1
|
Haltaufderhyde K, Gutiérrez AH, McAllister M, Boyle CM, Moise L, Martin W, De Groot AS. T-cell responses to highly conserved SARS-CoV-2 epitopes in Hispanic Americans receiving an mRNA COVID-19 vaccine. Hum Vaccin Immunother 2025; 21:2501844. [PMID: 40407705 PMCID: PMC12118426 DOI: 10.1080/21645515.2025.2501844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 04/02/2025] [Accepted: 05/01/2025] [Indexed: 06/01/2025] Open
Abstract
This study reports the pre-clinical evaluation of peptides from EPV-CoV-19, a T cell epitope-based SARS-CoV-2 vaccine candidate, following spike-mRNA vaccination of a predominantly Hispanic American cohort. EPV-COV-19 peptides' potential to boost T cell responses to spike protein vaccines was evaluated, confirming previously observed memory recall responses in donors with prior immunity to COVID-19. The vaccinated subjects' averaged immune responses to the 15-peptide EPV-CoV-19 pool achieved 85% of the observed response to a spike protein peptide array containing a 7-fold greater epitope content, suggesting that the EPV-CoV-19 peptides have a higher relative concentration of T cell epitope content per-peptide. Ten of the 15 peptides contained spike epitopes conserved in the majority of variants of concern (VOC) evaluated over the 2020-2024 period. While commercial vaccines exhibited gradual loss of T cell epitope conservation with VOC over time, the EPV-CoV-19 epitope-peptides maintained conservation until the XBB variant emerged. The addition of one new peptide to the vaccine design reestablished broad T cell epitope coverage. These findings underscore the importance of identifying highly conserved T cell epitopes for vaccine designs that target rapidly-mutating strains of emergent pathogens, while also documenting broad memory T cell response to the vaccine in a predominantly Hispanic American cohort.
Collapse
|
2
|
Wong YC, Hang Ho DH, Zhou R, Zhang R, Woo KF, Cheng WY, Wang T, Du Y, Polly Pang KP, Tai WK, Jin X, Chen Z, Ngai Hung IF. An open-label study on the safety and immunogenicity of a PD-1-enhanced DNA vaccine used as a T cell booster for COVID-19. EBioMedicine 2025; 115:105699. [PMID: 40245494 DOI: 10.1016/j.ebiom.2025.105699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 03/21/2025] [Accepted: 03/31/2025] [Indexed: 04/19/2025] Open
Abstract
BACKGROUND Inducing T cell responses by vaccines among elderly has been a long-standing challenge. There is a need for effective COVID-19 vaccines to boost waning immunity against emerging SARS-CoV-2 variants, especially for the elderly. This study investigated the safety and immunogenicity of a PD-1-enhanced COVID-19 DNA vaccine (ICCOV™), as a booster vaccine in healthy adults (aged 18-59 years) and elderly (aged 60-75 years). METHODS This open-label, non-randomised Phase 2 study enrolled healthy participants aged 18-75 years who had previously been vaccinated with Sinovac CoronaVac, Pfizer-bioNTech Comirnaty vaccines, or both. Participants were stratified into four cohorts according to age, primary vaccination, and COVID-19 infection history, namely Adult-CoronaVac, Adult-Comirnaty, Adult-Mixed, and Elderly-Mixed cohorts. Participants were administered with a single dose of 2 mg ICCOV intramuscularly followed by electroporation using the proprietary TERESA-EPT-I device. Participants were followed up for 60 days. The primary endpoint was T cell immunogenicity within 28 days post-ICCOV vaccination. The secondary endpoints were safety, T cell and antibody responses within 60 days post-vaccination (ClinicalTrials.govNCT05904054). FINDINGS The study was conducted at Gleneagles Hospital Hong Kong between 30 June and 30 November 2023. In total, 31 participants were enrolled across the Adult-Comirnaty (n = 4), Adult-Mixed (n = 15), and Elderly-Mixed (n = 12) cohorts. All enrolled participants completed the study and were included in safety and immunogenicity analyses. Among these participants, 2 from the Adult-Comirnaty cohort, 9 from the Adult-Mixed cohort, and 4 from the Elderly-Mixed cohort reported a total of 31 adverse events, all in grade 1-2. Pain at the administration site was the most frequently reported (38·7%). The proportion of participants demonstrating an increase of SARS-CoV-2-specific ELISpot T cell responses within 28 days post ICCOV vaccination was 100% (4/4), 80% (12/15), and 75% (9/12) in Adult-Comirnaty, Adult-Mixed, and Elderly-Mixed cohorts, respectively. Single ICCOV vaccination elicited SARS-CoV-2-specific, polyfunctional CD8+ and CD4+ T cells against both ancestral and Omicron strains in all cohorts. The magnitude of responses was not inferior in the elderly, compared to adults. No elevation of antibody responses was detected. INTERPRETATION Single PD-1-enhanced ICCOV booster DNA vaccination did not show major safety concerns. The ICCOV booster elicited cross-reactive T cell responses to multiple SARS-CoV-2 strains, including in the elderly. This report demonstrates the T-cell boosting immunogenicity of ICCOV in the susceptible elderly population. FUNDING Clinical Translational Catalyst, Hong Kong Science & Technology Parks Corporation.
Collapse
Affiliation(s)
- Yik Chun Wong
- AIDS Institute, Department of Microbiology and Pandemic Research Alliance Unit, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region of the People's Republic of China; Immuno Cure Holding (HK) Limited, Hong Kong Science Park, Hong Kong Special Administrative Region of the People's Republic of China
| | - Derek Hoi Hang Ho
- Immuno Cure Holding (HK) Limited, Hong Kong Science Park, Hong Kong Special Administrative Region of the People's Republic of China
| | - Runhong Zhou
- AIDS Institute, Department of Microbiology and Pandemic Research Alliance Unit, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region of the People's Republic of China
| | - Ruiqi Zhang
- Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region of the People's Republic of China
| | - Kin Fai Woo
- AIDS Institute, Department of Microbiology and Pandemic Research Alliance Unit, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region of the People's Republic of China; Immuno Cure Holding (HK) Limited, Hong Kong Science Park, Hong Kong Special Administrative Region of the People's Republic of China
| | - Wing Yin Cheng
- Immuno Cure Holding (HK) Limited, Hong Kong Science Park, Hong Kong Special Administrative Region of the People's Republic of China
| | - Ting Wang
- Immuno Cure Holding (HK) Limited, Hong Kong Science Park, Hong Kong Special Administrative Region of the People's Republic of China
| | - Yanhua Du
- Immuno Cure Holding (HK) Limited, Hong Kong Science Park, Hong Kong Special Administrative Region of the People's Republic of China
| | - Ka Po Polly Pang
- Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region of the People's Republic of China
| | - Wai Ki Tai
- Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region of the People's Republic of China
| | - Xia Jin
- Immuno Cure Holding (HK) Limited, Hong Kong Science Park, Hong Kong Special Administrative Region of the People's Republic of China.
| | - Zhiwei Chen
- AIDS Institute, Department of Microbiology and Pandemic Research Alliance Unit, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region of the People's Republic of China; State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region of the People's Republic of China; Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong Special Administrative Region of the People's Republic of China.
| | - Ivan Fan Ngai Hung
- Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region of the People's Republic of China.
| |
Collapse
|
3
|
Patel RS, Duque D, Bavananthasivam J, Hewitt M, Sandhu JK, Kumar R, Tran A, Agrawal B. Mixed lipopeptide-based mucosal vaccine candidate induces cross-variant immunity and protects against SARS-CoV-2 infection in hamsters. Immunohorizons 2025; 9:vlae011. [PMID: 39849995 PMCID: PMC11841972 DOI: 10.1093/immhor/vlae011] [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: 11/07/2024] [Accepted: 11/14/2024] [Indexed: 01/25/2025] Open
Abstract
The global dissemination of SARS-CoV-2 led to a worldwide pandemic in March 2020. Even after the official downgrading of the COVID-19 pandemic, infection with SARS-CoV-2 variants continues. The rapid development and deployment of SARS-CoV-2 vaccines helped to mitigate the pandemic to a great extent. However, the current vaccines are suboptimal; they elicit incomplete and short-lived protection and are ineffective against evolving virus variants. Updating the spike antigen according to the prevailing variant and repeated boosters is not the long-term solution. We have designed a lipopeptide-based, mucosal, pan-coronavirus vaccine candidate, derived from highly conserved and/or functional regions of the SARS-CoV-2 spike, nucleocapsid, and membrane proteins. Our studies demonstrate that the designed lipopeptides (LPMix) induced both cellular and humoral (mucosal and systemic) immune responses upon intranasal immunization in mice. Furthermore, the antibodies bound to the wild-type and mutated S proteins of SARS-CoV-2 variants of concern, including Alpha, Beta, Delta and Omicron, and also led to efficient neutralization in a surrogate viral neutralization assay. Our sequence alignment and 3-dimensional molecular modeling studies demonstrated that spike-derived epitopes, P1 and P2, are sequentially and/or structurally conserved among the SARS-CoV-2 variants. The addition of a novel mucosal adjuvant, heat-killed Caulobacter crescentus, to the lipopeptide vaccine significantly bolstered mucosal antibody responses. Finally, the lipopeptide-based intranasal vaccine demonstrated significant improvement in lung pathologies in a hamster model of SARS-CoV-2 infection. These studies are fundamentally important and open new avenues in the investigation of an innovative, broadly protective intranasal vaccine platform for SARS-CoV-2 and its variants.
