1
|
Qu Y, Wang D, Zhang Y, Shen F, Xia B, Xu Q, Wang Q, Kong H, Zhu Y, Wang L, Willner I, Yang X, Fan C, Sun L. DNA-Engineered Modular Nanovaccines Featuring Precise Topology for Enhanced Immunogenicity. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025:e2500577. [PMID: 40371439 DOI: 10.1002/adma.202500577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 04/17/2025] [Indexed: 05/16/2025]
Abstract
Multivalent display of antigens can boost subunit vaccine immunogenicity. However, owing to the inherent difficulty in programmatically controlling the topology of multivalent antigens, its impact on antigen immunogenicity remains elusive. In this study, DNA-mediated modular precision assembly is employed to organize SARS-CoV-2 receptor-binding domains (RBDs) with different topological connections while preserving their epitopes. It is found that branching-connected RBDs induced significantly higher IgG titers than linear-connected RBDs at higher antigen valency (≥4). This increase in IgG response is associated with stronger B cell proliferation, likely due to enhanced antigen-receptor synergistic interactions leading to enhanced B cell receptor signaling. Branching-connected RBDs also provided superior humoral immunity in mice and stronger protection in SARS-CoV-2-infected hamsters compared to adjuvanted RBD. This work highlights the role of antigen topology in vaccine design and offers a universal modular platform for producing more effective subunit vaccines.
Collapse
Affiliation(s)
- Yanfei Qu
- Institute of Materiobiology, Department of Chemistry, College of Science, Shanghai University, Shanghai, 200444, China
| | - Dawei Wang
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai, 200433, China
| | - Yunlong Zhang
- Institute of Materiobiology, Department of Chemistry, College of Science, Shanghai University, Shanghai, 200444, China
| | - Fengyun Shen
- Institute of Materiobiology, Department of Chemistry, College of Science, Shanghai University, Shanghai, 200444, China
| | - Binghui Xia
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai, 200433, China
| | - Qin Xu
- Shanghai Synchrotron Radiation Facility (SSRF), Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai, 201204, China
| | - Qisheng Wang
- Shanghai Synchrotron Radiation Facility (SSRF), Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai, 201204, China
| | - Huating Kong
- Shanghai Synchrotron Radiation Facility (SSRF), Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai, 201204, China
| | - Ying Zhu
- Institute of Materiobiology, Department of Chemistry, College of Science, Shanghai University, Shanghai, 200444, China
| | - Lihua Wang
- Institute of Materiobiology, Department of Chemistry, College of Science, Shanghai University, Shanghai, 200444, China
| | - Itamar Willner
- Institute of Chemistry, The Minerva Center for Bio-hybrid Complex Systems, The Hebrew University of Jerusalem, Jerusalem, 91904, Israel
| | - Xiurong Yang
- School of Chemistry and Chemical Engineering, New Cornerstone Science Laboratory, Frontiers Science Center for Transformative Molecules, Zhang jiang Institute for Advanced Study and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Chunhai Fan
- School of Chemistry and Chemical Engineering, New Cornerstone Science Laboratory, Frontiers Science Center for Transformative Molecules, Zhang jiang Institute for Advanced Study and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Lele Sun
- Institute of Materiobiology, Department of Chemistry, College of Science, Shanghai University, Shanghai, 200444, China
| |
Collapse
|
2
|
Burmas L, Lee WS, Kelly A, Webster R, Esterbauer R, Kent SJ, Wheatley AK, Juno JA, Tan HX. Modulation of germinal center and antibody dynamics via ipsilateral versus contralateral immunization against SARS-CoV-2. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025:vkae067. [PMID: 40073085 DOI: 10.1093/jimmun/vkae067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 12/15/2024] [Indexed: 03/14/2025]
Abstract
Human clinical trials have reported immunological outcomes can differ between ipsilateral (same side) and contralateral (alternate sides) prime-boost vaccination. However, our mechanistic understanding of how keeping or shifting the anatomical sites of immunization impacts the resultant germinal centers (GCs) and antibody responses is limited. Here, we use an adjuvanted SARS-CoV-2 spike vaccine to dissect GC dynamics in draining lymph nodes and serological outcomes following ipsilateral or contralateral prime-boost vaccination in C57BL/6 mice. Contralateral vaccination elicited independent GCs at distinct lymph nodes, where robust secondary GCs only appeared upon secondary distal vaccination, while ongoing GCs from the primary site were not boosted. In contrast, ipsilateral vaccination resulted in sustained GC activity. Ipsilateral vaccination accelerated the development of antibody titers against ancestral (wild-type [WT]), Beta, and BA.1 but were later comparable between ipsilateral and contralateral groups in terms of magnitude, durability, and neutralization capacity beyond 28 d. Using a heterologous SARS-CoV-2 WT/BA.1 spike prime-boost model, cross-reactive GC responses were generated against WT and BA.1 spike, with analogous serological and GC dynamics to our homologous model. Within the cross-reactive GC B cells, differential recognition of WT and BA.1 antigens was observed and were further compartmentalized in primary or secondary GCs, depending on ipsilateral or contralateral regimes. Collectively, maintaining a common prime-boost site augments the kinetics of memory B cell recall and transiently drive higher antibody titers, but longer-term serological outcomes are unaffected by the anatomical localization of immunization.
Collapse
Affiliation(s)
- Lauren Burmas
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Wen Shi Lee
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Andrew Kelly
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Rosela Webster
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Robyn Esterbauer
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Stephen J Kent
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
- Melbourne Sexual Health Centre and Department of Infectious Diseases, Alfred Hospital and Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Adam K Wheatley
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Jennifer A Juno
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Hyon-Xhi Tan
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| |
Collapse
|
3
|
Jiang Y, Sanyal M, Hussein NA, Baghdasaryan A, Zhang M, Wang F, Ren F, Li J, Zhu G, Meng Y, Adamska JZ, Mellins E, Dai H. A SARS-CoV-2 vaccine on an NIR-II/SWIR emitting nanoparticle platform. SCIENCE ADVANCES 2025; 11:eadp5539. [PMID: 39919189 PMCID: PMC11804919 DOI: 10.1126/sciadv.adp5539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Accepted: 01/07/2025] [Indexed: 02/09/2025]
Abstract
The COVID-19 pandemic caused a global health crisis that resulted in millions of deaths. Effective vaccines have played central roles in curtailing the pandemic. Here, we developed a down-converting near-infrared IIb (NIR-IIb; 1500 to 1700 nanometers) luminescent, pure NaErF4@NaYF4 rare-earth nanoparticle (pEr) as vaccine carriers. The pEr nanoparticles were coated with three layers of cross-linked biocompatible polymers (pEr-P3; ~55 nanometers) and conjugated to the receptor binding domain (RBD) of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike protein. Upon subcutaneous injection of the pEr-P3-RBD nanovaccine in mice, in vivo NIR-IIb imaging revealed active vaccine trafficking and migration to lymph nodes through lymphatic vessels. Two doses of the adjuvant-free vaccine elicited long-lasting (>7 months) high titers of serum viral neutralization antibody and anti-RBD immunoglobulin G, along with robust RBD-specific germinal center B cells and T follicular helper cells. We devised in vivo NIR-II molecular imaging of RBD-specific cells in lymph nodes, opening noninvasive assessments of vaccine-elicited immune responses longitudinally.
Collapse
Affiliation(s)
- Yingying Jiang
- Department of Chemistry and Bio-X, Stanford University, Stanford, CA, USA
- Department of Chemistry, The University of Hong Kong, Pokfulam, Hong Kong SAR
- Materials Innovation Institute for Life Sciences and Energy (MILES), HKU-SIRI, Shenzhen, P. R. China
| | - Mrinmoy Sanyal
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
- Department of Biochemistry, School of Medicine, Stanford University, Stanford, CA, USA
| | - Noor A. Hussein
- Department of Pediatrics, Stanford University, Stanford, CA, USA
| | - Ani Baghdasaryan
- Department of Chemistry and Bio-X, Stanford University, Stanford, CA, USA
| | - Mengzhen Zhang
- Department of Chemistry and Bio-X, Stanford University, Stanford, CA, USA
| | - Feifei Wang
- Department of Chemistry and Bio-X, Stanford University, Stanford, CA, USA
- Materials Innovation Institute for Life Sciences and Energy (MILES), HKU-SIRI, Shenzhen, P. R. China
- Department of Electrical and Electronic Engineering, The University of Hong Kong, Pokfulam, Hong Kong SAR
| | - Fuqiang Ren
- Department of Chemistry and Bio-X, Stanford University, Stanford, CA, USA
| | - Jiachen Li
- Department of Chemistry and Bio-X, Stanford University, Stanford, CA, USA
| | - Guanzhou Zhu
- Department of Chemistry and Bio-X, Stanford University, Stanford, CA, USA
| | - Yifan Meng
- Department of Chemistry and Bio-X, Stanford University, Stanford, CA, USA
| | - Julia Zofia Adamska
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
| | | | - Hongjie Dai
- Department of Chemistry and Bio-X, Stanford University, Stanford, CA, USA
- Department of Chemistry, The University of Hong Kong, Pokfulam, Hong Kong SAR
- Materials Innovation Institute for Life Sciences and Energy (MILES), HKU-SIRI, Shenzhen, P. R. China
| |
Collapse
|
4
|
Pilapitiya D, Lee WS, Vu MN, Kelly A, Webster RH, Koutsakos M, Kent SJ, Juno JA, Tan HX, Wheatley AK. Mucosal vaccination against SARS-CoV-2 using recombinant influenza viruses delivering self-assembling nanoparticles. Vaccine 2025; 46:126668. [PMID: 39740385 DOI: 10.1016/j.vaccine.2024.126668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 11/07/2024] [Accepted: 12/21/2024] [Indexed: 01/02/2025]
Abstract
Recombinant influenza viruses are promising vectors that can bolster antibody and resident lymphocyte responses within mucosal sites. This study evaluates recombinant influenza viruses with SARS-CoV-2 RBD genes in eliciting mucosal and systemic responses. Using reverse genetics, we generated replication-competent recombinant influenza viruses carrying heterologous RBD genes in monomeric, trimeric, or ferritin-based nanoparticle forms. Following intranasal immunisation, mice developed potent serological anti-RBD responses, with ferritin nanoparticles superseding monomeric or trimeric RBD responses. While parenteral and mucosal immunisation elicited robust anti-RBD IgG in serum, mucosal immunisation seeded respiratory IgA, RBD-specific lung-resident memory and germinal centre (GC) B cells. In animals with prior intramuscular vaccination, intranasal boosting with recombinant influenza vectors augmented mucosal IgG, IgA, GC and memory B cells, and SARS-CoV-2 lung neutralising titres. Recall of RBD-specific memory B cells via antigen re-exposure in the lung increased antibody-secreting cells in the lung-draining lymph nodes, with maintenance of lung GC B cells. Recombinant influenza-based vaccines effectively deliver highly immunogenic self-assembling nanoparticles, generating antibodies and B cells in the respiratory mucosa. This strategy provides a tractable pathway to augment lung-localised responses against recurrent respiratory viral infections.
Collapse
MESH Headings
- Animals
- Nanoparticles/administration & dosage
- Nanoparticles/chemistry
- SARS-CoV-2/immunology
- COVID-19/prevention & control
- COVID-19/immunology
- Antibodies, Viral/immunology
- Antibodies, Viral/blood
- Mice
- COVID-19 Vaccines/immunology
- COVID-19 Vaccines/administration & dosage
- Influenza Vaccines/immunology
- Influenza Vaccines/administration & dosage
- Female
- Administration, Intranasal
- Immunity, Mucosal
- Immunoglobulin G/immunology
- Vaccines, Synthetic/immunology
- Vaccines, Synthetic/administration & dosage
- Mice, Inbred BALB C
- Antibodies, Neutralizing/immunology
- Immunoglobulin A/immunology
- Spike Glycoprotein, Coronavirus/immunology
- Spike Glycoprotein, Coronavirus/genetics
- Vaccination/methods
- Lung/immunology
- B-Lymphocytes/immunology
- Humans
- Orthomyxoviridae/genetics
- Orthomyxoviridae/immunology
Collapse
Affiliation(s)
- Devaki Pilapitiya
- Department of Microbiology and Immunology, University of Melbourne, at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Wen Shi Lee
- Department of Microbiology and Immunology, University of Melbourne, at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Mai N Vu
- Department of Microbiology and Immunology, University of Melbourne, at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Andrew Kelly
- Department of Microbiology and Immunology, University of Melbourne, at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Rosela H Webster
- Department of Microbiology and Immunology, University of Melbourne, at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Marios Koutsakos
- Department of Microbiology and Immunology, University of Melbourne, at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Stephen J Kent
- Department of Microbiology and Immunology, University of Melbourne, at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia; Melbourne Sexual Health Centre and Department of Infectious Diseases, Alfred Hospital and Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Jennifer A Juno
- Department of Microbiology and Immunology, University of Melbourne, at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Hyon-Xhi Tan
- Department of Microbiology and Immunology, University of Melbourne, at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia.
| | - Adam K Wheatley
- Department of Microbiology and Immunology, University of Melbourne, at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia.
| |
Collapse
|
5
|
Doroud D, Daneshi M, Kazemi-Lomedash F, Eftekhari Z. Comprehensive review of preclinical evaluation strategies for COVID-19 vaccine candidates: assessing immunogenicity, toxicology, and safety profiles. IRANIAN JOURNAL OF MICROBIOLOGY 2025; 17:1-18. [PMID: 40330066 PMCID: PMC12049746 DOI: 10.18502/ijm.v17i1.17796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/08/2025]
Abstract
Following the worldwide spread of Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), there is a vital requirement for safe and effective vaccines against Coronavirus disease 2019 (COVID-19). Therefore, several vaccine-candidate platforms have been designed, tested, and developed. Based on guidelines, preclinical studies are recommended to assess the safety and potency of COVID-19 vaccines in appropriate in vitro and in vivo settings. These studies provide essential information to describe the potential toxic properties of a vaccine and the formulation of vaccine agents during the preclinical trial phase. In toxicology studies, several factors must be considered, such as the appropriate animal species and strains, dosing timetable, mode of administration, time of sampling for biochemistry and antibody evaluation, and necropsy. Pharmacokinetic/ biodistribution studies are not usually required for infectious disease prophylaxis vaccines unless the vaccine contains a novel substance. Evaluating their biodistribution is crucial for newly developed vaccines, such as lipid nanoparticles -messenger RNA (LNP-mRNA), DNA, and Viral vectors in non-replicated (VVnr), or recombinant virus vaccines. The review highlights the importance of preclinical studies in assessing the safety and efficacy of vaccine candidates. This guidance is essential for researchers and manufacturers to design effective vaccines that can progress to clinical trials safely.
Collapse
Affiliation(s)
- Delaram Doroud
- Department of Immunotherapy and Leishmania Vaccine Research, Pasteur Institute of Iran, Tehran, Iran
| | - Mojtaba Daneshi
- Department of Animal Sciences, Center for Nutrition and Pregnancy, North Dakota State University, Fargo,USA
| | - Fatemeh Kazemi-Lomedash
- Biotechnology Research Center, Department of Biotechnology, Pasteur Institute of Iran, Tehran, Iran
| | - Zohre Eftekhari
- Biotechnology Research Center, Department of Biotechnology, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
6
|
Maltseva M, Keeshan A, Cooper C, Langlois MA. Immune imprinting: The persisting influence of the first antigenic encounter with rapidly evolving viruses. Hum Vaccin Immunother 2024; 20:2384192. [PMID: 39149872 PMCID: PMC11328881 DOI: 10.1080/21645515.2024.2384192] [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: 05/02/2024] [Revised: 07/08/2024] [Accepted: 07/21/2024] [Indexed: 08/17/2024] Open
Abstract
Immune imprinting is a phenomenon that stems from the fundamentals of immunological memory. Upon recurrent exposures to an evolving pathogen, the immune system must weigh the benefits of rapidly recalling established antibody repertoires with greater affinity to the initial variant or invest additional time and energy in producing de novo responses specific to the emerging variant. In this review, we delve into the mechanistic complexities of immune imprinting and its role in shaping subsequent immune responses, both de novo and recall, against rapidly evolving respiratory viruses such as influenza and coronaviruses. By exploring the duality of immune imprinting, we examine its potential to both enhance or hinder immune protection against disease, while emphasizing the role of host and viral factors. Finally, we explore how different vaccine platforms may affect immune imprinting and comment on vaccine strategies that can favor de novo variant-specific antibody responses.
Collapse
Affiliation(s)
- Mariam Maltseva
- Department of Biochemistry, Microbiology & Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Alexa Keeshan
- School of Epidemiology and Public Health, University of Ottawa Faculty of Medicine, Ottawa, ON, Canada
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Curtis Cooper
- School of Epidemiology and Public Health, University of Ottawa Faculty of Medicine, Ottawa, ON, Canada
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Department of Medicine, University of Ottawa, Ottawa, ON, Canada
- Division of Infectious Diseases, Ottawa Hospital Research Institute Clinical Epidemiology Program, Ottawa, ON, Canada
| | - Marc-André Langlois
- Department of Biochemistry, Microbiology & Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Canada
- Center for Infection, Immunity, and Inflammation (CI3), University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
7
|
Hassanel DNBP, Pilkington EH, Ju Y, Kent SJ, Pouton CW, Truong NP. Replacing poly(ethylene glycol) with RAFT lipopolymers in mRNA lipid nanoparticle systems for effective gene delivery. Int J Pharm 2024; 665:124695. [PMID: 39288840 DOI: 10.1016/j.ijpharm.2024.124695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 08/07/2024] [Accepted: 09/09/2024] [Indexed: 09/19/2024]
Abstract
Lipid nanoparticles (LNPs) have emerged as promising carriers to efficiently transport mRNA into cells for protein translation, as seen with the mRNA vaccines used against COVID-19. However, they contain a widely used polymer - poly(ethylene glycol) (PEG) - which lacks the functionality to be easily modified (which could effectively control the physicochemical properties of the LNPs such as its charge), and is also known to be immunogenic. Thus, it is desirable to explore alternative polymers which can replace the PEG component in mRNA LNP vaccines and therapeutics, while still maintaining their efficacy. Herein, we employed reversible addition-fragmentation chain transfer (RAFT) polymerisation to synthesise five PEG-lipid alternatives that could stabilise LNPs encapsulating mRNA or pDNA molecules. Importantly, the resultant RAFT lipopolymer LNPs exhibit analogous or higher in vivo gene expression and antigen-specific antibody production compared to traditional PEG-based formulations. Our synthesis strategy which allows the introduction of positive charges along the lipopolymer backbone also significantly improved the in vivo gene expression. This work expands the potential of RAFT polymer-conjugated LNPs as promising mRNA carriers and offers an innovative strategy for the development of PEG-free mRNA vaccines and therapeutics.
Collapse
Affiliation(s)
| | - Emily H Pilkington
- Monash Institute of Pharmaceutical Sciences, Monash University, 399 Royal Parade, Parkville, VIC 3052, Australia; Peter Doherty Institute for Infection and Immunity, University of Melbourne, 792 Elizabeth St, Melbourne, VIC 3000, Australia
| | - Yi Ju
- Peter Doherty Institute for Infection and Immunity, University of Melbourne, 792 Elizabeth St, Melbourne, VIC 3000, Australia; School of Science and School of Health and Biomedical Sciences, RMIT University, 264 Plenty Rd, Mill Park, VIC 3083, Australia
| | - Stephen J Kent
- Peter Doherty Institute for Infection and Immunity, University of Melbourne, 792 Elizabeth St, Melbourne, VIC 3000, Australia
| | - Colin W Pouton
- Monash Institute of Pharmaceutical Sciences, Monash University, 399 Royal Parade, Parkville, VIC 3052, Australia
| | - Nghia P Truong
- Monash Institute of Pharmaceutical Sciences, Monash University, 399 Royal Parade, Parkville, VIC 3052, Australia.
| |
Collapse
|
8
|
Dvorscek AR, McKenzie CI, Stäheli VC, Ding Z, White J, Fabb SA, Lim L, O'Donnell K, Pitt C, Christ D, Hill DL, Pouton CW, Burnett DL, Brink R, Robinson MJ, Tarlinton DM, Quast I. Conversion of vaccines from low to high immunogenicity by antibodies with epitope complementarity. Immunity 2024; 57:2433-2452.e7. [PMID: 39305904 DOI: 10.1016/j.immuni.2024.08.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 05/06/2024] [Accepted: 08/27/2024] [Indexed: 10/11/2024]
Abstract
Existing antibodies (Abs) have varied effects on humoral immunity during subsequent infections. Here, we leveraged in vivo systems that allow precise control of antigen-specific Abs and B cells to examine the impact of Ab dose, affinity, and specificity in directing B cell activation and differentiation. Abs competing with the B cell receptor (BCR) epitope showed affinity-dependent suppression. By contrast, Abs targeting a complementary epitope, not overlapping with the BCR, shifted B cell differentiation toward Ab-secreting cells. Such Abs allowed for potent germinal center (GC) responses to otherwise poorly immunogenic sites by promoting antigen capture and presentation by low-affinity B cells. These mechanisms jointly diversified the B cell repertoire by facilitating the recruitment of high- and low-affinity B cells into Ab-secreting cell, GC, and memory B cell fates. Incorporation of small amounts of monoclonal Abs into protein- or mRNA-based vaccines enhanced immunogenicity and facilitated sustained immune responses, with implications for vaccine design and our understanding of protective immunity.
