, College of Basic Medicine, and
- Chongqing Key Laboratory of Basic and Translational Research of Tumor Immunology, Chongqing Medical University, Chongqing, China
| | - Chao Hu
- Department of Immunology, College of Basic Medicine, and
- Chongqing Key Laboratory of Basic and Translational Research of Tumor Immunology, Chongqing Medical University, Chongqing, China
| | | | | | | | - Yuchi Lu
- Shanghai Institute for Advanced Immunochemical Studies and School of Life Science and Technology, Shanghai Tech University, Shanghai, China
| | | | - Kai Wang
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Ni Tang
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Tengchuan Jin
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | | | - Shuhui Luo
- Mindao Haoyue Co., Ltd., Chongqing, China
| | - Cuicui Lin
- Mindao Haoyue Co., Ltd., Chongqing, China
| | | | - Bingxia Lu
- Mindao Haoyue Co., Ltd., Chongqing, China
| | - Yang Tian
- Mindao Haoyue Co., Ltd., Chongqing, China
| | | | | | - Haitao Yang
- Shanghai Institute for Advanced Immunochemical Studies and School of Life Science and Technology, Shanghai Tech University, Shanghai, China
- Shanghai Clinical Research and Trial Center, Shanghai, China
| | - Aishun Jin
- Department of Immunology, College of Basic Medicine, and
- Chongqing Key Laboratory of Basic and Translational Research of Tumor Immunology, Chongqing Medical University, Chongqing, China
| | - Xiaoyun Ji
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Institute of Viruses and Infectious Diseases, Chemistry and Biomedicine Innovation Center (ChemBIC), Institute of Artificial Intelligence Biomedicine, Nanjing University, Nanjing, China
- Institute of Life Sciences, and
- Engineering Research Center of Protein and Peptide Medicine, Ministry of Education, Nanjing, China
| | - Rui Gong
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, China
| | - Sandra Chiu
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Ailong Huang
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| |
Collapse
|
7
|
Yin Z, Chen JL, Lu Y, Wang B, Godfrey L, Mentzer AJ, Yao X, Liu G, Wellington D, Zhao Y, Wing PAC, Dejnirattisa W, Supasa P, Liu C, Hublitz P, Beveridge R, Waugh C, Clark SA, Clark K, Sopp P, Rostron T, Mongkolsapaya J, Screaton GR, Ogg G, Ewer K, Pollard AJ, Gilbert S, Knight JC, Lambe T, Smith GL, Dong T, Peng Y. Evaluation of T cell responses to naturally processed variant SARS-CoV-2 spike antigens in individuals following infection or vaccination. Cell Rep 2023; 42:112470. [PMID: 37141092 PMCID: PMC10121105 DOI: 10.1016/j.celrep.2023.112470] [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: 01/19/2023] [Revised: 03/20/2023] [Accepted: 04/18/2023] [Indexed: 05/05/2023] Open
Abstract
Most existing studies characterizing severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-specific T cell responses are peptide based. This does not allow evaluation of whether tested peptides are processed and presented canonically. In this study, we use recombinant vaccinia virus (rVACV)-mediated expression of SARS-CoV-2 spike protein and SARS-CoV-2 infection of angiotensin-converting enzyme (ACE)-2-transduced B cell lines to evaluate overall T cell responses in a small cohort of recovered COVID-19 patients and uninfected donors vaccinated with ChAdOx1 nCoV-19. We show that rVACV expression of SARS-CoV-2 antigen can be used as an alternative to SARS-CoV-2 infection to evaluate T cell responses to naturally processed spike antigens. In addition, the rVACV system can be used to evaluate the cross-reactivity of memory T cells to variants of concern (VOCs) and to identify epitope escape mutants. Finally, our data show that both natural infection and vaccination could induce multi-functional T cell responses with overall T cell responses remaining despite the identification of escape mutations.