Collapse
Affiliation(s)
- Raj S Patel
- Department of Surgery, Faculty of Medicine and Dentistry, College of Health Sciences, University of Alberta, Edmonton, AB, Canada
| | - Diana Duque
- Infectious Diseases, Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada
| | - Jegarubee Bavananthasivam
- Infectious Diseases, Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada
| | - Melissa Hewitt
- Preclinical Imaging, Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada
| | - Jagdeep K Sandhu
- Preclinical Imaging, Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada
| | | | - Anh Tran
- Infectious Diseases, Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - Babita Agrawal
- Department of Surgery, Faculty of Medicine and Dentistry, College of Health Sciences, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
4
|
Atanasova M, Dimitrov I, Ralchev N, Markovski A, Manoylov I, Bradyanova S, Mihaylova N, Tchorbanov A, Doytchinova I. Design, Development and Immunogenicity Study of a Multi-Epitope Vaccine Prototype Against SARS-CoV-2. Pharmaceuticals (Basel) 2024; 17:1498. [PMID: 39598409 PMCID: PMC11597159 DOI: 10.3390/ph17111498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 10/31/2024] [Accepted: 11/05/2024] [Indexed: 11/29/2024] Open
Abstract
Objectives: SARS-CoV-2 caused the COVID-19 pandemic, which overwhelmed global healthcare systems. Over 776 million COVID-19 cases and more than 7 million deaths were reported by WHO in September 2024. COVID-19 vaccination is crucial for preventing infection and controlling the pandemic. Here, we describe the design and development of a next-generation multi-epitope vaccine for SARS-CoV-2, consisting of T cell epitopes. Methods: Immunoinformatic methods were used to derive models for the selection of MHC binders specific for the mouse strain used in this study among a set of human SARS-CoV-2 T cell epitopes identified in convalescent patients with COVID-19. The immunogenicity of the vaccine prototype was tested on humanized-ACE2 transgenic B6.Cg-Tg(K18-ACE2)2Prlmn/J mice by in vitro, in vivo, and ex vivo immunoassays. Results: Eleven binders (two from the Envelope (E) protein; two from the Membrane (M) protein; three from the Spike (S) protein; and four from the Nucleocapsid (N) protein) were synthesized and included in a multi-epitope vaccine prototype. The animals were immunized with a mix of predicted MHC-I, MHC-II, or MHC-I/MHC-II peptide epitopes in Complete Freund's Adjuvant, and boosted with peptides in Incomplete Freund's Adjuvant. Immunization with SARS-CoV-2 epitopes remodeled the lymphocyte profile. A weak humoral response and the significant production of IL-4 and IFN-γ from T cells were found after the vaccination of the animals. Conclusions: The multi-epitope vaccine prototype presented in this study demonstrates immunogenicity in mice and shows potential for human vaccine construction.
Collapse
Affiliation(s)
- Mariyana Atanasova
- Drug Design and Bioinformatics Laboratory, Faculty of Pharmacy, Medical University of Sofia, 1000 Sofia, Bulgaria; (M.A.); (I.D.)
| | - Ivan Dimitrov
- Drug Design and Bioinformatics Laboratory, Faculty of Pharmacy, Medical University of Sofia, 1000 Sofia, Bulgaria; (M.A.); (I.D.)
| | - Nikola Ralchev
- Department of Immunology, Stefan Angelov Institute of Microbiology, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria; (N.R.); (A.M.); (I.M.); (S.B.); (N.M.)
| | - Aleksandar Markovski
- Department of Immunology, Stefan Angelov Institute of Microbiology, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria; (N.R.); (A.M.); (I.M.); (S.B.); (N.M.)
| | - Iliyan Manoylov
- Department of Immunology, Stefan Angelov Institute of Microbiology, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria; (N.R.); (A.M.); (I.M.); (S.B.); (N.M.)
| | - Silviya Bradyanova
- Department of Immunology, Stefan Angelov Institute of Microbiology, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria; (N.R.); (A.M.); (I.M.); (S.B.); (N.M.)
| | - Nikolina Mihaylova
- Department of Immunology, Stefan Angelov Institute of Microbiology, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria; (N.R.); (A.M.); (I.M.); (S.B.); (N.M.)
| | - Andrey Tchorbanov
- Department of Immunology, Stefan Angelov Institute of Microbiology, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria; (N.R.); (A.M.); (I.M.); (S.B.); (N.M.)
| | - Irini Doytchinova
- Drug Design and Bioinformatics Laboratory, Faculty of Pharmacy, Medical University of Sofia, 1000 Sofia, Bulgaria; (M.A.); (I.D.)
| |
Collapse
|
5
|
Perdiguero B, Álvarez E, Marcos-Villar L, Sin L, López-Bravo M, Valverde JR, Sorzano CÓS, Falqui M, Coloma R, Esteban M, Guerra S, Gómez CE. B and T Cell Bi-Cistronic Multiepitopic Vaccine Induces Broad Immunogenicity and Provides Protection Against SARS-CoV-2. Vaccines (Basel) 2024; 12:1213. [PMID: 39591118 PMCID: PMC11598604 DOI: 10.3390/vaccines12111213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 10/18/2024] [Accepted: 10/22/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND The COVID-19 pandemic, caused by SARS-CoV-2, has highlighted the need for vaccines targeting both neutralizing antibodies (NAbs) and long-lasting cross-reactive T cells covering multiple viral proteins to provide broad and durable protection against emerging variants. METHODS To address this, here we developed two vaccine candidates, namely (i) DNA-CoV2-TMEP, expressing the multiepitopic CoV2-TMEP protein containing immunodominant and conserved T cell regions from SARS-CoV-2 structural proteins, and (ii) MVA-CoV2-B2AT, encoding a bi-cistronic multiepitopic construct that combines conserved B and T cell overlapping regions from SARS-CoV-2 structural proteins. RESULTS Both candidates were assessed in vitro and in vivo demonstrating their ability to induce robust immune responses. In C57BL/6 mice, DNA-CoV2-TMEP enhanced the recruitment of innate immune cells and stimulated SARS-CoV-2-specific polyfunctional T cells targeting multiple viral proteins. MVA-CoV2-B2AT elicited NAbs against various SARS-CoV-2 variants of concern (VoCs) and reduced viral replication and viral yields against the Beta variant in susceptible K18-hACE2 mice. The combination of MVA-CoV2-B2AT with a mutated ISG15 form as an adjuvant further increased the magnitude, breadth and polyfunctional profile of the response. CONCLUSION These findings underscore the potential of these multiepitopic proteins when expressed from DNA or MVA vectors to provide protection against SARS-CoV-2 and its variants, supporting their further development as next-generation COVID-19 vaccines.
Collapse
Affiliation(s)
- Beatriz Perdiguero
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), 28049 Madrid, Spain; (E.Á.); (L.M.-V.); (M.E.)
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain;
| | - Enrique Álvarez
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), 28049 Madrid, Spain; (E.Á.); (L.M.-V.); (M.E.)
| | - Laura Marcos-Villar
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), 28049 Madrid, Spain; (E.Á.); (L.M.-V.); (M.E.)
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain;
| | - Laura Sin
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain;
| | - María López-Bravo
- Department of Microbial Biotechnology, CNB-CSIC, 28049 Madrid, Spain;
| | | | | | - Michela Falqui
- Department of Preventive Medicine, Public Health and Microbiology, Faculty of Medicine, Universidad Autónoma de Madrid, 28029 Madrid, Spain; (M.F.); (R.C.); (S.G.)
| | - Rocío Coloma
- Department of Preventive Medicine, Public Health and Microbiology, Faculty of Medicine, Universidad Autónoma de Madrid, 28029 Madrid, Spain; (M.F.); (R.C.); (S.G.)
| | - Mariano Esteban
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), 28049 Madrid, Spain; (E.Á.); (L.M.-V.); (M.E.)
| | - Susana Guerra
- Department of Preventive Medicine, Public Health and Microbiology, Faculty of Medicine, Universidad Autónoma de Madrid, 28029 Madrid, Spain; (M.F.); (R.C.); (S.G.)
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Carmen Elena Gómez
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), 28049 Madrid, Spain; (E.Á.); (L.M.-V.); (M.E.)
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain;
| |
Collapse
|
6
|
Bukhari SNH, Ogudo KA. Prediction of antigenic peptides of SARS- CoV-2 pathogen using machine learning. PeerJ Comput Sci 2024; 10:e2319. [PMID: 39650382 PMCID: PMC11623221 DOI: 10.7717/peerj-cs.2319] [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: 05/01/2024] [Accepted: 08/20/2024] [Indexed: 12/11/2024]
Abstract
Antigenic peptides (APs), also known as T-cell epitopes (TCEs), represent the immunogenic segment of pathogens capable of inducing an immune response, making them potential candidates for epitope-based vaccine (EBV) design. Traditional wet lab methods for identifying TCEs are expensive, challenging, and time-consuming. Alternatively, computational approaches employing machine learning (ML) techniques offer a faster and more cost-effective solution. In this study, we present a robust XGBoost ML model for predicting TCEs of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus as potential vaccine candidates. The peptide sequences comprising TCEs and non-TCEs retrieved from Immune Epitope Database Repository (IEDB) were subjected to feature extraction process to extract their physicochemical properties for model training. Upon evaluation using a test dataset, the model achieved an impressive accuracy of 97.6%, outperforming other ML classifiers. Employing a five-fold cross-validation a mean accuracy of 97.58% was recorded, indicating consistent and linear performance across all iterations. While the predicted epitopes show promise as vaccine candidates for SARS-CoV-2, further scientific examination through in vivo and in vitro studies is essential to validate their suitability.
Collapse
Affiliation(s)
| | - Kingsley A. Ogudo
- Department of Electrical & Electronics Engineering Faculty of Engineering and the Built Environment, University of Johannesburg, Johannesburg, South Africa
| |
Collapse
|
7
|
Jaishwal P, Jha K, Singh SP. Revisiting the dimensions of universal vaccine with special focus on COVID-19: Efficacy versus methods of designing. Int J Biol Macromol 2024; 277:134012. [PMID: 39048013 DOI: 10.1016/j.ijbiomac.2024.134012] [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: 10/28/2023] [Revised: 05/28/2024] [Accepted: 07/17/2024] [Indexed: 07/27/2024]
Abstract
Even though the use of SARS-CoV-2 vaccines during the COVID-19 pandemic showed unprecedented success in a short time, it also exposed a flaw in the current vaccine design strategy to offer broad protection against emerging variants of concern. However, developing broad-spectrum vaccines is still a challenge for immunologists. The development of universal vaccines against emerging pathogens and their variants appears to be a practical solution to mitigate the economic and physical effects of the pandemic on society. Very few reports are available to explain the basic concept of universal vaccine design and development. This review provides an overview of the innate and adaptive immune responses generated against vaccination and essential insight into immune mechanisms helpful in designing universal vaccines targeting influenza viruses and coronaviruses. In addition, the characteristics, safety, and factors affecting the efficacy of universal vaccines have been discussed. Furthermore, several advancements in methods worthy of designing universal vaccines are described, including chimeric immunogens, heterologous prime-boost vaccines, reverse vaccinology, structure-based antigen design, pan-reactive antibody vaccines, conserved neutralizing epitope-based vaccines, mosaic nanoparticle-based vaccines, etc. In addition to the several advantages, significant potential constraints, such as defocusing the immune response and subdominance, are also discussed.