Collapse
Affiliation(s)
- Alexandra R Dvorscek
- Department of Immunology, Monash University, 89 Commercial Rd, Melbourne, VIC 3004, Australia
| | - Craig I McKenzie
- Department of Immunology, Monash University, 89 Commercial Rd, Melbourne, VIC 3004, Australia
| | - Vera C Stäheli
- Department of Immunology, Monash University, 89 Commercial Rd, Melbourne, VIC 3004, Australia
| | - Zhoujie Ding
- Department of Immunology, Monash University, 89 Commercial Rd, Melbourne, VIC 3004, Australia
| | - Jacqueline White
- Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, NSW 2010, Australia; St. Vincent's Clinical School, University of New South Wales, Sydney, NSW 2010, Australia
| | - Stewart A Fabb
- Monash Institute of Pharmaceutical Sciences, Monash University, 399 Royal Parade, Parkville, VIC 3052, Australia
| | - Leonard Lim
- Monash Institute of Pharmaceutical Sciences, Monash University, 399 Royal Parade, Parkville, VIC 3052, Australia
| | - Kristy O'Donnell
- Department of Immunology, Monash University, 89 Commercial Rd, Melbourne, VIC 3004, Australia
| | - Catherine Pitt
- Department of Immunology, Monash University, 89 Commercial Rd, Melbourne, VIC 3004, Australia
| | - Daniel Christ
- Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, NSW 2010, Australia; St. Vincent's Clinical School, University of New South Wales, Sydney, NSW 2010, Australia
| | - Danika L Hill
- Department of Immunology, Monash University, 89 Commercial Rd, Melbourne, VIC 3004, Australia
| | - Colin W Pouton
- Monash Institute of Pharmaceutical Sciences, Monash University, 399 Royal Parade, Parkville, VIC 3052, Australia
| | - Deborah L Burnett
- Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, NSW 2010, Australia; School of Biomedical Sciences, University of New South Wales, Sydney, NSW 2010, Australia
| | - Robert Brink
- Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, NSW 2010, Australia; St. Vincent's Clinical School, University of New South Wales, Sydney, NSW 2010, Australia
| | - Marcus J Robinson
- Department of Immunology, Monash University, 89 Commercial Rd, Melbourne, VIC 3004, Australia
| | - David M Tarlinton
- Department of Immunology, Monash University, 89 Commercial Rd, Melbourne, VIC 3004, Australia
| | - Isaak Quast
- Department of Immunology, Monash University, 89 Commercial Rd, Melbourne, VIC 3004, Australia.
| |
Collapse
|
9
|
Safitri IA, Sugijo Y, Puspasari F, Masduki FF, Ihsanawati, Giri-Rachman EA, Artarini AA, Tan MI, Natalia D. Immunogenicity studies of recombinant RBD SARS-CoV-2 as a COVID-19 vaccine candidate produced in Escherichia coli. Vaccine X 2024; 16:100443. [PMID: 38304876 PMCID: PMC10832452 DOI: 10.1016/j.jvacx.2024.100443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 01/16/2024] [Accepted: 01/18/2024] [Indexed: 02/03/2024] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 -related global COVID-19 pandemic has been impacting millions of people since its outbreak in 2020. COVID-19 vaccination has proven highly efficient in reducing illness severity and preventing infection-related fatalities. The World Health Organization has granted emergency use approval to multiple, including protein subunit technology-based, COVID-19 vaccines. Foreseeably, additional COVID-19 subunit vaccine development would be essential to meet the accessible and growing demand for effective vaccines, especially for Low-Middle-Income Countries (LMIC). The SARS-CoV-2 spike protein receptor binding domain (RBD), as the primary target for neutralizing antibodies, holds significant potential for future COVID-19 subunit vaccine development. In this study, we developed a recombinant Escherichia coli-expressed RBD (rRBD) as a vaccine candidate and evaluated its immunogenicity and preliminary toxicity in BALB/c mice. The rRBD induced humoral immune response from day 7 post-vaccination and, following the booster doses, the IgG levels increased dramatically in mice. Interestingly, our vaccine candidate also significantly induced cellular immune response, indicated by the incrased IFN-ɣ-producing cell numbers. We observed no adverse effect or local reactogenicity either in control or treated mice. Taken together, our discoveries could potentially support efficient and cost-effective vaccine antigen production, from which LMICs could particularly benefit.
Collapse
Affiliation(s)
- Intan Aghniya Safitri
- Biology Department, School of Life Science and Technology, Bandung Institute of Technology, Bandung, Indonesia
| | - Yovin Sugijo
- Biochemistry Group, Department of Chemistry, Faculty of Mathematics and Natural Science, Bandung Institute of Technology, Bandung, Indonesia
| | - Fernita Puspasari
- Biochemistry Group, Department of Chemistry, Faculty of Mathematics and Natural Science, Bandung Institute of Technology, Bandung, Indonesia
| | - Fifi Fitriyah Masduki
- Biochemistry Group, Department of Chemistry, Faculty of Mathematics and Natural Science, Bandung Institute of Technology, Bandung, Indonesia
- Bioscience and Biotechnology Research Centre, Bandung Institute of Technology, Bandung, Indonesia
| | - Ihsanawati
- Biochemistry Group, Department of Chemistry, Faculty of Mathematics and Natural Science, Bandung Institute of Technology, Bandung, Indonesia
| | - Ernawati Arifin Giri-Rachman
- Biology Department, School of Life Science and Technology, Bandung Institute of Technology, Bandung, Indonesia
- Bioscience and Biotechnology Research Centre, Bandung Institute of Technology, Bandung, Indonesia
| | - Aluicia Anita Artarini
- Pharmaceutical Biotechnology Laboratory, Pharmaceutics Department, School of Pharmacy, Bandung Institute of Technology, Bandung, Indonesia
- Bioscience and Biotechnology Research Centre, Bandung Institute of Technology, Bandung, Indonesia
| | - Marselina Irasonia Tan
- Biology Department, School of Life Science and Technology, Bandung Institute of Technology, Bandung, Indonesia
- Bioscience and Biotechnology Research Centre, Bandung Institute of Technology, Bandung, Indonesia
| | - Dessy Natalia
- Biochemistry Group, Department of Chemistry, Faculty of Mathematics and Natural Science, Bandung Institute of Technology, Bandung, Indonesia
- Bioscience and Biotechnology Research Centre, Bandung Institute of Technology, Bandung, Indonesia
| |
Collapse
|
10
|
Lee IJ, Lan YH, Wu PY, Wu YW, Chen YH, Tseng SC, Kuo TJ, Sun CP, Jan JT, Ma HH, Liao CC, Liang JJ, Ko HY, Chang CS, Liu WC, Ko YA, Chen YH, Sie ZL, Tsung SI, Lin YL, Wang IH, Tao MH. A receptor-binding domain-based nanoparticle vaccine elicits durable neutralizing antibody responses against SARS-CoV-2 and variants of concern. Emerg Microbes Infect 2023; 12:2149353. [PMID: 36395071 PMCID: PMC9793938 DOI: 10.1080/22221751.2022.2149353] [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] [Indexed: 11/18/2022]
Abstract
Numerous vaccines have been developed to address the current COVID-19 pandemic, but safety, cross-neutralizing efficacy, and long-term protectivity of currently approved vaccines are still important issues. In this study, we developed a subunit vaccine, ASD254, by using a nanoparticle vaccine platform to encapsulate the SARS-CoV-2 spike receptor-binding domain (RBD) protein. As compared with the aluminum-adjuvant RBD vaccine, ASD254 induced higher titers of RBD-specific antibodies and generated 10- to 30-fold more neutralizing antibodies. Mice vaccinated with ASD254 showed protective immune responses against SARS-CoV-2 challenge, with undetectable infectious viral loads and reduced typical lesions in lung. Besides, neutralizing antibodies in vaccinated mice lasted for at least one year and were effective against various SARS-CoV-2 variants of concern, including B.1.1.7 (Alpha), B.1.351 (Beta), P.1 (Gamma), B.1.617.2 (Delta), and B.1.1.529 (Omicron). Furthermore, particle size, polydispersity index, and zeta-potential of ASD254 remained stable after 8-month storage at 4°C. Thus, ASD254 is a promising nanoparticle vaccine with good immunogenicity and stability to be developed as an effective vaccine option in controlling upcoming waves of COVID-19.
Collapse
Affiliation(s)
- I-Jung Lee
- Graduate Institute of Microbiology, College of Medicine, National Taiwan University, Taipei, Taiwan,Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Yu-Hua Lan
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Ping-Yi Wu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Yan-Wei Wu
- School of Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yu-Hung Chen
- School of Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Sheng-Che Tseng
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Tzu-Jiun Kuo
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Cheng-Pu Sun
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Jia-Tsrong Jan
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Hsiu-Hua Ma
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Chun-Che Liao
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Jian-Jong Liang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Hui-Ying Ko
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Chih-Shin Chang
- Biomedical Translation Research Center, Academia Sinica, Taipei, Taiwan
| | - Wen-Chun Liu
- Biomedical Translation Research Center, Academia Sinica, Taipei, Taiwan
| | - Yi-An Ko
- Biomedical Translation Research Center, Academia Sinica, Taipei, Taiwan
| | - Yen-Hui Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Zong-Lin Sie
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Szu-I Tsung
- Graduate Institute of Microbiology, College of Medicine, National Taiwan University, Taipei, Taiwan,Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Yi-Ling Lin
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan,Biomedical Translation Research Center, Academia Sinica, Taipei, Taiwan
| | - I-Hsuan Wang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Mi-Hua Tao
- Graduate Institute of Microbiology, College of Medicine, National Taiwan University, Taipei, Taiwan,Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan,Biomedical Translation Research Center, Academia Sinica, Taipei, Taiwan, Mi-Hua Tao Graduate Institute of Microbiology, College of Medicine, National Taiwan University, Taipei 100, Taiwan; Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan; Biomedical Translation Research Center, Academia Sinica, Taipei115, Taiwan
| |
Collapse
|
11
|
Shorayeva K, Nakhanov A, Nurpeisova A, Chervyakova O, Jekebekov K, Abay Z, Assanzhanova N, Sadikaliyeva S, Kalimolda E, Terebay A, Moldagulova S, Absatova Z, Tulendibayev A, Kopeyev S, Nakhanova G, Issabek A, Nurabayev S, Kerimbayev A, Kutumbetov L, Abduraimov Y, Kassenov M, Orynbayev M, Zakarya K. Pre-Clinical Safety and Immunogenicity Study of a Coronavirus Protein-Based Subunit Vaccine for COVID-19. Vaccines (Basel) 2023; 11:1771. [PMID: 38140175 PMCID: PMC10748237 DOI: 10.3390/vaccines11121771] [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: 09/21/2023] [Revised: 11/08/2023] [Accepted: 11/21/2023] [Indexed: 12/24/2023] Open
Abstract
Creating an effective and safe vaccine is critical to fighting the coronavirus infection successfully. Several types of COVID-19 vaccines exist, including inactivated, live attenuated, recombinant, synthetic peptide, virus-like particle-based, DNA and mRNA-based, and sub-unit vaccines containing purified immunogenic viral proteins. However, the scale and speed at which COVID-19 is spreading demonstrate a global public demand for an effective prophylaxis that must be supplied more. The developed products promise a bright future for SARS-CoV-2 prevention; however, evidence of safety and immunogenicity is mandatory before any vaccine can be produced. In this paper, we report on the results of our work examining the safety, toxicity, immunizing dose choice, and immunogenicity of QazCoVac-P, a Kazakhstan-made sub-unit vaccine for COVID-19. First, we looked into the product's safety profile by assessing its pyrogenicity in vaccinated rabbit models and using the LAL (limulus amebocyte lysate) test. We examined the vaccine's acute and sub-chronic toxicity on BALB/c mice and rats. The vaccine did not cause clinically significant toxicity-related changes or symptoms in our toxicity experiments. Finally, we performed a double immunization of mice, ferrets, Syrian hamsters, and rhesus macaques (Macaca mulatta). We used ELISA to measure antibody titers with the maximum mean geometric titer of antibodies in the animals' blood sera totaling approximately 8 log2. The results of this and other studies warrant recommending the QazCoVac-P vaccine for clinical trials.
Collapse
Affiliation(s)
| | | | - Ainur Nurpeisova
- Research Institute for Biological Safety Problems, The Ministry of Health of the Republic of Kazakhstan, Gvardeiskiy 080409, Kazakhstan (Z.A.); (E.K.); (Z.A.)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Vu MN, Pilkington EH, Lee WS, Tan H, Davis TP, Truong NP, Kent SJ, Wheatley AK. Engineered Ferritin Nanoparticle Vaccines Enable Rapid Screening of Antibody Functionalization to Boost Immune Responses. Adv Healthc Mater 2023; 12:e2202595. [PMID: 36786027 PMCID: PMC11469303 DOI: 10.1002/adhm.202202595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 01/16/2023] [Indexed: 02/15/2023]
Abstract
Employing monoclonal antibodies to target vaccine antigens to different immune cells within lymph nodes where adaptive immunity is initiated can provide a mechanism to fine-tune the magnitude or the quality of immune responses. However, studying the effects of different targeting antibodies head-to-head is challenging due to the lack of a feasible method that allows rapid screening of multiple antibodies for their impact on immunogenicity. Here self-assembling ferritin nanoparticles are prepared that co-display vaccine antigens and the Fc-binding domain of Staphylococcal protein A, allowing rapid attachment of soluble antibodies to the nanoparticle surface. Using this tunable system, ten antibodies targeting different immune cell subsets are screened, with targeting to Clec9a associated with higher serum antibody titers after immunization. Immune cell targeting using ferritin nanoparticles with anti-Clec9a antibodies drives concentrated deposition of antigens within germinal centers, boosting germinal center formation and robust antibody responses. However, the capacity to augment humoral immunity is antigen-dependent, with significant boosting observed for prototypic ovalbumin immunogens but reduced effectiveness with the SARS-CoV-2 RBD. This work provides a rapid platform for screening targeting antibodies, which will accelerate mechanistic insights into optimal delivery strategies for nanoparticle-based vaccines to maximize protective immunity.
Collapse
Affiliation(s)
- Mai N. Vu
- Peter Doherty Institute for Infection and ImmunityDepartment of Microbiology and ImmunologyThe University of MelbourneMelbourneVIC3010Australia
- Australian Research Council Centre of Excellence in Convergent Bio‐Nano Science and TechnologyParkvilleVIC3052Australia
- Monash Institute of Pharmaceutical SciencesMonash UniversityParkvilleVIC3052Australia
- Faculty of Pharmaceutics and Pharmaceutical TechnologyHanoi University of Pharmacy10000HanoiVietnam
| | - Emily H. Pilkington
- Peter Doherty Institute for Infection and ImmunityDepartment of Microbiology and ImmunologyThe University of MelbourneMelbourneVIC3010Australia
- Australian Research Council Centre of Excellence in Convergent Bio‐Nano Science and TechnologyParkvilleVIC3052Australia
- Monash Institute of Pharmaceutical SciencesMonash UniversityParkvilleVIC3052Australia
| | - Wen Shi Lee
- Peter Doherty Institute for Infection and ImmunityDepartment of Microbiology and ImmunologyThe University of MelbourneMelbourneVIC3010Australia
| | - Hyon‐Xhi Tan
- Peter Doherty Institute for Infection and ImmunityDepartment of Microbiology and ImmunologyThe University of MelbourneMelbourneVIC3010Australia
- Australian Research Council Centre of Excellence in Convergent Bio‐Nano Science and TechnologyParkvilleVIC3052Australia
| | - Thomas P. Davis
- Australian Research Council Centre of Excellence in Convergent Bio‐Nano Science and TechnologyParkvilleVIC3052Australia
- Monash Institute of Pharmaceutical SciencesMonash UniversityParkvilleVIC3052Australia
- Australia Institute of Bioengineering & NanotechnologyUniversity of QueenslandBrisbaneQLD4072Australia
| | - Nghia P. Truong
- Australian Research Council Centre of Excellence in Convergent Bio‐Nano Science and TechnologyParkvilleVIC3052Australia
- Monash Institute of Pharmaceutical SciencesMonash UniversityParkvilleVIC3052Australia
| | - Stephen J. Kent
- Peter Doherty Institute for Infection and ImmunityDepartment of Microbiology and ImmunologyThe University of MelbourneMelbourneVIC3010Australia
- Australian Research Council Centre of Excellence in Convergent Bio‐Nano Science and TechnologyParkvilleVIC3052Australia
- Melbourne Sexual Health Centre and Department of Infectious DiseasesAlfred Hospital and Central Clinical SchoolMonash UniversityMelbourneVIC3004Australia
| | - Adam K. Wheatley
- Peter Doherty Institute for Infection and ImmunityDepartment of Microbiology and ImmunologyThe University of MelbourneMelbourneVIC3010Australia
- Australian Research Council Centre of Excellence in Convergent Bio‐Nano Science and TechnologyParkvilleVIC3052Australia
| |
Collapse
|
13
|
Wang Z, Li Z, Shi W, Zhu D, Hu S, Dinh PUC, Cheng K. A SARS-CoV-2 and influenza double hit vaccine based on RBD-conjugated inactivated influenza A virus. SCIENCE ADVANCES 2023; 9:eabo4100. [PMID: 37352360 PMCID: PMC10289656 DOI: 10.1126/sciadv.abo4100] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 05/19/2023] [Indexed: 06/25/2023]
Abstract
The circulating flu viruses merging with the ongoing COVID-19 pandemic raises a more severe threat that promotes the infectivity of SARS-CoV-2 associated with higher mortality rates. Here, we conjugated recombinant receptor binding domain (RBD) of SARS-CoV-2 spike protein onto inactivated influenza A virus (Flu) to develop a SARS-CoV-2 virus-like particle (VLP) vaccine with two-hit protection. This double-hit vaccine (Flu-RBD) not only induced protective immunities against SARS-CoV-2 but also remained functional as a flu vaccine. The Flu core improved the retention and distribution of Flu-RBD vaccine in the draining lymph nodes, with enhanced immunogenicity. In a hamster model of live SARS-CoV-2 infection, two doses of Flu-RBD efficiently protected animals against viral infection. Furthermore, Flu-RBD VLP elicited a strong neutralization activity against both SARS-CoV-2 Delta pseudovirus and wild-type influenza A H1N1 inactivated virus in mice. Overall, the Flu-RBD VLP vaccine is a promising candidate for combating COVID-19, influenza A, and coinfection.
Collapse
Affiliation(s)
- Zhenzhen Wang
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC 27607, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, and North Carolina State University, Raleigh, NC 27606, USA
| | - Zhenhua Li
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC 27607, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, and North Carolina State University, Raleigh, NC 27606, USA
| | - Weiwei Shi
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC 27607, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, and North Carolina State University, Raleigh, NC 27606, USA
| | - Dashuai Zhu
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC 27607, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, and North Carolina State University, Raleigh, NC 27606, USA
| | - Shiqi Hu
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC 27607, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, and North Carolina State University, Raleigh, NC 27606, USA
| | - Phuong-Uyen C. Dinh
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC 27607, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, and North Carolina State University, Raleigh, NC 27606, USA
| | - Ke Cheng
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC 27607, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, and North Carolina State University, Raleigh, NC 27606, USA
- Division of Pharmacoengineering and Molecular Pharmaceutics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
14
|
Porello I, Cellesi F. Intracellular delivery of therapeutic proteins. New advancements and future directions. Front Bioeng Biotechnol 2023; 11:1211798. [PMID: 37304137 PMCID: PMC10247999 DOI: 10.3389/fbioe.2023.1211798] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 05/16/2023] [Indexed: 06/13/2023] Open
Abstract
Achieving the full potential of therapeutic proteins to access and target intracellular receptors will have enormous benefits in advancing human health and fighting disease. Existing strategies for intracellular protein delivery, such as chemical modification and nanocarrier-based protein delivery approaches, have shown promise but with limited efficiency and safety concerns. The development of more effective and versatile delivery tools is crucial for the safe and effective use of protein drugs. Nanosystems that can trigger endocytosis and endosomal disruption, or directly deliver proteins into the cytosol, are essential for successful therapeutic effects. This article aims to provide a brief overview of the current methods for intracellular protein delivery to mammalian cells, highlighting current challenges, new developments, and future research opportunities.
