1
|
Marcos-Villar L, Perdiguero B, López-Bravo M, Zamora C, Sin L, Álvarez E, Sorzano CÓS, Sánchez-Cordón PJ, Casasnovas JM, Astorgano D, García-Arriaza J, Anthiya S, Borrajo ML, Lou G, Cuesta B, Franceschini L, Gelpí JL, Thielemans K, Sisteré-Oró M, Meyerhans A, García F, Esteban I, López-Bigas N, Plana M, Alonso MJ, Esteban M, Gómez CE. Heterologous mRNA/MVA delivering trimeric-RBD as effective vaccination regimen against SARS-CoV-2: COVARNA Consortium. Emerg Microbes Infect 2024; 13:2387906. [PMID: 39087555 PMCID: PMC11313003 DOI: 10.1080/22221751.2024.2387906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 07/26/2024] [Accepted: 07/30/2024] [Indexed: 08/02/2024]
Abstract
Despite the high efficiency of current SARS-CoV-2 mRNA vaccines in reducing COVID-19 morbidity and mortality, waning immunity and the emergence of resistant variants underscore the need for novel vaccination strategies. This study explores a heterologous mRNA/Modified Vaccinia virus Ankara (MVA) prime/boost regimen employing a trimeric form of the receptor binding domain (RBD) of the SARS-CoV-2 spike (S) protein compared to a homologous MVA/MVA regimen. In C57BL/6 mice, the RBD was delivered during priming via an mRNA vector encapsulated in nanoemulsions (NE) or lipid nanoparticles (LNP), followed by a booster with a replication-deficient MVA-based recombinant virus (MVA-RBD). This heterologous mRNA/MVA regimen elicited strong anti-RBD binding and neutralizing antibodies (BAbs and NAbs) against both the ancestral SARS-CoV-2 strain and different variants of concern (VoCs). Additionally, this protocol induced robust and polyfunctional RBD-specific CD4 and CD8 T cell responses, particularly in animals primed with mLNP-RBD. In K18-hACE2 transgenic mice, the LNP-RBD/MVA combination provided complete protection from morbidity and mortality following a live SARS-CoV-2 challenge compared with the partial protection observed with mNE-RBD/MVA or MVA/MVA regimens. Although the mNE-RBD/MVA regimen only protects half of the animals, it was able to induce antibodies with Fc-mediated effector functions besides NAbs. Moreover, viral replication and viral load in the respiratory tract were markedly reduced and decreased pro-inflammatory cytokine levels were observed. These results support the efficacy of heterologous mRNA/MVA vaccine combinations over homologous MVA/MVA regimen, using alternative nanocarriers that circumvent intellectual property restrictions of current mRNA vaccine formulations.
Collapse
MESH Headings
- Animals
- COVID-19 Vaccines/immunology
- COVID-19 Vaccines/administration & dosage
- SARS-CoV-2/immunology
- SARS-CoV-2/genetics
- Mice
- Spike Glycoprotein, Coronavirus/immunology
- Spike Glycoprotein, Coronavirus/genetics
- COVID-19/prevention & control
- COVID-19/immunology
- Antibodies, Viral/immunology
- Antibodies, Viral/blood
- Antibodies, Neutralizing/immunology
- Mice, Inbred C57BL
- Vaccinia virus/genetics
- Vaccinia virus/immunology
- Humans
- Female
- Nanoparticles/administration & dosage
- Vaccination
- mRNA Vaccines/administration & dosage
- Mice, Transgenic
- Vaccines, Synthetic/immunology
- Vaccines, Synthetic/administration & dosage
- Vaccines, Synthetic/genetics
- CD8-Positive T-Lymphocytes/immunology
- Angiotensin-Converting Enzyme 2/immunology
- Angiotensin-Converting Enzyme 2/genetics
- Liposomes
Collapse
Affiliation(s)
- Laura Marcos-Villar
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Beatriz Perdiguero
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | | | - Carmen Zamora
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Laura Sin
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Enrique Álvarez
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | | | - Pedro J. Sánchez-Cordón
- Veterinary Pathology Department, Centro de Investigación en Sanidad Animal (CISA), Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), CSIC, Madrid, Spain
| | | | - David Astorgano
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Juan García-Arriaza
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Shubaash Anthiya
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Mireya L. Borrajo
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Gustavo Lou
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Belén Cuesta
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Lorenzo Franceschini
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Josep L. Gelpí
- Barcelona Supercomputing Center (BSC), Barcelona, Spain
- Department of Biochemistry and Molecular Biomedicine, University of Barcelona (UB), Barcelona, Spain
| | - Kris Thielemans
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Marta Sisteré-Oró
- Infection Biology Laboratory, Department of Medicine and Life Sciences, University Pompeu Fabra, Barcelona, Spain
| | - Andreas Meyerhans
- Infection Biology Laboratory, Department of Medicine and Life Sciences, University Pompeu Fabra, Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - Felipe García
- Infectious Diseases Department, Hospital Clínic, UB,Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, UB, Barcelona, Spain
| | - Ignasi Esteban
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, UB, Barcelona, Spain
| | - Núria López-Bigas
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
- Institute for Research in Biomedicine (IRB), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), ISCIII, Madrid, Spain
| | - Montserrat Plana
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, UB, Barcelona, Spain
| | - María J. Alonso
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
- Instituto de Investigaciones Sanitarias (IDIS), Santiago de Compostela, Spain
| | - Mariano Esteban
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Carmen Elena Gómez
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| |
Collapse
|
2
|
Doroud D, Sadat Larijani M, Biglari A, Ashrafian F, Sabouni T, Eybpoosh S, Verez-Bencomo V, Valdés-Balbín Y, García-Rivera D, Herrera-Rojas Y, Climent-Ruiz Y, Santana-Mederos D, Ramezani A. Comparative assessment of a COVID-19 vaccine after technology transfer to Iran from critical quality attributes to clinical and immunogenicity aspects. Sci Rep 2024; 14:26793. [PMID: 39501012 PMCID: PMC11538526 DOI: 10.1038/s41598-024-77331-8] [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: 02/26/2024] [Accepted: 10/22/2024] [Indexed: 11/08/2024] Open
Abstract
During COVID-19 pandemic, international pharmaceutical companies put effort to build global manufacturing networks for vaccines. Soberana Plus vaccine, a recombinant protein based vaccine (RBD dimer), with the trade name of PastoCovac Plus in Iran, is based on a protein subunit platform in Cuba and completed preclinical and toxicological assessments. This study aimed at presenting the steps of vaccine technology transfer from Cuba to Iran. This study provides the first practical comparability results in Iran to ensure the quality, safety and efficacy of a protein subunit vaccine against COVID-19 after a successful technology transfer from Cuba. PastoCovac Plus was transferred to Iran at the formulation stage. The assessment of the active ingredient pharmaceutical (API) was achieved through physicochemical and clinical data collection and tests to assure if there was any adverse impact on the vaccination results. In order to assess the quality of the vaccine product after technology transfer, we sought different properties including regulatory features, physicochemical quality, vaccine potency and stability as well as its immunogenicity and safety. Following the evaluation of the clinical quality attributes (CQAs) based on the standard protocols, the results showed that the two vaccines are highly similar and comparable, with no considerable effect on safety or efficacy profiles. The CQAs were all in the acceptance limits in terms of safety and efficacy as well as clinical evaluation results. The immunogenicity evaluation also confirmed no significant differences between the vaccines regarding reinfection (P = 0.199) or vaccine breakthrough (P = 0.176). Furthermore, the level of anti-spike and neutralizing antibodies in the both vaccine groups was not significantly different indicating the equality of performance between the two vaccines. According to the results of the quality and clinical assessment of this study, we achieved an acceptable quality attributes and acceptant limits in terms of safety and efficacy of the vaccines pre and post technology transfer.
Collapse
Affiliation(s)
- Delaram Doroud
- Quality Control Department, Production and Research Complex, Pasteur Institute of Iran, Tehran, 1316943551, Iran
| | - Mona Sadat Larijani
- Clinical Research Department, Pasteur Institute of Iran, No 69, Pasteur Ave., Tehran, 13164, Iran
| | - Alireza Biglari
- School of Medicine, Tehran University of Medical Sciences, P.O. BOX 14155-6559, Tehran, Tehran, Iran.
| | - Fatemeh Ashrafian
- Clinical Research Department, Pasteur Institute of Iran, No 69, Pasteur Ave., Tehran, 13164, Iran
| | - Talieh Sabouni
- Quality Control Department, Production and Research Complex, Pasteur Institute of Iran, Tehran, 1316943551, Iran
| | - Sana Eybpoosh
- Department of Epidemiology and Biostatistics, Pasteur Institute of Iran, Research Centre for Emerging and Reemerging Infectious Diseases, Tehran, Iran
| | | | | | | | | | | | | | - Amitis Ramezani
- Clinical Research Department, Pasteur Institute of Iran, No 69, Pasteur Ave., Tehran, 13164, Iran.
| |
Collapse
|
3
|
Chiu CT, Tsai HH, Chen JY, Hu CMJ, Chen HW. An Immunoreceptor-Targeting Strategy with Minimalistic C3b Peptide Fusion Enhances SARS-CoV-2 RBD mRNA Vaccine Immunogenicity. Int J Nanomedicine 2024; 19:7201-7214. [PMID: 39050877 PMCID: PMC11268571 DOI: 10.2147/ijn.s463546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 07/11/2024] [Indexed: 07/27/2024] Open
Abstract
Introduction The clinical success of mRNA vaccine during the COVID-19 pandemic has inspired emerging approaches to elevate mRNA vaccine immunogenicity. Among them, antigen fusion protein designs for improved immune cell targeting have been shown to augment humoral immunity against small antigen targets. Methods This research demonstrates that SARS-CoV-2 receptor binding domain (RBD) fusion with a minimalistic peptide segment of complement component 3b (C3b, residues 727-767) ligand can improve mRNA vaccine immunogenicity through antigen targeting to complement receptor 1 (CR1). We affirm vaccines' antigenicity and targeting ability towards specific receptors through Western blot and immunofluorescence assay. Furthermore, mice immunization studies help the investigation of the antibody responses. Results Using SARS-CoV-2 Omicron RBD antigen, we compare mRNA vaccine formulations expressing RBD fusion protein with mouse C3b peptide (RBD-mC3), RBD fusion protein with mouse Fc (RBD-Fc), and wild-type RBD. Our results confirm the proper antigenicity and normal functionality of RBD-mC3. Upon validating comparable antigen expression by the different vaccine formulations, receptor-targeting capability of the fusion antigens is further confirmed. In mouse immunization studies, we show that while both RBD-mC3 and RBD-Fc elevate vaccine immunogenicity, RBD-mC3 leads to more sustained RBD-specific titers over the RBD-Fc design, presumably due to reduced antigenic diversion by the minimalistic targeting ligand. Conclusion The study demonstrates a novel C3b-based antigen design strategy for immune cell targeting and mRNA vaccine enhancement.
Collapse
Affiliation(s)
- Chun-Ta Chiu
- Department of Veterinary Medicine, National Taiwan University, Taipei, 10617, Taiwan
| | - Hsiao-Han Tsai
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 115, Taiwan
| | - Jing-Yuan Chen
- Department of Veterinary Medicine, National Taiwan University, Taipei, 10617, Taiwan
- Animal Resource Center, National Taiwan University, Taipei, 10617, Taiwan
| | - Che-Ming Jack Hu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 115, Taiwan
| | - Hui-Wen Chen
- Department of Veterinary Medicine, National Taiwan University, Taipei, 10617, Taiwan
- Animal Resource Center, National Taiwan University, Taipei, 10617, Taiwan
| |
Collapse
|
4
|
Zhou J, Li C, Lu M, Jiang G, Chen S, Li H, Lu K. Pharmacological induction of autophagy reduces inflammation in macrophages by degrading immunoproteasome subunits. PLoS Biol 2024; 22:e3002537. [PMID: 38447109 PMCID: PMC10917451 DOI: 10.1371/journal.pbio.3002537] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Accepted: 02/05/2024] [Indexed: 03/08/2024] Open
Abstract
Defective autophagy is linked to proinflammatory diseases. However, the mechanisms by which autophagy limits inflammation remain elusive. Here, we found that the pan-FGFR inhibitor LY2874455 efficiently activated autophagy and suppressed expression of proinflammatory factors in macrophages stimulated by lipopolysaccharide (LPS). Multiplex proteomic profiling identified the immunoproteasome, which is a specific isoform of the 20s constitutive proteasome, as a substrate that is degraded by selective autophagy. SQSTM1/p62 was found to be a selective autophagy-related receptor that mediated this degradation. Autophagy deficiency or p62 knockdown blocked the effects of LY2874455, leading to the accumulation of immunoproteasomes and increases in inflammatory reactions. Expression of proinflammatory factors in autophagy-deficient macrophages could be reversed by immunoproteasome inhibitors, confirming the pivotal role of immunoproteasome turnover in the autophagy-mediated suppression on the expression of proinflammatory factors. In mice, LY2874455 protected against LPS-induced acute lung injury and dextran sulfate sodium (DSS)-induced colitis and caused low levels of proinflammatory cytokines and immunoproteasomes. These findings suggested that selective autophagy of the immunoproteasome was a key regulator of signaling via the innate immune system.