Collapse
Affiliation(s)
- Puja Jaishwal
- Department of Biotechnology, Mahatma Gandhi Central University, Motihari, India
| | - Kisalay Jha
- Department of Biotechnology, Mahatma Gandhi Central University, Motihari, India
| | | |
Collapse
|
8
|
Vargas-Bermudez DS, Gil-Silva AC, Naranjo-Ortíz MF, Mogollón JD, Gómez-Betancur JF, Estrada JF, Aldaz Á, Garzón-González H, Angulo J, Foss D, Gutierrez AH, Jaime J. Detection of PCV2d in Vaccinated Pigs in Colombia and Prediction of Vaccine T Cell Epitope Coverage against Circulating Strains Using EpiCC Analysis. Vaccines (Basel) 2024; 12:1119. [PMID: 39460286 PMCID: PMC11511301 DOI: 10.3390/vaccines12101119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 07/31/2024] [Accepted: 08/14/2024] [Indexed: 10/28/2024] Open
Abstract
Porcine circovirus type 2 (PCV2) is strongly linked to a group of syndromes referred to as porcine-circovirus-associated diseases (PCVADs), which are controlled through vaccination; however, this does not induce sterilizing immunity but is instead involved in the evolution of the virus and is considered a factor in vaccine failure. This study sampled 84 herds (167 pigs) vaccinated against PCV2 and with clinical signs of PCVADs in five provinces across Colombia. PCV2 was identified and further characterized at the molecular level via genotyping and phylogenetic reconstructions. In addition, PCV2-associated lesions were examined via histopathology. Furthermore, the PCV2-Cap sequences retrieved were compared with three vaccines via the EpiCC tool and T cell epitope coverage. The prevalence of PCV2 was 82% in pigs and 92.9% in herds. The highest viral loads were identified in lymphoid tissue, and PCV2d emerged as the most predominant in pigs and herds (93.4% and 92.3%). Sequences for PCV2-ORF2 (n = 57; 55 PCV2d and 2 PCV2a) were determined, and PCV2d sequences were highly similar. The most common pneumonia pattern was suppurative bronchopneumonia, while the most common lung lesion was exudation in the airways; in lymphoid tissue, there was lymphoid depletion. The bivalent vaccine (PCV2a and PCVb) exhibited a higher EpiCC score (8.36) and T cell epitope coverage (80.6%) than monovalent PCV2a vaccines. In conclusion, PCV2d currently circulates widely in Colombia. Despite vaccination, there are clinical cases of PCV2, and immunoinformatic analyses demonstrate that bivalent vaccines improved the average coverage.
Collapse
Affiliation(s)
- Diana S. Vargas-Bermudez
- Universidad Nacional de Colombia, Sede Bogotá, Facultad de Medicina Veterinaria y de Zootecnia, Centro de Investigación en Infectología e Inmunología Veterinaria (CI3V), Bogotá 111321, Colombia; (D.S.V.-B.); (J.D.M.)
| | - Alixs Constanza Gil-Silva
- Zoetis de Colombia, Titan Plaza Centro Empresarial, Bogotá 111021, Colombia; (A.C.G.-S.); (J.F.G.-B.); (J.F.E.)
| | - María F. Naranjo-Ortíz
- Universidad Nacional de Colombia, Sede Bogotá, Facultad de Medicina Veterinaria y de Zootecnia, Centro de Investigación en Infectología e Inmunología Veterinaria (CI3V), Bogotá 111321, Colombia; (D.S.V.-B.); (J.D.M.)
| | - José Darío Mogollón
- Universidad Nacional de Colombia, Sede Bogotá, Facultad de Medicina Veterinaria y de Zootecnia, Centro de Investigación en Infectología e Inmunología Veterinaria (CI3V), Bogotá 111321, Colombia; (D.S.V.-B.); (J.D.M.)
| | - Jair F. Gómez-Betancur
- Zoetis de Colombia, Titan Plaza Centro Empresarial, Bogotá 111021, Colombia; (A.C.G.-S.); (J.F.G.-B.); (J.F.E.)
| | - José F. Estrada
- Zoetis de Colombia, Titan Plaza Centro Empresarial, Bogotá 111021, Colombia; (A.C.G.-S.); (J.F.G.-B.); (J.F.E.)
| | - Álvaro Aldaz
- Zoetis Inc., 10 Sylvan Way, Parsippany, NJ 07054, USA;
| | - Harold Garzón-González
- Grupo de Investigación en Ciencias Biomédicas (GICBUPTC), Universidad Pedagógica y Tecnológica de Colombia (UPTC), Tunja 150003, BOY, Colombia;
| | - José Angulo
- Zoetis Inc., 1040 Swabia Ct, Durham, NC 27703, USA;
| | - Dennis Foss
- Zoetis Inc., 333 Portage St, Kalamazoo, MI 49007, USA;
| | | | - Jairo Jaime
- Universidad Nacional de Colombia, Sede Bogotá, Facultad de Medicina Veterinaria y de Zootecnia, Centro de Investigación en Infectología e Inmunología Veterinaria (CI3V), Bogotá 111321, Colombia; (D.S.V.-B.); (J.D.M.)
| |
Collapse
|
9
|
Yao D, Patel RS, Lam A, Glover Q, Srinivasan C, Herchen A, Ritchie B, Agrawal B. Antibody Responses in SARS-CoV-2-Exposed and/or Vaccinated Individuals Target Conserved Epitopes from Multiple CoV-2 Antigens. Int J Mol Sci 2024; 25:9814. [PMID: 39337303 PMCID: PMC11432605 DOI: 10.3390/ijms25189814] [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: 08/28/2024] [Revised: 09/06/2024] [Accepted: 09/09/2024] [Indexed: 09/30/2024] Open
Abstract
There is a need to investigate novel strategies in order to create an effective, broadly protective vaccine for current and future severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) outbreaks. The currently available vaccines demonstrate compromised efficacy against emerging SARS-CoV-2 variants of concern (VOCs), short-lived immunity, and susceptibility to immune imprinting due to frequent boosting practices. In this study, we examined the specificity of cross-reactive IgG antibody responses in mRNA-vaccinated, AstraZeneca-vaccinated, and unvaccinated donors to identify potentially conserved, cross-reactive epitopes to target in order to create a broadly protective SARS-CoV-2 vaccine. Our study provides evidence for cross-reactive IgG antibodies specific to eight different spike (S) variants. Furthermore, the specificities of these cross-variant IgG antibody titers were associated to some extent with spike S1- and S2-subunit-derived epitopes P1 and P2, respectively. In addition, nucleocapsid (N)- and membrane (M)-specific IgG antibody titers correlated with N- and M-derived epitopes conserved across beta-CoVs, P3-7. This study reveals conserved epitopes of viral antigens, targeted by natural and/or vaccine-induced human immunity, for future designs of next-generation COVID-19 vaccines.
Collapse
Affiliation(s)
- David Yao
- Department of Surgery, Faculty of Medicine and Dentistry, College of Health Sciences, University of Alberta, Edmonton, AB T6G 2R3, Canada; (D.Y.); (R.S.P.); (A.L.)
| | - Raj S. Patel
- Department of Surgery, Faculty of Medicine and Dentistry, College of Health Sciences, University of Alberta, Edmonton, AB T6G 2R3, Canada; (D.Y.); (R.S.P.); (A.L.)
| | - Adrien Lam
- Department of Surgery, Faculty of Medicine and Dentistry, College of Health Sciences, University of Alberta, Edmonton, AB T6G 2R3, Canada; (D.Y.); (R.S.P.); (A.L.)
| | - Quarshie Glover
- Department of Medicine, Faculty of Medicine and Dentistry, College of Health Sciences, University of Alberta, Edmonton, AB T6G 2R3, Canada; (Q.G.); (C.S.); (A.H.); (B.R.)
| | - Cindy Srinivasan
- Department of Medicine, Faculty of Medicine and Dentistry, College of Health Sciences, University of Alberta, Edmonton, AB T6G 2R3, Canada; (Q.G.); (C.S.); (A.H.); (B.R.)
| | - Alex Herchen
- Department of Medicine, Faculty of Medicine and Dentistry, College of Health Sciences, University of Alberta, Edmonton, AB T6G 2R3, Canada; (Q.G.); (C.S.); (A.H.); (B.R.)
| | - Bruce Ritchie
- Department of Medicine, Faculty of Medicine and Dentistry, College of Health Sciences, University of Alberta, Edmonton, AB T6G 2R3, Canada; (Q.G.); (C.S.); (A.H.); (B.R.)
| | - Babita Agrawal
- Department of Surgery, Faculty of Medicine and Dentistry, College of Health Sciences, University of Alberta, Edmonton, AB T6G 2R3, Canada; (D.Y.); (R.S.P.); (A.L.)
| |
Collapse
|
10
|
Bukhari SNH, Elshiekh E, Abbas M. Physicochemical properties-based hybrid machine learning technique for the prediction of SARS-CoV-2 T-cell epitopes as vaccine targets. PeerJ Comput Sci 2024; 10:e1980. [PMID: 38686005 PMCID: PMC11057572 DOI: 10.7717/peerj-cs.1980] [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: 01/19/2024] [Accepted: 03/15/2024] [Indexed: 05/02/2024]
Abstract
Majority of the existing SARS-CoV-2 vaccines work by presenting the whole pathogen in the attenuated form to immune system to invoke an immune response. On the other hand, the concept of a peptide based vaccine (PBV) is based on the identification and chemical synthesis of only immunodominant peptides known as T-cell epitopes (TCEs) to induce a specific immune response against a particular pathogen. However PBVs have received less attention despite holding huge untapped potential for boosting vaccine safety and immunogenicity. To identify these TCEs for designing PBV, wet-lab experiments are difficult, expensive, and time-consuming. Machine learning (ML) techniques can accurately predict TCEs, saving time and cost for speedy vaccine development. This work proposes novel hybrid ML techniques based on the physicochemical properties of peptides to predict SARS-CoV-2 TCEs. The proposed hybrid ML technique was evaluated using various ML model evaluation metrics and demonstrated promising results. The hybrid technique of decision tree classifier with chi-squared feature weighting technique and forward search optimal feature searching algorithm has been identified as the best model with an accuracy of 98.19%. Furthermore, K-fold cross-validation (KFCV) was performed to ensure that the model is reliable and the results indicate that the hybrid random forest model performs consistently well in terms of accuracy with respect to other hybrid approaches. The predicted TCEs are highly likely to serve as promising vaccine targets, subject to evaluations both in-vivo and in-vitro. This development could potentially save countless lives globally, prevent future epidemic-scale outbreaks, and reduce the risk of mutation escape.