Collapse
|
15
|
Deliyannis G, Gherardin NA, Wong CY, Grimley SL, Cooney JP, Redmond SJ, Ellenberg P, Davidson KC, Mordant FL, Smith T, Gillard M, Lopez E, McAuley J, Tan CW, Wang JJ, Zeng W, Littlejohn M, Zhou R, Fuk-Woo Chan J, Chen ZW, Hartwig AE, Bowen R, Mackenzie JM, Vincan E, Torresi J, Kedzierska K, Pouton CW, Gordon TP, Wang LF, Kent SJ, Wheatley AK, Lewin SR, Subbarao K, Chung AW, Pellegrini M, Munro T, Nolan T, Rockman S, Jackson DC, Purcell DFJ, Godfrey DI. Broad immunity to SARS-CoV-2 variants of concern mediated by a SARS-CoV-2 receptor-binding domain protein vaccine. EBioMedicine 2023; 92:104574. [PMID: 37148585 PMCID: PMC10159263 DOI: 10.1016/j.ebiom.2023.104574] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 03/02/2023] [Accepted: 04/01/2023] [Indexed: 05/08/2023] Open
Abstract
BACKGROUND The SARS-CoV-2 global pandemic has fuelled the generation of vaccines at an unprecedented pace and scale. However, many challenges remain, including: the emergence of vaccine-resistant mutant viruses, vaccine stability during storage and transport, waning vaccine-induced immunity, and concerns about infrequent adverse events associated with existing vaccines. METHODS We report on a protein subunit vaccine comprising the receptor-binding domain (RBD) of the ancestral SARS-CoV-2 spike protein, dimerised with an immunoglobulin IgG1 Fc domain. These were tested in conjunction with three different adjuvants: a TLR2 agonist R4-Pam2Cys, an NKT cell agonist glycolipid α-Galactosylceramide, or MF59® squalene oil-in-water adjuvant, using mice, rats and hamsters. We also developed an RBD-human IgG1 Fc vaccine with an RBD sequence of the immuno-evasive beta variant (N501Y, E484K, K417N). These vaccines were also tested as a heterologous third dose booster in mice, following priming with whole spike vaccine. FINDINGS Each formulation of the RBD-Fc vaccines drove strong neutralising antibody (nAb) responses and provided durable and highly protective immunity against lower and upper airway infection in mouse models of COVID-19. The 'beta variant' RBD vaccine, combined with MF59® adjuvant, induced strong protection in mice against the beta strain as well as the ancestral strain. Furthermore, when used as a heterologous third dose booster, the RBD-Fc vaccines combined with MF59® increased titres of nAb against other variants including alpha, delta, delta+, gamma, lambda, mu, and omicron BA.1, BA.2 and BA.5. INTERPRETATION These results demonstrated that an RBD-Fc protein subunit/MF59® adjuvanted vaccine can induce high levels of broadly reactive nAbs, including when used as a booster following prior immunisation of mice with whole ancestral-strain spike vaccines. This vaccine platform offers a potential approach to augment some of the currently approved vaccines in the face of emerging variants of concern, and it has now entered a phase I clinical trial. FUNDING This work was supported by grants from the Medical Research Future Fund (MRFF) (2005846), The Jack Ma Foundation, National Health and Medical Research Council of Australia (NHMRC; 1113293) and Singapore National Medical Research Council (MOH-COVID19RF-003). Individual researchers were supported by an NHMRC Senior Principal Research Fellowship (1117766), NHMRC Investigator Awards (2008913 and 1173871), Australian Research Council Discovery Early Career Research Award (ARC DECRA; DE210100705) and philanthropic awards from IFM investors and the A2 Milk Company.
Collapse
Affiliation(s)
- Georgia Deliyannis
- Department of Microbiology & Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Nicholas A Gherardin
- Department of Microbiology & Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Chinn Yi Wong
- Department of Microbiology & Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Samantha L Grimley
- Department of Microbiology & Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - James P Cooney
- Walter and Eliza Hall Institute, Infectious Diseases & Immune Defence Division, Parkville, Victoria 3052, Australia
| | - Samuel J Redmond
- Department of Microbiology & Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Paula Ellenberg
- Department of Microbiology & Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Kathryn C Davidson
- Walter and Eliza Hall Institute, Infectious Diseases & Immune Defence Division, Parkville, Victoria 3052, Australia
| | - Francesca L Mordant
- Department of Microbiology & Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Tim Smith
- Department of Microbiology & Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Marianne Gillard
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, Queensland, Australia
| | - Ester Lopez
- Department of Microbiology & Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Julie McAuley
- Department of Microbiology & Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Chee Wah Tan
- Duke NUS Medical School, Programme for Emerging Infectious Diseases, Singapore
| | - Jing J Wang
- Department of Immunology, Flinders University and SA Pathology, Flinders Medical Centre, Bedford Park, Adelaide, South Australia 5042, Australia
| | - Weiguang Zeng
- Department of Microbiology & Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Mason Littlejohn
- Doherty Directorate, The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Runhong Zhou
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Jasper Fuk-Woo Chan
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China; Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong Special Administrative Region, China
| | - Zhi-Wei Chen
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China; Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong Special Administrative Region, China
| | - Airn E Hartwig
- Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | - Richard Bowen
- Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | - Jason M Mackenzie
- Department of Microbiology & Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Elizabeth Vincan
- Victorian Infectious Diseases Reference Laboratory (VIDRL) at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia; Department of Infectious Diseases, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Joseph Torresi
- Department of Microbiology & Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Katherine Kedzierska
- Department of Microbiology & Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Colin W Pouton
- Monash Institute of Pharmaceutical Sciences, Parkville, Victoria 3052, Australia
| | - Tom P Gordon
- Department of Immunology, Flinders University and SA Pathology, Flinders Medical Centre, Bedford Park, Adelaide, South Australia 5042, Australia
| | - Lin-Fa Wang
- Duke NUS Medical School, Programme for Emerging Infectious Diseases, Singapore
| | - Stephen J Kent
- Department of Microbiology & Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Adam K Wheatley
- Department of Microbiology & Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Sharon R Lewin
- Department of Infectious Diseases, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia; Victorian Infectious Diseases Service, Royal Melbourne Hospital at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia; Department of Infectious Diseases, The Alfred Hospital and Monash University, Melbourne, 3010 Australia
| | - Kanta Subbarao
- Department of Microbiology & Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia; WHO Collaborating Centre for Reference and Research on Influenza at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Amy W Chung
- Department of Microbiology & Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Marc Pellegrini
- Walter and Eliza Hall Institute, Infectious Diseases & Immune Defence Division, Parkville, Victoria 3052, Australia
| | - Trent Munro
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, Queensland, Australia
| | - Terry Nolan
- Department of Infectious Diseases, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia; Vaccine and Immunisation Research Group (VIRGo), Department of Infectious Disease, Peter Doherty Institute for Infection and Immunity, University of Melbourne, and Murdoch Children's Research Institute, Victoria 3010, Australia
| | - Steven Rockman
- Department of Microbiology & Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia; Seqirus, Vaccine Innovation Unit, Parkville, Victoria, 3052, Australia
| | - David C Jackson
- Department of Microbiology & Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Damian F J Purcell
- Department of Microbiology & Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Dale I Godfrey
- Department of Microbiology & Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia.
| |
Collapse
|
16
|
Phoolcharoen W, Shanmugaraj B, Khorattanakulchai N, Sunyakumthorn P, Pichyangkul S, Taepavarapruk P, Praserthsee W, Malaivijitnond S, Manopwisedjaroen S, Thitithanyanont A, Srisutthisamphan K, Jongkaewwattana A, Tomai M, Fox CB, Taychakhoonavudh S. Preclinical evaluation of immunogenicity, efficacy and safety of a recombinant plant-based SARS-CoV-2 RBD vaccine formulated with 3M-052-Alum adjuvant. Vaccine 2023; 41:2781-2792. [PMID: 36963999 PMCID: PMC10027959 DOI: 10.1016/j.vaccine.2023.03.027] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Revised: 03/06/2023] [Accepted: 03/14/2023] [Indexed: 03/24/2023]
Abstract
Cost-effective, and accessible vaccines are needed for mass immunization to control the ongoing coronavirus disease 2019 (COVID-19), especially in low- and middle-income countries (LMIC).A plant-based vaccine is an attractive technology platform since the recombinant proteins can be easily produced at large scale and low cost. For the recombinant subunit-based vaccines, effective adjuvants are crucial to enhance the magnitude and breadth of immune responses elicited by the vaccine. In this study, we report a preclinical evaluation of the immunogenicity, efficacy and safety of a recombinant plant-based SARS-CoV-2 RBD vaccine formulated with 3M-052 (TLR7/8 agonist)-Alum adjuvant. This vaccine formulation, named Baiya SARS-CoV-2 Vax 2, induced significant levels of RBD-specific IgG and neutralizing antibody responses in mice. A viral challenge study using humanized K18-hACE2 mice has shown that animals vaccinated with two doses of Baiya SARS-CoV-2 Vax 2 established immune protection against SARS-CoV-2. A study in nonhuman primates (cynomolgus monkeys) indicated that immunization with two doses of Baiya SARS-CoV-2 Vax 2 was safe, well tolerated, and induced neutralizing antibodies against the prototype virus and other viral variants (Alpha, Beta, Gamma, Delta, and Omicron subvariants). The toxicity of Baiya SARS-CoV-2 Vax 2 was further investigated in Jcl:SD rats, which demonstrated that a single dose and repeated doses of Baiya SARS-CoV-2 Vax 2 were well tolerated and no mortality or unanticipated findings were observed. Overall, these preclinical findings support further clinical development of Baiya SARS-CoV-2 Vax 2.
Collapse
Affiliation(s)
- Waranyoo Phoolcharoen
- Center of Excellence in Plant-produced Pharmaceuticals, Chulalongkorn University, Bangkok 10330, Thailand; Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand.
| | | | - Narach Khorattanakulchai
- Center of Excellence in Plant-produced Pharmaceuticals, Chulalongkorn University, Bangkok 10330, Thailand; Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | | | - Sathit Pichyangkul
- US Armed Forces Research Institute of Medical Sciences, Bangkok 10400, Thailand
| | - Pornnarin Taepavarapruk
- Center for Animal Research and Department of Physiology, Faculty of Medical Science, Naresuan University, Pitsanulok 65000, Thailand
| | | | - Suchinda Malaivijitnond
- National Primate Research Center of Thailand-Chulalongkorn University, Saraburi 18110, Thailand
| | | | - Arunee Thitithanyanont
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | - Kanjana Srisutthisamphan
- Virology and Cell Technology Laboratory, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency, Pathumthani, Thailand
| | - Anan Jongkaewwattana
- Virology and Cell Technology Laboratory, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency, Pathumthani, Thailand
| | - Mark Tomai
- 3M Healthcare, 3M Center, Bldg 270-4N-04, St. Paul, MN 55144-1000, USA
| | - Christopher B Fox
- Access to Advanced Health Institute (AAHI), 1616 Eastlake Ave E, Ste 400, Seattle, WA 98102, USA
| | - Suthira Taychakhoonavudh
- Department of Social and Administrative Pharmacy, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| |
Collapse
|
17
|
Kwak HW, Park HJ, Jung SY, Oh EY, Park SI, Kim Y, Park HJ, Park S, Kim YJ, Ko HL, Lee JA, Won H, Hwang YH, Kim SY, Kim SE, Bae SE, Yoon M, Kim JO, Song M, Lee SJ, Seo KW, Lee K, Kim D, Kim H, Lee SM, Hong SH, Nam JH. Recombinant measles virus encoding the spike protein of SARS-CoV-2 efficiently induces Th1 responses and neutralizing antibodies that block SARS-CoV-2 variants. Vaccine 2023; 41:1892-1901. [PMID: 36792434 PMCID: PMC9902292 DOI: 10.1016/j.vaccine.2023.02.005] [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: 06/23/2022] [Revised: 02/03/2023] [Accepted: 02/04/2023] [Indexed: 02/10/2023]
Abstract
Owing to the rapid spread of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and its variants, the development of effective and safe vaccines has become a priority. The measles virus (MeV) vaccine is an attractive vaccine platform as it has been administered to children for more than 40 years in over 100 countries. In this study, we developed a recombinant MeV expressing the full-length SARS-CoV-2 spike protein (rMeV-S) and tested its efficacy using mouse and hamster models. In hCD46Tg mice, two-dose rMeV-S vaccination induced higher Th1 secretion and humoral responses than one-dose vaccination. Interestingly, neutralizing antibodies induced by one-dose and two-dose rMeV-S immunization effectively blocked the entry of the α, β, γ, and δ variants of SARS-CoV-2. Furthermore, two-dose rMeV-S immunization provided complete protection against SARS-CoV-2 in the hamster model. These results suggest the potential of rMeV-S as a vaccine candidate for targeting SARS-CoV-2 and its variants.
Collapse
Affiliation(s)
- Hye Won Kwak
- Department of Medical and Biological Sciences, The Catholic University of Korea, Gyeonggi-do, Bucheon, Republic of Korea; BK Plus Department of Biotechnology, The Catholic University of Korea, Gyeonggi-do, Bucheon, Republic of Korea; SML biopharm, Gyeonggi-do, Bucheon, Republic of Korea
| | - Hyo-Jung Park
- Department of Medical and Biological Sciences, The Catholic University of Korea, Gyeonggi-do, Bucheon, Republic of Korea; BK Plus Department of Biotechnology, The Catholic University of Korea, Gyeonggi-do, Bucheon, Republic of Korea
| | - Seo-Yeon Jung
- Department of R&D, SK bioscience, Pangyoro, Bundang-gu, Republic of Korea
| | - Eun Young Oh
- College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Republic of Korea
| | - Sang-In Park
- Division of Research Program, Scripps Korea Antibody Institute, Chuncheon, Kangwon-do, Republic of Korea
| | - Yeonhwa Kim
- College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Republic of Korea
| | - Hyeong-Jun Park
- Department of Medical and Biological Sciences, The Catholic University of Korea, Gyeonggi-do, Bucheon, Republic of Korea; BK Plus Department of Biotechnology, The Catholic University of Korea, Gyeonggi-do, Bucheon, Republic of Korea; SML biopharm, Gyeonggi-do, Bucheon, Republic of Korea
| | - Sohyun Park
- College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Republic of Korea
| | - You-Jin Kim
- Division of Infectious Disease Vaccine Research, Center for Vaccine Research, National Institute of Infectious Diseases, National Institute of Health, Republic of Korea
| | - Hae Li Ko
- Division of Research Program, Scripps Korea Antibody Institute, Chuncheon, Kangwon-do, Republic of Korea
| | - Jung-Ah Lee
- Division of Infectious Disease Vaccine Research, Center for Vaccine Research, National Institute of Infectious Diseases, National Institute of Health, Republic of Korea
| | - Hyeran Won
- Division of Infectious Disease Vaccine Research, Center for Vaccine Research, National Institute of Infectious Diseases, National Institute of Health, Republic of Korea
| | - Yun-Ho Hwang
- Division of Infectious Disease Vaccine Research, Center for Vaccine Research, National Institute of Infectious Diseases, National Institute of Health, Republic of Korea
| | - Seo Yeon Kim
- Division of Infectious Disease Vaccine Research, Center for Vaccine Research, National Institute of Infectious Diseases, National Institute of Health, Republic of Korea
| | - Se Eun Kim
- Division of Infectious Disease Vaccine Research, Center for Vaccine Research, National Institute of Infectious Diseases, National Institute of Health, Republic of Korea
| | - Seoung Eun Bae
- Science Unit, International Vaccine Institute, Seoul 08826, Republic of Korea
| | - Minhyuk Yoon
- Science Unit, International Vaccine Institute, Seoul 08826, Republic of Korea
| | - Jae-Ouk Kim
- Science Unit, International Vaccine Institute, Seoul 08826, Republic of Korea
| | - Manki Song
- Science Unit, International Vaccine Institute, Seoul 08826, Republic of Korea
| | - Su Jeen Lee
- Department of R&D, SK bioscience, Pangyoro, Bundang-gu, Republic of Korea
| | - Ki-Weon Seo
- Department of R&D, SK bioscience, Pangyoro, Bundang-gu, Republic of Korea
| | - Kunse Lee
- Department of R&D, SK bioscience, Pangyoro, Bundang-gu, Republic of Korea
| | - Dokeun Kim
- Division of Infectious Disease Vaccine Research, Center for Vaccine Research, National Institute of Infectious Diseases, National Institute of Health, Republic of Korea
| | - Hun Kim
- Department of R&D, SK bioscience, Pangyoro, Bundang-gu, Republic of Korea
| | - Sang-Myeong Lee
- College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Republic of Korea.
| | - So-Hee Hong
- Department of Microbiology, College of Medicine, Ewha Womans University, Seoul 07804, Republic of Korea.
| | - Jae-Hwan Nam
- Department of Medical and Biological Sciences, The Catholic University of Korea, Gyeonggi-do, Bucheon, Republic of Korea; BK Plus Department of Biotechnology, The Catholic University of Korea, Gyeonggi-do, Bucheon, Republic of Korea.
| |
Collapse
|
18
|
Ahuja R, Srichandan S, Meena J, Biswal BK, Panda AK. Immunogenicity Evaluation of Thermostable Microparticles Entrapping Receptor Binding Domain of SARS-CoV-2 by Single Point Administration. J Pharm Sci 2023; 112:1664-1670. [PMID: 36736778 PMCID: PMC9891792 DOI: 10.1016/j.xphs.2023.01.024] [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: 07/12/2022] [Revised: 01/25/2023] [Accepted: 01/25/2023] [Indexed: 02/04/2023]
Abstract
Receptor binding domain (RBD) of SARS-CoV-2 is a prime vaccine target against which neutralizing antibody responses are directed. Purified RBD as a vaccine candidate warrants administration of multiple doses along with adjuvants and use of delivery systems to improve its immunogenicity. The present investigation examines the immunogenicity of RBD delivered by biodegradable polymer particles from single dose administration. Mice upon single point immunization of RBD entrapped microparticles generated improved antibody response. The polymer microparticles showed better temperature stability and could be stored at 37 degrees for one month without any considerable loss of immunogenicity. Further, immunization with microparticles could elicit memory antibody response upon challenge after four months of single dose administration. Thus, using microparticles entrapping RBD as a vaccine candidate confer improved immunogenicity, temperature stability and recall response. These thermostable microparticles seem to be a potentially cost-effective approach which can help in dose reduction, provide a wider access of vaccines and accelerate the end of global pandemic.
Collapse
Affiliation(s)
- Rahul Ahuja
- Product Development Cell, National Institute of Immunology, New Delhi, 110067, India; Infection and Immunology Laboratory, Translational Health Science & Technology Institute, Faridabad, Haryana, 120001, India.
| | - Sudeepa Srichandan
- Product Development Cell, National Institute of Immunology, New Delhi, 110067, India
| | - Jairam Meena
- Product Development Cell, National Institute of Immunology, New Delhi, 110067, India; Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Uttar Pradesh, 221005, India
| | - Bichitra Kumar Biswal
- Structural and Functional Laboratory, National Institute of Immunology, New Delhi, 110067, India
| | - Amulya K Panda
- Product Development Cell, National Institute of Immunology, New Delhi, 110067, India.
| |
Collapse
|
19
|
Rodriguez-Aponte SA, Dalvie NC, Wong TY, Johnston RS, Naranjo CA, Bajoria S, Kumru OS, Kaur K, Russ BP, Lee KS, Cyphert HA, Barbier M, Rao HD, Rajurkar MP, Lothe RR, Shaligram US, Batwal S, Chandrasekaran R, Nagar G, Kleanthous H, Biswas S, Bevere JR, Joshi SB, Volkin DB, Damron FH, Love JC. Molecular engineering of a cryptic epitope in Spike RBD improves manufacturability and neutralizing breadth against SARS-CoV-2 variants. Vaccine 2023; 41:1108-1118. [PMID: 36610932 PMCID: PMC9797419 DOI: 10.1016/j.vaccine.2022.12.062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 12/22/2022] [Accepted: 12/25/2022] [Indexed: 12/30/2022]
Abstract
There is a continued need for sarbecovirus vaccines that can be manufactured and distributed in low- and middle-income countries (LMICs). Subunit protein vaccines are manufactured at large scales at low costs, have less stringent temperature requirements for distribution in LMICs, and several candidates have shown protection against SARS-CoV-2. We previously reported an engineered variant of the SARS-CoV-2 Spike protein receptor binding domain antigen (RBD-L452K-F490W; RBD-J) with enhanced manufacturability and immunogenicity compared to the ancestral RBD. Here, we report a second-generation engineered RBD antigen (RBD-J6) with two additional mutations to a hydrophobic cryptic epitope in the RBD core, S383D and L518D, that further improved expression titers and biophysical stability. RBD-J6 retained binding affinity to human convalescent sera and to all tested neutralizing antibodies except antibodies that target the class IV epitope on the RBD core. K18-hACE2 transgenic mice immunized with three doses of a Beta variant of RBD-J6 displayed on a virus-like particle (VLP) generated neutralizing antibodies (nAb) to nine SARS-CoV-2 variants of concern at similar levels as two doses of Comirnaty. The vaccinated mice were also protected from challenge with Alpha or Beta SARS-CoV-2. This engineered antigen could be useful for modular RBD-based subunit vaccines to enhance manufacturability and global access, or for further development of variant-specific or broadly acting booster vaccines.