Collapse
Affiliation(s)
- Zixi Yin
- Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, Oxford OX3 7FZ, UK; MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DS, UK
| | - Ji-Li Chen
- Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, Oxford OX3 7FZ, UK; MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DS, UK
| | - Yongxu Lu
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK
| | - Beibei Wang
- Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, Oxford OX3 7FZ, UK
| | - Leila Godfrey
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford OX3 7LE, UK
| | - Alexander J Mentzer
- Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK; Nuffield Department of Medicine, University of Oxford, Oxford OX3 7BN, UK
| | - Xuan Yao
- Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, Oxford OX3 7FZ, UK
| | - Guihai Liu
- Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, Oxford OX3 7FZ, UK; Beijing You'an Hospital, Capital Medical University, Beijing 100069, China
| | - Dannielle Wellington
- Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, Oxford OX3 7FZ, UK
| | - Yiqi Zhao
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK
| | - Peter A C Wing
- Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, Oxford OX3 7FZ, UK; Nuffield Department of Medicine, University of Oxford, Oxford OX3 7BN, UK
| | - Wanwisa Dejnirattisa
- Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK; Nuffield Department of Medicine, University of Oxford, Oxford OX3 7BN, UK; Division of Emerging Infectious Disease, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Piyada Supasa
- Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK; Nuffield Department of Medicine, University of Oxford, Oxford OX3 7BN, UK
| | - Chang Liu
- Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, Oxford OX3 7FZ, UK; Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK; Nuffield Department of Medicine, University of Oxford, Oxford OX3 7BN, UK
| | - Philip Hublitz
- Genome Engineering Facility, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK
| | - Ryan Beveridge
- Screening Facility, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK
| | - Craig Waugh
- Flow Cytometry Facility, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK
| | - Sally-Ann Clark
- Flow Cytometry Facility, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK
| | - Kevin Clark
- Flow Cytometry Facility, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK
| | - Paul Sopp
- Flow Cytometry Facility, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK
| | - Timothy Rostron
- Sequencing Facility, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK
| | - Juthathip Mongkolsapaya
- Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, Oxford OX3 7FZ, UK; Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK; Nuffield Department of Medicine, University of Oxford, Oxford OX3 7BN, UK
| | - Gavin R Screaton
- Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, Oxford OX3 7FZ, UK; Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK; Nuffield Department of Medicine, University of Oxford, Oxford OX3 7BN, UK
| | - Graham Ogg
- Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, Oxford OX3 7FZ, UK; MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DS, UK
| | - Katie Ewer
- Nuffield Department of Medicine, University of Oxford, Oxford OX3 7BN, UK
| | - Andrew J Pollard
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford OX3 7LE, UK; Pandemic Sciences Institute, University of Oxford, Oxford, UK; National Institute for Health Research Oxford Biomedical Research Center, Oxford, UK
| | - Sarah Gilbert
- Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, Oxford OX3 7FZ, UK; Pandemic Sciences Institute, University of Oxford, Oxford, UK
| | - Julian C Knight
- Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, Oxford OX3 7FZ, UK; Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK; Nuffield Department of Medicine, University of Oxford, Oxford OX3 7BN, UK
| | - Teresa Lambe
- Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, Oxford OX3 7FZ, UK; Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford OX3 7LE, UK; Pandemic Sciences Institute, University of Oxford, Oxford, UK.
| | - Geoffrey L Smith
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK.
| | - Tao Dong
- Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, Oxford OX3 7FZ, UK; MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DS, UK.
| | - Yanchun Peng
- Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, Oxford OX3 7FZ, UK; MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DS, UK.