Collapse
Affiliation(s)
- Syed Nisar Hussain Bukhari
- National Institute of Electronics and Information Technology (NIELIT), Srinagar, Jammu and Kashmir, India
| | - E. Elshiekh
- Department of Radiological Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Mohamed Abbas
- Electrical Engineering Department, College of Engineering, King Khalid University, Abha, Saudi Arabia
| |
Collapse
|
11
|
Tan D, Kang N, Zhu Y, Hou J, Wang H, Xu H, Zu C, Gao Z, Liu M, Liu N, Deng Q, Lu H, Liu J, Xie Y. Construction and efficacy testing of DNA vaccines containing HLA-A*02:01-restricted SARS-CoV-2 T-cell epitopes predicted by immunoinformatics. Acta Biochim Biophys Sin (Shanghai) 2024; 56:986-996. [PMID: 38655616 PMCID: PMC11322877 DOI: 10.3724/abbs.2024039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Accepted: 01/31/2024] [Indexed: 04/26/2024] Open
Abstract
Vaccines play essential roles in the fight against the COVID-19 pandemic. The development and assessment of COVID-19 vaccines have generally focused on the induction and boosting of neutralizing antibodies targeting the SARS-CoV-2 spike (S) protein. Due to rapid and continuous variation in the S protein, such vaccines need to be regularly updated to match newly emerged dominant variants. T-cell vaccines that target MHC I- or II-restricted epitopes in both structural and non-structural viral proteins have the potential to induce broadly cross-protective and long-lasting responses. In this work, the entire proteome encoded by SARS-CoV-2 (Wuhan-hu-1) is subjected to immunoinformatics-based prediction of HLA-A*02:01-restricted epitopes. The immunogenicity of the predicted epitopes is evaluated using peripheral blood mononuclear cells from convalescent Wuhan-hu-1-infected patients. Furthermore, predicted epitopes that are conserved across major SARS-CoV-2 lineages and variants are used to construct DNA vaccines expressing multi-epitope polypeptides. Most importantly, two DNA vaccine constructs induce epitope-specific CD8 + T-cell responses in a mouse model of HLA-A*02:01 restriction and protect immunized mice from challenge with Wuhan-hu-1 virus after hACE2 transduction. These data provide candidate T-cell epitopes useful for the development of T-cell vaccines against SARS-CoV-2 and demonstrate a strategy for quick T-cell vaccine candidate development applicable to other emerging pathogens.
Collapse
Affiliation(s)
- Dan Tan
- Key Laboratory of Medical Molecular Virology (NHC & MOE & CAMS)Shanghai Institute of Infectious Diseases and BiosecurityDepartment of Medical Microbiology and ParasitologySchool of Basic Medical SciencesShanghai Medical CollegeFudan UniversityShanghai200031China
| | - Ning Kang
- Key Laboratory of Medical Molecular Virology (NHC & MOE & CAMS)Shanghai Institute of Infectious Diseases and BiosecurityDepartment of Medical Microbiology and ParasitologySchool of Basic Medical SciencesShanghai Medical CollegeFudan UniversityShanghai200031China
| | - Yuanfei Zhu
- Key Laboratory of Medical Molecular Virology (NHC & MOE & CAMS)Shanghai Institute of Infectious Diseases and BiosecurityDepartment of Medical Microbiology and ParasitologySchool of Basic Medical SciencesShanghai Medical CollegeFudan UniversityShanghai200031China
| | - Jia Hou
- Key Laboratory of Medical Molecular Virology (NHC & MOE & CAMS)Shanghai Institute of Infectious Diseases and BiosecurityDepartment of Medical Microbiology and ParasitologySchool of Basic Medical SciencesShanghai Medical CollegeFudan UniversityShanghai200031China
| | - Hanqing Wang
- Key Laboratory of Medical Molecular Virology (NHC & MOE & CAMS)Shanghai Institute of Infectious Diseases and BiosecurityDepartment of Medical Microbiology and ParasitologySchool of Basic Medical SciencesShanghai Medical CollegeFudan UniversityShanghai200031China
| | - Huijun Xu
- Key Laboratory of Medical Molecular Virology (NHC & MOE & CAMS)Shanghai Institute of Infectious Diseases and BiosecurityDepartment of Medical Microbiology and ParasitologySchool of Basic Medical SciencesShanghai Medical CollegeFudan UniversityShanghai200031China
| | - Cheng Zu
- Key Laboratory of Medical Molecular Virology (NHC & MOE & CAMS)Shanghai Institute of Infectious Diseases and BiosecurityDepartment of Medical Microbiology and ParasitologySchool of Basic Medical SciencesShanghai Medical CollegeFudan UniversityShanghai200031China
| | - Zixiang Gao
- Key Laboratory of Medical Molecular Virology (NHC & MOE & CAMS)Shanghai Institute of Infectious Diseases and BiosecurityDepartment of Medical Microbiology and ParasitologySchool of Basic Medical SciencesShanghai Medical CollegeFudan UniversityShanghai200031China
| | - Mu Liu
- Key Laboratory of Medical Molecular Virology (NHC & MOE & CAMS)Shanghai Institute of Infectious Diseases and BiosecurityDepartment of Medical Microbiology and ParasitologySchool of Basic Medical SciencesShanghai Medical CollegeFudan UniversityShanghai200031China
| | - Nannan Liu
- Key Laboratory of Medical Molecular Virology (NHC & MOE & CAMS)Shanghai Institute of Infectious Diseases and BiosecurityDepartment of Medical Microbiology and ParasitologySchool of Basic Medical SciencesShanghai Medical CollegeFudan UniversityShanghai200031China
| | - Qiang Deng
- Key Laboratory of Medical Molecular Virology (NHC & MOE & CAMS)Shanghai Institute of Infectious Diseases and BiosecurityDepartment of Medical Microbiology and ParasitologySchool of Basic Medical SciencesShanghai Medical CollegeFudan UniversityShanghai200031China
| | - Hongzhou Lu
- Shanghai Public Health Clinical CenterFudan UniversityShanghai201508China
- National Clinical Research Centre for Infectious Diseasesthe Third People’s Hospital of ShenzhenThe Second Affiliated Hospital of Southern University of Science and TechnologyShenzhen518112China
| | - Jing Liu
- Key Laboratory of Medical Molecular Virology (NHC & MOE & CAMS)Shanghai Institute of Infectious Diseases and BiosecurityDepartment of Medical Microbiology and ParasitologySchool of Basic Medical SciencesShanghai Medical CollegeFudan UniversityShanghai200031China
| | - Youhua Xie
- Key Laboratory of Medical Molecular Virology (NHC & MOE & CAMS)Shanghai Institute of Infectious Diseases and BiosecurityDepartment of Medical Microbiology and ParasitologySchool of Basic Medical SciencesShanghai Medical CollegeFudan UniversityShanghai200031China
- Shanghai Public Health Clinical CenterFudan UniversityShanghai201508China
| |
Collapse
|
12
|
Ratishvili T, Quach HQ, Haralambieva IH, Suryawanshi YR, Ovsyannikova IG, Kennedy RB, Poland GA. A multifaceted approach for identification, validation, and immunogenicity of naturally processed and in silico-predicted highly conserved SARS-CoV-2 peptides. Vaccine 2024; 42:162-174. [PMID: 38105139 DOI: 10.1016/j.vaccine.2023.12.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 11/19/2023] [Accepted: 12/05/2023] [Indexed: 12/19/2023]
Abstract
SARS-CoV-2 remains a major global public health concern. Antibody waning and immune escape variant emergence necessitate the development of next generation vaccines that induce cross-reactive durable immune responses. T cell responses to SARS-CoV-2 demonstrate higher conservation, antigenic breadth, and longevity than antibody responses. Therefore, we sought to identify pathogen-derived T cell epitopes for a potential peptide-based vaccine. We pursued an approach leveraging: 1) liquid chromatography and tandem mass spectrometry (LC-MS/MS)-based identification of peptides from ancestral SARS-CoV-2-infected cell lines, 2) epitope prediction algorithms, and 3) overlapping peptide libraries. From this strategy, we identified 380 unique SARS-CoV-2-derived peptide sequences, including 53 antigenic HLA class I and class II peptides from multiple structural and non-structural/accessory viral proteins. These peptide sequences were highly conserved across variants of concern/interest (VoC/VoIs), and are estimated to achieve coverage of >96% of the world population. Our findings validate this discovery pipeline for peptide identification and immunogenicity testing, and are a crucial step toward the development of a next-generation multi-epitope SARS-CoV-2 peptide vaccine, and a novel vaccine platform methodology.
Collapse
Affiliation(s)
- Tamar Ratishvili
- Mayo Clinic Vaccine Research Group, Department of General Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Huy Quang Quach
- Mayo Clinic Vaccine Research Group, Department of General Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Iana H Haralambieva
- Mayo Clinic Vaccine Research Group, Department of General Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Yogesh R Suryawanshi
- Mayo Clinic Vaccine Research Group, Department of General Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Inna G Ovsyannikova
- Mayo Clinic Vaccine Research Group, Department of General Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Richard B Kennedy
- Mayo Clinic Vaccine Research Group, Department of General Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Gregory A Poland
- Mayo Clinic Vaccine Research Group, Department of General Internal Medicine, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
13
|
Mohapatra S, Kumar S, Kumar S, Singh AK, Nayak B. Immunodominant conserved moieties on spike protein of SARS-CoV-2 renders virulence factor for the design of epitope-based peptide vaccines. Virusdisease 2023; 34:456-482. [PMID: 38046066 PMCID: PMC10686954 DOI: 10.1007/s13337-023-00852-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 11/03/2023] [Indexed: 12/05/2023] Open
Abstract
UNLABELLED The outbreak of novel SARS-CoV-2 virion has wreaked havoc with a high prevalence of respiratory illness and high transmission due to a vague understanding of the viral antigenicity, augmenting the dire challenge to public health globally. This viral member necessitates the expansion of diagnostic and therapeutic tools to track its transmission and confront it through vaccine development. Therefore, prophylactic strategies are mandatory. Virulent spike proteins can be the most desirable candidate for the computational design of vaccines targeting SARS-CoV-2, followed by the meteoric development of immune epitopes. Spike protein was characterized using existing bioinformatics tools with a unique roadmap related to the immunological profile of SARS-CoV-2 to predict immunogenic virulence epitopes based on antigenicity, allergenicity, toxicity, immunogenicity, and population coverage. Applying in silico approaches, a set of twenty-four B lymphocyte-based epitopes and forty-six T lymphocyte-based epitopes were selected. The predicted epitopes were evaluated for their intrinsic properties. The physico-chemical characterization of epitopes qualifies them for further in vitro and in vivo analysis and pre-requisite vaccine development. This study presents a set of screened epitopes that bind to HLA-specific allelic proteins and can be employed for designing a peptide vaccine construct against SARS-CoV-2 that will confer vaccine-induced protective immunity due to its structural stability. SUPPLEMENTARY INFORMATION The online version contains supplementary material available at 10.1007/s13337-023-00852-9.