Collapse
Affiliation(s)
- Sergio A Rodriguez-Aponte
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Neil C Dalvie
- The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Ting Y Wong
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV 26506, USA; Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, WV 26506, USA
| | - Ryan S Johnston
- The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Christopher A Naranjo
- The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Sakshi Bajoria
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS 66047, USA
| | - Ozan S Kumru
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS 66047, USA
| | - Kawaljit Kaur
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS 66047, USA
| | - Brynnan P Russ
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV 26506, USA; Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, WV 26506, USA
| | - Katherine S Lee
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV 26506, USA; Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, WV 26506, USA
| | - Holly A Cyphert
- Department of Biological Sciences, Marshall University, Huntington, WV 26506, USA
| | - Mariette Barbier
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV 26506, USA; Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, WV 26506, USA
| | - Harish D Rao
- Serum Institute of India Pvt. Ltd., Pune 411028, India
| | | | | | | | | | | | - Gaurav Nagar
- Serum Institute of India Pvt. Ltd., Pune 411028, India
| | | | - Sumi Biswas
- SpyBiotech Limited, Oxford Business Park North, Oxford OX4 2JZ, UK
| | - Justin R Bevere
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV 26506, USA; Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, WV 26506, USA
| | - Sangeeta B Joshi
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS 66047, USA
| | - David B Volkin
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS 66047, USA
| | - F Heath Damron
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV 26506, USA; Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, WV 26506, USA
| | - J Christopher Love
- The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA.
| |
Collapse
|
20
|
Guan X, Yang Y, Du L. Advances in SARS-CoV-2 receptor-binding domain-based COVID-19 vaccines. Expert Rev Vaccines 2023; 22:422-439. [PMID: 37161869 PMCID: PMC10355161 DOI: 10.1080/14760584.2023.2211153] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 05/03/2023] [Indexed: 05/11/2023]
Abstract
INTRODUCTION The Coronavirus Disease 2019 (COVID-19) pandemic has caused devastating human and economic costs. Vaccination is an important step in controlling the pandemic. Severe acute respiratory coronavirus-2 (SARS-CoV-2), the causative agent of COVID-19, infects cells by binding a cellular receptor through the receptor-binding domain (RBD) within the S1 subunit of the spike (S) protein. Viral entry and membrane fusion are mediated by the S2 subunit. AREAS COVERED SARS-CoV-2 S protein, particularly RBD, serves as an important target for vaccines. Here we review the structure and function of SARS-CoV-2 S protein and its RBD, summarize current COVID-19 vaccines targeting the RBD, and outline potential strategies for improving RBD-based vaccines. Overall, this review provides important information that will facilitate rational design and development of safer and more effective COVID-19 vaccines. EXPERT OPINION The S protein of SARS-CoV-2 harbors numerous mutations, mostly in the RBD, resulting in multiple variant strains. Although many COVID-19 vaccines targeting the RBD of original virus strain (and previous variants) can prevent infection of these strains, their ability against recent dominant variants, particularly Omicron and its offspring, is significantly reduced. Collective efforts are needed to develop effective broad-spectrum vaccines to control current and future variants that have pandemic potential.
Collapse
Affiliation(s)
- Xiaoqing Guan
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Yang Yang
- Roy J. Carver Department of Biochemistry, Biophysics and Molecular Biology, Iowa State University, Ames, IA, USA
| | - Lanying Du
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| |
Collapse
|
21
|
Characterization of Systemic and Mucosal Humoral Immune Responses to an Adjuvanted Intranasal SARS-CoV-2 Protein Subunit Vaccine Candidate in Mice. Vaccines (Basel) 2022; 11:vaccines11010030. [PMID: 36679875 PMCID: PMC9865305 DOI: 10.3390/vaccines11010030] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 12/17/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022] Open
Abstract
Continuous viral evolution of SARS-CoV-2 has resulted in variants capable of immune evasion, vaccine breakthrough infections and increased transmissibility. New vaccines that invoke mucosal immunity may provide a solution to reducing virus transmission. Here, we evaluated the immunogenicity of intranasally administered subunit protein vaccines composed of a stabilized SARS-CoV-2 spike trimer or the receptor binding domain (RBD) adjuvanted with either cholera toxin (CT) or an archaeal lipid mucosal adjuvant (AMVAD). We show robust induction of immunoglobulin (Ig) G and IgA responses in plasma, nasal wash and bronchoalveolar lavage in mice only when adjuvant is used in the vaccine formulation. While the AMVAD adjuvant was more effective at inducing systemic antibodies against the RBD antigen than CT, CT was generally more effective at inducing overall higher IgA and IgG titers against the spike antigen in both systemic and mucosal compartments. Furthermore, vaccination with adjuvanted spike led to superior mucosal IgA responses than with the RBD antigen and produced broadly targeting neutralizing plasma antibodies against ancestral, Delta and Omicron variants in vitro; whereas adjuvanted RBD elicited a narrower antibody response with neutralizing activity only against ancestral and Delta variants. Our study demonstrates that intranasal administration of an adjuvanted protein subunit vaccine in immunologically naïve mice induced both systemic and mucosal neutralizing antibody responses that were most effective at neutralizing SARS-CoV-2 variants when the trimeric spike was used as an antigen compared to RBD.
Collapse
|
22
|
Pymm P, Redmond SJ, Dolezal O, Mordant F, Lopez E, Cooney JP, Davidson KC, Haycroft ER, Tan CW, Seneviratna R, Grimley SL, Purcell DF, Kent SJ, Wheatley AK, Wang LF, Leis A, Glukhova A, Pellegrini M, Chung AW, Subbarao K, Uldrich AP, Tham WH, Godfrey DI, Gherardin NA. Biparatopic nanobodies targeting the receptor binding domain efficiently neutralize SARS-CoV-2. iScience 2022; 25:105259. [PMID: 36213007 PMCID: PMC9529347 DOI: 10.1016/j.isci.2022.105259] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 08/15/2022] [Accepted: 09/28/2022] [Indexed: 11/17/2022] Open
Abstract
The development of therapeutics to prevent or treat COVID-19 remains an area of intense focus. Protein biologics, including monoclonal antibodies and nanobodies that neutralize virus, have potential for the treatment of active disease. Here, we have used yeast display of a synthetic nanobody library to isolate nanobodies that bind the receptor-binding domain (RBD) of SARS-CoV-2 and neutralize the virus. We show that combining two clones with distinct binding epitopes within the RBD into a single protein construct to generate biparatopic reagents dramatically enhances their neutralizing capacity. Furthermore, the biparatopic nanobodies exhibit enhanced control over clinically relevant RBD variants that escaped recognition by the individual nanobodies. Structural analysis of biparatopic binding to spike (S) protein revealed a unique binding mode whereby the two nanobody paratopes bridge RBDs encoded by distinct S trimers. Accordingly, biparatopic nanobodies offer a way to rapidly generate powerful viral neutralizers with enhanced ability to control viral escape mutants.
Collapse
Affiliation(s)
- Phillip Pymm
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia,Department of Medical Biology, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Samuel J. Redmond
- Department of Microbiology & Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, VIC 3000, Australia
| | - Olan Dolezal
- Commonwealth Scientific and Industrial Research Organisation (CSIRO) Biomedical Program, Clayton, VIC 3168, Australia
| | - Francesca Mordant
- Department of Microbiology & Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, VIC 3000, Australia
| | - Ester Lopez
- Department of Microbiology & Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, VIC 3000, Australia
| | - James P. Cooney
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia,Department of Medical Biology, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Kathryn C. Davidson
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia,Department of Medical Biology, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Ebene R. Haycroft
- Department of Microbiology & Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, VIC 3000, Australia
| | - Chee Wah Tan
- Programme in Emerging Infectious Diseases, Duke NUS Medical School, Singapore 169857, Singapore
| | - Rebecca Seneviratna
- Department of Microbiology & Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, VIC 3000, Australia
| | - Samantha L. Grimley
- Department of Microbiology & Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, VIC 3000, Australia
| | - Damian F.J. Purcell
- Department of Microbiology & Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, VIC 3000, Australia
| | - Stephen J. Kent
- Department of Microbiology & Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, VIC 3000, Australia,Australian Research Council Centre for Excellence in Convergent Bio-Nano Science and Technology, University of Melbourne, Melbourne VIC 3010, Australia
| | - Adam K. Wheatley
- Department of Microbiology & Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, VIC 3000, Australia,Programme in Emerging Infectious Diseases, Duke NUS Medical School, Singapore 169857, Singapore
| | - Lin-Fa Wang
- Programme in Emerging Infectious Diseases, Duke NUS Medical School, Singapore 169857, Singapore
| | - Andrew Leis
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Alisa Glukhova
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia,Department of Medical Biology, University of Melbourne, Melbourne, VIC 3010, Australia,Drug Discovery Biology, Monash Faculty of Pharmacy and Pharmaceutical Sciences, Monash University, Parkville 3052 VIC, Australia,WHO Collaborating Centre for Reference and Research on Influenza, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC 3000, Australia
| | - Marc Pellegrini
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia,Department of Medical Biology, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Amy W. Chung
- Department of Microbiology & Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, VIC 3000, Australia
| | - Kanta Subbarao
- Department of Microbiology & Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, VIC 3000, Australia
| | - Adam P. Uldrich
- Department of Microbiology & Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, VIC 3000, Australia
| | - Wai-Hong Tham
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia,Department of Medical Biology, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Dale I. Godfrey
- Department of Microbiology & Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, VIC 3000, Australia,Corresponding author
| | - Nicholas A. Gherardin
- Department of Microbiology & Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, VIC 3000, Australia,Corresponding author
| |
Collapse
|
23
|
Chi X, Gu J, Ma X. Characteristics and Roles of T Follicular Helper Cells in SARS-CoV-2 Vaccine Response. Vaccines (Basel) 2022; 10:vaccines10101623. [PMID: 36298488 PMCID: PMC9611968 DOI: 10.3390/vaccines10101623] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 09/26/2022] [Accepted: 09/26/2022] [Indexed: 11/17/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccination is critical to controlling the coronavirus disease 2019 (COVID-19) pandemic. However, a weak response to the vaccine and insufficient persistence of specific antibodies may threaten the global impact of mass vaccination campaigns. This study summarizes the internal factors of the body that affect the effectiveness of the SARS-CoV-2 vaccine. T follicular helper (Tfh) cells support germinal center B cells to produce vaccine-specific immunoglobulins. A reduction in the Tfh cell number and a shift in the subset phenotypes caused by multiple factors may impair the production and persistence of high-affinity antibodies. Besides efficacy differences caused by the different types of vaccines, the factors that affect vaccine effectiveness by intervening in the Tfh cell response also include age-related defects, the polarity of the body microenvironment, repeated immunization, immunodeficiency, and immunosuppressive treatments. Assessing the phenotypic distribution and activation levels of Tfh cell subsets after vaccination is helpful in predicting vaccine responses and may identify potential targets for improving vaccine effectiveness.
Collapse
Affiliation(s)
- Xuyang Chi
- Department of Pediatrics, The First Hospital of China Medical University, Shenyang 110001, China
| | - Jia Gu
- Department of Pediatrics, The First Hospital of China Medical University, Shenyang 110001, China
| | - Xiaoxue Ma
- Department of Pediatrics, The First Hospital of China Medical University, Shenyang 110001, China
- Department of Microbiology & Immunology and Pediatrics, Dalhousie University, and Canadian Center for Vaccinology, IWK Health Centre, Halifax, NS B3K 6R8, Canada
- Correspondence: ; Tel.: +86-024-83282527
| |
Collapse
|
24
|
Harhala MA, Gembara K, Nelson DC, Miernikiewicz P, Dąbrowska K. Immunogenicity of Endolysin PlyC. Antibiotics (Basel) 2022; 11:966. [PMID: 35884219 PMCID: PMC9312349 DOI: 10.3390/antibiotics11070966] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/14/2022] [Accepted: 07/16/2022] [Indexed: 12/10/2022] Open
Abstract
Endolysins are bacteriolytic enzymes derived from bacteriophages. They represent an alternative to antibiotics, since they are not susceptible to conventional antimicrobial resistance mechanisms. Since non-human proteins are efficient inducers of specific immune responses, including the IgG response or the development of an allergic response mediated by IgE, we evaluated the general immunogenicity of the highly active antibacterial enzyme, PlyC, in a human population and in a mouse model. The study includes the identification of molecular epitopes of PlyC. The overall assessment of potential hypersensitivity to this protein and PlyC-specific IgE testing was also conducted in mice. PlyC induced efficient IgG production in mice, and the molecular analysis revealed that PlyC-specific IgG interacted with four immunogenic regions identified within the PlyCA subunit. In humans, approximately 10% of the population demonstrated IgG reactivity to the PlyCB subunit only, which is attributed to cross-reactions since this was a naïve serum. Of note, in spite of being immunogenic, PlyC induced a normal immune response, without hypersensitivity, since both the animals challenged with PlyC and in the human population PlyC-specific IgE was not detected.
Collapse
Affiliation(s)
- Marek Adam Harhala
- Laboratory of Phage Molecular Biology, Department of Phage Therapy, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland; (M.A.H.); (K.G.); (P.M.)
- Research & Development Center, Regional Specialist Hospital, 53-114 Wroclaw, Poland
| | - Katarzyna Gembara
- Laboratory of Phage Molecular Biology, Department of Phage Therapy, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland; (M.A.H.); (K.G.); (P.M.)
- Research & Development Center, Regional Specialist Hospital, 53-114 Wroclaw, Poland
| | - Daniel C. Nelson
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, MD 20853, USA;
| | - Paulina Miernikiewicz
- Laboratory of Phage Molecular Biology, Department of Phage Therapy, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland; (M.A.H.); (K.G.); (P.M.)
| | - Krystyna Dąbrowska
- Laboratory of Phage Molecular Biology, Department of Phage Therapy, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland; (M.A.H.); (K.G.); (P.M.)
- Research & Development Center, Regional Specialist Hospital, 53-114 Wroclaw, Poland
| |
Collapse
|
25
|
Blanas A, Karsjens H, de Ligt A, Huijbers EJ, van Loon K, Denisov SS, Durukan C, Engbersen DJ, Groen J, Hennig S, Hackeng TM, van Beijnum JR, Griffioen AW. Vaccination with a bacterial peptide conjugated to SARS-CoV-2 RBD accelerates immunity and protects against COVID-19. iScience 2022; 25:104719. [PMID: 35813877 PMCID: PMC9252865 DOI: 10.1016/j.isci.2022.104719] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 05/31/2022] [Accepted: 06/29/2022] [Indexed: 11/16/2022] Open
Abstract
Poor immunogenicity of critical epitopes can hamper vaccine efficacy. To boost immune recognition of non- or low-immunogenic antigens, we developed a vaccine platform based on the conjugation of a target protein to a chimeric designer peptide (CDP) of bacterial origin. Here, we exploited this immune Boost (iBoost) technology to enhance the immune response against the receptor-binding domain (RBD) of the SARS-CoV-2 spike glycoprotein. Despite its fundamental role during viral infection, RBD is only moderately immunogenic. Immunization studies in mice showed that the conjugation of CDP to RBD induced superior immune responses compared to RBD alone. CDP-RBD elicited cross-reactive antibodies against the variants of concern Delta and Omicron. Furthermore, hamsters vaccinated with CDP-RBD developed potent neutralizing antibody responses and were fully protected from lung lesion formation upon challenge with SARS-CoV-2. In sum, we show that the iBoost conjugate vaccine technology provides a valuable tool for both quantitatively and qualitatively enhancing anti-viral immunity. An iBoost-based CDP-RBD conjugate vaccine against SARS-CoV-2 Induction of potent RBD-specific humoral and cellular responses CDP-RBD vaccination protects hamsters from lung lesion formation
Collapse
Affiliation(s)
- Athanasios Blanas
- Angiogenesis Laboratory, Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | - Haiko Karsjens
- Angiogenesis Laboratory, Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | - Aafke de Ligt
- Angiogenesis Laboratory, Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | - Elisabeth J.M. Huijbers
- Angiogenesis Laboratory, Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | - Karlijn van Loon
- Angiogenesis Laboratory, Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | - Stepan S. Denisov
- School for Cardiovascular Sciences, Department of Biochemistry, Maastricht University, Maastricht, the Netherlands
| | - Canan Durukan
- Department of Chemistry & Pharmaceutical Sciences, Amsterdam Institute of Molecules, Medicines and Systems, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | | | - Jan Groen
- Intravacc, Institute for Translational Vaccinology, Bilthoven, the Netherlands
| | - Sven Hennig
- Department of Chemistry & Pharmaceutical Sciences, Amsterdam Institute of Molecules, Medicines and Systems, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Tilman M. Hackeng
- School for Cardiovascular Sciences, Department of Biochemistry, Maastricht University, Maastricht, the Netherlands
| | | | - Arjan W. Griffioen
- Angiogenesis Laboratory, Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
- Corresponding author
| |
Collapse
|
26
|
Ávalos I, Lao T, Rodríguez EM, Zamora Y, Rodríguez A, Ramón A, Lemos G, Cabrales A, Bequet-Romero M, Casillas D, Andújar I, Espinosa LA, González LJ, Alvarez Y, Carpio Y, Estrada MP. Chimeric Antigen by the Fusion of SARS-CoV-2 Receptor Binding Domain with the Extracellular Domain of Human CD154: A Promising Improved Vaccine Candidate. Vaccines (Basel) 2022; 10:897. [PMID: 35746505 PMCID: PMC9228316 DOI: 10.3390/vaccines10060897] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 05/26/2022] [Accepted: 05/27/2022] [Indexed: 02/04/2023] Open
Abstract
COVID-19 is a respiratory viral disease caused by a new coronavirus called SARS-CoV-2. This disease has spread rapidly worldwide with a high rate of morbidity and mortality. The receptor-binding domain (RBD) of protein spike (S) mediates the attachment of the virus to the host's cellular receptor. The RBD domain constitutes a very attractive target for subunit vaccine development due to its ability to induce a neutralizing antibody response against the virus. With the aim of boosting the immunogenicity of RBD, it was fused to the extracellular domain of CD154, an immune system modulator molecule. To obtain the chimeric protein, stable transduction of HEK-293 was carried out with recombinant lentivirus and polyclonal populations and cell clones were obtained. RBD-CD was purified from culture supernatant and further characterized by several techniques. RBD-CD immunogenicity evaluated in mice and non-human primates (NHP) indicated that recombinant protein was able to induce a specific and high IgG response after two doses. NHP sera also neutralize SARS-CoV-2 infection of Vero E6 cells. RBD-CD could improve the current vaccines against COVID-19, based in the enhancement of the host humoral and cellular response. Further experiments are necessary to confirm the utility of RBD-CD as a prophylactic vaccine and/or booster purpose.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Yamila Carpio
- Center for Genetic Engineering and Biotechnology, CIGB, Ave. 31 E/158 y 190, Havana 10600, Cuba; (I.Á.); (T.L.); (E.M.R.); (Y.Z.); (A.R.); (A.R.); (G.L.); (A.C.); (M.B.-R.); (D.C.); (I.A.); (L.A.E.); (L.J.G.); (Y.A.)
| | - Mario Pablo Estrada
- Center for Genetic Engineering and Biotechnology, CIGB, Ave. 31 E/158 y 190, Havana 10600, Cuba; (I.Á.); (T.L.); (E.M.R.); (Y.Z.); (A.R.); (A.R.); (G.L.); (A.C.); (M.B.-R.); (D.C.); (I.A.); (L.A.E.); (L.J.G.); (Y.A.)
| |
Collapse
|
27
|
Chen N, He Y, Zang M, Zhang Y, Lu H, Zhao Q, Wang S, Gao Y. Approaches and materials for endocytosis-independent intracellular delivery of proteins. Biomaterials 2022; 286:121567. [DOI: 10.1016/j.biomaterials.2022.121567] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 04/26/2022] [Accepted: 05/03/2022] [Indexed: 12/12/2022]
|
28
|
Willcox AC, Sung K, Garrett ME, Galloway JG, Erasmus JH, Logue JK, Hawman DW, Chu HY, Hasenkrug KJ, Fuller DH, Matsen IV FA, Overbaugh J. Detailed analysis of antibody responses to SARS-CoV-2 vaccination and infection in macaques. PLoS Pathog 2022; 18:e1010155. [PMID: 35404959 PMCID: PMC9022802 DOI: 10.1371/journal.ppat.1010155] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 04/21/2022] [Accepted: 03/21/2022] [Indexed: 02/02/2023] Open
Abstract
Macaques are a commonly used model for studying immunity to human viruses, including for studies of SARS-CoV-2 infection and vaccination. However, it is unknown whether macaque antibody responses resemble the response in humans. To answer this question, we employed a phage-based deep mutational scanning approach (Phage-DMS) to compare which linear epitopes are targeted on the SARS-CoV-2 Spike protein in convalescent humans, convalescent (re-infected) rhesus macaques, mRNA-vaccinated humans, and repRNA-vaccinated pigtail macaques. We also used Phage-DMS to determine antibody escape pathways within each epitope, enabling a granular comparison of antibody binding specificities at the locus level. Overall, we identified some common epitope targets in both macaques and humans, including in the fusion peptide (FP) and stem helix-heptad repeat 2 (SH-H) regions. Differences between groups included a response to epitopes in the N-terminal domain (NTD) and C-terminal domain (CTD) in vaccinated humans but not vaccinated macaques, as well as recognition of a CTD epitope and epitopes flanking the FP in convalescent macaques but not convalescent humans. There was also considerable variability in the escape pathways among individuals within each group. Sera from convalescent macaques showed the least variability in escape overall and converged on a common response with vaccinated humans in the SH-H epitope region, suggesting highly similar antibodies were elicited. Collectively, these findings suggest that the antibody response to SARS-CoV-2 in macaques shares many features with humans, but with substantial differences in the recognition of certain epitopes and considerable individual variability in antibody escape profiles, suggesting a diverse repertoire of antibodies that can respond to major epitopes in both humans and macaques. Differences in macaque species and exposure type may also contribute to these findings.