Collapse
Affiliation(s)
- Jiao Zhou
- Department of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and the Research Units of West China, Chinese Academy of Medical Sciences, Chengdu, China
- National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Chunxia Li
- Department of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and the Research Units of West China, Chinese Academy of Medical Sciences, Chengdu, China
| | - Meng Lu
- Department of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and the Research Units of West China, Chinese Academy of Medical Sciences, Chengdu, China
| | - Gaoyue Jiang
- Department of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and the Research Units of West China, Chinese Academy of Medical Sciences, Chengdu, China
| | - Shanze Chen
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medical College of Jinan University, Shenzhen People’s Hospital, Shenzhen Institute of Respiratory Diseases, Shenzhen, China
| | - Huihui Li
- West China Second University Hospital, Sichuan University, Chengdu, China
| | - Kefeng Lu
- Department of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and the Research Units of West China, Chinese Academy of Medical Sciences, Chengdu, China
| |
Collapse
|
5
|
Dulin H, Barre RS, Xu D, Neal A, Vizcarra E, Chavez J, Ulu A, Yang MS, Khan SR, Wuang K, Bhakta N, Chea C, Wilson EH, Martinez-Sobrido L, Hai R. Harnessing preexisting influenza virus-specific immunity increases antibody responses against SARS-CoV-2. J Virol 2024; 98:e0157123. [PMID: 38206036 PMCID: PMC10878257 DOI: 10.1128/jvi.01571-23] [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: 10/09/2023] [Accepted: 11/13/2023] [Indexed: 01/12/2024] Open
Abstract
In pandemic scenarios involving novel human pathogenic viruses, it is highly desirable that vaccines induce strong neutralizing antibodies as quickly as possible. However, current vaccine strategies require multiple immunization doses to produce high titers of neutralizing antibodies and are poorly protective after a single vaccination. We therefore wished to design a vaccine candidate that would induce increased protective immune responses following the first vaccine dose. We hypothesized that antibodies against the receptor-binding domain (RBD) of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike glycoprotein could be increased by drawing upon immunity to a previous infection. We generated a fusion protein containing the influenza H1N1 PR8 virus nucleoprotein (NP) and the SARS-CoV-2 spike RBD. Mice with or without preexisting immunity to PR8 were then vaccinated with NP/RBD. We observed significantly increased SARS-CoV-2 neutralizing antibodies in mice with PR8 immunity compared to mice without preexisting PR8 immunity. Vaccination with NP/RBD protected mice from SARS-CoV-2-induced morbidity and mortality after a single dose. Additionally, we compared SARS-CoV-2 virus titers in the lungs and nasal turbinates 4 days post-challenge of mice vaccinated with NP/RBD. SARS-CoV-2 virus was detectable in the lungs and nasal turbinate of mice without preexisting PR8 immunity, while SARS-CoV-2 virus was completely undetectable in mice with preexisting PR8 immunity. We also found that CD4-positive T cells in mice with preexisting immunity to PR8 play an essential role in producing the increased antibody response against RBD. This vaccine strategy potentially can be modified to target other pathogens of concern and offers extra value in future pandemic scenarios.IMPORTANCEIncreased globalization and changes in human interactions with wild animals has increased the likelihood of the emergence of novel viruses with pandemic potential. Vaccines can be effective in preventing severe disease caused by pandemic viruses. However, it takes time to develop protective immunity via prime-boost vaccination. More effective vaccine designs should quickly induce protective immunity. We propose leveraging preexisting immunity to a different pathogen to boost protection against emerging viruses. We targeted SARS-CoV-2 as a representative pandemic virus and generated a fusion protein vaccine that combines the nucleoprotein from influenza A virus and the receptor-binding domain (RBD) of the SARS-CoV-2 spike protein. Our vaccine design significantly increased the production of RBD-specific antibodies in mice that had previously been exposed to influenza virus, compared to those without previous exposure. This enhanced immunity reduced SARS-CoV-2 replication in mice. Our results offer a vaccine design that could be valuable in a future pandemic setting.
Collapse
Affiliation(s)
- Harrison Dulin
- Department of Microbiology and Plant Pathology, University of California, Riverside, California, USA
- Cell, Molecular, and Developmental Biology Graduate Program, University of California, Riverside, California, USA
| | - Ramya S. Barre
- Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Duo Xu
- Department of Microbiology and Plant Pathology, University of California, Riverside, California, USA
| | - Arrmund Neal
- Department of Microbiology and Plant Pathology, University of California, Riverside, California, USA
| | - Edward Vizcarra
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, California, USA
| | - Jerald Chavez
- Department of Microbiology and Plant Pathology, University of California, Riverside, California, USA
| | - Arzu Ulu
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, California, USA
| | - Myeon-Sik Yang
- Texas Biomedical Research Institute, San Antonio, Texas, USA
| | | | - Keidy Wuang
- Department of Microbiology and Plant Pathology, University of California, Riverside, California, USA
| | - Nikhil Bhakta
- Department of Microbiology and Plant Pathology, University of California, Riverside, California, USA
| | - Chanvoraboth Chea
- Department of Microbiology and Plant Pathology, University of California, Riverside, California, USA
| | - Emma H. Wilson
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, California, USA
| | | | - Rong Hai
- Department of Microbiology and Plant Pathology, University of California, Riverside, California, USA
- Cell, Molecular, and Developmental Biology Graduate Program, University of California, Riverside, California, USA
| |
Collapse
|
6
|
Wu D, Cong J, Wei J, Hu J, Sun W, Ran W, Liao C, Zheng H, Ye L. A Naïve Phage Display Library-Derived Nanobody Neutralizes SARS-CoV-2 and Three Variants of Concern. Int J Nanomedicine 2023; 18:5781-5795. [PMID: 37869063 PMCID: PMC10588750 DOI: 10.2147/ijn.s427990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 10/07/2023] [Indexed: 10/24/2023] Open
Abstract
Background The emergence of the coronavirus disease 2019 (COVID-19) pandemic and the new severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) variants of concern (VOCs) requires the continuous development of safe, effective, and affordable prevention and therapeutics. Nanobodies have demonstrated antiviral activity against a variety of viruses, providing a new candidate for the prevention and treatment of SARS-CoV-2 and its variants. Methods SARS-CoV-2 glycoprotein spike 1 subunit (S1) was selected as the target antigen for nanobody screening of a naïve phage display library. We obtained a nanobody, named Nb-H6, and then determined its affinity, inhibition, and stability by ELISA, Competitive ELISA, and Biolayer Interferometry (BLI). Infection assays of authentic and pseudotyped SARS-CoV-2 were performed to evaluate the neutralization of Nb-H6. The structure and mechanism of action were investigated by AlphaFold, docking, and residue mutation assays. Results We isolated and characterized a nanobody, Nb-H6, which exhibits a broad affinity for S1 and the receptor binding domain (RBD) of SARS-CoV-2, or Alpha (B.1.1.7), Delta (B.1.617.2), Lambda (C.37), and Omicron (BA.2 and BA.5), and blocks receptor angiotensin-converting enzyme 2 (ACE2) binding. Moreover, Nb-H6 can retain its binding capability after pH or thermal treatment and effectively neutralize both pseudotyped and authentic SARS-CoV-2, as well as VOC Alpha (B.1.1.7), Delta (B.1.617.2), and Omicron (BA.2 and BA.5) pseudoviruses. We also confirmed that Nb-H6 binds two distinct amino acid residues of the RBD, preventing SARS-CoV-2 from interacting with the host receptor. Conclusion Our study highlights a novel nanobody, Nb-H6, that may be useful therapeutically in SARS-CoV-2 and VOC outbreaks and pandemics. These findings also provide a molecular foundation for further studies into how nanobodies neutralize SARS-CoV-2 and variants and imply potential therapeutic targets for the treatment of COVID-19.
Collapse
Affiliation(s)
- Dandan Wu
- Department of Immunology, International Cancer Center, Shenzhen University Health Science Center, Shenzhen, People’s Republic of China
| | - Junxiao Cong
- Department of Immunology, International Cancer Center, Shenzhen University Health Science Center, Shenzhen, People’s Republic of China
| | - Jiali Wei
- Department of Immunology, International Cancer Center, Shenzhen University Health Science Center, Shenzhen, People’s Republic of China
| | - Jing Hu
- Department of Immunology, International Cancer Center, Shenzhen University Health Science Center, Shenzhen, People’s Republic of China
| | - Wenhao Sun
- Department of Immunology, International Cancer Center, Shenzhen University Health Science Center, Shenzhen, People’s Republic of China
| | - Wei Ran
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, People’s Republic of China
| | - Chenghui Liao
- Department of Immunology, International Cancer Center, Shenzhen University Health Science Center, Shenzhen, People’s Republic of China
| | - Housheng Zheng
- Department of Immunology, International Cancer Center, Shenzhen University Health Science Center, Shenzhen, People’s Republic of China
| | - Liang Ye
- Department of Immunology, International Cancer Center, Shenzhen University Health Science Center, Shenzhen, People’s Republic of China
| |
Collapse
|
7
|
Wang J, Zhang Y, Liu C, Zha W, Dong S, Wang Y, Jiang Y, Xing H, Li X. Trivalent mRNA Vaccine against SARS-CoV-2 and Variants with Effective Immunization. Mol Pharm 2023; 20:4971-4983. [PMID: 37699256 DOI: 10.1021/acs.molpharmaceut.2c00860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/14/2023]
Abstract
mRNA vaccines encoding a single spike protein effectively prevent severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. However, the emergence of SARS-CoV-2 variants leads to a wide range of immune evasion. Herein, a unique trivalent mRNA vaccine based on ancestral SARS-CoV-2, Delta, and Omicron variant spike receptor-binding domain (RBD) mRNAs was developed to tackle the immune evasion of the variants. First, three RBD mRNAs of SARS-CoV-2, Delta, and Omicron were coencapsulated into lipid nanoparticles (LNPs) by using microfluidic technology. After that, the physicochemical properties and time-dependent storage stability of the trivalent mRNA vaccine nanoformulation were tested by using dynamic light scattering (DLS). In vitro, the trivalent mRNA vaccine exhibited better lysosomal escape ability, transfection efficiency, and biocompatibility than did the commercial transfection reagent Lipo3000. In addition, Western blot analyses confirmed that the three RBD proteins can be detected in cells transfected with the trivalent mRNA vaccine. Furthermore, ex vivo imaging analysis indicated that the livers of BALB/c mice had the strongest protein expression levels after intramuscular (IM) injection. Using a prime-boost strategy, this trivalent vaccine elicited robust humoral and T-cell immune responses in both the high-dose and low-dose groups and showed no toxicity in BALB/c mice. Three specific IgG antibodies in the high-dose group against SARS-CoV-2, Delta, and Omicron variants approached ∼1/1,833,333, ∼1/1,866,667, and ∼1/925,000, respectively. Taken together, two doses of inoculation with the trivalent mRNA vaccine may provide broad and effective immunization responses against SARS-CoV-2 and variants.
Collapse
Affiliation(s)
- Ji Wang
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 14122, PR China
| | - Yanhao Zhang
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 14122, PR China
| | - Chao Liu
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 14122, PR China
| | - Wenhui Zha
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 14122, PR China
| | - Shuo Dong
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 14122, PR China
| | - Yang Wang
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 14122, PR China
| | - Yuhao Jiang
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 14122, PR China
| | - Hanlei Xing
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 14122, PR China
| | - Xinsong Li
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 14122, PR China
| |
Collapse
|
8
|
Gromova MS, Gromov AV, Grunina TM, Lyashchuk AM, Galushkina ZM, Subbotina ME, Esmagambetov IB, Ryabova EI, Prokofiev VV, Kovyrshina AV, Ilyukhina AA, Shelkov AY, Karyagina AS, Lunin VG. Recombinant RBD of the SARS-CoV-2 Spike Protein: Production in Escherichia coli Cells, Binding to Antibodies, and Antiviral Activity. MOLECULAR GENETICS, MICROBIOLOGY AND VIROLOGY 2023; 38:86-94. [DOI: 10.3103/s0891416823020052] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/07/2022] [Accepted: 12/14/2022] [Indexed: 01/04/2025]
|
9
|
Dormeshkin D, Katsin M, Stegantseva M, Golenchenko S, Shapira M, Dubovik S, Lutskovich D, Kavaleuski A, Meleshko A. Design and Immunogenicity of SARS-CoV-2 DNA Vaccine Encoding RBD-PVXCP Fusion Protein. Vaccines (Basel) 2023; 11:1014. [PMID: 37376403 DOI: 10.3390/vaccines11061014] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/16/2023] [Accepted: 03/21/2023] [Indexed: 06/29/2023] Open
Abstract
The potential of immune-evasive mutation accumulation in the SARS-CoV-2 virus has led to its rapid spread, causing over 600 million confirmed cases and more than 6.5 million confirmed deaths. The huge demand for the rapid development and deployment of low-cost and effective vaccines against emerging variants has renewed interest in DNA vaccine technology. Here, we report the rapid generation and immunological evaluation of novel DNA vaccine candidates against the Wuhan-Hu-1 and Omicron variants based on the RBD protein fused with the Potato virus X coat protein (PVXCP). The delivery of DNA vaccines using electroporation in a two-dose regimen induced high-antibody titers and profound cellular responses in mice. The antibody titers induced against the Omicron variant of the vaccine were sufficient for effective protection against both Omicron and Wuhan-Hu-1 virus infections. The PVXCP protein in the vaccine construct shifted the immune response to the favorable Th1-like type and provided the oligomerization of RBD-PVXCP protein. Naked DNA delivery by needle-free injection allowed us to achieve antibody titers comparable with mRNA-LNP delivery in rabbits. These data identify the RBD-PVXCP DNA vaccine platform as a promising solution for robust and effective SARS-CoV-2 protection, supporting further translational study.