Collapse
Affiliation(s)
- Subhashree Mohapatra
- Immunology and Molecular Medicine Laboratory, Department of Life Science, National Institute of Technology Rourkela, Rourkela, Odisha 769008 India
| | - Santosh Kumar
- RNA Biology Laboratory, Department of Life Science, National Institute of Technology Rourkela, Rourkela, Odisha 769008 India
| | - Shashank Kumar
- Molecular Signalling and Drug Discovery Laboratory, Department of Biochemistry, Central University of Punjab, Bathinda, Punjab 151401 India
| | - Atul Kumar Singh
- Molecular Signalling and Drug Discovery Laboratory, Department of Biochemistry, Central University of Punjab, Bathinda, Punjab 151401 India
| | - Bismita Nayak
- Immunology and Molecular Medicine Laboratory, Department of Life Science, National Institute of Technology Rourkela, Rourkela, Odisha 769008 India
| |
Collapse
|
14
|
Miah SMS, Lelias S, Gutierrez AH, McAllister M, Boyle CM, Moise L, De Groot AS. A SARS-CoV-2 NSP7 homolog of a Treg epitope suppresses CD4+ and CD8+ T cell memory responses. Front Immunol 2023; 14:1290688. [PMID: 38124752 PMCID: PMC10731459 DOI: 10.3389/fimmu.2023.1290688] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 11/03/2023] [Indexed: 12/23/2023] Open
Abstract
Pathogens escape host defenses by T-cell epitope mutation or deletion (immune escape) and by simulating the appearance of human T cell epitopes (immune camouflage). We identified a highly conserved, human-like T cell epitope in non-structural protein 7 (NSP7) of SARS-CoV-2, RNA-dependent RNA polymerase (RdRp) hetero-tetramer complex. Remarkably, this T cell epitope has significant homology to a T regulatory cell epitope (Tregitope) previously identified in the Fc region of human immunoglobulin G (IgG) (Tregitope 289). We hypothesized that the SARS-CoV-2 NSP7 epitope (NSP7-289) may induce suppressive responses by engaging and activating pre-existing regulatory T cells. We therefore compared NSP7-289 and IgG Tregitopes (289 and 289z, a shorter version of 289 that isolates the shared NSP7 epitope) in vitro. Tregitope peptides 289, 289z and NSP7-289 bound to multiple HLA-DRB1 alleles in vitro and suppressed CD4+ and CD8+ T cell memory responses. Identification and in vitro validation of SARS-CoV-2 NSP7-289 provides further evidence of immune camouflage and suggests that pathogens can use human-like epitopes to evade immune response and potentially enhance host tolerance. Further exploration of the role of cross-conserved Tregs in human immune responses to pathogens such as SARS-CoV-2 is warranted.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Anne S. De Groot
- EpiVax, Inc., Providence, RI, United States
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, United States
| |
Collapse
|
15
|
Eickhoff CS, Meza KA, Terry FE, Colbert CG, Blazevic A, Gutiérrez AH, Stone ET, Brien JD, Pinto AK, El Sahly HM, Mulligan MJ, Rouphael N, Alcaide ML, Tomashek KM, Focht C, Martin WD, Moise L, De Groot AS, Hoft DF. Identification of immunodominant T cell epitopes induced by natural Zika virus infection. Front Immunol 2023; 14:1247876. [PMID: 37705976 PMCID: PMC10497216 DOI: 10.3389/fimmu.2023.1247876] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 08/07/2023] [Indexed: 09/15/2023] Open
Abstract
Zika virus (ZIKV) is a flavivirus primarily transmitted by Aedes species mosquitoes, first discovered in Africa in 1947, that disseminated through Southeast Asia and the Pacific Islands in the 2000s. The first ZIKV infections in the Americas were identified in 2014, and infections exploded through populations in Brazil and other countries in 2015/16. ZIKV infection during pregnancy can cause severe brain and eye defects in offspring, and infection in adults has been associated with higher risks of Guillain-Barré syndrome. We initiated a study to describe the natural history of Zika (the disease) and the immune response to infection, for which some results have been reported. In this paper, we identify ZIKV-specific CD4+ and CD8+ T cell epitopes that induce responses during infection. Two screening approaches were utilized: an untargeted approach with overlapping peptide arrays spanning the entire viral genome, and a targeted approach utilizing peptides predicted to bind human MHC molecules. Immunoinformatic tools were used to identify conserved MHC class I supertype binders and promiscuous class II binding peptide clusters predicted to bind 9 common class II alleles. T cell responses were evaluated in overnight IFN-γ ELISPOT assays. We found that MHC supertype binding predictions outperformed the bulk overlapping peptide approach. Diverse CD4+ T cell responses were observed in most ZIKV-infected participants, while responses to CD8+ T cell epitopes were more limited. Most individuals developed a robust T cell response against epitopes restricted to a single MHC class I supertype and only a single or few CD8+ T cell epitopes overall, suggesting a strong immunodominance phenomenon. Noteworthy is that many epitopes were commonly immunodominant across persons expressing the same class I supertype. Nearly all of the identified epitopes are unique to ZIKV and are not present in Dengue viruses. Collectively, we identified 31 immunogenic peptides restricted by the 6 major class I supertypes and 27 promiscuous class II epitopes. These sequences are highly relevant for design of T cell-targeted ZIKV vaccines and monitoring T cell responses to Zika virus infection and vaccination.
Collapse
Affiliation(s)
- Christopher S. Eickhoff
- Department of Internal Medicine, Saint Louis University, Division of Infectious Diseases, Allergy, and Immunology, Saint Louis, MO, United States
| | - Krystal A. Meza
- Department of Internal Medicine, Saint Louis University, Division of Infectious Diseases, Allergy, and Immunology, Saint Louis, MO, United States
| | | | - Chase G. Colbert
- Department of Internal Medicine, Saint Louis University, Division of Infectious Diseases, Allergy, and Immunology, Saint Louis, MO, United States
| | - Azra Blazevic
- Department of Internal Medicine, Saint Louis University, Division of Infectious Diseases, Allergy, and Immunology, Saint Louis, MO, United States
| | | | - E. Taylor Stone
- Department of Molecular Microbiology and Immunology, Saint Louis University, Saint Louis, MO, United States
| | - James D. Brien
- Department of Molecular Microbiology and Immunology, Saint Louis University, Saint Louis, MO, United States
| | - Amelia K. Pinto
- Department of Molecular Microbiology and Immunology, Saint Louis University, Saint Louis, MO, United States
| | - Hana M. El Sahly
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States
| | - Mark J. Mulligan
- New York University Grossman School of Medicine, Division of Infectious Diseases and Immunology, New York, NY, United States
| | - Nadine Rouphael
- Emory University School of Medicine, Division of Infectious Diseases, Department of Internal Medicine, Atlanta, GA, United States
| | - Maria L. Alcaide
- University of Miami, Division of Infectious Diseases, Miller School of Medicine, Miami, FL, United States
| | - Kay M. Tomashek
- Division of Microbiology, Immunology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Chris Focht
- The Emmes Company, LLC., Rockville, MD, United States
| | | | | | - Anne S. De Groot
- EpiVax, Inc., Providence, RI, United States
- University of Georgia Center for Vaccines and Immunology, Athens, GA, United States
| | - Daniel F. Hoft
- Department of Internal Medicine, Saint Louis University, Division of Infectious Diseases, Allergy, and Immunology, Saint Louis, MO, United States
- Department of Molecular Microbiology and Immunology, Saint Louis University, Saint Louis, MO, United States
| |
Collapse
|
16
|
Szardenings M, Delaroque N, Kern K, Ramirez-Caballero L, Puder M, Ehrentreich-Förster E, Beige J, Zürner S, Popp G, Wolf J, Borte S. Detection of Antibodies against Endemic and SARS-CoV-2 Coronaviruses with Short Peptide Epitopes. Vaccines (Basel) 2023; 11:1403. [PMID: 37766081 PMCID: PMC10535424 DOI: 10.3390/vaccines11091403] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 08/08/2023] [Accepted: 08/17/2023] [Indexed: 09/29/2023] Open
Abstract
(1) Background: Coronavirus proteins are quite conserved amongst endemic strains (eCoV) and SARS-CoV-2. We aimed to evaluate whether peptide epitopes might serve as useful diagnostic biomarkers to stratify previous infections and COVID-19. (2) Methods: Peptide epitopes were identified at an amino acid resolution that applied a novel statistical approach to generate data sets of potential antibody binding peptides. (3) Results: Data sets from more than 120 COVID-19 or eCoV-infected patients, as well as vaccinated persons, have been used to generate data sets that have been used to search in silico for potential epitopes in proteins of SARS-CoV-2 and eCoV. Peptide epitopes were validated with >300 serum samples in synthetic peptide micro arrays and epitopes specific for different viruses, in addition to the identified cross reactive epitopes. (4) Conclusions: Most patients develop antibodies against non-structural proteins, which are useful general markers for recent infections. However, there are differences in the epitope patterns of COVID-19, and eCoV, and the S-protein vaccine, which can only be explained by a high degree of cross-reactivity between the viruses, a pre-existing immune response against some epitopes, and even an alternate processing of the vaccine proteins.
Collapse
Affiliation(s)
- Michael Szardenings
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Perlickstr. 1, 04103 Leipzig, Germany; (N.D.); (K.K.); (S.Z.); (G.P.)
- epitopic GmbH, Deutscher Platz 5e, 04103 Leipzig, Germany;
| | - Nicolas Delaroque
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Perlickstr. 1, 04103 Leipzig, Germany; (N.D.); (K.K.); (S.Z.); (G.P.)
| | - Karolin Kern
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Perlickstr. 1, 04103 Leipzig, Germany; (N.D.); (K.K.); (S.Z.); (G.P.)