Collapse
Affiliation(s)
- Alexandra C. Willcox
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- Medical Scientist Training Program, University of Washington, Seattle, Washington, United States of America
- Molecular and Cellular Biology Program, University of Washington, Seattle, Washington, United States of America
| | - Kevin Sung
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Meghan E. Garrett
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- Molecular and Cellular Biology Program, University of Washington, Seattle, Washington, United States of America
| | - Jared G. Galloway
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Jesse H. Erasmus
- Department of Microbiology, University of Washington, Seattle, Washington, United States of America
- HDT Bio, Seattle, Washington, United States of America
| | - Jennifer K. Logue
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - David W. Hawman
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America
| | - Helen Y. Chu
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Kim J. Hasenkrug
- Laboratory of Persistent Viral Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America
| | - Deborah H. Fuller
- Department of Microbiology, University of Washington, Seattle, Washington, United States of America
- Infectious Diseases and Translational Medicine, Washington National Primate Research Center, Seattle, Washington, United States of America
- Center for Innate Immunity and Immune Disease, University of Washington, Seattle, Washington, United States of America
| | - Frederick A. Matsen IV
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Julie Overbaugh
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| |
Collapse
|
29
|
Wambani J, Okoth P. Scope of SARS-CoV-2 variants, mutations, and vaccine technologies. THE EGYPTIAN JOURNAL OF INTERNAL MEDICINE 2022; 34:34. [PMID: 35368846 PMCID: PMC8962228 DOI: 10.1186/s43162-022-00121-z] [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: 02/10/2022] [Accepted: 03/07/2022] [Indexed: 12/23/2022] Open
Abstract
Background The COVID-19 pandemic is caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). SARS-CoV-2 is disseminated by respiratory aerosols. The virus uses the spike protein to target epithelial cells by binding to the ACE2 receptor on the host cells. As a result, effective vaccines must target the viral spike glycoprotein. However, the appearance of an Omicron variant with 32 mutations in its spike protein raises questions about the vaccine's efficacy. Vaccines are critical in boosting immunity, lowering COVID-19-related illnesses, reducing the infectious burden on the healthcare system, and reducing economic loss, according to current data. An efficient vaccination campaign is projected to increase innate and adaptive immune responses, offering better protection against SARS-CoV-2 variants. Main body The presence of altered SARS-CoV-2 variants circulating around the world puts the effectiveness of vaccines already on the market at risk. The problem is made even worse by the Omicron variant, which has 32 mutations in its spike protein. Experts are currently examining the potential consequences of commercial vaccines on variants. However, there are worries about the vaccines' safety, the protection they provide, and whether future structural changes are required for these vaccines to be more effective. As a result of these concerns, new vaccines based on modern technology should be developed to guard against the growing SARS-CoV-2 variations. Conclusion The choice of a particular vaccine is influenced by several factors including mode of action, storage conditions, group of the vaccinee, immune response mounted, cost, dosage protocol, age, and side effects. Currently, seven SARS-CoV-2 vaccine platforms have been developed. This comprises of inactivated viruses, messenger RNA (mRNA), DNA vaccines, protein subunits, nonreplicating and replicating vector viral-like particles (VLP), and live attenuated vaccines. This review focuses on the SARS-CoV-2 mutations, variants of concern (VOCs), and advances in vaccine technologies.
Collapse
Affiliation(s)
- Josephine Wambani
- Kenya Medical Research Institute (KEMRI) HIV Laboratory-Alupe, P.O Box 3-50400, Busia, Kenya
- Department of Medical Laboratory Sciences, School of Public Health, Biomedical Sciences and Technology, Masinde Muliro University of Science and Technology, P.O Box 190, Kakamega, 50100 Kenya
| | - Patrick Okoth
- Department of Biological Sciences, School of Natural Sciences, Masinde Muliro University of Science and Technology, P. O Box 190, Kakamega, 50100 Kenya
| |
Collapse
|
30
|
Kovalenko AO, Ryabchevskaya EM, Evtushenko EA, Manukhova TI, Kondakova OA, Ivanov PA, Arkhipenko MV, Gushchin VA, Nikitin NA, Karpova OV. Vaccine Candidate Against COVID-19 Based on Structurally Modified Plant Virus as an Adjuvant. Front Microbiol 2022; 13:845316. [PMID: 35295298 PMCID: PMC8919459 DOI: 10.3389/fmicb.2022.845316] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 02/04/2022] [Indexed: 12/24/2022] Open
Abstract
A recombinant vaccine candidate has been developed based on the major coronaviruses’ antigen (S protein) fragments and a novel adjuvant—spherical particles (SPs) formed during tobacco mosaic virus thermal remodeling. The receptor-binding domain and the highly conserved antigenic fragments of the S2 protein subunit were chosen for the design of recombinant coronavirus antigens. The set of three antigens (Co1, CoF, and PE) was developed and used to create a vaccine candidate composed of antigens and SPs (SPs + 3AG). Recognition of SPs + 3AG compositions by commercially available antibodies against spike proteins of SARS-CoV and SARS-CoV-2 was confirmed. The immunogenicity testing of these compositions in a mouse model showed that SPs improved immune response to the CoF and PE antigens. Total IgG titers against both proteins were 9–16 times higher than those to SPs. Neutralizing activity against SARS-CoV-2 in serum samples collected from hamsters immunized with the SPs + 3AG was demonstrated.
Collapse
Affiliation(s)
- Angelina O Kovalenko
- Department of Virology, Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | | | - Ekaterina A Evtushenko
- Department of Virology, Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Tatiana I Manukhova
- Department of Virology, Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Olga A Kondakova
- Department of Virology, Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Peter A Ivanov
- Department of Virology, Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Marina V Arkhipenko
- Department of Virology, Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Vladimir A Gushchin
- Department of Virology, Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia.,N.F. Gamaleya National Research Center for Epidemiology and Microbiology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Nikolai A Nikitin
- Department of Virology, Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Olga V Karpova
- Department of Virology, Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| |
Collapse
|
31
|
Dalvie NC, Tostanoski LH, Rodriguez-Aponte SA, Kaur K, Bajoria S, Kumru OS, Martinot AJ, Chandrashekar A, McMahan K, Mercado NB, Yu J, Chang A, Giffin VM, Nampanya F, Patel S, Bowman L, Naranjo CA, Yun D, Flinchbaugh Z, Pessaint L, Brown R, Velasco J, Teow E, Cook A, Andersen H, Lewis MG, Camp DL, Silverman JM, Nagar GS, Rao HD, Lothe RR, Chandrasekharan R, Rajurkar MP, Shaligram US, Kleanthous H, Joshi SB, Volkin DB, Biswas S, Love JC, Barouch DH. SARS-CoV-2 receptor binding domain displayed on HBsAg virus-like particles elicits protective immunity in macaques. SCIENCE ADVANCES 2022; 8:eabl6015. [PMID: 35294244 PMCID: PMC8926328 DOI: 10.1126/sciadv.abl6015] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 01/25/2022] [Indexed: 05/16/2023]
Abstract
Authorized vaccines against SARS-CoV-2 remain less available in low- and middle-income countries due to insufficient supply, high costs, and storage requirements. Global immunity could still benefit from new vaccines using widely available, safe adjuvants, such as alum and protein subunits, suited to low-cost production in existing manufacturing facilities. Here, a clinical-stage vaccine candidate comprising a SARS-CoV-2 receptor binding domain-hepatitis B surface antigen virus-like particle elicited protective immunity in cynomolgus macaques. Titers of neutralizing antibodies (>104) induced by this candidate were above the range of protection for other licensed vaccines in nonhuman primates. Including CpG 1018 did not significantly improve the immunological responses. Vaccinated animals challenged with SARS-CoV-2 showed reduced median viral loads in bronchoalveolar lavage (~3.4 log10) and nasal mucosa (~2.9 log10) versus sham controls. These data support the potential benefit of this design for a low-cost modular vaccine platform for SARS-CoV-2 and other variants of concern or betacoronaviruses.
Collapse
Affiliation(s)
- Neil C. Dalvie
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Lisa H. Tostanoski
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Sergio A. Rodriguez-Aponte
- The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Kawaljit Kaur
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS 66047, USA
| | - Sakshi Bajoria
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS 66047, USA
| | - Ozan S. Kumru
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS 66047, USA
| | - Amanda J. Martinot
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
- Departments of Infectious Diseases and Global Health and Comparative Pathobiology, Cummings School of Veterinary Medicine, Tufts University, North Grafton, MA 01536, USA
| | - Abishek Chandrashekar
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Katherine McMahan
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Noe B. Mercado
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Jingyou Yu
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Aiquan Chang
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Victoria M. Giffin
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Felix Nampanya
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Shivani Patel
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Lesley Bowman
- SpyBiotech Limited, Oxford Business Park North, Oxford OX4 2JZ, UK
| | - Christopher A. Naranjo
- The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Dongsoo Yun
- The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | | | | | | | | | | | | | | | | - Danielle L. Camp
- The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | | | | | | | | | | | | | | | | - Sangeeta B. Joshi
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS 66047, USA
| | - David B. Volkin
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS 66047, USA
| | - Sumi Biswas
- SpyBiotech Limited, Oxford Business Park North, Oxford OX4 2JZ, UK
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | - J. Christopher Love
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Dan H. Barouch
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02115, USA
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
- Massachusetts Consortium on Pathogen Readiness, Boston, MA 02115, USA
| |
Collapse
|
32
|
Coria LM, Saposnik LM, Pueblas Castro C, Castro EF, Bruno LA, Stone WB, Pérez PS, Darriba ML, Chemes LB, Alcain J, Mazzitelli I, Varese A, Salvatori M, Auguste AJ, Álvarez DE, Pasquevich KA, Cassataro J. A Novel Bacterial Protease Inhibitor Adjuvant in RBD-Based COVID-19 Vaccine Formulations Containing Alum Increases Neutralizing Antibodies, Specific Germinal Center B Cells and Confers Protection Against SARS-CoV-2 Infection in Mice. Front Immunol 2022; 13:844837. [PMID: 35296091 PMCID: PMC8919065 DOI: 10.3389/fimmu.2022.844837] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 02/01/2022] [Indexed: 01/18/2023] Open
Abstract
In this work, we evaluated recombinant receptor binding domain (RBD)-based vaccine formulation prototypes with potential for further clinical development. We assessed different formulations containing RBD plus alum, AddaS03, AddaVax, or the combination of alum and U-Omp19: a novel Brucella spp. protease inhibitor vaccine adjuvant. Results show that the vaccine formulation composed of U-Omp19 and alum as adjuvants has a better performance: it significantly increased mucosal and systemic neutralizing antibodies in comparison to antigen plus alum, AddaVax, or AddaS03. Antibodies induced with the formulation containing U-Omp19 and alum not only increased their neutralization capacity against the ancestral virus but also cross-neutralized alpha, lambda, and gamma variants with similar potency. Furthermore, the addition of U-Omp19 to alum vaccine formulation increased the frequency of RBD-specific geminal center B cells and plasmablasts. Additionally, U-Omp19+alum formulation induced RBD-specific Th1 and CD8+ T-cell responses in spleens and lungs. Finally, this vaccine formulation conferred protection against an intranasal severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) challenge of K18-hACE2 mice.
Collapse
Affiliation(s)
- Lorena M. Coria
- Instituto de Investigaciones Biotecnológicas Dr. Rodolfo A. Ugalde, Universidad Nacional de San Martín, Consejo Nacional de Investigaciones Científicas y Técnicas (UNSAM-CONICET), San Martín, Argentina
| | - Lucas M. Saposnik
- Instituto de Investigaciones Biotecnológicas Dr. Rodolfo A. Ugalde, Universidad Nacional de San Martín, Consejo Nacional de Investigaciones Científicas y Técnicas (UNSAM-CONICET), San Martín, Argentina
| | - Celeste Pueblas Castro
- Instituto de Investigaciones Biotecnológicas Dr. Rodolfo A. Ugalde, Universidad Nacional de San Martín, Consejo Nacional de Investigaciones Científicas y Técnicas (UNSAM-CONICET), San Martín, Argentina
| | - Eliana F. Castro
- Instituto de Investigaciones Biotecnológicas Dr. Rodolfo A. Ugalde, Universidad Nacional de San Martín, Consejo Nacional de Investigaciones Científicas y Técnicas (UNSAM-CONICET), San Martín, Argentina
- Instituto de Virología e Innovaciones Tecnológicas (IVIT), Centro de Investigaciones en Ciencias Veterinarias y Agronómicas (CICVyA), Instituto Nacional de Tecnología Agropecuaria (INTA)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Laura A. Bruno
- Instituto de Investigaciones Biotecnológicas Dr. Rodolfo A. Ugalde, Universidad Nacional de San Martín, Consejo Nacional de Investigaciones Científicas y Técnicas (UNSAM-CONICET), San Martín, Argentina
| | - William B. Stone
- Department of Entomology, College of Agriculture and Life Sciences, Fralin Life Science Institute, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Paula S. Pérez
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS, Universidad de Buenos Aires-CONICET), Buenos Aires, Argentina
| | - Maria Laura Darriba
- Instituto de Investigaciones Biotecnológicas Dr. Rodolfo A. Ugalde, Universidad Nacional de San Martín, Consejo Nacional de Investigaciones Científicas y Técnicas (UNSAM-CONICET), San Martín, Argentina
| | - Lucia B. Chemes
- Instituto de Investigaciones Biotecnológicas Dr. Rodolfo A. Ugalde, Universidad Nacional de San Martín, Consejo Nacional de Investigaciones Científicas y Técnicas (UNSAM-CONICET), San Martín, Argentina
| | - Julieta Alcain
- Instituto de Investigaciones Biotecnológicas Dr. Rodolfo A. Ugalde, Universidad Nacional de San Martín, Consejo Nacional de Investigaciones Científicas y Técnicas (UNSAM-CONICET), San Martín, Argentina
| | - Ignacio Mazzitelli
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS, Universidad de Buenos Aires-CONICET), Buenos Aires, Argentina
| | - Augusto Varese
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS, Universidad de Buenos Aires-CONICET), Buenos Aires, Argentina
| | - Melina Salvatori
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS, Universidad de Buenos Aires-CONICET), Buenos Aires, Argentina
| | - Albert J. Auguste
- Department of Entomology, College of Agriculture and Life Sciences, Fralin Life Science Institute, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
- Center for Emerging, Zoonotic, and Arthropod-borne Pathogens, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Diego E. Álvarez
- Instituto de Investigaciones Biotecnológicas Dr. Rodolfo A. Ugalde, Universidad Nacional de San Martín, Consejo Nacional de Investigaciones Científicas y Técnicas (UNSAM-CONICET), San Martín, Argentina
| | - Karina A. Pasquevich
- Instituto de Investigaciones Biotecnológicas Dr. Rodolfo A. Ugalde, Universidad Nacional de San Martín, Consejo Nacional de Investigaciones Científicas y Técnicas (UNSAM-CONICET), San Martín, Argentina
- *Correspondence: Karina A. Pasquevich, ; Juliana Cassataro,
| | - Juliana Cassataro
- Instituto de Investigaciones Biotecnológicas Dr. Rodolfo A. Ugalde, Universidad Nacional de San Martín, Consejo Nacional de Investigaciones Científicas y Técnicas (UNSAM-CONICET), San Martín, Argentina
- *Correspondence: Karina A. Pasquevich, ; Juliana Cassataro,
| |
Collapse
|
33
|
Pitcovski J, Gruzdev N, Abzach A, Katz C, Ben-Adiva R, Brand-Shwartz M, Yadid I, Ratzon-Ashkenazi E, Emquies K, Israeli H, Haviv H, Rapoport I, Bloch I, Shadmon R, Eitan Z, Eliahu D, Hilel T, Laster M, Kremer-Tal S, Byk-Tennenbaum T, Shahar E. Oral subunit SARS-CoV-2 vaccine induces systemic neutralizing IgG, IgA and cellular immune responses and can boost neutralizing antibody responses primed by an injected vaccine. Vaccine 2022; 40:1098-1107. [PMID: 35078662 PMCID: PMC8768024 DOI: 10.1016/j.vaccine.2022.01.025] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 11/30/2021] [Accepted: 01/13/2022] [Indexed: 12/15/2022]
Abstract
The rapid spread of the COVID-19 pandemic, with its devastating medical and economic impacts, triggered an unprecedented race toward development of effective vaccines. The commercialized vaccines are parenterally administered, which poses logistic challenges, while adequate protection at the mucosal sites of virus entry is questionable. Furthermore, essentially all vaccine candidates target the viral spike (S) protein, a surface protein that undergoes significant antigenic drift. This work aimed to develop an oral multi-antigen SARS-CoV-2 vaccine comprised of the receptor binding domain (RBD) of the viral S protein, two domains of the viral nucleocapsid protein (N), and heat-labile enterotoxin B (LTB), a potent mucosal adjuvant. The humoral, mucosal and cell-mediated immune responses of both a three-dose vaccination schedule and a heterologous subcutaneous prime and oral booster regimen were assessed in mice and rats, respectively. Mice receiving the oral vaccine compared to control mice showed significantly enhanced post-dose-3 virus-neutralizing antibody, anti-S IgG and IgA production and N-protein-stimulated IFN-γ and IL-2 secretion by T cells. When administered as a booster to rats following parenteral priming with the viral S1 protein, the oral vaccine elicited markedly higher neutralizing antibody titres than did oral placebo booster. A single oral booster following two subcutaneous priming doses elicited serum IgG and mucosal IgA levels similar to those raised by three subcutaneous doses. In conclusion, the oral LTB-adjuvanted multi-epitope SARS-CoV-2 vaccine triggered versatile humoral, cellular and mucosal immune responses, which are likely to provide protection, while also minimizing technical hurdles presently limiting global vaccination, whether by priming or booster programs.