Collapse
Affiliation(s)
- Dmitri Dormeshkin
- Institute of Bioorganic Chemistry of the National Academy of Sciences of Belarus, 220084 Minsk, Belarus
| | - Mikalai Katsin
- Immunofusion, LLC, 210004 Vitebsk, Belarus
- Imunovakcina, UAB, LT-08102 Vilnius, Lithuania
| | | | | | - Michail Shapira
- Institute of Bioorganic Chemistry of the National Academy of Sciences of Belarus, 220084 Minsk, Belarus
| | - Simon Dubovik
- Institute of Bioorganic Chemistry of the National Academy of Sciences of Belarus, 220084 Minsk, Belarus
| | | | - Anton Kavaleuski
- Institute of Science and Technology Austria, 3400 Klosterneuburg, Austria
| | - Alexander Meleshko
- Immunofusion, LLC, 210004 Vitebsk, Belarus
- Imunovakcina, UAB, LT-08102 Vilnius, Lithuania
| |
Collapse
|
10
|
Volosnikova EA, Merkuleva IA, Esina TI, Shcherbakov DN, Borgoyakova MB, Isaeva AA, Nesmeyanova VS, Volkova NV, Belenkaya SV, Zaykovskaya AV, Pyankov OV, Starostina EV, Zadorozhny AM, Zaitsev BN, Karpenko LI, Ilyichev AA, Danilenko ED. SARS-CoV-2 RBD Conjugated to Polyglucin, Spermidine, and dsRNA Elicits a Strong Immune Response in Mice. Vaccines (Basel) 2023; 11:vaccines11040808. [PMID: 37112720 PMCID: PMC10146165 DOI: 10.3390/vaccines11040808] [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: 03/05/2023] [Revised: 03/31/2023] [Accepted: 04/04/2023] [Indexed: 04/29/2023] Open
Abstract
Despite the rapid development and approval of several COVID vaccines based on the full-length spike protein, there is a need for safe, potent, and high-volume vaccines. Considering the predominance of the production of neutralizing antibodies targeting the receptor-binding domain (RBD) of S-protein after natural infection or vaccination, it makes sense to choose RBD as a vaccine immunogen. However, due to its small size, RBD exhibits relatively poor immunogenicity. Searching for novel adjuvants for RBD-based vaccine formulations is considered a good strategy for enhancing its immunogenicity. Herein, we assess the immunogenicity of severe acute respiratory syndrome coronavirus 2 RBD conjugated to a polyglucin:spermidine complex (PGS) and dsRNA (RBD-PGS + dsRNA) in a mouse model. BALB/c mice were immunized intramuscularly twice, with a 2-week interval, with 50 µg of RBD, RBD with Al(OH)3, or conjugated RBD. A comparative analysis of serum RBD-specific IgG and neutralizing antibody titers showed that PGS, PGS + dsRNA, and Al(OH)3 enhanced the specific humoral response in animals. There was no significant difference between the groups immunized with RBD-PGS + dsRNA and RBD with Al(OH)3. Additionally, the study of the T-cell response in animals showed that, unlike adjuvants, the RBD-PGS + dsRNA conjugate stimulates the production of specific CD4+ and CD8+ T cells in animals.
Collapse
Affiliation(s)
- Ekaterina A Volosnikova
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, 630559 Koltsovo, Russia
| | - Iuliia A Merkuleva
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, 630559 Koltsovo, Russia
| | - Tatiana I Esina
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, 630559 Koltsovo, Russia
| | - Dmitry N Shcherbakov
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, 630559 Koltsovo, Russia
| | - Mariya B Borgoyakova
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, 630559 Koltsovo, Russia
| | - Anastasiya A Isaeva
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, 630559 Koltsovo, Russia
| | - Valentina S Nesmeyanova
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, 630559 Koltsovo, Russia
| | - Natalia V Volkova
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, 630559 Koltsovo, Russia
| | - Svetlana V Belenkaya
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, 630559 Koltsovo, Russia
| | - Anna V Zaykovskaya
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, 630559 Koltsovo, Russia
| | - Oleg V Pyankov
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, 630559 Koltsovo, Russia
| | - Ekaterina V Starostina
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, 630559 Koltsovo, Russia
| | - Alexey M Zadorozhny
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, 630559 Koltsovo, Russia
| | - Boris N Zaitsev
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, 630559 Koltsovo, Russia
| | - Larisa I Karpenko
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, 630559 Koltsovo, Russia
| | - Alexander A Ilyichev
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, 630559 Koltsovo, Russia
| | - Elena D Danilenko
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, 630559 Koltsovo, Russia
| |
Collapse
|
11
|
Thermophilic Filamentous Fungus C1-Cell-Cloned SARS-CoV-2-Spike-RBD-Subunit-Vaccine Adjuvanted with Aldydrogel ®85 Protects K18-hACE2 Mice against Lethal Virus Challenge. Vaccines (Basel) 2022; 10:vaccines10122119. [PMID: 36560529 PMCID: PMC9783968 DOI: 10.3390/vaccines10122119] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/30/2022] [Accepted: 11/30/2022] [Indexed: 12/14/2022] Open
Abstract
SARS-CoV-2 is evolving with increased transmission, host range, pathogenicity, and virulence. The original and mutant viruses escape host innate (Interferon) immunity and adaptive (Antibody) immunity, emphasizing unmet needs for high-yield, commercial-scale manufacturing to produce inexpensive vaccines/boosters for global/equitable distribution. We developed DYAI-100A85, a SARS-CoV-2 spike receptor binding domain (RBD) subunit antigen vaccine expressed in genetically modified thermophilic filamentous fungus, Thermothelomyces heterothallica C1, and secreted at high levels into fermentation medium. The RBD-C-tag antigen strongly binds ACE2 receptors in vitro. Alhydrogel®'85'-adjuvanted RDB-C-tag-based vaccine candidate (DYAI-100A85) demonstrates strong immunogenicity, and antiviral efficacy, including in vivo protection against lethal intranasal SARS-CoV-2 (D614G) challenge in human ACE2-transgenic mice. No loss of body weight or adverse events occurred. DYAI-100A85 also demonstrates excellent safety profile in repeat-dose GLP toxicity study. In summary, subcutaneous prime/boost DYAI-100A85 inoculation induces high titers of RBD-specific neutralizing antibodies and protection of hACE2-transgenic mice against lethal challenge with SARS-CoV-2. Given its demonstrated safety, efficacy, and low production cost, vaccine candidate DYAI-100 received regulatory approval to initiate a Phase 1 clinical trial to demonstrate its safety and efficacy in humans.
Collapse
|
12
|
Wo J, Lv ZY, Sun JN, Tang H, Qi N, Ye BC. Engineering probiotic-derived outer membrane vesicles as functional vaccine carriers to enhance immunity against SARS-CoV-2. iScience 2022; 26:105772. [PMID: 36510593 PMCID: PMC9729586 DOI: 10.1016/j.isci.2022.105772] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 11/10/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022] Open
Abstract
Because of the continued emergence of SARS-CoV-2 variants, there has been considerable interest in how to display multivalent antigens efficiently. Bacterial outer membrane vesicles (OMVs) can serve as an attractive vaccine delivery system because of their self-adjuvant properties and the ability to be decorated with antigens. Here we set up a bivalent antigen display platform based on engineered OMVs using mCherry and GFP and demonstrated that two different antigens of SARS-CoV-2 could be presented simultaneously in the lumen and on the surface of OMVs. Comparing immunogenicity, ClyA-NG06 fusion and the receptor-binding domain (RBD) of the spike protein in the OMV lumen elicited a stronger humoral response in mice than OMVs presenting either the ClyA-NG06 fusion or RBD alone. Taken together, we provided an efficient approach to display SARS-CoV-2 antigens in the lumen and on the surface of the same OMV and highlighted the potential of OMVs as general multi-antigen carriers.
Collapse
Affiliation(s)
- Jing Wo
- Institute of Engineering Biology and Health, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014 Zhejiang, China
| | - Zhao-Yong Lv
- Institute of Engineering Biology and Health, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014 Zhejiang, China
| | - Jia-Nan Sun
- Institute of Engineering Biology and Health, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014 Zhejiang, China
| | - Hao Tang
- Institute of Engineering Biology and Health, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014 Zhejiang, China
| | - Nan Qi
- Institute of Engineering Biology and Health, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014 Zhejiang, China
| | - Bang-Ce Ye
- Institute of Engineering Biology and Health, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014 Zhejiang, China,Lab of Biosystem and Microanalysis, State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China,Corresponding author
| |
Collapse
|
13
|
Park JH, Cha MJ, Choi H, Kim MC, Chung JW, Lee KS, Jeong DG, Baek MS, Kim WY, Lim Y, Yoon SW, Choi SH. Relationship between SARS-CoV-2 antibody titer and the severity of COVID-19. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2022; 55:1094-1100. [PMID: 35570185 PMCID: PMC9069977 DOI: 10.1016/j.jmii.2022.04.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 02/14/2022] [Accepted: 04/01/2022] [Indexed: 12/27/2022]
Abstract
BACKGROUND It remains unclear whether high titers of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) antibodies aggravate clinical manifestations in patients or whether severe clinical manifestations result in high antibody titers. Thus, we investigated the cause-effect relationship between SARS-CoV-2 antibody titers and disease severity. METHODS We prospectively enrolled patients admitted with the diagnosis of coronavirus disease-19 (COVID-19) from February 2020 to August 2020. We measured SARS-CoV-2 antibody titers, namely anti-receptor-binding domain (RBD) antibody and neutralizing antibody (NAb), from blood samples and calculated the chest radiograph (CXR) scores of the patients to evaluate the severity of COVID-19. RESULTS Overall, 40 patients with COVID-19 were enrolled. Pneumonia was observed in more than half of the patients (25/40, 60%). SARS-CoV-2 antibody titers were higher in patients who were aged >60 years (anti-RBD antibodies, P = 0.003 and NAb, P = 0.009), presented with pneumonia (P = 0.006 and 0.007, respectively), and required oxygen therapy (P = 0.003 and 0.004, respectively) than in those who were not. CXR scores peaked (at 15-21 days after the onset of symptoms) statistically significantly earlier than SARS-CoV-2 antibody titers (at 22-30 days for NAb and at 31-70 days for anti-RBD antibody). There was a close correlation between the maximum CXR score and the maximum SAR-CoV-2 antibody titer. CONCLUSIONS Based on the comparison of the peak time of SARS-CoV-2 antibody titers with the CXR score after symptom onset, we suggest that severe clinical manifestations result in high titers of SARS-CoV-2 antibodies.
Collapse
Affiliation(s)
- Joung Ha Park
- Division of Infectious Diseases, Department of Internal Medicine, Chung-Ang University Gwangmyeong Hospital, Gwangmyeong, South Korea
| | - Min Jae Cha
- Department of Radiology, Chung-Ang University Hospital, Seoul, South Korea
| | - Hyewon Choi
- Department of Radiology, Chung-Ang University Hospital, Seoul, South Korea
| | - Min-Chul Kim
- Division of Infectious Diseases, Department of Internal Medicine, Chung-Ang University Gwangmyeong Hospital, Gwangmyeong, South Korea
| | - Jin-Won Chung
- Division of Infectious Diseases, Department of Internal Medicine, Chung-Ang University Hospital, Seoul, South Korea
| | - Kyu-Sun Lee
- Bionanotechnology Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea
| | - Dae Gwin Jeong
- Bionanotechnology Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea
| | - Moon Seong Baek
- Department of Internal Medicine, Chung-Ang University Hospital, Seoul, South Korea
| | - Won-Young Kim
- Department of Internal Medicine, Chung-Ang University Hospital, Seoul, South Korea
| | - Yaeji Lim
- Department of Applied Statistics, Chung-Ang University, Seoul, South Korea
| | - Sun Woo Yoon
- Bionanotechnology Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea,Corresponding author. Bionanotechnology Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea
| | - Seong-Ho Choi
- Division of Infectious Diseases, Department of Internal Medicine, Chung-Ang University Hospital, Seoul, South Korea,Corresponding author. Fax: +82 2 6299 2064
| |
Collapse
|
14
|
Development of an LNP-Encapsulated mRNA-RBD Vaccine against SARS-CoV-2 and Its Variants. Pharmaceutics 2022; 14:pharmaceutics14051101. [PMID: 35631687 PMCID: PMC9143166 DOI: 10.3390/pharmaceutics14051101] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 04/15/2022] [Accepted: 04/26/2022] [Indexed: 11/30/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is undoubtedly the most challenging pandemic in the current century and remains a global health emergency. As the number of COVID-19 cases in the world is on the rise and variants continue to emerge, there is an urgent need for vaccines. Among all immunization approaches, mRNA vaccines have demonstrated more promising results in response to this challenge. Herein, we designed an mRNA-based vaccine encoding the receptor-binding domain (RBD) of SARS-CoV-2 encapsulated in lipid nanoparticles (LNPs). Intramuscular (i.m.) administration of the mRNA-RBD vaccine elicited broad-spectrum neutralizing antibodies and cellular responses against not only the wild-type SARS-CoV-2 virus but also Delta and Omicron variants. These results indicated that two doses of mRNA-RBD immunization conferred a strong immune response in mice against the wild-type SARS-CoV-2, while the booster dose provided a sufficient immunity against SARS-CoV-2 and its variants. Taken together, the three-dose regimen strategy of the mRNA-RBD vaccine proposed in the present study appears to be a promising reference for the development of mRNA vaccines targeting SARS-CoV-2 variants.