- epitopic GmbH, Deutscher Platz 5e, 04103 Leipzig, Germany;
| | - Lisbeth Ramirez-Caballero
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Perlickstr. 1, 04103 Leipzig, Germany; (N.D.); (K.K.); (S.Z.); (G.P.)
| | - Marcus Puder
- epitopic GmbH, Deutscher Platz 5e, 04103 Leipzig, Germany;
| | - Eva Ehrentreich-Förster
- Fraunhofer Institute for Cell Therapy and Immunology, Branch Bioanalytics and Bioprocesses IZI-BB, Am Mühlenberg 13, 14476 Potsdam, Germany;
| | - Joachim Beige
- Martin-Luther-University Halle/Wittenberg, Medical Clinic 2, 06112 Halle, Germany;
| | - Sebastian Zürner
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Perlickstr. 1, 04103 Leipzig, Germany; (N.D.); (K.K.); (S.Z.); (G.P.)
- WINF/Informationsmanagement, University Leipzig, Grimmaische Straße 12, 04109 Leipzig, Germany
| | - Georg Popp
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Perlickstr. 1, 04103 Leipzig, Germany; (N.D.); (K.K.); (S.Z.); (G.P.)
| | - Johannes Wolf
- Department of Laboratory Medicine, Hospital St. Georg, Delitzscher Strasse 141, 04129 Leipzig, Germany; (J.W.); (S.B.)
- ImmunoDeficiencyCenter Leipzig (IDCL), Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiency Diseases, Hospital St. Georg, Delitzscher Strasse 141, 04129 Leipzig, Germany
| | - Stephan Borte
- Department of Laboratory Medicine, Hospital St. Georg, Delitzscher Strasse 141, 04129 Leipzig, Germany; (J.W.); (S.B.)
- ImmunoDeficiencyCenter Leipzig (IDCL), Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiency Diseases, Hospital St. Georg, Delitzscher Strasse 141, 04129 Leipzig, Germany
| |
Collapse
|
17
|
Jin X, Liu X, Shen C. A systemic review of T-cell epitopes defined from the proteome of SARS-CoV-2. Virus Res 2023; 324:199024. [PMID: 36526016 PMCID: PMC9757803 DOI: 10.1016/j.virusres.2022.199024] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 12/10/2022] [Accepted: 12/12/2022] [Indexed: 12/15/2022]
Abstract
Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection remains in a global pandemic, and no eradicative therapy is currently available. Host T cells have been shown to play a crucial role in the antiviral immune protection and pathology in Coronavirus disease 2019 (COVID-19) patients; thus, identifying sufficient T-cell epitopes from the SARS-CoV-2 proteome can contribute greatly to the development of T-cell epitope vaccines and the precise evaluation of host SARS-CoV-2-specific cellular immunity. This review presents a comprehensive map of T-cell epitopes functionally validated from SARS-CoV-2 antigens, the human leukocyte antigen (HLA) supertypes to present these epitopes, and the strategies to screen and identify T-cell epitopes. To the best of our knowledge, a total of 1349 CD8+ T-cell epitopes and 790 CD4+ T-cell epitopes have been defined by functional experiments thus far, but most are presented by approximately twenty common HLA supertypes, such as HLA-A0201, A2402, B0702, DR15, DR7 and DR11 molecules, and 74-80% of the T-cell epitopes are derived from S protein and nonstructural protein. These data provide useful insight into the development of vaccines and specific T-cell detection systems. However, the currently defined T-cell epitope repertoire cannot cover the HLA polymorphism of major populations in an indicated geographic region. More research is needed to depict an overall landscape of T-cell epitopes, which covers the overall SARS-CoV-2 proteome and global patients.
Collapse
Affiliation(s)
- Xiaoxiao Jin
- Northern Jiangsu People's Hospital, Yangzhou, Jiangsu, China 225002; Department of Microbiology and Immunology, Medical School of Southeast University, Nanjing, Jiangsu, China 210009
| | - Xiaotao Liu
- Department of Microbiology and Immunology, Medical School of Southeast University, Nanjing, Jiangsu, China 210009
| | - Chuanlai Shen
- Department of Microbiology and Immunology, Medical School of Southeast University, Nanjing, Jiangsu, China 210009.
| |
Collapse
|
18
|
Persson G, Restori KH, Emdrup JH, Schussek S, Klausen MS, Nicol MJ, Katkere B, Rønø B, Kirimanjeswara G, Sørensen AB. DNA immunization with in silico predicted T-cell epitopes protects against lethal SARS-CoV-2 infection in K18-hACE2 mice. Front Immunol 2023; 14:1166546. [PMID: 37114047 PMCID: PMC10126292 DOI: 10.3389/fimmu.2023.1166546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 03/16/2023] [Indexed: 04/29/2023] Open
Abstract
The global SARS-CoV-2 pandemic caused significant social and economic disruption worldwide, despite highly effective vaccines being developed at an unprecedented speed. Because the first licensed vaccines target only single B-cell antigens, antigenic drift could lead to loss of efficacy against emerging SARS-CoV-2 variants. Improving B-cell vaccines by including multiple T-cell epitopes could solve this problem. Here, we show that in silico predicted MHC class I/II ligands induce robust T-cell responses and protect against severe disease in genetically modified K18-hACE2/BL6 mice susceptible to SARS-CoV-2 infection.
Collapse
Affiliation(s)
| | - Katherine H. Restori
- Department of Veterinary and Biomedical Sciences, Pennsylvania State University, University Park, PA, United States
| | | | | | | | - McKayla J. Nicol
- Department of Veterinary and Biomedical Sciences, Pennsylvania State University, University Park, PA, United States
| | - Bhuvana Katkere
- Department of Veterinary and Biomedical Sciences, Pennsylvania State University, University Park, PA, United States
| | | | - Girish Kirimanjeswara
- Department of Veterinary and Biomedical Sciences, Pennsylvania State University, University Park, PA, United States
| | | |
Collapse
|
19
|
Huffman A, Ong E, Hur J, D’Mello A, Tettelin H, He Y. COVID-19 vaccine design using reverse and structural vaccinology, ontology-based literature mining and machine learning. Brief Bioinform 2022; 23:bbac190. [PMID: 35649389 PMCID: PMC9294427 DOI: 10.1093/bib/bbac190] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 04/13/2022] [Accepted: 04/26/2022] [Indexed: 12/11/2022] Open
Abstract
Rational vaccine design, especially vaccine antigen identification and optimization, is critical to successful and efficient vaccine development against various infectious diseases including coronavirus disease 2019 (COVID-19). In general, computational vaccine design includes three major stages: (i) identification and annotation of experimentally verified gold standard protective antigens through literature mining, (ii) rational vaccine design using reverse vaccinology (RV) and structural vaccinology (SV) and (iii) post-licensure vaccine success and adverse event surveillance and its usage for vaccine design. Protegen is a database of experimentally verified protective antigens, which can be used as gold standard data for rational vaccine design. RV predicts protective antigen targets primarily from genome sequence analysis. SV refines antigens through structural engineering. Recently, RV and SV approaches, with the support of various machine learning methods, have been applied to COVID-19 vaccine design. The analysis of post-licensure vaccine adverse event report data also provides valuable results in terms of vaccine safety and how vaccines should be used or paused. Ontology standardizes and incorporates heterogeneous data and knowledge in a human- and computer-interpretable manner, further supporting machine learning and vaccine design. Future directions on rational vaccine design are discussed.
Collapse
Affiliation(s)
- Anthony Huffman
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, Michigan 48109, USA
| | - Edison Ong
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, Michigan 48109, USA
| | - Junguk Hur
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota 58202, USA
| | - Adonis D’Mello
- Department of Microbiology and Immunology, Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Hervé Tettelin
- Department of Microbiology and Immunology, Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Yongqun He
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, Michigan 48109, USA
- Unit for Laboratory Animal Medicine, Department of Microbiology and Immunology, Center for Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, Michigan 48109, USA
| |
Collapse
|
20
|
Longitudinal Assessment of SARS-CoV-2-Specific T Cell Cytokine-Producing Responses for 1 Year Reveals Persistence of Multicytokine Proliferative Responses, with Greater Immunity Associated with Disease Severity. J Virol 2022; 96:e0050922. [PMID: 35699447 PMCID: PMC9278147 DOI: 10.1128/jvi.00509-22] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Cell-mediated immunity is critical for long-term protection against most viral infections, including coronaviruses. We studied 23 severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-infected survivors over a 1-year post-symptom onset (PSO) interval by ex vivo cytokine enzyme-linked immunosorbent spot assay (ELISpot) assay. All subjects demonstrated SARS-CoV-2-specific gamma interferon (IFN-γ), interleukin 2 (IL-2), and granzyme B (GzmB) T cell responses at presentation, with greater frequencies in severe disease. Cytokines, mainly produced by CD4+ T cells, targeted all structural proteins (nucleocapsid, membrane, and spike) except envelope, with GzmB and IL-2 greater than IFN-γ. Mathematical modeling predicted that (i) cytokine responses peaked at 6 days for IFN-γ, 36 days for IL-2, and 7 days for GzmB, (ii) severe illness was associated with reduced IFN-γ and GzmB but increased IL-2 production rates, and (iii) males displayed greater production of IFN-γ, whereas females produced more GzmB. Ex vivo responses declined over time, with persistence of IL-2 in 86% and of IFN-γ and GzmB in 70% of subjects at a median of 336 days PSO. The average half-life of SARS-CoV-2-specific cytokine-producing cells was modeled to be 139 days (~4.6 months). Potent T cell proliferative responses persisted throughout observation, were CD4 dominant, and were capable of producing all 3 cytokines. Several immunodominant CD4 and CD8 epitopes identified in this study were shared by seasonal coronaviruses or SARS-CoV-1 in the nucleocapsid and membrane regions. Both SARS-CoV-2-specific CD4+ and CD8+ T cell clones were able to kill target cells, though CD8 tended to be more potent. IMPORTANCE Our findings highlight the relative importance of SARS-CoV-2-specific GzmB-producing T cell responses in SARS-CoV-2 control and shared CD4 and CD8 immunodominant epitopes in seasonal coronaviruses or SARS-CoV-1, and they indicate robust persistence of T cell memory at least 1 year after infection. Our findings should inform future strategies to induce T cell vaccines against SARS-CoV-2 and other coronaviruses.