Collapse
Affiliation(s)
- Jacob Pitcovski
- MIGAL Research Institute in the Galilee, Kiryat Shmona, Israel; Tel-Hai Academic College, Upper Galilee, Israel
| | - Nady Gruzdev
- MIGAL Research Institute in the Galilee, Kiryat Shmona, Israel
| | - Anna Abzach
- MIGAL Research Institute in the Galilee, Kiryat Shmona, Israel
| | - Chen Katz
- MIGAL Research Institute in the Galilee, Kiryat Shmona, Israel
| | - Ran Ben-Adiva
- MIGAL Research Institute in the Galilee, Kiryat Shmona, Israel
| | | | - Itamar Yadid
- MIGAL Research Institute in the Galilee, Kiryat Shmona, Israel; Tel-Hai Academic College, Upper Galilee, Israel
| | | | - Ken Emquies
- MIGAL Research Institute in the Galilee, Kiryat Shmona, Israel
| | - Hadasa Israeli
- MIGAL Research Institute in the Galilee, Kiryat Shmona, Israel
| | - Hadar Haviv
- MIGAL Research Institute in the Galilee, Kiryat Shmona, Israel
| | - Irena Rapoport
- MIGAL Research Institute in the Galilee, Kiryat Shmona, Israel
| | - Itai Bloch
- MIGAL Research Institute in the Galilee, Kiryat Shmona, Israel
| | | | | | - Dalia Eliahu
- MIGAL Research Institute in the Galilee, Kiryat Shmona, Israel
| | - Talia Hilel
- MIGAL Research Institute in the Galilee, Kiryat Shmona, Israel
| | | | | | | | - Ehud Shahar
- MIGAL Research Institute in the Galilee, Kiryat Shmona, Israel; Tel-Hai Academic College, Upper Galilee, Israel.
| |
Collapse
|
34
|
Immunization with synthetic SARS-CoV-2 S glycoprotein virus-like particles protects macaques from infection. Cell Rep Med 2022; 3:100528. [PMID: 35233549 PMCID: PMC8784613 DOI: 10.1016/j.xcrm.2022.100528] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 11/26/2021] [Accepted: 01/19/2022] [Indexed: 11/20/2022]
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic has caused an ongoing global health crisis. Here, we present as a vaccine candidate synthetic SARS-CoV-2 spike (S) glycoprotein-coated lipid vesicles that resemble virus-like particles. Soluble S glycoprotein trimer stabilization by formaldehyde cross-linking introduces two major inter-protomer cross-links that keep all receptor-binding domains in the “down” conformation. Immunization of cynomolgus macaques with S coated onto lipid vesicles (S-LVs) induces high antibody titers with potent neutralizing activity against the vaccine strain, Alpha, Beta, and Gamma variants as well as T helper (Th)1 CD4+-biased T cell responses. Although anti-receptor-binding domain (RBD)-specific antibody responses are initially predominant, the third immunization boosts significant non-RBD antibody titers. Challenging vaccinated animals with SARS-CoV-2 shows a complete protection through sterilizing immunity, which correlates with the presence of nasopharyngeal anti-S immunoglobulin G (IgG) and IgA titers. Thus, the S-LV approach is an efficient and safe vaccine candidate based on a proven classical approach for further development and clinical testing. S glycoprotein formaldehyde cross-linking stabilizes S in the prefusion conformation Vaccination of cynomolgus macaques with S lipid particles induces neutralization Vaccination protects macaques against a SARS-CoV-2 challenge Sterilizing protection correlates with nasopharyngeal anti-S IgG and IgA titers
Collapse
|
35
|
Staufer O, Gupta K, Hernandez Bücher JE, Kohler F, Sigl C, Singh G, Vasileiou K, Yagüe Relimpio A, Macher M, Fabritz S, Dietz H, Cavalcanti Adam EA, Schaffitzel C, Ruggieri A, Platzman I, Berger I, Spatz JP. Synthetic virions reveal fatty acid-coupled adaptive immunogenicity of SARS-CoV-2 spike glycoprotein. Nat Commun 2022; 13:868. [PMID: 35165285 PMCID: PMC8844029 DOI: 10.1038/s41467-022-28446-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 01/24/2022] [Indexed: 12/15/2022] Open
Abstract
SARS-CoV-2 infection is a major global public health concern with incompletely understood pathogenesis. The SARS-CoV-2 spike (S) glycoprotein comprises a highly conserved free fatty acid binding pocket (FABP) with unknown function and evolutionary selection advantage1,2. Deciphering FABP impact on COVID-19 progression is challenged by the heterogenous nature and large molecular variability of live virus. Here we create synthetic minimal virions (MiniVs) of wild-type and mutant SARS-CoV-2 with precise molecular composition and programmable complexity by bottom-up assembly. MiniV-based systematic assessment of S free fatty acid (FFA) binding reveals that FABP functions as an allosteric regulatory site enabling adaptation of SARS-CoV-2 immunogenicity to inflammation states via binding of pro-inflammatory FFAs. This is achieved by regulation of the S open-to-close equilibrium and the exposure of both, the receptor binding domain (RBD) and the SARS-CoV-2 RGD motif that is responsible for integrin co-receptor engagement. We find that the FDA-approved drugs vitamin K and dexamethasone modulate S-based cell binding in an FABP-like manner. In inflammatory FFA environments, neutralizing immunoglobulins from human convalescent COVID-19 donors lose neutralization activity. Empowered by our MiniV technology, we suggest a conserved mechanism by which SARS-CoV-2 dynamically couples its immunogenicity to the host immune response.
Collapse
Affiliation(s)
- Oskar Staufer
- Department for Cellular Biophysics, Max Planck Institute for Medical Research, Jahnstraße 29, 69120, Heidelberg, Germany.
- Institute for Molecular Systems Engineering, University of Heidelberg, Im Neuenheimer Feld 253, 69120, Heidelberg, Germany.
- Max Planck-Bristol Center for Minimal Biology, University of Bristol, 1 Tankard's Close, Bristol, BS8 1TD, UK.
- Max Planck School Matter to Life, Jahnstraße 29, 69120, Heidelberg, Germany.
| | - Kapil Gupta
- School of Biochemistry, Biomedical Sciences, University of Bristol, 1 Tankard's Close, Bristol, BS8 1TD, UK
- Bristol Synthetic Biology Centre BrisSynBio, University of Bristol, 4 Tyndall Ave, Bristol, BS8 1TQ, UK
| | - Jochen Estebano Hernandez Bücher
- Department for Cellular Biophysics, Max Planck Institute for Medical Research, Jahnstraße 29, 69120, Heidelberg, Germany
- Institute for Molecular Systems Engineering, University of Heidelberg, Im Neuenheimer Feld 253, 69120, Heidelberg, Germany
| | - Fabian Kohler
- Department of Physics, Technical University of Munich, 85748, Garching, Germany
| | - Christian Sigl
- Department of Physics, Technical University of Munich, 85748, Garching, Germany
| | - Gunjita Singh
- School of Biochemistry, Biomedical Sciences, University of Bristol, 1 Tankard's Close, Bristol, BS8 1TD, UK
| | - Kate Vasileiou
- School of Biochemistry, Biomedical Sciences, University of Bristol, 1 Tankard's Close, Bristol, BS8 1TD, UK
| | - Ana Yagüe Relimpio
- Department for Cellular Biophysics, Max Planck Institute for Medical Research, Jahnstraße 29, 69120, Heidelberg, Germany
- Institute for Molecular Systems Engineering, University of Heidelberg, Im Neuenheimer Feld 253, 69120, Heidelberg, Germany
| | - Meline Macher
- Department for Cellular Biophysics, Max Planck Institute for Medical Research, Jahnstraße 29, 69120, Heidelberg, Germany
- Institute for Molecular Systems Engineering, University of Heidelberg, Im Neuenheimer Feld 253, 69120, Heidelberg, Germany
- Max Planck School Matter to Life, Jahnstraße 29, 69120, Heidelberg, Germany
| | - Sebastian Fabritz
- Department for Chemical Biology, Max Planck Institute for Medical Research, Jahnstraße 29, 69120, Heidelberg, Germany
| | - Hendrik Dietz
- Max Planck School Matter to Life, Jahnstraße 29, 69120, Heidelberg, Germany
- Department of Physics, Technical University of Munich, 85748, Garching, Germany
| | - Elisabetta Ada Cavalcanti Adam
- Department for Cellular Biophysics, Max Planck Institute for Medical Research, Jahnstraße 29, 69120, Heidelberg, Germany
- Max Planck School Matter to Life, Jahnstraße 29, 69120, Heidelberg, Germany
| | - Christiane Schaffitzel
- School of Biochemistry, Biomedical Sciences, University of Bristol, 1 Tankard's Close, Bristol, BS8 1TD, UK
- Bristol Synthetic Biology Centre BrisSynBio, University of Bristol, 4 Tyndall Ave, Bristol, BS8 1TQ, UK
- Halo Therapeutics Ltd, Science Creates, Albert Road St. Philips Central, Bristol, BS2 0XJ, UK
| | - Alessia Ruggieri
- Department of Infectious Diseases, Molecular Virology, Center for Integrated Infectious Disease Research, University of Heidelberg, Im Neuenheimer Feld 344, 69120, Heidelberg, Germany
| | - Ilia Platzman
- Department for Cellular Biophysics, Max Planck Institute for Medical Research, Jahnstraße 29, 69120, Heidelberg, Germany
- Institute for Molecular Systems Engineering, University of Heidelberg, Im Neuenheimer Feld 253, 69120, Heidelberg, Germany
- Max Planck-Bristol Center for Minimal Biology, University of Bristol, 1 Tankard's Close, Bristol, BS8 1TD, UK
| | - Imre Berger
- Max Planck-Bristol Center for Minimal Biology, University of Bristol, 1 Tankard's Close, Bristol, BS8 1TD, UK.
- School of Biochemistry, Biomedical Sciences, University of Bristol, 1 Tankard's Close, Bristol, BS8 1TD, UK.
- Bristol Synthetic Biology Centre BrisSynBio, University of Bristol, 4 Tyndall Ave, Bristol, BS8 1TQ, UK.
- Halo Therapeutics Ltd, Science Creates, Albert Road St. Philips Central, Bristol, BS2 0XJ, UK.
| | - Joachim P Spatz
- Department for Cellular Biophysics, Max Planck Institute for Medical Research, Jahnstraße 29, 69120, Heidelberg, Germany.
- Institute for Molecular Systems Engineering, University of Heidelberg, Im Neuenheimer Feld 253, 69120, Heidelberg, Germany.
- Max Planck-Bristol Center for Minimal Biology, University of Bristol, 1 Tankard's Close, Bristol, BS8 1TD, UK.
- Max Planck School Matter to Life, Jahnstraße 29, 69120, Heidelberg, Germany.
| |
Collapse
|
36
|
Masuda A, Lee JM, Miyata T, Mon H, Sato K, Oyama K, Sakurai Y, Yasuda J, Takahashi D, Ueda T, Kato Y, Nishida M, Karasaki N, Kakino K, Ebihara T, Nagasato T, Hino M, Nakashima A, Suzuki K, Tonooka Y, Tanaka M, Moriyama T, Nakatake H, Fujita R, Kusakabe T. Optimization of SARS-CoV-2 Spike Protein Expression in the Silkworm and Induction of Efficient Protective Immunity by Inoculation With Alum Adjuvants. Front Immunol 2022; 12:803647. [PMID: 35095889 PMCID: PMC8789674 DOI: 10.3389/fimmu.2021.803647] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 12/20/2021] [Indexed: 11/13/2022] Open
Abstract
The newly emerged severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is causing a spread of coronavirus disease 2019 (COVID-19) globally. In order to end the COVID-19 pandemic, an effective vaccine against SARS-CoV-2 must be produced at low cost and disseminated worldwide. The spike (S) protein of coronaviruses plays a pivotal role in the infection to host cells. Therefore, targeting the S protein is one of the most rational approaches in developing vaccines and therapeutic agents. In this study, we optimized the expression of secreted trimerized S protein of SARS-CoV-2 using a silkworm-baculovirus expression vector system and evaluated its immunogenicity in mice. The results showed that the S protein forming the trimeric structure was the most stable when the chicken cartilage matrix protein was used as the trimeric motif and could be purified in large amounts from the serum of silkworm larvae. The purified S protein efficiently induced antigen-specific antibodies in mouse serum without adjuvant, but its ability to induce neutralizing antibodies was low. After examining several adjuvants, the use of Alum adjuvant was the most effective in inducing strong neutralizing antibody induction. We also examined the adjuvant effect of paramylon from Euglena gracilis when administered with the S protein. Our results highlight the effectiveness and suitable construct design of the S protein produced in silkworms for the subunit vaccine development against SARS-CoV-2.
Collapse
Affiliation(s)
- Akitsu Masuda
- Laboratory of Insect Genome Science, Graduate School of Bioresource and Bioenvironmental Sciences, Kyushu University, Fukuoka, Japan
| | - Jae Man Lee
- Laboratory of Creative Science for Insect Industries, Graduate School of Bioresource and Bioenvironmental Sciences, Kyushu University, Fukuoka, Japan
| | - Takeshi Miyata
- Department of Biochemistry and Biotechnology, Faculty of Agriculture, Kagoshima University, Kagoshima, Japan
| | - Hiroaki Mon
- Laboratory of Insect Genome Science, Graduate School of Bioresource and Bioenvironmental Sciences, Kyushu University, Fukuoka, Japan
| | - Keita Sato
- Department of Biochemistry and Biotechnology, Faculty of Agriculture, Kagoshima University, Kagoshima, Japan
| | - Kosuke Oyama
- Laboratory of Protein Structure, Function and Design, Faculty of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Yasuteru Sakurai
- Department of Emerging Infectious Diseases, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Japan.,National Research Center for the Control and Prevention of Infectious Diseases, Nagasaki University, Nagasaki, Japan
| | - Jiro Yasuda
- Department of Emerging Infectious Diseases, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Japan.,National Research Center for the Control and Prevention of Infectious Diseases, Nagasaki University, Nagasaki, Japan
| | - Daisuke Takahashi
- Laboratory of Protein Structure, Function and Design, Faculty of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Tadashi Ueda
- Laboratory of Protein Structure, Function and Design, Faculty of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Yuri Kato
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Motohiro Nishida
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Noriko Karasaki
- Laboratory of Insect Genome Science, Graduate School of Bioresource and Bioenvironmental Sciences, Kyushu University, Fukuoka, Japan
| | - Kohei Kakino
- Laboratory of Insect Genome Science, Graduate School of Bioresource and Bioenvironmental Sciences, Kyushu University, Fukuoka, Japan
| | - Takeru Ebihara
- Laboratory of Insect Genome Science, Graduate School of Bioresource and Bioenvironmental Sciences, Kyushu University, Fukuoka, Japan
| | - Takumi Nagasato
- Laboratory of Insect Genome Science, Graduate School of Bioresource and Bioenvironmental Sciences, Kyushu University, Fukuoka, Japan
| | - Masato Hino
- Laboratory of Sanitary Entomology, Graduate School of Bioresource and Bioenvironmental Sciences, Kyushu University, Fukuoka, Japan
| | - Ayaka Nakashima
- The Research and Development Department, Euglena Co., Ltd, Tokyo, Japan
| | - Kengo Suzuki
- The Research and Development Department, Euglena Co., Ltd, Tokyo, Japan
| | - Yoshino Tonooka
- Laboratory of Insect Genome Science, Graduate School of Bioresource and Bioenvironmental Sciences, Kyushu University, Fukuoka, Japan
| | - Miyu Tanaka
- Laboratory of Insect Genome Science, Graduate School of Bioresource and Bioenvironmental Sciences, Kyushu University, Fukuoka, Japan
| | - Takato Moriyama
- Laboratory of Insect Genome Science, Graduate School of Bioresource and Bioenvironmental Sciences, Kyushu University, Fukuoka, Japan
| | | | - Ryosuke Fujita
- Laboratory of Sanitary Entomology, Graduate School of Bioresource and Bioenvironmental Sciences, Kyushu University, Fukuoka, Japan
| | - Takahiro Kusakabe
- Laboratory of Insect Genome Science, Graduate School of Bioresource and Bioenvironmental Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
37
|
Liu J, Budylowski P, Samson R, Griffin BD, Babuadze G, Rathod B, Colwill K, Abioye JA, Schwartz JA, Law R, Yip L, Ahn SK, Chau S, Naghibosadat M, Arita Y, Hu Q, Yue FY, Banerjee A, Hardy WR, Mossman K, Mubareka S, Kozak RA, Pollanen MS, Martin Orozco N, Gingras AC, Marcusson EG, Ostrowski MA. Preclinical evaluation of a SARS-CoV-2 mRNA vaccine PTX-COVID19-B. SCIENCE ADVANCES 2022; 8:eabj9815. [PMID: 35044832 PMCID: PMC8769538 DOI: 10.1126/sciadv.abj9815] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 11/24/2021] [Indexed: 06/01/2023]
Abstract
Safe and effective vaccines are needed to end the COVID-19 pandemic. Here, we report the preclinical development of a lipid nanoparticle–formulated SARS-CoV-2 mRNA vaccine, PTX-COVID19-B. PTX-COVID19-B was chosen among three candidates after the initial mouse vaccination results showed that it elicited the strongest neutralizing antibody response against SARS-CoV-2. Further tests in mice and hamsters indicated that PTX-COVID19-B induced robust humoral and cellular immune responses and completely protected the vaccinated animals from SARS-CoV-2 infection in the lung. Studies in hamsters also showed that PTX-COVID19-B protected the upper respiratory tract from SARS-CoV-2 infection. Mouse immune sera elicited by PTX-COVID19-B vaccination were able to neutralize SARS-CoV-2 variants of concern, including the Alpha, Beta, Gamma, and Delta lineages. No adverse effects were induced by PTX-COVID19-B in either mice or hamsters. Based on these results, PTX-COVID19-B was authorized by Health Canada to enter clinical trials in December 2020 with a phase 2 clinical trial ongoing.
Collapse
MESH Headings
- Animals
- Antibodies, Neutralizing/blood
- Antibodies, Viral/blood
- CD4 Lymphocyte Count
- CD8-Positive T-Lymphocytes/immunology
- COVID-19/immunology
- COVID-19/prevention & control
- COVID-19 Vaccines/adverse effects
- COVID-19 Vaccines/immunology
- Canada
- Cell Line
- Cricetinae
- Drug Evaluation, Preclinical
- Female
- HEK293 Cells
- Humans
- Immunity, Cellular/immunology
- Immunity, Humoral/immunology
- Liposomes/pharmacology
- Male
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Nanoparticles
- SARS-CoV-2/immunology
- Spike Glycoprotein, Coronavirus/genetics
- Spike Glycoprotein, Coronavirus/immunology
- Th1 Cells/immunology
- Vaccines, Synthetic/immunology
- mRNA Vaccines/immunology
Collapse
Affiliation(s)
- Jun Liu
- Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Patrick Budylowski
- Department of Medicine, University of Toronto, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Reuben Samson
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- Lunenfeld-Tanenbaum Research Institute at Mount Sinai Hospital, Sinai Health System, Toronto, ON, Canada
| | | | | | - Bhavisha Rathod
- Lunenfeld-Tanenbaum Research Institute at Mount Sinai Hospital, Sinai Health System, Toronto, ON, Canada
| | - Karen Colwill
- Lunenfeld-Tanenbaum Research Institute at Mount Sinai Hospital, Sinai Health System, Toronto, ON, Canada
| | | | | | - Ryan Law
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Lily Yip
- Sunnybrook Research Institute, Toronto, ON, Canada
| | - Sang Kyun Ahn
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Serena Chau
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | | | - Yuko Arita
- Providence Therapeutics Holdings Inc., Calgary, AB, Canada
| | - Queenie Hu
- Lunenfeld-Tanenbaum Research Institute at Mount Sinai Hospital, Sinai Health System, Toronto, ON, Canada
| | - Feng Yun Yue
- Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Arinjay Banerjee
- Vaccine and Infectious Disease Organization, University of Saskatchewan, Saskatoon, SK, Canada
- Department of Veterinary Microbiology, University of Saskatchewan, Saskatoon, SK, Canada
- Department of Biology, University of Waterloo, Waterloo, ON, Canada
| | - W. Rod Hardy
- Lunenfeld-Tanenbaum Research Institute at Mount Sinai Hospital, Sinai Health System, Toronto, ON, Canada
| | - Karen Mossman
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Samira Mubareka
- Sunnybrook Research Institute, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathology, University of Toronto, Toronto, ON, Canada
| | | | - Michael S. Pollanen
- Department of Laboratory Medicine and Pathology, University of Toronto, Toronto, ON, Canada
| | | | - Anne-Claude Gingras
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- Lunenfeld-Tanenbaum Research Institute at Mount Sinai Hospital, Sinai Health System, Toronto, ON, Canada
| | - Eric G. Marcusson
- Providence Therapeutics Holdings Inc., Calgary, AB, Canada
- Marcusson Consulting, San Francisco, CA, USA
| | - Mario A. Ostrowski
- Department of Medicine, University of Toronto, Toronto, ON, Canada
- Department of Immunology, University of Toronto, Toronto, ON, Canada
- Keenan Research Centre for Biomedical Science of St. Michael’s Hospital, Unity Health Toronto, Toronto, ON, Canada
| |
Collapse
|
38
|
Blanco OR, Dorta D, Hernández CA, Abreu D, Domínguez AG, Luna Y, Valdivia O, Pérez-Bernal M, Tamayo C, Lemos G, Pasarón IM, Pérez JJ, Benítez L, Bequet-Romero M, Fragas A, Cabrera Y, Pérez ER. Murine monoclonal antibodies against RBD of the SARS-CoV-2 spike protein as useful analytical tools for subunit vaccine development and clinical trials. J Immunol Methods 2022; 500:113195. [PMID: 34843713 PMCID: PMC8619880 DOI: 10.1016/j.jim.2021.113195] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 11/19/2021] [Accepted: 11/23/2021] [Indexed: 12/16/2022]
Abstract
COVID-19 pandemic poses a serious threat to human health; it has completely disrupted global stability, making vaccine development an important goal to achieve. Monoclonal antibodies play an important role in subunit vaccines strategies. In this work, nine murine MAbs against the RBD of the SARS-CoV-2 spike protein were obtained by hybridoma technology. Characterization of purified antibodies demonstrated that five of them have affinities in the order of 108 L/mol. Six MAbs showed specific recognition of different recombinant RBD-S antigens in solution. Studies of the additivity index of anti-RBD antibodies, by using a novel procedure to determine the additivity cut point, showed recognition of at least five different epitopes. The MAbs CBSSRBD-S.11 and CBSSRBD-S.8 revealed significant neutralizing capacity against SARS-CoV-2 in an ACE2-RBD binding inhibition assay (IC50 = 85.5pM and IC50 = 122.7pM, respectively) and in a virus neutralizing test with intact SARS-CoV-2 (VN50 = 0.552 nM and VN50 = 4.854 nM, respectively) when D614G strain was used to infect Vero cells. Also CBSSRBD-S.11 neutralized the SARS-CoV-2 strains Alpha and Beta: VN50 = 0.707 nM and VN50 = 0.132 nM, respectively. The high affinity CBSSRBD-S.8 and CBSSRBD-S.7 recognized different epitopes, so they are suitable for the development of a sandwich ELISA to quantitate RBD-S recombinant antigens in biomanufacturing processes, as well as in pharmacokinetic studies in clinical and preclinical trials.