Collapse
|
15
|
Abstract
Antibody-dependent enhancement (ADE) of infection is generally known for many viruses. A potential risk of ADE in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection has also been discussed since the beginning of the coronavirus disease 2019 (COVID-19) pandemic; however, clinical evidence of the presence of antibodies with ADE potential is limited. Here, we show that ADE antibodies are produced by SARS-CoV-2 infection and the ADE process can be mediated by at least two different host factors, Fcγ receptor (FcγR) and complement component C1q. Of 89 serum samples collected from acute or convalescent COVID-19 patients, 62.9% were found to be positive for SARS-CoV-2-specific IgG. FcγR- and/or C1q-mediated ADE were detected in 50% of the IgG-positive sera, whereas most of them showed neutralizing activity in the absence of FcγR and C1q. Importantly, ADE antibodies were found in 41.4% of the acute COVID-19 patients. Neutralizing activity was also detected in most of the IgG-positive sera, but it was counteracted by ADE in subneutralizing conditions in the presence of FcγR or C1q. Although the clinical importance of ADE needs to be further investigated with larger numbers of COVID-19 patient samples, our data suggest that SARS-CoV-2 utilizes multiple mechanisms of ADE. C1q-mediated ADE may particularly have a clinical impact since C1q is present at high concentrations in plasma and its receptors are ubiquitously expressed on the surfaces of many types of cells, including respiratory epithelial cells, which SARS-CoV-2 primarily infects. IMPORTANCE Potential risks of antibody-dependent enhancement (ADE) in the coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection has been discussed and the proposed mechanism mostly depends on the Fc gamma receptor (FcγR). However, since FcγRs are exclusively expressed on immune cells, which are not primary targets of SARS-CoV-2, the clinical importance of ADE of SARS-CoV-2 infection remains controversial. Our study demonstrates that SARS-CoV-2 infection induces antibodies that increase SARS-CoV-2 infection through another ADE mechanism in which complement component C1q mediates the enhancement. Although neutralizing activity was also detected in the serum samples, it was counteracted by ADE in the presence of FcγR or C1q. Considering the ubiquity of C1q and its cellular receptors, C1q-mediated ADE may more likely occur in respiratory epithelial cells, which SARS-CoV-2 primarily infects. Our data highlight the importance of careful monitoring of the antibody properties in COVID-19 convalescent and vaccinated individuals.
Collapse
|
16
|
Chourasia R, Padhi S, Phukon LC, Abedin MM, Sirohi R, Singh SP, Rai AK. Peptide candidates for the development of therapeutics and vaccines against β-coronavirus infection. Bioengineered 2022; 13:9435-9454. [PMID: 35387556 PMCID: PMC9161909 DOI: 10.1080/21655979.2022.2060453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/16/2022] [Accepted: 03/17/2022] [Indexed: 01/18/2023] Open
Abstract
Betacoronaviruses (β-CoVs) have caused major viral outbreaks in the last two decades in the world. The mutation and recombination abilities in β-CoVs resulted in zoonotic diseases in humans. Proteins responsible for viral attachment and replication are highly conserved in β-CoVs. These conserved proteins have been extensively studied as targets for preventing infection and the spread of β-CoVs. Peptides are among the most promising candidates for developing vaccines and therapeutics against viral pathogens. The immunostimulatory and viral inhibitory potential of natural and synthetic peptides has been extensively studied since the SARS-CoV outbreak. Food-derived peptides demonstrating high antiviral activity can be used to develop effective therapeutics against β-CoVs. Specificity, tolerability, and customizability of peptides can be explored to develop potent drugs against β-CoVs. However, the proteolytic susceptibility and low bioavailability of peptides pose challenges for the development of therapeutics. This review illustrates the potential role of peptides in eliciting an adaptive immune response and inhibiting different stages of the β-CoV life cycle. Further, the challenges and future directions associated with developing peptide-based therapeutics and vaccines against existing and future β-CoV pathogens have been discussed.
Collapse
Affiliation(s)
- Rounak Chourasia
- Institute of Bioresources and Sustainable Development (DBT-IBSD), Regional Centre, Tadong- 737102, India
| | - Srichandan Padhi
- Institute of Bioresources and Sustainable Development (DBT-IBSD), Regional Centre, Tadong- 737102, India
| | - Loreni Chiring Phukon
- Institute of Bioresources and Sustainable Development (DBT-IBSD), Regional Centre, Tadong- 737102, India
| | - Md Minhajul Abedin
- Institute of Bioresources and Sustainable Development (DBT-IBSD), Regional Centre, Tadong- 737102, India
| | - Ranjana Sirohi
- Department of Chemical and Biological Engineering, Korea University, 145, Anam-ro, Seongbuk-gu, 02841, Republic of Korea
| | - Sudhir P Singh
- Centre of Innovative and Applied Bioprocessing (DBT-CIAB), Sector-81, S.A.S. Nagar, Mohali- 140306, India
| | - Amit Kumar Rai
- Institute of Bioresources and Sustainable Development (DBT-IBSD), Regional Centre, Tadong- 737102, India
- Institute of Bioresources and Sustainable Development (DBT-IBSD), Mizoram Node, Aizawl, India
| |
Collapse
|
17
|
Cobb RR, Nkolola J, Gilchuk P, Chandrashekar A, Yu J, House RV, Earnhart CG, Dorsey NM, Hopkins SA, Snow DM, Chen RE, VanBlargan LA, Hechenblaickner M, Hoppe B, Collins L, Tomic MT, Nonet GH, Hackett K, Slaughter JC, Lewis MG, Andersen H, Cook A, Diamond MS, Carnahan RH, Barouch DH, Crowe JE. A combination of two human neutralizing antibodies prevents SARS-CoV-2 infection in cynomolgus macaques. MED 2022; 3:188-203.e4. [PMID: 35132398 PMCID: PMC8810411 DOI: 10.1016/j.medj.2022.01.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 12/13/2021] [Accepted: 01/20/2022] [Indexed: 02/07/2023]
Abstract
BACKGROUND Human monoclonal antibody (mAb) treatments are promising for COVID-19 prevention or therapy. The pre-exposure prophylactic efficacy of neutralizing antibodies that are engineered with mutations to extend their persistence in human serum and the neutralizing antibody titer in serum required for protection against SARS-CoV-2 infection remain poorly characterized. METHODS The Fc region of two neutralizing mAbs (COV2-2130 and COV2-2381) targeting non-overlapping epitopes on the receptor binding domain of SARS-CoV-2 spike protein was engineered to extend their persistence in humans and reduce interactions with Fc gamma receptors. We assessed protection by individual antibodies or a combination of the two antibodies (designated ADM03820) given prophylactically by an intravenous or intramuscular route in a non-human primate (NHP) model of SARS-CoV-2 infection. FINDINGS Passive transfer of individual mAbs or ADM03820 conferred virological protection in the NHP respiratory tract in a dose-dependent manner, and ADM03820 potently neutralized SARS-CoV-2 variants of concern in vitro. We defined a protective serum-neutralizing antibody titer and concentration in NHPs for passively transferred human antibodies that acted by direct viral neutralization. CONCLUSIONS In summary, we demonstrate that neutralizing antibodies with extended half-life and lacking Fc-mediated effector functions are efficient for pre-exposure prophylaxis of SARS-CoV-2 infection in NHPs. These results support clinical development of ADM03820 for COVID-19 prevention. FUNDING This research was supported by a contract from the JPEO-CBRND (W911QY-20-9-003, 20-05); the Joint Sciences and Technology Office and Joint Program Executive Office (MCDC-16-01-002 JSTO, JPEO); a DARPA grant (HR0011-18-2-0001); an NIH grant (R01 AI157155); and the 2019 Future Insight Prize from Merck KGaA.
Collapse
Affiliation(s)
- Ronald R Cobb
- Ology Bioservices, Process Development and Manufacturing, Alachua, FL 32615, USA
| | - Joseph Nkolola
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Pavlo Gilchuk
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Abishek Chandrashekar
- 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
| | | | - Christopher G Earnhart
- Joint Program Executive Office for Chemical, Biological, Radiological, and Nuclear Defense, US Department of Defense, Frederick, MD 21703, USA
| | - Nicole M Dorsey
- Joint Program Executive Office for Chemical, Biological, Radiological, and Nuclear Defense, US Department of Defense, Frederick, MD 21703, USA
| | | | | | - Rita E Chen
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Laura A VanBlargan
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | - Brian Hoppe
- Ology Bioservices, Process Development and Manufacturing, Alachua, FL 32615, USA
| | - Laura Collins
- Ology Bioservices, Process Development and Manufacturing, Alachua, FL 32615, USA
| | - Milan T Tomic
- Research and Development, Ology Bioservices, Inc., Alameda 94501, CA, USA
| | - Genevieve H Nonet
- Research and Development, Ology Bioservices, Inc., Alameda 94501, CA, USA
| | | | - James C Slaughter
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | | | | | | | - Michael S Diamond
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Robert H Carnahan
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Dan H Barouch
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - James E Crowe
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| |
Collapse
|
18
|
Clark NM, Janaka SK, Hartman W, Stramer S, Goodhue E, Weiss J, Evans DT, Connor JP. Anti-SARS-CoV-2 IgG and IgA antibodies in COVID-19 convalescent plasma do not enhance viral infection. PLoS One 2022; 17:e0257930. [PMID: 35259162 PMCID: PMC8903276 DOI: 10.1371/journal.pone.0257930] [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: 09/08/2021] [Accepted: 02/18/2022] [Indexed: 12/02/2022] Open
Abstract
The novel coronavirus, SARS-CoV-2 that causes COVID-19 has resulted in the death of nearly 4 million people within the last 18 months. While preventive vaccination, and monoclonal antibody therapies have been rapidly developed and deployed, early in the pandemic the use of COVID-19 convalescent plasma (CCP) was a common means of passive immunization with a theoretical risk of antibody-dependent enhancement (ADE) of viral infection. Though vaccines elicit a strong and protective immune response and transfusion of CCP with high titers of neutralization activity are correlated with better clinical outcomes, the question of whether antibodies in CCP can enhance infection of SARS-CoV-2 has not been directly addressed. In this study, we analyzed for and observed passive transfer of neutralization activity with CCP transfusion. Furthermore, to specifically understand if antibodies against the spike protein (S) enhance infection, we measured the anti-S IgG, IgA, and IgM responses and adapted retroviral-pseudotypes to measure virus neutralization with target cells expressing the ACE2 virus receptor and the Fc alpha receptor (FcαR) or Fc gamma receptor IIA (FcγRIIA). Whereas neutralizing activity of CCP correlated best with higher titers of anti-S IgG antibodies, the neutralizing titer was not affected when Fc receptors were present on target cells. These observations support the absence of antibody-dependent enhancement of infection (ADE) by IgG and IgA isotypes found in CCP. The results presented, therefore, not only supports the therapeutic use of currently available antibody-based treatment, including the continuation of CCP transfusion strategies, but also the use of various vaccine platforms in a prophylactic approach.