| |
Collapse
|
8
|
Pomatto MAC, Gai C, Negro F, Massari L, Deregibus MC, Grange C, De Rosa FG, Camussi G. Plant-Derived Extracellular Vesicles as a Delivery Platform for RNA-Based Vaccine: Feasibility Study of an Oral and Intranasal SARS-CoV-2 Vaccine. Pharmaceutics 2023; 15:pharmaceutics15030974. [PMID: 36986835 PMCID: PMC10058531 DOI: 10.3390/pharmaceutics15030974] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/10/2023] [Accepted: 03/16/2023] [Indexed: 03/19/2023] Open
Abstract
Plant-derived extracellular vesicles (EVs) may represent a platform for the delivery of RNA-based vaccines, exploiting their natural membrane envelope to protect and deliver nucleic acids. Here, EVs extracted from orange (Citrus sinensis) juice (oEVs) were investigated as carriers for oral and intranasal SARS-CoV-2 mRNA vaccine. oEVs were efficiently loaded with different mRNA molecules (coding N, subunit 1 and full S proteins) and the mRNA was protected from degrading stress (including RNase and simulated gastric fluid), delivered to target cells and translated into protein. APC cells stimulated with oEVs loaded with mRNAs induced T lymphocyte activation in vitro. The immunization of mice with oEVs loaded with S1 mRNA via different routes of administration including intramuscular, oral and intranasal stimulated a humoral immune response with production of specific IgM and IgG blocking antibodies and a T cell immune response, as suggested by IFN-γ production by spleen lymphocytes stimulated with S peptide. Oral and intranasal administration also triggered the production of specific IgA, the mucosal barrier in the adaptive immune response. In conclusion, plant-derived EVs represent a useful platform for mRNA-based vaccines administered not only parentally but also orally and intranasally.
Collapse
Affiliation(s)
- Margherita A. C. Pomatto
- EvoBiotech s.r.l., 10122 Turin, Italy
- Department of Medical Science, University of Turin, A.O.U. Città della Salute e della Scienza di Torino, 10126 Turin, Italy
- Correspondence: (M.A.C.P.); (G.C.)
| | - Chiara Gai
- EvoBiotech s.r.l., 10122 Turin, Italy
- Department of Medical Science, University of Turin, A.O.U. Città della Salute e della Scienza di Torino, 10126 Turin, Italy
| | | | | | - Maria Chiara Deregibus
- Department of Medical Science, University of Turin, A.O.U. Città della Salute e della Scienza di Torino, 10126 Turin, Italy
| | - Cristina Grange
- Department of Medical Science, University of Turin, A.O.U. Città della Salute e della Scienza di Torino, 10126 Turin, Italy
| | - Francesco Giuseppe De Rosa
- Department of Medical Science, University of Turin, A.O.U. Città della Salute e della Scienza di Torino, 10126 Turin, Italy
| | - Giovanni Camussi
- EvoBiotech s.r.l., 10122 Turin, Italy
- Department of Medical Science, University of Turin, A.O.U. Città della Salute e della Scienza di Torino, 10126 Turin, Italy
- Correspondence: (M.A.C.P.); (G.C.)
| |
Collapse
|
9
|
Cireli E, Çavaş L. A Sample Guideline for Reverse Vaccinology Approach for the Development of Subunit Vaccine Using Varicella Zoster as a Model Disease. Methods Mol Biol 2023; 2673:453-474. [PMID: 37258932 DOI: 10.1007/978-1-0716-3239-0_30] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
For the development of multi-peptide vaccine, identification of antigenic epitopes is crucial. If it is done using wet lab techniques, the identification process can be time-consuming, laborious, and cost-intensive. In silico tools, on the other hand, enable researchers to predict potential epitopes with little to no cost for further in vivo and in vitro testing. The rapid identification process using in silico tools helps in responding to health emergencies faster. Developing an efficient and high coverage vaccine is one of the ways to reduce morbidity and mortality rates of the diseases and protect the affected populations. In this chapter, we introduce the necessary tools and methodology for the identification and characterization of antigenic epitopes to design a multi-epitope vaccine using varicella-zoster virus as an example vector model.
Collapse
Affiliation(s)
- Elif Cireli
- Department of Life Sciences and Chemistry, Constructor University Bremen, Bremen, Germany
| | - Levent Çavaş
- Dokuz Eylül University, Faculty of Science, Department of Chemistry (Biochemistry Division), İzmir, Turkey.
| |
Collapse
|