Collapse
|
21
|
Narayanan DKL, Djearamane S, Fuloria S, Kayarohanam S, Subramaniyan V, Sekar M, Fuloria NK. A Review on DNA Vaccines in Pre-Clinical Trials Against SARS-CoV-2. JOURNAL OF EXPERIMENTAL BIOLOGY AND AGRICULTURAL SCIENCES 2022; 10:487-493. [DOI: 10.18006/2022.10(3).487.493] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2024]
Abstract
COVID 19 Pandemic is caused by the viral pathogen severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Scientific fraternity worldwide swiftly developed various types of vaccines for the prevention and as mitigation measures for curbing the pandemic. Traditional inactivated vaccines, mRNA vaccines (protein subunits such as spike proteins), and viral vector vaccines (non-replicating vectors with protein subunits) have been approved by World Health Organisation (WHO) for emergency use. The emergence of many mutated variants has been a worrying factor in the fight against the pandemic. There has been continuous research in the quest for more therapeutics, especially vaccines to curb and stop the pandemic. According to WHO, there are 194 vaccines in pre-clinical trials belonging to various types out of which sixteen is DNA vaccines. In this review, we have discussed the advantages and disadvantages of the DNA vaccines for Covid - 19. This article tried to explore the available information on DNA vaccines and their current status against Covid – 19 which are in pre-clinical trials.
Collapse
|
22
|
Hassan J, Haigh C, Ahmed T, Uddin MJ, Das DB. Potential of Microneedle Systems for COVID-19 Vaccination: Current Trends and Challenges. Pharmaceutics 2022; 14:1066. [PMID: 35631652 PMCID: PMC9144974 DOI: 10.3390/pharmaceutics14051066] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Revised: 04/27/2022] [Accepted: 05/09/2022] [Indexed: 12/12/2022] Open
Abstract
To prevent the coronavirus disease 2019 (COVID-19) pandemic and aid restoration to prepandemic normality, global mass vaccination is urgently needed. Inducing herd immunity through mass vaccination has proven to be a highly effective strategy for preventing the spread of many infectious diseases, which protects the most vulnerable population groups that are unable to develop immunity, such as people with immunodeficiencies or weakened immune systems due to underlying medical or debilitating conditions. In achieving global outreach, the maintenance of the vaccine potency, transportation, and needle waste generation become major issues. Moreover, needle phobia and vaccine hesitancy act as hurdles to successful mass vaccination. The use of dissolvable microneedles for COVID-19 vaccination could act as a major paradigm shift in attaining the desired goal to vaccinate billions in the shortest time possible. In addressing these points, we discuss the potential of the use of dissolvable microneedles for COVID-19 vaccination based on the current literature.
Collapse
Affiliation(s)
- Jasmin Hassan
- Drug Delivery & Therapeutics Lab, Dhaka 1212, Bangladesh; (J.H.); (T.A.)
| | - Charlotte Haigh
- Department of Chemical Engineering, Loughborough University, Epinal Way, Loughborough LE11 3TU, UK;
| | - Tanvir Ahmed
- Drug Delivery & Therapeutics Lab, Dhaka 1212, Bangladesh; (J.H.); (T.A.)
| | - Md Jasim Uddin
- Drug Delivery & Therapeutics Lab, Dhaka 1212, Bangladesh; (J.H.); (T.A.)
- Faculty of Engineering and Science, University of Greenwich, Chatham Maritime, Kent ME4 4TB, UK
- Department of Pharmacy, Brac University, 66 Mohakhali, Dhaka 1212, Bangladesh
| | - Diganta B. Das
- Department of Chemical Engineering, Loughborough University, Epinal Way, Loughborough LE11 3TU, UK;
| |
Collapse
|
23
|
Designing AbhiSCoVac - A single potential vaccine for all ‘corona culprits’: Immunoinformatics and immune simulation approaches. J Mol Liq 2022; 351:118633. [PMID: 35125571 PMCID: PMC8801591 DOI: 10.1016/j.molliq.2022.118633] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 01/21/2022] [Accepted: 01/26/2022] [Indexed: 12/12/2022]
|
24
|
Naranbhai V, Nathan A, Kaseke C, Berrios C, Khatri A, Choi S, Getz MA, Tano-Menka R, Ofoman O, Gayton A, Senjobe F, Zhao Z, St Denis KJ, Lam EC, Carrington M, Garcia-Beltran WF, Balazs AB, Walker BD, Iafrate AJ, Gaiha GD. T cell reactivity to the SARS-CoV-2 Omicron variant is preserved in most but not all individuals. Cell 2022; 185:1041-1051.e6. [PMID: 35202566 PMCID: PMC8810349 DOI: 10.1016/j.cell.2022.01.029] [Citation(s) in RCA: 194] [Impact Index Per Article: 64.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 01/04/2022] [Accepted: 01/28/2022] [Indexed: 01/11/2023]
Abstract
The SARS-CoV-2 Omicron variant (B.1.1.529) contains mutations that mediate escape from antibody responses, although the extent to which these substitutions in spike and non-spike proteins affect T cell recognition is unknown. In this study, we show that T cell responses in individuals with prior infection, vaccination, both prior infection and vaccination, and boosted vaccination are largely preserved to Omicron spike and non-spike proteins. However, we also identify a subset of individuals (∼21%) with a >50% reduction in T cell reactivity to the Omicron spike. Evaluation of functional CD4+ and CD8+ memory T cell responses confirmed these findings and revealed that reduced recognition to Omicron spike is primarily observed within the CD8+ T cell compartment potentially due to escape from HLA binding. Booster vaccination enhanced T cell responses to Omicron spike. In contrast to neutralizing immunity, these findings suggest preservation of T cell responses to the Omicron variant, although with reduced reactivity in some individuals.
Collapse
Affiliation(s)
- Vivek Naranbhai
- Massachusetts General Hospital Cancer Center, Massachusetts General Hospital, Boston, MA 02114, USA; Dana-Farber Cancer Institute, Boston, MA 02215, USA; Center for the AIDS Programme of Research in South Africa, Durban 4001, South Africa.
| | - Anusha Nathan
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA; Program in Health Sciences & Technology, Harvard Medical School, Massachusetts Institute of Technology, Boston, MA 02115, USA
| | - Clarety Kaseke
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Cristhian Berrios
- Department of Pathology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Ashok Khatri
- Massachusetts General Hospital Endocrine Division and Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Shawn Choi
- Massachusetts General Hospital Endocrine Division and Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Matthew A Getz
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Rhoda Tano-Menka
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Onosereme Ofoman
- Department of Pathology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Alton Gayton
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Fernando Senjobe
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Zezhou Zhao
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA; Program in Health Sciences & Technology, Harvard Medical School, Massachusetts Institute of Technology, Boston, MA 02115, USA
| | - Kerri J St Denis
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Evan C Lam
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Mary Carrington
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA; Basic Science Program, Frederick National Laboratory for Cancer Research in the Laboratory of Integrative Cancer Immunology, National Cancer Institute, Bethesda, MD 20892, USA
| | - Wilfredo F Garcia-Beltran
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA; Department of Pathology, Massachusetts General Hospital, Boston, MA 02114, USA
| | | | - Bruce D Walker
- Center for the AIDS Programme of Research in South Africa, Durban 4001, South Africa; Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA; The Broad Institute, Cambridge, MA 02142, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA; Institute for Medical Engineering and Science, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - A John Iafrate
- Department of Pathology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Gaurav D Gaiha
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA; Division of Gastroenterology, Massachusetts General Hospital, Boston, MA 02114, USA.
| |
Collapse
|
25
|
Saini S, Agarwal M, Pradhan A, Pareek S, Singh AK, Dhawan G, Dhawan U, Kumar Y. Exploring the role of framework mutations in enabling breadth of a cross-reactive antibody (CR3022) against the SARS-CoV-2 RBD and its variants of concern. J Biomol Struct Dyn 2022; 41:2341-2354. [PMID: 35098888 DOI: 10.1080/07391102.2022.2030800] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cross-reactive and broadly neutralizing antibodies against surface proteins of diverse strains of rapidly evolving viral pathogens like SARS-CoV-2 can prevent infection and therefore are crucial for the development of effective universal vaccines. While antibodies typically incorporate mutations in their complementarity determining regions during affinity maturation, mutations in the framework regions have been reported as players in determining properties of broadly neutralizing antibodies against HIV and the Influenza virus. We propose an increase in the cross-reactive potential of CR3022 against the emerging SARS- CoV-2 variants of concern through enhanced conformational flexibility. In this study, we use molecular dynamics simulations, in silico mutagenesis, structural modeling, and docking to explore the role of light chain FWR mutations in CR3022, a SARS-CoV anti-spike (S)-protein antibody cross-reactive to the S-protein receptor binding domain of SARS-CoV-2. Our study shows that single substitutions in the light chain framework region of CR3022 with conserved epitopes across SARS-CoV strains allow targeting of diverse antibody epitope footprints that align with the epitopes of recently-categorized neutralizing antibody classes while enabling binding to more than one strain of SARS-CoV-2. Our study has implications for rapid and evolution-based engineering of broadly neutralizing antibodies and reaffirms the role of framework mutations in effective change of antibody orientation and conformation via improved flexibility.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Samvedna Saini
- Department of Biological Sciences and Engineering, Netaji Subhas University of Technology, New Delhi, India
| | - Manusmriti Agarwal
- Department of Biological Sciences and Engineering, Netaji Subhas University of Technology, New Delhi, India.,Faculty of Technology, University of Delhi, New Delhi, India
| | - Amartya Pradhan
- Department of Biological Sciences and Engineering, Netaji Subhas University of Technology, New Delhi, India.,Faculty of Technology, University of Delhi, New Delhi, India.,Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| | - Savitha Pareek
- High Performance Computing (HPC) & AI Innovation Lab, Dell EMC, Bengaluru, India
| | - Ashish K Singh
- High Performance Computing (HPC) & AI Innovation Lab, Dell EMC, Bengaluru, India
| | - Gagan Dhawan
- Department of Biomedical Science, Acharya Narendra Dev College, University of Delhi, New Delhi, India
| | - Uma Dhawan
- Department of Biomedical Science, Bhaskaracharya College of Applied Sciences, University of Delhi, New Delhi, India
| | - Yatender Kumar
- Department of Biological Sciences and Engineering, Netaji Subhas University of Technology, New Delhi, India
| |
Collapse
|
26
|
Naranbhai V, Nathan A, Kaseke C, Berrios C, Khatri A, Choi S, Getz MA, Tano-Menka R, Ofoman O, Gayton A, Senjobe F, Denis KJS, Lam EC, Garcia-Beltran WF, Balazs AB, Walker BD, Iafrate AJ, Gaiha GD. T cell reactivity to the SARS-CoV-2 Omicron variant is preserved in most but not all prior infected and vaccinated individuals. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2022:2022.01.04.21268586. [PMID: 35018386 PMCID: PMC8750712 DOI: 10.1101/2022.01.04.21268586] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The SARS-CoV-2 Omicron variant (B.1.1.529) contains mutations that mediate escape from infection and vaccine-induced antibody responses, although the extent to which these substitutions in spike and non-spike proteins affect T cell recognition is unknown. Here we show that T cell responses in individuals with prior infection, vaccination, both prior infection and vaccination, and boosted vaccination are largely preserved to Omicron spike and non-spike proteins. However, we also identify a subset of individuals (∼21%) with a >50% reduction in T cell reactivity to the Omicron spike. Evaluation of functional CD4 + and CD8 + memory T cell responses confirmed these findings and reveal that reduced recognition to Omicron spike is primarily observed within the CD8 + T cell compartment. Booster vaccination substantially enhanced T cell responses to Omicron spike. In contrast to neutralizing immunity, these findings suggest preservation of T cell responses to the Omicron variant, although with reduced reactivity in some individuals.