Collapse
Affiliation(s)
- Natasha M. Clark
- Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Sanath Kumar Janaka
- Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, Wisconsin, United States of America
| | - William Hartman
- Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Susan Stramer
- American Red Cross, Washington, DC, United States of America
| | - Erin Goodhue
- American Red Cross, Washington, DC, United States of America
| | - John Weiss
- American Red Cross, Washington, DC, United States of America
| | - David T. Evans
- Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, Wisconsin, United States of America
- Wisconsin National Primate Research Center, Madison, Wisconsin, United States of America
| | - Joseph P. Connor
- Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, Wisconsin, United States of America
| |
Collapse
|
19
|
Chen GL, Li XF, Dai XH, Li N, Cheng ML, Huang Z, Shen J, Ge YH, Shen ZW, Deng YQ, Yang SY, Zhao H, Zhang NN, Zhang YF, Wei L, Wu KQ, Zhu MF, Peng CG, Jiang Q, Cao SC, Li YH, Zhao DH, Wu XH, Ni L, Shen HH, Dong C, Ying B, Sheng GP, Qin CF, Gao HN, Li LJ. Safety and immunogenicity of the SARS-CoV-2 ARCoV mRNA vaccine in Chinese adults: a randomised, double-blind, placebo-controlled, phase 1 trial. THE LANCET. MICROBE 2022; 3:e193-e202. [PMID: 35098177 PMCID: PMC8786321 DOI: 10.1016/s2666-5247(21)00280-9] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
BACKGROUND Safe and effective vaccines are urgently needed to end the COVID-19 pandemic caused by SARS-CoV-2 infection. We aimed to assess the preliminary safety, tolerability, and immunogenicity of an mRNA vaccine ARCoV, which encodes the SARS-CoV-2 spike protein receptor-binding domain (RBD). METHODS This single centre, double-blind, randomised, placebo-controlled, dose-escalation, phase 1 trial of ARCoV was conducted at Shulan (Hangzhou) hospital in Hangzhou, Zhejiang province, China. Healthy adults aged 18-59 years negative for SARS-CoV-2 infection were enrolled and randomly assigned using block randomisation to receive an intramuscular injection of vaccine or placebo. Vaccine doses were 5 μg, 10 μg, 15 μg, 20 μg, and 25 μg. The first six participants in each block were sentinels and along with the remaining 18 participants, were randomly assigned to groups (5:1). In block 1 sentinels were given the lowest vaccine dose and after a 4-day observation with confirmed safety analyses, the remaining 18 participants in the same dose group proceeded and sentinels in block 2 were given their first administration on a two-dose schedule, 28 days apart. All participants, investigators, and staff doing laboratory analyses were masked to treatment allocation. Humoral responses were assessed by measuring anti-SARS-CoV-2 RBD IgG using a standardised ELISA and neutralising antibodies using pseudovirus-based and live SARS-CoV-2 neutralisation assays. SARS-CoV-2 RBD-specific T-cell responses, including IFN-γ and IL-2 production, were assessed using an enzyme-linked immunospot (ELISpot) assay. The primary outcome for safety was incidence of adverse events or adverse reactions within 60 min, and at days 7, 14, and 28 after each vaccine dose. The secondary safety outcome was abnormal changes detected by laboratory tests at days 1, 4, 7, and 28 after each vaccine dose. For immunogenicity, the secondary outcome was humoral immune responses: titres of neutralising antibodies to live SARS-CoV-2, neutralising antibodies to pseudovirus, and RBD-specific IgG at baseline and 28 days after first vaccination and at days 7, 15, and 28 after second vaccination. The exploratory outcome was SARS-CoV-2-specific T-cell responses at 7 days after the first vaccination and at days 7 and 15 after the second vaccination. This trial is registered with www.chictr.org.cn (ChiCTR2000039212). FINDINGS Between Oct 30 and Dec 2, 2020, 230 individuals were screened and 120 eligible participants were randomly assigned to receive five-dose levels of ARCoV or a placebo (20 per group). All participants received the first vaccination and 118 received the second dose. No serious adverse events were reported within 56 days after vaccination and the majority of adverse events were mild or moderate. Fever was the most common systemic adverse reaction (one [5%] of 20 in the 5 μg group, 13 [65%] of 20 in the 10 μg group, 17 [85%] of 20 in the 15 μg group, 19 [95%] of 20 in the 20 μg group, 16 [100%] of 16 in the 25 μg group; p<0·0001). The incidence of grade 3 systemic adverse events were none (0%) of 20 in the 5 μg group, three (15%) of 20 in the 10 μg group, six (30%) of 20 in the 15 μg group, seven (35%) of 20 in the 20 μg group, five (31%) of 16 in the 25 μg group, and none (0%) of 20 in the placebo group (p=0·0013). As expected, the majority of fever resolved in the first 2 days after vaccination for all groups. The incidence of solicited systemic adverse events was similar after administration of ARCoV as a first or second vaccination. Humoral immune responses including anti-RBD IgG and neutralising antibodies increased significantly 7 days after the second dose and peaked between 14 and 28 days thereafter. Specific T-cell response peaked between 7 and 14 days after full vaccination. 15 μg induced the highest titre of neutralising antibodies, which was about twofold more than the antibody titre of convalescent patients with COVID-19. INTERPRETATION ARCoV was safe and well tolerated at all five doses. The acceptable safety profile, together with the induction of strong humoral and cellular immune responses, support further clinical testing of ARCoV at a large scale. FUNDING National Key Research and Development Project of China, Academy of Medical Sciences China, National Natural Science Foundation China, and Chinese Academy of Medical Sciences.
Collapse
Affiliation(s)
| | - Xiao-Feng Li
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
| | | | - Nan Li
- Shulan (Hangzhou) Hospital, Hangzhou, China
| | - Meng-Li Cheng
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
| | | | - Jian Shen
- 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, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yu-Hua Ge
- Shulan (Hangzhou) Hospital, Hangzhou, China
| | - Zhen-Wei Shen
- Zhejiang Shuren University Shulan International Medical College, Hangzhou, China
| | - Yong-Qiang Deng
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
| | | | - Hui Zhao
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
| | - Na-Na Zhang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
| | - Yi-Fei Zhang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
| | - Ling Wei
- Suzhou Abogen Biosciences, Suzhou, China
| | - Kai-Qi Wu
- Shulan (Hangzhou) Hospital, Hangzhou, China
| | | | | | - Qi Jiang
- Shulan (Hangzhou) Hospital, Hangzhou, China
| | - Shou-Chun Cao
- National Institutes for Food and Drug Control, Beijing, China
| | - Yu-Hua Li
- National Institutes for Food and Drug Control, Beijing, China
| | - Dan-Hua Zhao
- National Institutes for Food and Drug Control, Beijing, China
| | - Xiao-Hong Wu
- National Institutes for Food and Drug Control, Beijing, China
| | - Ling Ni
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China
- Center for Human Disease Immuno-monitoring, Beijing Friendship Hospital, Beijing, China
| | - Hua-Hao Shen
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Chen Dong
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China
- Center for Human Disease Immuno-monitoring, Beijing Friendship Hospital, Beijing, China
| | - Bo Ying
- Suzhou Abogen Biosciences, Suzhou, China
| | | | - Cheng-Feng Qin
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
- Research Unit of Discovery and Tracing of Natural Focus Diseases, Chinese Academy of Medical Sciences, Beijing China
| | - Hai-Nv Gao
- Shulan (Hangzhou) Hospital, Hangzhou, China
| | - Lan-Juan Li
- 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, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
20
|
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: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [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
|
21
|
Merkuleva IA, Shcherbakov DN, Borgoyakova MB, Shanshin DV, Rudometov AP, Karpenko LI, Belenkaya SV, Isaeva AA, Nesmeyanova VS, Kazachinskaia EI, Volosnikova EA, Esina TI, Zaykovskaya AV, Pyankov OV, Borisevich SS, Shelemba AA, Chikaev AN, Ilyichev AA. Comparative Immunogenicity of the Recombinant Receptor-Binding Domain of Protein S SARS-CoV-2 Obtained in Prokaryotic and Mammalian Expression Systems. Vaccines (Basel) 2022; 10:vaccines10010096. [PMID: 35062757 PMCID: PMC8779843 DOI: 10.3390/vaccines10010096] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 01/02/2022] [Accepted: 01/04/2022] [Indexed: 01/05/2023] Open
Abstract
The receptor-binding domain (RBD) of the protein S SARS-CoV-2 is considered to be one of the appealing targets for developing a vaccine against COVID-19. The choice of an expression system is essential when developing subunit vaccines, as it ensures the effective synthesis of the correctly folded target protein, and maintains its antigenic and immunogenic properties. Here, we describe the production of a recombinant RBD protein using prokaryotic (pRBD) and mammalian (mRBD) expression systems, and compare the immunogenicity of prokaryotic and mammalian-expressed RBD using a BALB/c mice model. An analysis of the sera from mice immunized with both variants of the protein revealed that the mRBD expressed in CHO cells provides a significantly stronger humoral immune response compared with the RBD expressed in E.coli cells. A specific antibody titer of sera from mice immunized with mRBD was ten-fold higher than the sera from the mice that received pRBD in ELISA, and about 100-fold higher in a neutralization test. The data obtained suggests that mRBD is capable of inducing neutralizing antibodies against SARS-CoV-2.
Collapse
Affiliation(s)
- Iuliia A. Merkuleva
- State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, World-Class Genomic Research Center for Biological Safety and Technological Independence, Federal Scientific and Technical Program on the Development of Genetic Technologies, 630559 Novosibirsk, Russia; (I.A.M.); (M.B.B.); (D.V.S.); (A.P.R.); (L.I.K.); (S.V.B.); (A.A.I.); (V.S.N.); (E.I.K.); (E.A.V.); (T.I.E.); (A.V.Z.); (O.V.P.); (A.A.I.)
| | - Dmitry N. Shcherbakov
- State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, World-Class Genomic Research Center for Biological Safety and Technological Independence, Federal Scientific and Technical Program on the Development of Genetic Technologies, 630559 Novosibirsk, Russia; (I.A.M.); (M.B.B.); (D.V.S.); (A.P.R.); (L.I.K.); (S.V.B.); (A.A.I.); (V.S.N.); (E.I.K.); (E.A.V.); (T.I.E.); (A.V.Z.); (O.V.P.); (A.A.I.)
- Correspondence: ; Tel.: +7-383-363-47-00 (ext. 2007)
| | - Mariya B. Borgoyakova
- State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, World-Class Genomic Research Center for Biological Safety and Technological Independence, Federal Scientific and Technical Program on the Development of Genetic Technologies, 630559 Novosibirsk, Russia; (I.A.M.); (M.B.B.); (D.V.S.); (A.P.R.); (L.I.K.); (S.V.B.); (A.A.I.); (V.S.N.); (E.I.K.); (E.A.V.); (T.I.E.); (A.V.Z.); (O.V.P.); (A.A.I.)
| | - Daniil V. Shanshin
- State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, World-Class Genomic Research Center for Biological Safety and Technological Independence, Federal Scientific and Technical Program on the Development of Genetic Technologies, 630559 Novosibirsk, Russia; (I.A.M.); (M.B.B.); (D.V.S.); (A.P.R.); (L.I.K.); (S.V.B.); (A.A.I.); (V.S.N.); (E.I.K.); (E.A.V.); (T.I.E.); (A.V.Z.); (O.V.P.); (A.A.I.)
| | - Andrey P. Rudometov
- State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, World-Class Genomic Research Center for Biological Safety and Technological Independence, Federal Scientific and Technical Program on the Development of Genetic Technologies, 630559 Novosibirsk, Russia; (I.A.M.); (M.B.B.); (D.V.S.); (A.P.R.); (L.I.K.); (S.V.B.); (A.A.I.); (V.S.N.); (E.I.K.); (E.A.V.); (T.I.E.); (A.V.Z.); (O.V.P.); (A.A.I.)
| | - Larisa I. Karpenko
- State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, World-Class Genomic Research Center for Biological Safety and Technological Independence, Federal Scientific and Technical Program on the Development of Genetic Technologies, 630559 Novosibirsk, Russia; (I.A.M.); (M.B.B.); (D.V.S.); (A.P.R.); (L.I.K.); (S.V.B.); (A.A.I.); (V.S.N.); (E.I.K.); (E.A.V.); (T.I.E.); (A.V.Z.); (O.V.P.); (A.A.I.)
| | - Svetlana V. Belenkaya
- State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, World-Class Genomic Research Center for Biological Safety and Technological Independence, Federal Scientific and Technical Program on the Development of Genetic Technologies, 630559 Novosibirsk, Russia; (I.A.M.); (M.B.B.); (D.V.S.); (A.P.R.); (L.I.K.); (S.V.B.); (A.A.I.); (V.S.N.); (E.I.K.); (E.A.V.); (T.I.E.); (A.V.Z.); (O.V.P.); (A.A.I.)
| | - Anastasiya A. Isaeva
- State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, World-Class Genomic Research Center for Biological Safety and Technological Independence, Federal Scientific and Technical Program on the Development of Genetic Technologies, 630559 Novosibirsk, Russia; (I.A.M.); (M.B.B.); (D.V.S.); (A.P.R.); (L.I.K.); (S.V.B.); (A.A.I.); (V.S.N.); (E.I.K.); (E.A.V.); (T.I.E.); (A.V.Z.); (O.V.P.); (A.A.I.)
| | - Valentina S. Nesmeyanova
- State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, World-Class Genomic Research Center for Biological Safety and Technological Independence, Federal Scientific and Technical Program on the Development of Genetic Technologies, 630559 Novosibirsk, Russia; (I.A.M.); (M.B.B.); (D.V.S.); (A.P.R.); (L.I.K.); (S.V.B.); (A.A.I.); (V.S.N.); (E.I.K.); (E.A.V.); (T.I.E.); (A.V.Z.); (O.V.P.); (A.A.I.)
| | - Elena I. Kazachinskaia
- State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, World-Class Genomic Research Center for Biological Safety and Technological Independence, Federal Scientific and Technical Program on the Development of Genetic Technologies, 630559 Novosibirsk, Russia; (I.A.M.); (M.B.B.); (D.V.S.); (A.P.R.); (L.I.K.); (S.V.B.); (A.A.I.); (V.S.N.); (E.I.K.); (E.A.V.); (T.I.E.); (A.V.Z.); (O.V.P.); (A.A.I.)