Collapse
Affiliation(s)
- Omar R Blanco
- Center for Genetic Engineering and Biotechnology of Sancti Spíritus, Circunvalante Norte, Olivos III, Sancti Spíritus, Cuba
| | - Dayamí Dorta
- Center for Genetic Engineering and Biotechnology of Sancti Spíritus, Circunvalante Norte, Olivos III, Sancti Spíritus, Cuba
| | - Carlos A Hernández
- Center for Genetic Engineering and Biotechnology of Sancti Spíritus, Circunvalante Norte, Olivos III, Sancti Spíritus, Cuba
| | - Daymí Abreu
- Center for Genetic Engineering and Biotechnology of Sancti Spíritus, Circunvalante Norte, Olivos III, Sancti Spíritus, Cuba
| | - Andy G Domínguez
- Center for Genetic Engineering and Biotechnology of Sancti Spíritus, Circunvalante Norte, Olivos III, Sancti Spíritus, Cuba
| | - Yaramis Luna
- Center for Genetic Engineering and Biotechnology of Sancti Spíritus, Circunvalante Norte, Olivos III, Sancti Spíritus, Cuba
| | - Onel Valdivia
- Center for Genetic Engineering and Biotechnology of Sancti Spíritus, Circunvalante Norte, Olivos III, Sancti Spíritus, Cuba
| | - Maylín Pérez-Bernal
- Center for Genetic Engineering and Biotechnology of Sancti Spíritus, Circunvalante Norte, Olivos III, Sancti Spíritus, Cuba
| | - Celia Tamayo
- Center for Genetic Engineering and Biotechnology of Sancti Spíritus, Circunvalante Norte, Olivos III, Sancti Spíritus, Cuba
| | - Gilda Lemos
- Center for Genetic Engineering and Biotechnology, Ave. 31 e/ 158 y 190, Playa, Havana, Cuba
| | - Ivis M Pasarón
- Center for Genetic Engineering and Biotechnology of Sancti Spíritus, Circunvalante Norte, Olivos III, Sancti Spíritus, Cuba
| | - Joel J Pérez
- Center for Genetic Engineering and Biotechnology of Sancti Spíritus, Circunvalante Norte, Olivos III, Sancti Spíritus, Cuba
| | - Liudmila Benítez
- Center for Genetic Engineering and Biotechnology of Sancti Spíritus, Circunvalante Norte, Olivos III, Sancti Spíritus, Cuba
| | - Mónica Bequet-Romero
- Center for Genetic Engineering and Biotechnology, Ave. 31 e/ 158 y 190, Playa, Havana, Cuba
| | - Anitza Fragas
- Civilian Defense Scientific Research Center, Carretera de Jamaica y Autopista 15 Nacional, San José de las Lajas, Mayabeque, Cuba
| | - Yeosvany Cabrera
- Center for Genetic Engineering and Biotechnology of Sancti Spíritus, Circunvalante Norte, Olivos III, Sancti Spíritus, Cuba.
| | - Enrique R Pérez
- Center for Genetic Engineering and Biotechnology of Sancti Spíritus, Circunvalante Norte, Olivos III, Sancti Spíritus, Cuba
| |
Collapse
|
39
|
Kantarcioglu B, Iqbal O, Lewis J, Carter CA, Singh M, Lievano F, Ligocki M, Jeske W, Adiguzel C, Gerotziafas GT, Fareed J. An Update on the Status of Vaccine Development for SARS-CoV-2 Including Variants. Practical Considerations for COVID-19 Special Populations. Clin Appl Thromb Hemost 2022; 28:10760296211056648. [PMID: 35167393 PMCID: PMC8851053 DOI: 10.1177/10760296211056648] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/25/2021] [Accepted: 10/13/2021] [Indexed: 01/09/2023] Open
Abstract
The progress in the development of various vaccine platforms against SARS-CoV-2 have been rather remarkable owing to advancement in molecular and biologic sciences. Most of the current vaccines and those in development focus on targeting the viral spike proteins by generating antibodies of varying spectrum. These vaccines represent a variety of platforms including whole virus vaccines, viral vector vaccines, nucleic acid vaccines representing RNA, DNA, and their hybrid forms.The therapeutic efficacy of these vaccines varies owing to their pharmacodynamic individualities. COVID-19 variants are capable of inducing different pathologic responses and some of which may be resistant to antibodies generated by current vaccines. The current clinical use of these vaccines has been through emergency use authorization until recently. Moreover, the efficacy and safety of these vaccines have been tested in substantial numbers of individuals but studies in special populations that better reflect the global population are pending results. These specialized populations include young children, immunocompromised patients, pregnant individuals, and other specialized groups. Combination approaches, molecularly modified vaccination approaches, and vaccines conferring longer periods of immunity are being currently being investigated, as well as pharmacovigilance studies.The continual transformation of SARS-CoV-2 and its variants are of concern along with the breakthrough infections. These considerations pose new challenges for the development of vaccination platforms. For this purpose, booster doses, combination vaccine approaches, and other modalities are being discussed. This review provides an updated account of currently available vaccines and those in advanced development with reference to their composition and mechanisms of action.A discussion on the use of vaccines in special populations including immunocompromised patients, pregnant women and other specialized populations are also included.
Collapse
Affiliation(s)
- Bulent Kantarcioglu
- Cardiovascular Research Institute, Loyola University Chicago, Health Sciences Division, Maywood, IL, USA
| | - Omer Iqbal
- Cardiovascular Research Institute, Loyola University Chicago, Health Sciences Division, Maywood, IL, USA
| | - Joseph Lewis
- Cardiovascular Research Institute, Loyola University Chicago, Health Sciences Division, Maywood, IL, USA
| | - Charles A. Carter
- Campbell University College of Pharmacy and Health Sciences, Campbell University, Buies Creek, NC, USA
| | - Meharvan Singh
- Loyola University Chicago Stritch School of Medicine, Maywood, IL, USA
| | | | | | - Walter Jeske
- Cardiovascular Research Institute, Loyola University Chicago, Health Sciences Division, Maywood, IL, USA
| | | | - Grigoris T. Gerotziafas
- Sorbonne Université, Assistance Publique-Hôpitaux de Paris, Thrombosis Center, Service D’Hématologie Biologique Hôpital Tenon, Paris, France
| | - Jawed Fareed
- Cardiovascular Research Institute, Loyola University Chicago, Health Sciences Division, Maywood, IL, USA
| |
Collapse
|
40
|
Mekonnen D, Mengist HM, Jin T. SARS-CoV-2 subunit vaccine adjuvants and their signaling pathways. Expert Rev Vaccines 2022; 21:69-81. [PMID: 34633259 PMCID: PMC8567292 DOI: 10.1080/14760584.2021.1991794] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 10/07/2021] [Indexed: 12/23/2022]
Abstract
INTRODUCTION Vaccines are the agreed upon weapon against the COVID-19 pandemic. This review discusses about COVID-19 subunit vaccines adjuvants and their signaling pathways, which could provide a glimpse into the selection of appropriate adjuvants for prospective vaccine development studies. AREAS COVERED In the introduction, a brief background about the SARS-CoV-2 pandemic, the vaccine development race and classes of vaccine adjuvants were provided. . The antigen, trial stage, and types of adjuvants were extracted from the included articles and thun assimilated. Finally, the pattern recognition receptors (PRRs), their classes, cognate adjuvants, and potential signaling pathways were comprehended. EXPERT OPINION Adjuvants are unsung heroes of subunit vaccines. The in silico studies are very vital in avoiding several costly trial errors and save much work times. The majority of the (pre)clinical studies are promising. It is encouraging that most of the selected adjuvants are novel. Much emphasis must be paid to the optimal paring of antigen-adjuvant-PRRs for obtaining the desired vaccine effect. A good subunit vaccine/adjuvant is one that has high efficacy, safety, dose sparing, and rapid seroconversion rate and broad spectrum of immune response. In the years to come, COVID-19 adjuvanted subunit vaccines are expected to have superior utility than any other vaccines for various reasons.
Collapse
Affiliation(s)
- Daniel Mekonnen
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Hylemariam Mihiretie Mengist
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, Cas Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Department of Medical Laboratory Science, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Tengchuan Jin
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, Cas Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
41
|
Rezaei A, Nazarian S, Samiei Abianeh H, Kordbacheh E, Alizadeh Z, Mousavi Gargari SL. Antibodies Produced Toward Recombinant RBD and Nucleocapsid Neutralize SARS-COV-2. Avicenna J Med Biotechnol 2022; 14:270-277. [PMID: 36504571 PMCID: PMC9706246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 05/30/2022] [Indexed: 12/15/2022] Open
Abstract
Background The highly contagious SARS-COV-2 virus spread rapidly from China and formed a global pandemic. The virus has infected over 509 million people worldwide and killed about 6.32 million up to date. Up on invasion, the Receptor Binding Domain (RBD) of Spike protein plays a crucial role in the entry of the virus into the host cell. The virus N protein is another protein that has a critical role for genome packaging. Methods As bioinformatics approaches, the cassette design, codon adaptation, and protein stability were investigated in this study. Synthetic genes of RBD and N were cloned separately in pET28a + expression vector. They were transferred into Escherichia coli (E. coli) BL21 DE3 host cell, and expression of recombinant proteins was induced with IPTG. The recombinant proteins were purified by column chromatography and approved by Western blotting. Animal immunization was performed with each of the recombinant proteins individually and in combination of the two. The antibody titer of the blood serum from control and immunized mice groups was determined by ELISA technique. Finally, the anti-spike neutralization test was performed. Results The expression and purification of RBD protein were monitored on SDS-PAGE, two bands of about 28 and 45 kDa for RBD and N appeared on gel distinctly, which were further validated by Western blotting. According to ELISA results, related antibodies were traced to a dilution of 1/64000 in immunized sera. The neutralization test exhibited produced antibodies' potency to bind the virus proteins. Using SPSS software, statistical analysis was performed by Duncan's test and T-test. Conclusion According to the present study, recombinant proteins, either RBD alone or in combination with N adequately stimulated the immune response, and the raised antibodies could neutralize the virus in in vitro test.
Collapse
Affiliation(s)
- Amir Rezaei
- Department of Biology, Shahed University, Tehran, Iran
| | - Shahram Nazarian
- Molecular Biotechnology Research Center and Department of Biology, Imam Hussein University, Tehran, Iran,Corresponding authors: Shahram Nazarian, Ph.D., Molecular Biotechnology Research Center and Department of Biology, Imam Hussein University, Tehran, Iran; Seyed Latif Mousavi Gargari, Ph.D., Department of Biology, Shahed University, Tehran, Iran, Tel: +98 21 51212232, Fax: +98 21 51212232, E-mail:,
| | | | - Emad Kordbacheh
- Department of Biology, Imam Hussein University, Tehran, Iran
| | | | | |
Collapse
|
42
|
Gartlan C, Tipton T, Salguero FJ, Sattentau Q, Gorringe A, Carroll MW. Vaccine-Associated Enhanced Disease and Pathogenic Human Coronaviruses. Front Immunol 2022; 13:882972. [PMID: 35444667 PMCID: PMC9014240 DOI: 10.3389/fimmu.2022.882972] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 03/14/2022] [Indexed: 01/14/2023] Open
Abstract
Vaccine-associated enhanced disease (VAED) is a difficult phenomenon to define and can be confused with vaccine failure. Using studies on respiratory syncytial virus (RSV) vaccination and dengue virus infection, we highlight known and theoretical mechanisms of VAED, including antibody-dependent enhancement (ADE), antibody-enhanced disease (AED) and Th2-mediated pathology. We also critically review the literature surrounding this phenomenon in pathogenic human coronaviruses, including MERS-CoV, SARS-CoV-1 and SARS-CoV-2. Poor quality histopathological data and a lack of consistency in defining severe pathology and VAED in preclinical studies of MERS-CoV and SARS-CoV-1 vaccines in particular make it difficult to interrogate potential cases of VAED. Fortuitously, there have been only few reports of mild VAED in SARS-CoV-2 vaccination in preclinical models and no observations in their clinical use. We describe the problem areas and discuss methods to improve the characterisation of VAED in the future.
Collapse
Affiliation(s)
- Cillian Gartlan
- Wellcome Centre for Human Genetics and Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Tom Tipton
- Wellcome Centre for Human Genetics and Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Francisco J Salguero
- Research and Evaluation, UK Health Security Agency, Porton Down, Salisbury, United Kingdom
| | - Quentin Sattentau
- The Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Andrew Gorringe
- Research and Evaluation, UK Health Security Agency, Porton Down, Salisbury, United Kingdom
| | - Miles W Carroll
- Wellcome Centre for Human Genetics and Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
43
|
Willcox AC, Sung K, Garrett ME, Galloway JG, O’Connor MA, Erasmus JH, Logue JK, Hawman DW, Chu HY, Hasenkrug KJ, Fuller DH, Matsen FA, Overbaugh J. Macaque-human differences in SARS-CoV-2 Spike antibody response elicited by vaccination or infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021:2021.12.01.470697. [PMID: 34909774 PMCID: PMC8669841 DOI: 10.1101/2021.12.01.470697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Macaques are a commonly used model for studying immunity to human viruses, including for studies of SARS-CoV-2 infection and vaccination. However, it is unknown whether macaque antibody responses recapitulate, and thus appropriately model, the response in humans. To answer this question, we employed a phage-based deep mutational scanning approach (Phage-DMS) to compare which linear epitopes are targeted on the SARS-CoV-2 Spike protein in humans and macaques following either vaccination or infection. We also used Phage-DMS to determine antibody escape pathways within each epitope, enabling a granular comparison of antibody binding specificities at the locus level. Overall, we identified some common epitope targets in both macaques and humans, including in the fusion peptide (FP) and stem helix-heptad repeat 2 (SH-H) regions. Differences between groups included a response to epitopes in the N-terminal domain (NTD) and C-terminal domain (CTD) in vaccinated humans but not vaccinated macaques, as well as recognition of a CTD epitope and epitopes flanking the FP in convalescent macaques but not convalescent humans. There was also considerable variability in the escape pathways among individuals within each group. Sera from convalescent macaques showed the least variability in escape overall and converged on a common response with vaccinated humans in the SH-H epitope region, suggesting highly similar antibodies were elicited. Collectively, these findings suggest that the antibody response to SARS-CoV-2 in macaques shares many features with humans, but with substantial differences in the recognition of certain epitopes and considerable individual variability in antibody escape profiles, suggesting a diverse repertoire of antibodies that can respond to major epitopes in both humans and macaques.
Collapse
Affiliation(s)
- Alexandra C. Willcox
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Medical Scientist Training Program, University of Washington, Seattle, WA, USA
- Molecular and Cellular Biology Program, University of Washington, Seattle, WA, USA
| | - Kevin Sung
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Meghan E. Garrett
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Molecular and Cellular Biology Program, University of Washington, Seattle, WA, USA
| | - Jared G. Galloway
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Megan A. O’Connor
- Department of Microbiology, University of Washington, Seattle, WA, USA
- Infectious Diseases and Translational Medicine, Washington National Primate Research Center, Seattle, WA, USA
| | - Jesse H. Erasmus
- Department of Microbiology, University of Washington, Seattle, WA, USA
- HDT Bio, Seattle, WA, USA
| | | | - David W. Hawman
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Helen Y. Chu
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Kim J. Hasenkrug
- Laboratory of Persistent Viral Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Deborah H. Fuller
- Department of Microbiology, University of Washington, Seattle, WA, USA
- Infectious Diseases and Translational Medicine, Washington National Primate Research Center, Seattle, WA, USA
- Center for Innate Immunity and Immune Disease, University of Washington, Seattle, WA, USA
| | - Frederick A. Matsen
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Julie Overbaugh
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| |
Collapse
|
44
|
Fulford TS, Van H, Gherardin NA, Zheng S, Ciula M, Drummer HE, Redmond S, Tan HX, Boo I, Center RJ, Li F, Grimley SL, Wines BD, Nguyen THO, Mordant FL, Ellenberg P, Rowntree LC, Kedzierski L, Cheng AC, Doolan DL, Matthews G, Bond K, Hogarth PM, McQuilten Z, Subbarao K, Kedzierska K, Juno JA, Wheatley AK, Kent SJ, Williamson DA, Purcell DFJ, Anderson DA, Godfrey DI. A point-of-care lateral flow assay for neutralising antibodies against SARS-CoV-2. EBioMedicine 2021; 74:103729. [PMID: 34871960 PMCID: PMC8641961 DOI: 10.1016/j.ebiom.2021.103729] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 11/12/2021] [Accepted: 11/18/2021] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND As vaccines against SARS-CoV-2 are now being rolled out, a better understanding of immunity to the virus, whether from infection, or passive or active immunisation, and the durability of this protection is required. This will benefit from the ability to measure antibody-based protection to SARS-CoV-2, ideally with rapid turnaround and without the need for laboratory-based testing. METHODS We have developed a lateral flow POC test that can measure levels of RBD-ACE2 neutralising antibody (NAb) from whole blood, with a result that can be determined by eye or quantitatively on a small instrument. We compared our lateral flow test with the gold-standard microneutralisation assay, using samples from convalescent and vaccinated donors, as well as immunised macaques. FINDINGS We show a high correlation between our lateral flow test with conventional neutralisation and that this test is applicable with animal samples. We also show that this assay is readily adaptable to test for protection to newly emerging SARS-CoV-2 variants, including the beta variant which revealed a marked reduction in NAb activity. Lastly, using a cohort of vaccinated humans, we demonstrate that our whole-blood test correlates closely with microneutralisation assay data (specificity 100% and sensitivity 96% at a microneutralisation cutoff of 1:40) and that fingerprick whole blood samples are sufficient for this test. INTERPRETATION Taken together, the COVID-19 NAb-testTM device described here provides a rapid readout of NAb based protection to SARS-CoV-2 at the point of care. FUNDING Support was received from the Victorian Operational Infrastructure Support Program and the Australian Government Department of Health. This work was supported by grants from the Department of Health and Human Services of the Victorian State Government; the ARC (CE140100011, CE140100036), the NHMRC (1113293, 2002317 and 1116530), and Medical Research Future Fund Awards (2005544, 2002073, 2002132). Individual researchers were supported by an NHMRC Emerging Leadership Level 1 Investigator Grants (1194036), NHMRC APPRISE Research Fellowship (1116530), NHMRC Leadership Investigator Grant (1173871), NHMRC Principal Research Fellowship (1137285), NHMRC Investigator Grants (1177174 and 1174555) and NHMRC Senior Principal Research Fellowships (1117766 and 1136322). Grateful support was also received from the A2 Milk Company and the Jack Ma Foundation.