Collapse
|
27
|
Yilmaz IC, Ipekoglu EM, Bulbul A, Turay N, Yildirim M, Evcili I, Yilmaz NS, Guvencli N, Aydin Y, Gungor B, Saraydar B, Bartan AG, Ibibik B, Bildik T, Baydemir İ, Sanli HA, Kayaoglu B, Ceylan Y, Yildirim T, Abras I, Ayanoglu IC, Cam SB, Ciftci Dede E, Gizer M, Erganis O, Sarac F, Uzar S, Enul H, Adiay C, Aykut G, Polat H, Yildirim IS, Tekin S, Korukluoglu G, Zeytin HE, Korkusuz P, Gursel I, Gursel M. Development and preclinical evaluation of virus-like particle vaccine against COVID-19 infection. Allergy 2022; 77:258-270. [PMID: 34519053 PMCID: PMC8653174 DOI: 10.1111/all.15091] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Revised: 08/02/2021] [Accepted: 08/15/2021] [Indexed: 12/16/2022]
Abstract
Background Vaccines that incorporate multiple SARS‐CoV‐2 antigens can further broaden the breadth of virus‐specific cellular and humoral immunity. This study describes the development and immunogenicity of SARS‐CoV‐2 VLP vaccine that incorporates the four structural proteins of SARS‐CoV‐2. Methods VLPs were generated in transiently transfected HEK293 cells, purified by multimodal chromatography, and characterized by tunable‐resistive pulse sensing, AFM, SEM, and TEM. Immunoblotting studies verified the protein identities of VLPs. Cellular and humoral immune responses of immunized animals demonstrated the immune potency of the formulated VLP vaccine. Results Transiently transfected HEK293 cells reproducibly generated vesicular VLPs that were similar in size to and expressing all four structural proteins of SARS‐CoV‐2. Alum adsorbed, K3‐CpG ODN‐adjuvanted VLPs elicited high titer anti‐S, anti‐RBD, anti‐N IgG, triggered multifunctional Th1‐biased T‐cell responses, reduced virus load, and prevented lung pathology upon live virus challenge in vaccinated animals. Conclusion These data suggest that VLPs expressing all four structural protein antigens of SARS‐CoV‐2 are immunogenic and can protect animals from developing COVID‐19 infection following vaccination.
Collapse
|
28
|
Richard G, Princiotta MF, Bridon D, Martin WD, Steinberg GD, De Groot AS. Neoantigen-based personalized cancer vaccines: the emergence of precision cancer immunotherapy. Expert Rev Vaccines 2021; 21:173-184. [PMID: 34882038 DOI: 10.1080/14760584.2022.2012456] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
INTRODUCTION The field of cancer therapy has undergone a major transformation in less than a decade due to the introduction of checkpoint inhibitors, the advent of next generation sequencing and the discovery of neoantigens. The key observation that the breadth of each patient's immune response to the unique mutations or neoantigens present in their tumor is directly related to their survival has led oncologists to focus on driving immune responses to neoantigens through vaccination. Oncology has entered the era of precision immunotherapy, and cancer vaccine development is undergoing a paradigm shift. AREAS COVERED Neoantigens are short peptide sequences found in tumors, but not noncancerous tissues, the vast majority of which are unique to each patient. In addition to providing a description of the distinguishing features of neoantigen discovery platforms, this review will address cross-cutting personalized cancer vaccine design themes and developmental stumbling blocks. EXPERT OPINION Immunoinformatic pipelines that can rapidly scan cancer genomes and identify 'the best' neoantigens are in high demand. Despite the need for such tools, immunoinformatic methods for identifying neoepitopes in cancer genomes are diverse and have not been well-validated. Validation of 'personalized vaccine design pipelines' will bring about a revolution in neoantigen-based vaccine design and delivery.
Collapse
Affiliation(s)
| | | | | | | | - Gary D Steinberg
- EpiVax Therapeutics, Inc., Providence, RI, USA.,Perlmutter Cancer Center, Department of Urology at NYU Grossman School of Medicine, NYU Langone Health, New York, NY, USA
| | - Anne S De Groot
- EpiVax, Inc., Providence, RI, USA.,Center for Vaccines and Immunology, University of Georgia, Athens, GA, USA
| |
Collapse
|
29
|
Ferreira CS, Martins YC, Souza RC, Vasconcelos ATR. EpiCurator: an immunoinformatic workflow to predict and prioritize SARS-CoV-2 epitopes. PeerJ 2021; 9:e12548. [PMID: 34909278 PMCID: PMC8641484 DOI: 10.7717/peerj.12548] [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: 07/03/2021] [Accepted: 11/04/2021] [Indexed: 12/12/2022] Open
Abstract
The ongoing coronavirus 2019 (COVID-19) pandemic, triggered by the emerging SARS-CoV-2 virus, represents a global public health challenge. Therefore, the development of effective vaccines is an urgent need to prevent and control virus spread. One of the vaccine production strategies uses the in silico epitope prediction from the virus genome by immunoinformatic approaches, which assist in selecting candidate epitopes for in vitro and clinical trials research. This study introduces the EpiCurator workflow to predict and prioritize epitopes from SARS-CoV-2 genomes by combining a series of computational filtering tools. To validate the workflow effectiveness, SARS-CoV-2 genomes retrieved from the GISAID database were analyzed. We identified 11 epitopes in the receptor-binding domain (RBD) of Spike glycoprotein, an important antigenic determinant, not previously described in the literature or published on the Immune Epitope Database (IEDB). Interestingly, these epitopes have a combination of important properties: recognized in sequences of the current variants of concern, present high antigenicity, conservancy, and broad population coverage. The RBD epitopes were the source for a multi-epitope design to in silico validation of their immunogenic potential. The multi-epitope overall quality was computationally validated, endorsing its efficiency to trigger an effective immune response since it has stability, high antigenicity and strong interactions with Toll-Like Receptors (TLR). Taken together, the findings in the current study demonstrated the efficacy of the workflow for epitopes discovery, providing target candidates for immunogen development.
Collapse
Affiliation(s)
- Cristina S. Ferreira
- Bioinformatics Laboratory, National Laboratory of Scientific Computation, Petrópolis, Rio de Janeiro, Brazil
| | - Yasmmin C. Martins
- Bioinformatics Laboratory, National Laboratory of Scientific Computation, Petrópolis, Rio de Janeiro, Brazil
| | - Rangel Celso Souza
- Bioinformatics Laboratory, National Laboratory of Scientific Computation, Petrópolis, Rio de Janeiro, Brazil
| | - Ana Tereza R. Vasconcelos
- Bioinformatics Laboratory, National Laboratory of Scientific Computation, Petrópolis, Rio de Janeiro, Brazil
| |
Collapse
|
30
|
Bukhari SNH, Jain A, Haq E, Mehbodniya A, Webber J. Ensemble Machine Learning Model to Predict SARS-CoV-2 T-Cell Epitopes as Potential Vaccine Targets. Diagnostics (Basel) 2021; 11:diagnostics11111990. [PMID: 34829338 PMCID: PMC8617960 DOI: 10.3390/diagnostics11111990] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 10/20/2021] [Accepted: 10/21/2021] [Indexed: 01/03/2023] Open
Abstract
An ongoing outbreak of coronavirus disease 2019 (COVID-19), caused by a single-stranded RNA virus called severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has caused a worldwide pandemic that continues to date. Vaccination has proven to be the most effective technique, by far, for the treatment of COVID-19 and to combat the outbreak. Among all vaccine types, epitope-based peptide vaccines have received less attention and hold a large untapped potential for boosting vaccine safety and immunogenicity. Peptides used in such vaccine technology are chemically synthesized based on the amino acid sequences of antigenic proteins (T-cell epitopes) of the target pathogen. Using wet-lab experiments to identify antigenic proteins is very difficult, expensive, and time-consuming. We hereby propose an ensemble machine learning (ML) model for the prediction of T-cell epitopes (also known as immune relevant determinants or antigenic determinants) against SARS-CoV-2, utilizing physicochemical properties of amino acids. To train the model, we retrieved the experimentally determined SARS-CoV-2 T-cell epitopes from Immune Epitope Database and Analysis Resource (IEDB) repository. The model so developed achieved accuracy, AUC (Area under the ROC curve), Gini, specificity, sensitivity, F-score, and precision of 98.20%, 0.991, 0.994, 0.971, 0.982, 0.990, and 0.981, respectively, using a test set consisting of SARS-CoV-2 peptides (T-cell epitopes and non-epitopes) obtained from IEDB. The average accuracy of 97.98% was recorded in repeated 5-fold cross validation. Its comparison with 05 robust machine learning classifiers and existing T-cell epitope prediction techniques, such as NetMHC and CTLpred, suggest the proposed work as a better model. The predicted epitopes from the current model could possess a high probability to act as potential peptide vaccine candidates subjected to in vitro and in vivo scientific assessments. The model developed would help scientific community working in vaccine development save time to screen the active T-cell epitope candidates of SARS-CoV-2 against the inactive ones.
Collapse
Affiliation(s)
- Syed Nisar Hussain Bukhari
- University Institute of Computing, Chandigarh University, NH-95, Chandigarh-Ludhiana Highway, Mohali 140413, India;
- Correspondence:
| | - Amit Jain
- University Institute of Computing, Chandigarh University, NH-95, Chandigarh-Ludhiana Highway, Mohali 140413, India;
| | - Ehtishamul Haq
- Department of Biotechnology, University of Kashmir, Srinagar 190006, India;
| | - Abolfazl Mehbodniya
- Department of Electronics and Communication Engineering, Kuwait College of Science and Technology, Kuwait City 13133, Kuwait;
| | - Julian Webber
- Graduate School of Engineering Science, Osaka University, Toyonaka, Osaka 560-8531, Japan;
| |
Collapse
|