| | - Ekaterina A. Volosnikova
- State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, World-Class Genomic Research Center for Biological Safety and Technological Independence, Federal Scientific and Technical Program on the Development of Genetic Technologies, 630559 Novosibirsk, Russia; (I.A.M.); (M.B.B.); (D.V.S.); (A.P.R.); (L.I.K.); (S.V.B.); (A.A.I.); (V.S.N.); (E.I.K.); (E.A.V.); (T.I.E.); (A.V.Z.); (O.V.P.); (A.A.I.)
| | - Tatiana I. Esina
- State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, World-Class Genomic Research Center for Biological Safety and Technological Independence, Federal Scientific and Technical Program on the Development of Genetic Technologies, 630559 Novosibirsk, Russia; (I.A.M.); (M.B.B.); (D.V.S.); (A.P.R.); (L.I.K.); (S.V.B.); (A.A.I.); (V.S.N.); (E.I.K.); (E.A.V.); (T.I.E.); (A.V.Z.); (O.V.P.); (A.A.I.)
| | - Anna V. Zaykovskaya
- State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, World-Class Genomic Research Center for Biological Safety and Technological Independence, Federal Scientific and Technical Program on the Development of Genetic Technologies, 630559 Novosibirsk, Russia; (I.A.M.); (M.B.B.); (D.V.S.); (A.P.R.); (L.I.K.); (S.V.B.); (A.A.I.); (V.S.N.); (E.I.K.); (E.A.V.); (T.I.E.); (A.V.Z.); (O.V.P.); (A.A.I.)
| | - Oleg V. Pyankov
- State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, World-Class Genomic Research Center for Biological Safety and Technological Independence, Federal Scientific and Technical Program on the Development of Genetic Technologies, 630559 Novosibirsk, Russia; (I.A.M.); (M.B.B.); (D.V.S.); (A.P.R.); (L.I.K.); (S.V.B.); (A.A.I.); (V.S.N.); (E.I.K.); (E.A.V.); (T.I.E.); (A.V.Z.); (O.V.P.); (A.A.I.)
| | - Sophia S. Borisevich
- Laboratory of Chemical Physics, Ufa Institute of Chemistry, Ufa Federal Research Center, 450078 Ufa, Russia;
| | - Arseniya A. Shelemba
- Federal Research Center of Fundamental and Translational Medicine, 630060 Novosibirsk, Russia;
| | - Anton N. Chikaev
- Institute of Molecular and Cellular Biology, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia;
| | - Alexander A. Ilyichev
- State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, World-Class Genomic Research Center for Biological Safety and Technological Independence, Federal Scientific and Technical Program on the Development of Genetic Technologies, 630559 Novosibirsk, Russia; (I.A.M.); (M.B.B.); (D.V.S.); (A.P.R.); (L.I.K.); (S.V.B.); (A.A.I.); (V.S.N.); (E.I.K.); (E.A.V.); (T.I.E.); (A.V.Z.); (O.V.P.); (A.A.I.)
| |
Collapse
|
22
|
Krasilnikov IV, Kudriavtsev AV, Vakhrusheva AV, Frolova ME, Ivanov AV, Stukova MA, Romanovskaya-Romanko EA, Vasilyev KA, Mushenkova NV, Isaev AA. Design and Immunological Properties of the Novel Subunit Virus-like Vaccine against SARS-CoV-2. Vaccines (Basel) 2022; 10:vaccines10010069. [PMID: 35062730 PMCID: PMC8782008 DOI: 10.3390/vaccines10010069] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 12/23/2021] [Accepted: 12/27/2021] [Indexed: 02/06/2023] Open
Abstract
The COVID-19 pandemic is ongoing, and the need for safe and effective vaccines to prevent infection and to control spread of the virus remains urgent. Here, we report the development of a SARS-CoV-2 subunit vaccine candidate (Betuvax-CoV-2) based on RBD and SD1 domains of the spike (S) protein fused to a human IgG1 Fc fragment. The antigen is adsorbed on betulin adjuvant, forming spherical particles with a size of 100–180 nm, mimicking the size of viral particles. Here we confirm the potent immunostimulatory activity of betulin adjuvant, and demonstrate that two immunizations of mice with Betuvax-CoV-2 elicited high titers of RBD-specific antibodies. The candidate vaccine was also effective in stimulating a neutralizing antibody response and T cell immunity. The results indicate that Betuvax-CoV-2 has good potential for further development as an effective vaccine against SARS-CoV-2.
Collapse
Affiliation(s)
- Igor V. Krasilnikov
- Department of Vaccinology, Smorodintsev Research Institute of Influenza of the Ministry of Health of the Russian Federation, 197376 Saint Petersburg, Russia; (I.V.K.); (M.A.S.); (E.A.R.-R.); (K.A.V.)
| | | | | | - Maria E. Frolova
- PJSC Human Stem Cells Institute, 129110 Moscow, Russia; (M.E.F.); or
| | | | - Marina A. Stukova
- Department of Vaccinology, Smorodintsev Research Institute of Influenza of the Ministry of Health of the Russian Federation, 197376 Saint Petersburg, Russia; (I.V.K.); (M.A.S.); (E.A.R.-R.); (K.A.V.)
| | - Ekaterina A. Romanovskaya-Romanko
- Department of Vaccinology, Smorodintsev Research Institute of Influenza of the Ministry of Health of the Russian Federation, 197376 Saint Petersburg, Russia; (I.V.K.); (M.A.S.); (E.A.R.-R.); (K.A.V.)
| | - Kirill A. Vasilyev
- Department of Vaccinology, Smorodintsev Research Institute of Influenza of the Ministry of Health of the Russian Federation, 197376 Saint Petersburg, Russia; (I.V.K.); (M.A.S.); (E.A.R.-R.); (K.A.V.)
| | | | - Artur A. Isaev
- PJSC Human Stem Cells Institute, 129110 Moscow, Russia; (M.E.F.); or
- Center of Genetics and Reproductive Medicine “Genetico”, 119333 Moscow, Russia
| |
Collapse
|
23
|
Liu Q, Ding Z, Lan J, Wong G. Design of Replication-Competent VSV- and Ervebo-Vectored Vaccines Against SARS-CoV-2. Methods Mol Biol 2022; 2410:193-208. [PMID: 34914048 DOI: 10.1007/978-1-0716-1884-4_9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is a global public health emergency. Several vaccine candidates have been developed in response to the COVID-19 pandemic. One approach is to construct live-recombinant viruses expressing the SARS-CoV-2 spike protein (S) as vaccine candidates. The vesicular stomatitis virus (VSV) vector is a mature vaccine platform which was successfully developed as a vaccine against Ebola virus (EBOV), leading to its licensure by the Food and Drug Administration (FDA) in December 2019. Based on this work, we developed two live, replication-competent VSV-vectored vaccines against SARS-CoV-2: (1) a VSV expressing the S protein of SARS-CoV-2 and (2) a bivalent VSV expressing the S protein of SARS-CoV-2 and the glycoprotein (GP) of EBOV. This protocol describes the methodologies for the design, cloning, rescue, and preparation of these recombinant VSV vaccines.
Collapse
Affiliation(s)
- Qixing Liu
- Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Zhe Ding
- Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Jiaming Lan
- Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Gary Wong
- Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China.
- Département de microbiologie-infectiologie et d'immunologie, Université Laval, Québec, QC, Canada.
| |
Collapse
|
24
|
Santana-Mederos D, Perez-Nicado R, Climent Y, Rodriguez L, Ramirez BS, Perez-Rodriguez S, Rodriguez M, Labrada C, Hernandez T, Diaz M, Orosa I, Ramirez U, Oliva R, Garrido R, Cardoso F, Landys M, Martinez R, Gonzalez H, Hernandez T, Ochoa-Azze R, Perez JL, Enriquez J, Gonzalez N, Infante Y, Espinosa LA, Ramos Y, González LJ, Valenzuela C, Casadesus AV, Fernandez B, Rojas G, Pérez-Massón B, Tundidor Y, Bermudez E, Plasencia CA, Boggiano T, Ojito E, Chiodo F, Fernandez S, Paquet F, Fang C, Chen GW, Rivera DG, Valdes-Balbin Y, Garcia-Rivera D, Verez Bencomo V. A COVID-19 vaccine candidate composed of the SARS-CoV-2 RBD dimer and Neisseria meningitidis outer membrane vesicles. RSC Chem Biol 2022; 3:242-249. [PMID: 35360883 PMCID: PMC8826971 DOI: 10.1039/d1cb00200g] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 12/01/2021] [Indexed: 02/05/2023] Open
Abstract
Soberana01 is composed of the SARS-CoV-2 dimeric RBD and Neisseria meningitidis outer membrane vesicles (OMVs) adsorbed on alum. This vaccine induces a potent neutralizing immune response and shows potential against SARS-CoV-2 variants of concern.
Collapse
Affiliation(s)
| | | | - Yanet Climent
- Finlay Vaccine Institute, 200 and 21 Street, Havana 11600, Cuba
| | - Laura Rodriguez
- Finlay Vaccine Institute, 200 and 21 Street, Havana 11600, Cuba
| | | | | | - Meybi Rodriguez
- Finlay Vaccine Institute, 200 and 21 Street, Havana 11600, Cuba
| | - Claudia Labrada
- Finlay Vaccine Institute, 200 and 21 Street, Havana 11600, Cuba
| | - Tays Hernandez
- Center of Molecular Immunology, P.O. Box 16040, 216 St., Havana, Cuba
| | - Marianniz Diaz
- Center of Molecular Immunology, P.O. Box 16040, 216 St., Havana, Cuba
| | - Ivette Orosa
- Center of Molecular Immunology, P.O. Box 16040, 216 St., Havana, Cuba
| | - Ubel Ramirez
- Finlay Vaccine Institute, 200 and 21 Street, Havana 11600, Cuba
| | - Reynaldo Oliva
- Finlay Vaccine Institute, 200 and 21 Street, Havana 11600, Cuba
| | - Raine Garrido
- Finlay Vaccine Institute, 200 and 21 Street, Havana 11600, Cuba
| | - Felix Cardoso
- Finlay Vaccine Institute, 200 and 21 Street, Havana 11600, Cuba
| | - Mario Landys
- Finlay Vaccine Institute, 200 and 21 Street, Havana 11600, Cuba
| | | | | | | | | | - Jose L. Perez
- Finlay Vaccine Institute, 200 and 21 Street, Havana 11600, Cuba
| | - Juliet Enriquez
- National Civil Defense Research Laboratory, Mayabeque 32700, Cuba
| | - Nibaldo Gonzalez
- National Civil Defense Research Laboratory, Mayabeque 32700, Cuba
| | - Yenicet Infante
- National Civil Defense Research Laboratory, Mayabeque 32700, Cuba
| | - Luis A. Espinosa
- Center for Genetic Engineering and Biotechnology, Ave 31 e/158 y 190, Havana 10600, Cuba
| | - Yassel Ramos
- Center for Genetic Engineering and Biotechnology, Ave 31 e/158 y 190, Havana 10600, Cuba
| | - Luis Javier González
- Center for Genetic Engineering and Biotechnology, Ave 31 e/158 y 190, Havana 10600, Cuba
| | - Carmen Valenzuela
- Institute of Cybernetics, Mathematics and Physics, Havana 10400, Cuba
| | | | - Briandy Fernandez
- Center of Molecular Immunology, P.O. Box 16040, 216 St., Havana, Cuba
| | - Gertrudis Rojas
- Center of Molecular Immunology, P.O. Box 16040, 216 St., Havana, Cuba
| | | | - Yaima Tundidor
- Center of Molecular Immunology, P.O. Box 16040, 216 St., Havana, Cuba
| | - Ernesto Bermudez
- Center of Molecular Immunology, P.O. Box 16040, 216 St., Havana, Cuba
| | | | - Tammy Boggiano
- Center of Molecular Immunology, P.O. Box 16040, 216 St., Havana, Cuba
| | - Eduardo Ojito
- Center of Molecular Immunology, P.O. Box 16040, 216 St., Havana, Cuba
| | - Fabrizio Chiodo
- Finlay Vaccine Institute, 200 and 21 Street, Havana 11600, Cuba
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands and Institute of Biomolecular Chemistry, National Research Council (CNR), Pozzuoli, Napoli, Italy
| | | | - Françoise Paquet
- Centre de Biophysique Moléculaire, CNRS UPR 4301, rue Charles Sadron, F-45071, Orléans Cedex 2, France
| | - Cheng Fang
- Shanghai Fenglin Glycodrug Promotion Center, Shanghai 200032, China
| | - Guang-Wu Chen
- Chengdu Olisynn Biotech. Co. Ltd., and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China
| | - Daniel G. Rivera
- Laboratory of Synthetic and Biomolecular Chemistry, Faculty of Chemistry, University of Havana, Zapata y G, Havana 10400, Cuba
| | | | | | | |
Collapse
|
25
|
Long-term stability and protection efficacy of the RBD-targeting COVID-19 mRNA vaccine in nonhuman primates. Signal Transduct Target Ther 2021; 6:438. [PMID: 34952914 PMCID: PMC8703211 DOI: 10.1038/s41392-021-00861-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 10/28/2021] [Accepted: 12/07/2021] [Indexed: 01/08/2023] Open
Abstract
Messenger RNA (mRNA) vaccine technology has shown its power in preventing the ongoing COVID-19 pandemic. Two mRNA vaccines targeting the full-length S protein of SARS-CoV-2 have been authorized for emergency use. Recently, we have developed a lipid nanoparticle-encapsulated mRNA (mRNA-LNP) encoding the receptor-binding domain (RBD) of SARS-CoV-2 (termed ARCoV), which confers complete protection in mouse model. Herein, we further characterized the protection efficacy of ARCoV in nonhuman primates and the long-term stability under normal refrigerator temperature. Intramuscular immunization of two doses of ARCoV elicited robust neutralizing antibodies as well as cellular response against SARS-CoV-2 in cynomolgus macaques. More importantly, ARCoV vaccination in macaques significantly protected animals from acute lung lesions caused by SARS-CoV-2, and viral replication in lungs and secretion in nasal swabs were completely cleared in all animals immunized with low or high doses of ARCoV. No evidence of antibody-dependent enhancement of infection was observed throughout the study. Finally, extensive stability assays showed that ARCoV can be stored at 2-8 °C for at least 6 months without decrease of immunogenicity. All these promising results strongly support the ongoing clinical trial.