Collapse
Affiliation(s)
- Thomas S Fulford
- Department of Microbiology and Immunology, University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Huy Van
- Burnet Institute, Melbourne, Victoria, Australia
| | - Nicholas A Gherardin
- Department of Microbiology and Immunology, University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia; Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Melbourne, Victoria, Australia
| | | | - Marcin Ciula
- Department of Microbiology and Immunology, University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Heidi E Drummer
- Department of Microbiology and Immunology, University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia; Burnet Institute, Melbourne, Victoria, Australia; Department of Microbiology, Monash University, Australia
| | - Samuel Redmond
- Department of Microbiology and Immunology, University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Hyon-Xhi Tan
- Department of Microbiology and Immunology, University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Irene Boo
- Burnet Institute, Melbourne, Victoria, Australia
| | - Rob J Center
- Department of Microbiology and Immunology, University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia; Burnet Institute, Melbourne, Victoria, Australia
| | - Fan Li
- Burnet Institute, Melbourne, Victoria, Australia
| | - Samantha L Grimley
- Department of Microbiology and Immunology, University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Bruce D Wines
- Immune therapies Laboratory, Burnet Institute, Melbourne, VIC, Australia,; Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC, Australia; Department of Clinical Pathology, The University of Melbourne, Parkville, VIC, Australia
| | - Thi H O Nguyen
- Department of Microbiology and Immunology, University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Francesca L Mordant
- Department of Microbiology and Immunology, University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Paula Ellenberg
- Department of Microbiology and Immunology, University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Louise C Rowntree
- Department of Microbiology and Immunology, University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Lukasz Kedzierski
- Department of Microbiology and Immunology, University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Allen C Cheng
- Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Denise L Doolan
- Australian Institute of Tropical Health & Medicine, James Cook University, Cairns, Queensland, Australia
| | - Gail Matthews
- Kirby Institute, University of NSW, Sydney, NSW, Australia
| | - Katherine Bond
- Department of Microbiology and Immunology, University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia; Department of Microbiology, Royal Melbourne Hospital, Melbourne, Australia
| | - P Mark Hogarth
- Immune therapies Laboratory, Burnet Institute, Melbourne, VIC, Australia,; Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC, Australia; Department of Clinical Pathology, The University of Melbourne, Parkville, VIC, Australia
| | - Zoe McQuilten
- Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Kanta Subbarao
- Department of Microbiology and Immunology, University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia; WHO Collaborating Centre for Reference and Research on Influenza at the Peter Doherty Institute for Infection and Immunity
| | - Katherine Kedzierska
- Department of Microbiology and Immunology, University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Jennifer A Juno
- Department of Microbiology and Immunology, University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Adam K Wheatley
- Department of Microbiology and Immunology, University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia; Melbourne Sexual Health Centre and Department of Infectious Diseases, Alfred Hospital and Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Stephen J Kent
- Department of Microbiology and Immunology, University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia; Melbourne Sexual Health Centre and Department of Infectious Diseases, Alfred Hospital and Central Clinical School, Monash University, Melbourne, Victoria, Australia; Australian Research Council Centre for Excellence in Convergent Bio-Nano Science and Technology, University of Melbourne, Melbourne, Victoria, Australia
| | - Deborah A Williamson
- Department of Microbiology and Immunology, University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia; Department of Microbiology, Royal Melbourne Hospital, Melbourne, Australia
| | - Damian F J Purcell
- Department of Microbiology and Immunology, University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | | | - Dale I Godfrey
- Department of Microbiology and Immunology, University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia; Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Melbourne, Victoria, Australia.
| |
Collapse
|
45
|
Zhou Y, Wu Y, Ding L, Huang X, Xiong Y. Point-of-care COVID-19 diagnostics powered by lateral flow assay. Trends Analyt Chem 2021; 145:116452. [PMID: 34629572 PMCID: PMC8487324 DOI: 10.1016/j.trac.2021.116452] [Citation(s) in RCA: 101] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Since its first discovery in December 2019, the global coronavirus disease 2019 (COVID-19) pandemic caused by the novel coronavirus (SARS-CoV-2) has been posing a serious threat to human life and health. Diagnostic testing is critical for the control and management of the COVID-19 pandemic. In particular, diagnostic testing at the point of care (POC) has been widely accepted as part of the post restriction COVID-19 control strategy. Lateral flow assay (LFA) is a popular POC diagnostic platform that plays an important role in controlling the COVID-19 pandemic in industrialized countries and resource-limited settings. Numerous pioneering studies on the design and development of diverse LFA-based diagnostic technologies for the rapid diagnosis of COVID-19 have been done and reported by researchers. Hundreds of LFA-based diagnostic prototypes have sprung up, some of which have been developed into commercial test kits for the rapid diagnosis of COVID-19. In this review, we summarize the crucial role of rapid diagnostic tests using LFA in targeting SARS-CoV-2-specific RNA, antibodies, antigens, and whole virus. Then, we discuss the design principle and working mechanisms of these available LFA methods, emphasizing their clinical diagnostic efficiency. Ultimately, we elaborate the challenges of current LFA diagnostics for COVID-19 and highlight the need for continuous improvement in rapid diagnostic tests.
Collapse
Affiliation(s)
- Yaofeng Zhou
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, 330047, PR China
- School of Food Science and Technology, Nanchang University, Nanchang, 330047, PR China
| | - Yuhao Wu
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, 330047, PR China
- School of Food Science and Technology, Nanchang University, Nanchang, 330047, PR China
| | - Lu Ding
- Hypertension Research Institute of Jiangxi Province, Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, PR China
| | - Xiaolin Huang
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, 330047, PR China
- School of Food Science and Technology, Nanchang University, Nanchang, 330047, PR China
| | - Yonghua Xiong
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, 330047, PR China
- School of Food Science and Technology, Nanchang University, Nanchang, 330047, PR China
| |
Collapse
|
46
|
Juno JA, Hill DL. T follicular helper cells and their impact on humoral responses during pathogen and vaccine challenge. Curr Opin Immunol 2021; 74:112-117. [PMID: 34861545 DOI: 10.1016/j.coi.2021.11.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 11/05/2021] [Accepted: 11/08/2021] [Indexed: 12/23/2022]
Abstract
T follicular helper (Tfh) cells are essential for the establishment, maintenance and output of the germinal centre (GC) response. The transient nature of this response, and its location within secondary lymphoid tissues have hampered our understanding of this critical cell type, particularly in humans. A counterpart of GC Tfh cells in peripheral blood has enabled recent discoveries in disease and vaccination settings, while direct sampling of lymph nodes provides exciting new avenues to study GC responses directly in vivo. Tfh differentiation is shaped by the cytokine milieu during inflammation, vaccination and with age, and disease-specific patterns are emerging. An improved understanding of how to support a Tfh response remains key to enhancing vaccine immunity across the lifespan.
Collapse
Affiliation(s)
- Jennifer A Juno
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, 792 Elizabeth St., Melbourne 3000, Victoria, Australia.
| | - Danika L Hill
- Department of Immunology and Pathology, Monash University, 89 Commercial Rd., Melbourne 3004, Victoria, Australia.
| |
Collapse
|
47
|
Liu H, Zhou C, An J, Song Y, Yu P, Li J, Gu C, Hu D, Jiang Y, Zhang L, Huang C, Zhang C, Yang Y, Zhu Q, Wang D, Liu Y, Miao C, Cao X, Ding L, Zhu Y, Zhu H, Bao L, Zhou L, Yan H, Fan J, Xu J, Hu Z, Xie Y, Liu J, Liu G. Development of recombinant COVID-19 vaccine based on CHO-produced, prefusion spike trimer and alum/CpG adjuvants. Vaccine 2021; 39:7001-7011. [PMID: 34750014 PMCID: PMC8556577 DOI: 10.1016/j.vaccine.2021.10.066] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 10/22/2021] [Accepted: 10/25/2021] [Indexed: 12/23/2022]
Abstract
COVID-19 pandemic has severely impacted the public health and social economy worldwide. A safe, effective, and affordable vaccine against SARS-CoV-2 infections/diseases is urgently needed. We have been developing a recombinant vaccine based on a prefusion-stabilized spike trimer of SARS-CoV-2 and formulated with aluminium hydroxide and CpG 7909. The spike protein was expressed in Chinese hamster ovary (CHO) cells, purified, and prepared as a stable formulation with the dual adjuvant. Immunogenicity studies showed that candidate vaccines elicited robust neutralizing antibody responses and substantial CD4+ T cell responses in both mice and non-human primates. And vaccine-induced neutralizing antibodies persisted at high level for at least 6 months. Challenge studies demonstrated that candidate vaccine reduced the viral loads and inflammation in the lungs of SARS-CoV-2 infected golden Syrian hamsters significantly. In addition, the vaccine-induced antibodies showed cross-neutralization activity against B.1.1.7 and B.1.351 variants. These data suggest candidate vaccine is efficacious in preventing SARS-CoV-2 infections and associated pneumonia, thereby justifying ongoing phase I/II clinical studies in China (NCT04982068 and NCT04990544).
Collapse
Affiliation(s)
- Haitao Liu
- Shanghai Zerun Biotechnology Co., Ltd., Shanghai, China
| | | | - Jiao An
- Shanghai Zerun Biotechnology Co., Ltd., Shanghai, China
| | - Yujiao Song
- Shanghai Zerun Biotechnology Co., Ltd., Shanghai, China
| | - Pin Yu
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China
| | - Jiadai Li
- Shanghai Zerun Biotechnology Co., Ltd., Shanghai, China
| | - Chenjian Gu
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Dongdong Hu
- Shanghai Zerun Biotechnology Co., Ltd., Shanghai, China
| | | | - Lingli Zhang
- Shanghai Zerun Biotechnology Co., Ltd., Shanghai, China
| | - Chuanqi Huang
- Shanghai Zerun Biotechnology Co., Ltd., Shanghai, China
| | - Chao Zhang
- Shanghai Zerun Biotechnology Co., Ltd., Shanghai, China
| | - Yunqi Yang
- Shanghai Zerun Biotechnology Co., Ltd., Shanghai, China
| | - Qianjun Zhu
- Shanghai Zerun Biotechnology Co., Ltd., Shanghai, China
| | - Dekui Wang
- Shanghai Zerun Biotechnology Co., Ltd., Shanghai, China
| | - Yuqiang Liu
- Shanghai Zerun Biotechnology Co., Ltd., Shanghai, China
| | - Chenyang Miao
- Shanghai Zerun Biotechnology Co., Ltd., Shanghai, China
| | - Xiayao Cao
- Shanghai Zerun Biotechnology Co., Ltd., Shanghai, China
| | - Longfei Ding
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| | - Yuanfei Zhu
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hua Zhu
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China
| | - Linlin Bao
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China
| | - Lingyun Zhou
- Shanghai Zerun Biotechnology Co., Ltd., Shanghai, China
| | - Huan Yan
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Jiang Fan
- Shanghai Zerun Biotechnology Co., Ltd., Shanghai, China
| | - Jianqing Xu
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| | - Zhongyu Hu
- National Institute for Food and Drug Control (NIFDC), Beijing, China.
| | - Youhua Xie
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China.
| | - Jiangning Liu
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China.
| | - Ge Liu
- Shanghai Zerun Biotechnology Co., Ltd., Shanghai, China.
| |
Collapse
|
48
|
Langellotto F, Dellacherie MO, Yeager C, Ijaz H, Yu J, Cheng C, Dimitrakakis N, Seiler BT, Gebre MS, Gilboa T, Johnson R, Storm N, Bardales S, Graveline A, White D, Tringides CM, Cartwright MJ, Doherty EJ, Honko A, Griffiths A, Barouch DH, Walt DR, Mooney DJ. A Modular Biomaterial Scaffold-Based Vaccine Elicits Durable Adaptive Immunity to Subunit SARS-CoV-2 Antigens. Adv Healthc Mater 2021; 10:e2101370. [PMID: 34605223 PMCID: PMC8652677 DOI: 10.1002/adhm.202101370] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 09/07/2021] [Indexed: 12/14/2022]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic demonstrates the importance of generating safe and efficacious vaccines that can be rapidly deployed against emerging pathogens. Subunit vaccines are considered among the safest, but proteins used in these typically lack strong immunogenicity, leading to poor immune responses. Here, a biomaterial COVID-19 vaccine based on a mesoporous silica rods (MSRs) platform is described. MSRs loaded with granulocyte-macrophage colony-stimulating factor (GM-CSF), the toll-like receptor 4 (TLR-4) agonist monophosphoryl lipid A (MPLA), and SARS-CoV-2 viral protein antigens slowly release their cargo and form subcutaneous scaffolds that locally recruit and activate antigen-presenting cells (APCs) for the generation of adaptive immunity. MSR-based vaccines generate robust and durable cellular and humoral responses against SARS-CoV-2 antigens, including the poorly immunogenic receptor binding domain (RBD) of the spike (S) protein. Persistent antibodies over the course of 8 months are found in all vaccine configurations tested and robust in vitro viral neutralization is observed both in a prime-boost and a single-dose regimen. These vaccines can be fully formulated ahead of time or stored lyophilized and reconstituted with an antigen mixture moments before injection, which can facilitate its rapid deployment against emerging SARS-CoV-2 variants or new pathogens. Together, the data show a promising COVID-19 vaccine candidate and a generally adaptable vaccine platform against infectious pathogens.
Collapse
Affiliation(s)
- Fernanda Langellotto
- Wyss Institute for Biologically Inspired EngineeringHarvard UniversityBostonMA02115USA
| | - Maxence O. Dellacherie
- Wyss Institute for Biologically Inspired EngineeringHarvard UniversityBostonMA02115USA
- Harvard John A. Paulson School of Engineering and Applied SciencesHarvard UniversityCambridgeMA02138USA
| | - Chyenne Yeager
- Wyss Institute for Biologically Inspired EngineeringHarvard UniversityBostonMA02115USA
| | - Hamza Ijaz
- Wyss Institute for Biologically Inspired EngineeringHarvard UniversityBostonMA02115USA
| | - Jingyou Yu
- Center for Virology and Vaccine ResearchBeth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMA02115USA
| | - Chi‐An Cheng
- Wyss Institute for Biologically Inspired EngineeringHarvard UniversityBostonMA02115USA
- Department of PathologyBrigham and Women's HospitalBostonMA02115USA
- Harvard Medical SchoolBostonMA02115USA
| | - Nikolaos Dimitrakakis
- Wyss Institute for Biologically Inspired EngineeringHarvard UniversityBostonMA02115USA
| | - Benjamin T. Seiler
- Wyss Institute for Biologically Inspired EngineeringHarvard UniversityBostonMA02115USA
| | - Makda S. Gebre
- Center for Virology and Vaccine ResearchBeth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMA02115USA
| | - Tal Gilboa
- Wyss Institute for Biologically Inspired EngineeringHarvard UniversityBostonMA02115USA
- Department of PathologyBrigham and Women's HospitalBostonMA02115USA
- Harvard Medical SchoolBostonMA02115USA
| | - Rebecca Johnson
- Department of MicrobiologyBoston University School of Medicine and National Emerging Infectious Diseases LaboratoriesBostonMA02118USA
| | - Nadia Storm
- Department of MicrobiologyBoston University School of Medicine and National Emerging Infectious Diseases LaboratoriesBostonMA02118USA
| | - Sarai Bardales
- Wyss Institute for Biologically Inspired EngineeringHarvard UniversityBostonMA02115USA
| | - Amanda Graveline
- Wyss Institute for Biologically Inspired EngineeringHarvard UniversityBostonMA02115USA
| | - Des White
- Wyss Institute for Biologically Inspired EngineeringHarvard UniversityBostonMA02115USA
| | - Christina M. Tringides
- Wyss Institute for Biologically Inspired EngineeringHarvard UniversityBostonMA02115USA
- Harvard Program in BiophysicsHarvard UniversityCambridgeMA02138USA
- Harvard–MIT Division in Health Sciences and TechnologyMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Mark J. Cartwright
- Wyss Institute for Biologically Inspired EngineeringHarvard UniversityBostonMA02115USA
| | - Edward J. Doherty
- Wyss Institute for Biologically Inspired EngineeringHarvard UniversityBostonMA02115USA
| | - Anna Honko
- Department of MicrobiologyBoston University School of Medicine and National Emerging Infectious Diseases LaboratoriesBostonMA02118USA
| | - Anthony Griffiths
- Department of MicrobiologyBoston University School of Medicine and National Emerging Infectious Diseases LaboratoriesBostonMA02118USA
| | - Dan H. Barouch
- Center for Virology and Vaccine ResearchBeth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMA02115USA
- Ragon Institute of MGHMIT, and HarvardCambridgeMA02139USA
- Massachusetts Consortium on Pathogen ReadinessBostonMA02215USA
| | - David R. Walt
- Wyss Institute for Biologically Inspired EngineeringHarvard UniversityBostonMA02115USA
- Department of PathologyBrigham and Women's HospitalBostonMA02115USA
- Harvard Medical SchoolBostonMA02115USA
| | - David J. Mooney
- Wyss Institute for Biologically Inspired EngineeringHarvard UniversityBostonMA02115USA
- Harvard John A. Paulson School of Engineering and Applied SciencesHarvard UniversityCambridgeMA02138USA
| |
Collapse
|
49
|
Schardt JS, Pornnoppadol G, Desai AA, Park KS, Zupancic JM, Makowski EK, Smith MD, Chen H, Garcia de Mattos Barbosa M, Cascalho M, Lanigan TM, Moon JJ, Tessier PM. Discovery and characterization of high-affinity, potent SARS-CoV-2 neutralizing antibodies via single B cell screening. Sci Rep 2021; 11:20738. [PMID: 34671080 PMCID: PMC8528929 DOI: 10.1038/s41598-021-99401-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 09/22/2021] [Indexed: 12/17/2022] Open
Abstract
Monoclonal antibodies that target SARS-CoV-2 with high affinity are valuable for a wide range of biomedical applications involving novel coronavirus disease (COVID-19) diagnosis, treatment, and prophylactic intervention. Strategies for the rapid and reliable isolation of these antibodies, especially potent neutralizing antibodies, are critical toward improved COVID-19 response and informed future response to emergent infectious diseases. In this study, single B cell screening was used to interrogate antibody repertoires of immunized mice and isolate antigen-specific IgG1+ memory B cells. Using these methods, high-affinity, potent neutralizing antibodies were identified that target the receptor-binding domain of SARS-CoV-2. Further engineering of the identified molecules to increase valency resulted in enhanced neutralizing activity. Mechanistic investigation revealed that these antibodies compete with ACE2 for binding to the receptor-binding domain of SARS-CoV-2. These antibodies may warrant further development for urgent COVID-19 applications. Overall, these results highlight the potential of single B cell screening for the rapid and reliable identification of high-affinity, potent neutralizing antibodies for infectious disease applications.
Collapse
Affiliation(s)
- John S. Schardt
- grid.214458.e0000000086837370Departments of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109 USA ,grid.214458.e0000000086837370Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109 USA ,grid.214458.e0000000086837370Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109 USA
| | - Ghasidit Pornnoppadol
- grid.214458.e0000000086837370Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109 USA ,grid.214458.e0000000086837370Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109 USA
| | - Alec A. Desai
- grid.214458.e0000000086837370Departments of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109 USA ,grid.214458.e0000000086837370Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109 USA
| | - Kyung Soo Park
- grid.214458.e0000000086837370Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109 USA ,grid.214458.e0000000086837370Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109 USA
| | - Jennifer M. Zupancic
- grid.214458.e0000000086837370Departments of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109 USA ,grid.214458.e0000000086837370Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109 USA
| | - Emily K. Makowski
- grid.214458.e0000000086837370Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109 USA ,grid.214458.e0000000086837370Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109 USA
| | - Matthew D. Smith
- grid.214458.e0000000086837370Departments of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109 USA ,grid.214458.e0000000086837370Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109 USA
| | - Hongwei Chen
- grid.214458.e0000000086837370Departments of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109 USA ,grid.214458.e0000000086837370Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109 USA ,grid.214458.e0000000086837370Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109 USA
| | | | - Marilia Cascalho
- grid.214458.e0000000086837370Department of Surgery, University of Michigan, Ann Arbor, MI 48109 USA ,grid.214458.e0000000086837370Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI 48109 USA
| | - Thomas M. Lanigan
- grid.214458.e0000000086837370Division of Rheumatology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI USA
| | - James J. Moon
- grid.214458.e0000000086837370Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109 USA ,grid.214458.e0000000086837370Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109 USA ,grid.214458.e0000000086837370Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109 USA
| | - Peter M. Tessier
- grid.214458.e0000000086837370Departments of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109 USA ,grid.214458.e0000000086837370Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109 USA ,grid.214458.e0000000086837370Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109 USA ,grid.214458.e0000000086837370Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109 USA ,grid.214458.e0000000086837370University of Michigan, North Campus Research Complex, B10-179, 2800 Plymouth Road, Ann Arbor, MI 48109 USA
| |
Collapse
|
50
|
Cui D, Tang Y, Jiang Q, Jiang D, Zhang Y, Lv Y, Xu D, Wu J, Xie J, Wen C, Lu L. Follicular Helper T Cells in the Immunopathogenesis of SARS-CoV-2 Infection. Front Immunol 2021; 12:731100. [PMID: 34603308 PMCID: PMC8481693 DOI: 10.3389/fimmu.2021.731100] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Accepted: 09/01/2021] [Indexed: 12/21/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19), caused by the novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is a serious infectious disease that has led to a global pandemic with high morbidity and mortality. High-affinity neutralizing antibody is important for controlling infection, which is closely regulated by follicular helper T (Tfh) cells. Tfh cells play a central role in promoting germinal center reactions and driving cognate B cell differentiation for antibody secretion. Available studies indicate a close relationship between virus-specific Tfh cell-mediated immunity and SARS-CoV-2 infection progression. Although several lines of evidence have suggested that Tfh cells contribute to the control of SARS-CoV-2 infection by eliciting neutralizing antibody productions, further studies are needed to elucidate Tfh-mediated effector mechanisms in anti-SARS-CoV-2 immunity. Here, we summarize the functional features and roles of virus-specific Tfh cells in the immunopathogenesis of SARS-CoV-2 infection and in COVID-19 vaccines, and highlight the potential of targeting Tfh cells as therapeutic strategy against SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Dawei Cui
- Department of Blood Transfusion, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yuan Tang
- Department of Pathology and Shenzhen Institute of Research and Innovation, The University of Hong Kong, Hong Kong, Hong Kong, SAR China.,Chongqing International Institute for Immunology, Chongqing, China
| | - Qi Jiang
- Department of Blood Transfusion, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, China
| | - Daixi Jiang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yun Zhang
- School of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yan Lv
- Department of Blood Transfusion, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Dandan Xu
- Department of Blood Transfusion, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jian Wu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jue Xie
- Department of Blood Transfusion, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chengping Wen
- School of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Liwei Lu
- Department of Pathology and Shenzhen Institute of Research and Innovation, The University of Hong Kong, Hong Kong, Hong Kong, SAR China.,Chongqing International Institute for Immunology, Chongqing, China
| |
Collapse
|