Collapse
|
26
|
Xu K, An Y, Li Q, Huang W, Han Y, Zheng T, Fang F, Liu H, Liu C, Gao P, Xu S, Liu X, Zhang R, Zhao X, Liu WJ, Bi Y, Wang Y, Zhou D, Wang Q, Hou W, Xia Q, Gao GF, Dai L. Recombinant chimpanzee adenovirus AdC7 expressing dimeric tandem-repeat spike protein RBD protects mice against COVID-19. Emerg Microbes Infect 2021; 10:1574-1588. [PMID: 34289779 PMCID: PMC8366625 DOI: 10.1080/22221751.2021.1959270] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 06/29/2021] [Accepted: 07/19/2021] [Indexed: 12/19/2022]
Abstract
A safe and effective vaccine is urgently needed to control the unprecedented COVID-19 pandemic. Four adenovirus-vectored vaccines expressing spike (S) protein have been approved for use. Here, we generated several recombinant chimpanzee adenovirus (AdC7) vaccines expressing S, receptor-binding domain (RBD), or tandem-repeat dimeric RBD (RBD-tr2). We found vaccination via either intramuscular or intranasal route was highly immunogenic in mice to elicit both humoral and cellular immune responses. AdC7-RBD-tr2 showed higher antibody responses compared to either AdC7-S or AdC7-RBD. Intranasal administration of AdC7-RBD-tr2 additionally induced mucosal immunity with neutralizing activity in bronchoalveolar lavage fluid. Either single-dose or two-dose mucosal administration of AdC7-RBD-tr2 protected mice against SARS-CoV-2 challenge, with undetectable subgenomic RNA in lung and relieved lung injury. AdC7-RBD-tr2-elicted sera preserved the neutralizing activity against the circulating variants, especially the Delta variant. These results support AdC7-RBD-tr2 as a promising COVID-19 vaccine candidate.
Collapse
Affiliation(s)
- Kun Xu
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, School of Tropical Medicine and Laboratory Medicine, The First Affiliated Hospital, Hainan Medical University, Haikou, People’s Republic of China
- Research Network of Immunity and Health (RNIH), Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Yaling An
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Qunlong Li
- Chengdu Kanghua Biological Products Co., Ltd, Chengdu, People’s Republic of China
| | - Weijin Huang
- Division of HIV/AIDS and Sex-Transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing, People’s Republic of China
| | - Yuxuan Han
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Tianyi Zheng
- Research Network of Immunity and Health (RNIH), Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Fang Fang
- Chengdu Kanghua Biological Products Co., Ltd, Chengdu, People’s Republic of China
| | - Hui Liu
- Chengdu Kanghua Biological Products Co., Ltd, Chengdu, People’s Republic of China
| | - Chuanyu Liu
- Laboratory of Animal Infectious Diseases, College of Animal Sciences and Veterinary Medicine, Guangxi University, Nanning, People’s Republic of China
| | - Ping Gao
- Research Network of Immunity and Health (RNIH), Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Senyu Xu
- Research Network of Immunity and Health (RNIH), Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Xueyuan Liu
- School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, People’s Republic of China
| | - Rong Zhang
- Laboratory of Animal Infectious Diseases, College of Animal Sciences and Veterinary Medicine, Guangxi University, Nanning, People’s Republic of China
| | - Xin Zhao
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, People’s Republic of China
- CAS Center for Influenza Research and Early-Warning (CASCIRE), CAS-TWAS Center of Excellence for Emerging Infectious Diseases (CEEID), Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - William J. Liu
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, People’s Republic of China
| | - Yuhai Bi
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, People’s Republic of China
- CAS Center for Influenza Research and Early-Warning (CASCIRE), CAS-TWAS Center of Excellence for Emerging Infectious Diseases (CEEID), Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Youchun Wang
- Division of HIV/AIDS and Sex-Transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing, People’s Republic of China
| | - Dongming Zhou
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, People’s Republic of China
| | - Qinghan Wang
- Chengdu Kanghua Biological Products Co., Ltd, Chengdu, People’s Republic of China
| | - Wenli Hou
- Chengdu Kanghua Biological Products Co., Ltd, Chengdu, People’s Republic of China
| | - Qianfeng Xia
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, School of Tropical Medicine and Laboratory Medicine, The First Affiliated Hospital, Hainan Medical University, Haikou, People’s Republic of China
| | - George F. Gao
- Research Network of Immunity and Health (RNIH), Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing, People’s Republic of China
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, People’s Republic of China
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, People’s Republic of China
| | - Lianpan Dai
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, School of Tropical Medicine and Laboratory Medicine, The First Affiliated Hospital, Hainan Medical University, Haikou, People’s Republic of China
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, People’s Republic of China
| |
Collapse
|
27
|
Ortega-Rivera OA, Shukla S, Shin MD, Chen A, Beiss V, Moreno-Gonzalez MA, Zheng Y, Clark AE, Carlin AF, Pokorski JK, Steinmetz NF. Cowpea Mosaic Virus Nanoparticle Vaccine Candidates Displaying Peptide Epitopes Can Neutralize the Severe Acute Respiratory Syndrome Coronavirus. ACS Infect Dis 2021; 7:3096-3110. [PMID: 34672530 PMCID: PMC8547496 DOI: 10.1021/acsinfecdis.1c00410] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Indexed: 12/28/2022]
Abstract
The development of vaccines against coronaviruses has focused on the spike (S) protein, which is required for the recognition of host-cell receptors and thus elicits neutralizing antibodies. Targeting conserved epitopes on the S protein offers the potential for pan-beta-coronavirus vaccines that could prevent future pandemics. We displayed five B-cell epitopes, originally identified in the convalescent sera from recovered severe acute respiratory syndrome (SARS) patients, on the surface of the cowpea mosaic virus (CPMV) and evaluated these formulations as vaccines. Prime-boost immunization of mice with three of these candidate vaccines, CPMV-988, CPMV-1173, and CPMV-1209, elicited high antibody titers that neutralized the severe acute respiratory syndrome coronavirus (SARS-CoV) in vitro and showed an early Th1-biased profile (2-4 weeks) transitioning to a slightly Th2-biased profile just after the second boost (6 weeks). A pentavalent slow-release implant comprising all five peptides displayed on the CPMV elicited anti-S protein and epitope-specific antibody titers, albeit at a lower magnitude compared to the soluble formulations. While the CPMV remained intact when released from the PLGA implants, processing results in loss of RNA, which acts as an adjuvant. Loss of RNA may be a reason for the lower efficacy of the implants. Finally, although the three epitopes (988, 1173, and 1209) that were found to be neutralizing the SARS-CoV were 100% identical to the SARS-CoV-2, none of the vaccine candidates neutralized the SARS-CoV-2 in vitro suggesting differences in the natural epitope perhaps caused by conformational changes or the presence of N-linked glycans. While a cross-protective vaccine candidate was not developed, a multivalent SARS vaccine was developed. The technology discussed here is a versatile vaccination platform that can be pivoted toward other diseases and applications that are not limited to infectious diseases.
Collapse
Affiliation(s)
- Oscar A. Ortega-Rivera
- Department of NanoEngineering, University
of California-San Diego, La Jolla, California 92039, United
States
- Center for Nano-ImmunoEngineering,
University of California-San Diego, La Jolla, California
92039, United States
| | - Sourabh Shukla
- Department of NanoEngineering, University
of California-San Diego, La Jolla, California 92039, United
States
- Center for Nano-ImmunoEngineering,
University of California-San Diego, La Jolla, California
92039, United States
| | - Matthew D. Shin
- Department of NanoEngineering, University
of California-San Diego, La Jolla, California 92039, United
States
- Center for Nano-ImmunoEngineering,
University of California-San Diego, La Jolla, California
92039, United States
| | - Angela Chen
- Department of NanoEngineering, University
of California-San Diego, La Jolla, California 92039, United
States
- Center for Nano-ImmunoEngineering,
University of California-San Diego, La Jolla, California
92039, United States
| | - Veronique Beiss
- Department of NanoEngineering, University
of California-San Diego, La Jolla, California 92039, United
States
- Center for Nano-ImmunoEngineering,
University of California-San Diego, La Jolla, California
92039, United States
| | - Miguel A. Moreno-Gonzalez
- Department of NanoEngineering, University
of California-San Diego, La Jolla, California 92039, United
States
- Center for Nano-ImmunoEngineering,
University of California-San Diego, La Jolla, California
92039, United States
| | - Yi Zheng
- Department of NanoEngineering, University
of California-San Diego, La Jolla, California 92039, United
States
- Center for Nano-ImmunoEngineering,
University of California-San Diego, La Jolla, California
92039, United States
| | - Alex E. Clark
- Department of Medicine, University of
California-San Diego, La Jolla, California 92039, United
States
| | - Aaron F. Carlin
- Department of Medicine, University of
California-San Diego, La Jolla, California 92039, United
States
| | - Jonathan K. Pokorski
- Department of NanoEngineering, University
of California-San Diego, La Jolla, California 92039, United
States
- Center for Nano-ImmunoEngineering,
University of California-San Diego, La Jolla, California
92039, United States
- Institute for Materials Discovery and Design,
University of California-San Diego, La Jolla, California
92039, United States
| | - Nicole F. Steinmetz
- Department of NanoEngineering, University
of California-San Diego, La Jolla, California 92039, United
States
- Center for Nano-ImmunoEngineering,
University of California-San Diego, La Jolla, California
92039, United States
- Institute for Materials Discovery and Design,
University of California-San Diego, La Jolla, California
92039, United States
- Department of Bioengineering, University
of California-San Diego, La Jolla, California 92039, United
States
- Department of Radiology, University of
California-San Diego, La Jolla, California 92039, United
States
- Moores Cancer Center, University of
California-San Diego, La Jolla, California 92039, United
States
| |
Collapse
|
28
|
Clark NM, Janaka SK, Hartman W, Stramer S, Goodhue E, Weiss J, Evans DT, Connor JP. Anti-SARS-CoV-2 IgG and IgA antibodies in COVID-19 convalescent plasma do not facilitate antibody-dependent enhance of viral infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021. [PMID: 34545365 PMCID: PMC8452094 DOI: 10.1101/2021.09.14.460394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The novel coronavirus SARS-CoV2, which causes COVID-19, has resulted in the death of nearly 4 million people within the last 18 months. While preventive vaccination and monoclonal antibody therapies have been rapidly developed and deployed, early in the pandemic the use of COVID-19 convalescent plasma (CCP) was a common means of passive immunization, with the theoretical risk of antibody-dependent enhancement (ADE) of viral infection remaining undetermined. Though vaccines elicit a strong and protective immune response, and transfusion of CCP with high titers of neutralization activity are correlated with better clinical outcomes, the question of whether antibodies in CCP can enhance infection of SARS-CoV2 has not been directly addressed. In this study, we analyzed for and observed passive transfer of neutralization activity with CCP transfusion. Furthermore, to specifically understand if antibodies against the spike protein (S) enhance infection, we measured the anti-S IgG, IgA, and IgM responses and adapted retroviral-pseudotypes to measure virus neutralization with target cells expressing the ACE2 virus receptor and the Fc alpha receptor (FcαR) or Fc gamma receptor IIA (FcγRIIA). Whereas neutralizing activity of CCP correlated best with higher titers of anti-S IgG antibodies, the neutralizing titer was not affected when Fc receptors were present on target cells. These observations support the absence of antibody-dependent enhancement of infection (ADE) by IgG and IgA isotypes found in CCP. The results presented, therefore, support the clinical use of currently available antibody-based treatment including the continued study of CCP transfusion strategies.
Collapse
|
29
|
Lv H, Tsang OT, So RTY, Wang Y, Yuan M, Liu H, Yip GK, Teo QW, Lin Y, Liang W, Wang J, Ng WW, Wilson IA, Peiris JSM, Wu NC, Mok CKP. Homologous and heterologous serological response to the N-terminal domain of SARS-CoV-2 in humans and mice. Eur J Immunol 2021; 51:2296-2305. [PMID: 34089541 PMCID: PMC8237060 DOI: 10.1002/eji.202149234] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 04/14/2021] [Accepted: 05/31/2021] [Indexed: 12/23/2022]
Abstract
The increasing numbers of infected cases of coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) poses serious threats to public health and the global economy. Most SARS-CoV-2 neutralizing antibodies target the receptor binding domain (RBD) and some the N-terminal domain (NTD) of the spike protein, which is the major antigen of SARS-CoV-2. While the antibody response to RBD has been extensively characterized, the antigenicity and immunogenicity of the NTD protein are less well studied. Using 227 plasma samples from COVID-19 patients, we showed that SARS-CoV-2 NTD-specific antibodies could be induced during infection. As compared to the results of SARS-CoV-2 RBD, the serological response of SARS-CoV-2 NTD is less cross-reactive with SARS-CoV, a pandemic strain that was identified in 2003. Furthermore, neutralizing antibodies are rarely elicited in a mice model when NTD is used as an immunogen. We subsequently demonstrate that NTD has an altered antigenicity when expressed alone. Overall, our results suggest that while NTD offers a supplementary strategy for serology testing, it may not be suitable as an immunogen for vaccine development.
Collapse
Affiliation(s)
- Huibin Lv
- HKU‐Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of MedicineThe University of Hong KongHong KongSARChina
| | - Owen Tak‐Yin Tsang
- Infectious Diseases Centre, Princess Margaret HospitalHospital Authority of Hong Kong
| | - Ray T. Y. So
- HKU‐Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of MedicineThe University of Hong KongHong KongSARChina
- School of Public Health, Li Ka Shing Faculty of MedicineThe University of Hong KongHong KongSARChina
| | - Yiquan Wang
- HKU‐Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of MedicineThe University of Hong KongHong KongSARChina
- Department of BiochemistryUniversity of Illinois at Urbana‐ChampaignUrbanaIllinoisUSA
| | - Meng Yuan
- Department of Integrative Structural and Computational BiologyThe Scripps Research InstituteLa JollaCaliforniaUSA
| | - Hejun Liu
- Department of Integrative Structural and Computational BiologyThe Scripps Research InstituteLa JollaCaliforniaUSA
| | - Garrick K. Yip
- HKU‐Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of MedicineThe University of Hong KongHong KongSARChina
| | - Qi Wen Teo
- HKU‐Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of MedicineThe University of Hong KongHong KongSARChina
| | - Yihan Lin
- HKU‐Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of MedicineThe University of Hong KongHong KongSARChina
| | - Weiwen Liang
- HKU‐Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of MedicineThe University of Hong KongHong KongSARChina
| | - Jinlin Wang
- HKU‐Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of MedicineThe University of Hong KongHong KongSARChina
| | - Wilson W. Ng
- HKU‐Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of MedicineThe University of Hong KongHong KongSARChina
| | - Ian A. Wilson
- Department of Integrative Structural and Computational BiologyThe Scripps Research InstituteLa JollaCaliforniaUSA
- The Skaggs Institute for Chemical BiologyThe Scripps Research InstituteLa JollaCaliforniaUSA
| | - J. S. Malik Peiris
- HKU‐Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of MedicineThe University of Hong KongHong KongSARChina
- School of Public Health, Li Ka Shing Faculty of MedicineThe University of Hong KongHong KongSARChina
| | - Nicholas C. Wu
- Department of BiochemistryUniversity of Illinois at Urbana‐ChampaignUrbanaIllinoisUSA
- Carl R. Woese Institute for Genomic BiologyUniversity of Illinois at Urbana‐ChampaignUrbanaIllinoisUSA
- Center for Biophysics and Quantitative BiologyUniversity of Illinois at Urbana‐ChampaignUrbanaIllinoisUSA
| | - Chris K. P. Mok
- HKU‐Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of MedicineThe University of Hong KongHong KongSARChina
- Li Ka Shing Institute of Health SciencesFaculty of MedicineThe Chinese University of Hong KongShatinHong Kong SARChina
- The Jockey Club School of Public Health and Primary CareThe Chinese University of Hong KongHong KongSARChina
| |
Collapse
|
30
|
Núñez-Muñoz L, Marcelino-Pérez G, Calderón-Pérez B, Pérez-Saldívar M, Acosta-Virgen K, González-Conchillos H, Vargas-Hernández B, Olivares-Martínez A, Ruiz-Medrano R, Roa-Velázquez D, Morales-Ríos E, Ramos-Flores J, Torres-Franco G, Peláez-González D, Fernández-Hernández J, Espinosa-Cantellano M, Tapia-Sidas D, Ramírez-Pool JA, Padilla-Viveros A, Xoconostle-Cázares B. Recombinant Antigens Based on Non-Glycosylated Regions from RBD SARS-CoV-2 as Potential Vaccine Candidates against COVID-19. Vaccines (Basel) 2021; 9:928. [PMID: 34452053 PMCID: PMC8402574 DOI: 10.3390/vaccines9080928] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 08/17/2021] [Accepted: 08/18/2021] [Indexed: 01/22/2023] Open
Abstract
The Receptor-Binding Domain (RBD) of the Spike (S) protein from Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) has glycosylation sites which can limit the production of reliable antigens expressed in prokaryotic platforms, due to glycan-mediated evasion of the host immune response. However, protein regions without glycosylated residues capable of inducing neutralizing antibodies could be useful for antigen production in systems that do not carry the glycosylation machinery. To test this hypothesis, the potential antigens NG06 and NG19, located within the non-glycosylated S-RBD region, were selected and expressed in Escherichia coli, purified by FPLC and employed to determine their immunogenic potential through detection of antibodies in serum from immunized rabbits, mice, and COVID-19 patients. IgG antibodies from sera of COVID-19-recovered patients detected the recombinant antigens NG06 and NG19 (A450 nm = 0.80 ± 0.33; 1.13 ± 0.33; and 0.11 ± 0.08 for and negatives controls, respectively). Also, the purified antigens were able to raise polyclonal antibodies in animal models evoking a strong immune response with neutralizing activity in mice model. This research highlights the usefulness of antigens based on the non-N-glycosylated region of RBD from SARS-CoV-2 for candidate vaccine development.
Collapse
Affiliation(s)
- Leandro Núñez-Muñoz
- Department of Biotechnology and Bioengineering, Centro de Investigación y de Estudios Avanzados (CINVESTAV), Av. Instituto Politécnico Nacional 2508, México City 07360, Mexico; (L.N.-M.); (G.M.-P.); (B.C.-P.); (B.V.-H.); (A.O.-M.); (R.R.-M.); (D.T.-S.); (J.A.R.-P.)
| | - Gabriel Marcelino-Pérez
- Department of Biotechnology and Bioengineering, Centro de Investigación y de Estudios Avanzados (CINVESTAV), Av. Instituto Politécnico Nacional 2508, México City 07360, Mexico; (L.N.-M.); (G.M.-P.); (B.C.-P.); (B.V.-H.); (A.O.-M.); (R.R.-M.); (D.T.-S.); (J.A.R.-P.)
- Doctoral Program in Nanosciences and Nanotechnology, Centro de Investigación y de Estudios Avanzados (CINVESTAV), Av. Instituto Politécnico Nacional 2508, México City 07360, Mexico;
| | - Berenice Calderón-Pérez
- Department of Biotechnology and Bioengineering, Centro de Investigación y de Estudios Avanzados (CINVESTAV), Av. Instituto Politécnico Nacional 2508, México City 07360, Mexico; (L.N.-M.); (G.M.-P.); (B.C.-P.); (B.V.-H.); (A.O.-M.); (R.R.-M.); (D.T.-S.); (J.A.R.-P.)
| | - Miriam Pérez-Saldívar
- Department of Infectomics and Molecular Pathogenesis, Centro de Investigación y de Estudios Avanzados (CINVESTAV), Av. Instituto Politécnico Nacional 2508, México City 07360, Mexico; (M.P.-S.); (K.A.-V.); (H.G.-C.); (M.E.-C.)
| | - Karla Acosta-Virgen
- Department of Infectomics and Molecular Pathogenesis, Centro de Investigación y de Estudios Avanzados (CINVESTAV), Av. Instituto Politécnico Nacional 2508, México City 07360, Mexico; (M.P.-S.); (K.A.-V.); (H.G.-C.); (M.E.-C.)
| | - Hugo González-Conchillos
- Department of Infectomics and Molecular Pathogenesis, Centro de Investigación y de Estudios Avanzados (CINVESTAV), Av. Instituto Politécnico Nacional 2508, México City 07360, Mexico; (M.P.-S.); (K.A.-V.); (H.G.-C.); (M.E.-C.)
| | - Brenda Vargas-Hernández
- Department of Biotechnology and Bioengineering, Centro de Investigación y de Estudios Avanzados (CINVESTAV), Av. Instituto Politécnico Nacional 2508, México City 07360, Mexico; (L.N.-M.); (G.M.-P.); (B.C.-P.); (B.V.-H.); (A.O.-M.); (R.R.-M.); (D.T.-S.); (J.A.R.-P.)
| | - Ana Olivares-Martínez
- Department of Biotechnology and Bioengineering, Centro de Investigación y de Estudios Avanzados (CINVESTAV), Av. Instituto Politécnico Nacional 2508, México City 07360, Mexico; (L.N.-M.); (G.M.-P.); (B.C.-P.); (B.V.-H.); (A.O.-M.); (R.R.-M.); (D.T.-S.); (J.A.R.-P.)
| | - Roberto Ruiz-Medrano
- Department of Biotechnology and Bioengineering, Centro de Investigación y de Estudios Avanzados (CINVESTAV), Av. Instituto Politécnico Nacional 2508, México City 07360, Mexico; (L.N.-M.); (G.M.-P.); (B.C.-P.); (B.V.-H.); (A.O.-M.); (R.R.-M.); (D.T.-S.); (J.A.R.-P.)
| | - Daniela Roa-Velázquez
- Doctoral Program in Nanosciences and Nanotechnology, Centro de Investigación y de Estudios Avanzados (CINVESTAV), Av. Instituto Politécnico Nacional 2508, México City 07360, Mexico;
- Department of Biochemistry, Centro de Investigación y de Estudios Avanzados (CINVESTAV), Av. Instituto Politécnico Nacional 2508, México City 07360, Mexico;
| | - Edgar Morales-Ríos
- Department of Biochemistry, Centro de Investigación y de Estudios Avanzados (CINVESTAV), Av. Instituto Politécnico Nacional 2508, México City 07360, Mexico;
| | - Jorge Ramos-Flores
- Laboratory Animal Production and Experimentation Unit, Centro de Investigación y de Estudios Avanzados (CINVESTAV), Av. Instituto Politécnico Nacional 2508, México City 07360, Mexico; (J.R.-F.); (G.T.-F.); (D.P.-G.); (J.F.-H.)
| | - Gustavo Torres-Franco
- Laboratory Animal Production and Experimentation Unit, Centro de Investigación y de Estudios Avanzados (CINVESTAV), Av. Instituto Politécnico Nacional 2508, México City 07360, Mexico; (J.R.-F.); (G.T.-F.); (D.P.-G.); (J.F.-H.)
| | - Diana Peláez-González
- Laboratory Animal Production and Experimentation Unit, Centro de Investigación y de Estudios Avanzados (CINVESTAV), Av. Instituto Politécnico Nacional 2508, México City 07360, Mexico; (J.R.-F.); (G.T.-F.); (D.P.-G.); (J.F.-H.)
| | - Jorge Fernández-Hernández
- Laboratory Animal Production and Experimentation Unit, Centro de Investigación y de Estudios Avanzados (CINVESTAV), Av. Instituto Politécnico Nacional 2508, México City 07360, Mexico; (J.R.-F.); (G.T.-F.); (D.P.-G.); (J.F.-H.)
| | - Martha Espinosa-Cantellano
- Department of Infectomics and Molecular Pathogenesis, Centro de Investigación y de Estudios Avanzados (CINVESTAV), Av. Instituto Politécnico Nacional 2508, México City 07360, Mexico; (M.P.-S.); (K.A.-V.); (H.G.-C.); (M.E.-C.)
| | - Diana Tapia-Sidas
- Department of Biotechnology and Bioengineering, Centro de Investigación y de Estudios Avanzados (CINVESTAV), Av. Instituto Politécnico Nacional 2508, México City 07360, Mexico; (L.N.-M.); (G.M.-P.); (B.C.-P.); (B.V.-H.); (A.O.-M.); (R.R.-M.); (D.T.-S.); (J.A.R.-P.)
| | - José Abrahan Ramírez-Pool
- Department of Biotechnology and Bioengineering, Centro de Investigación y de Estudios Avanzados (CINVESTAV), Av. Instituto Politécnico Nacional 2508, México City 07360, Mexico; (L.N.-M.); (G.M.-P.); (B.C.-P.); (B.V.-H.); (A.O.-M.); (R.R.-M.); (D.T.-S.); (J.A.R.-P.)
| | - América Padilla-Viveros
- Transdisciplinary Doctoral Program in Scientific and Technological Development for Society, Centro de Investigación y de Estudios Avanzados (CINVESTAV), Av. Instituto Politécnico Nacional 2508, México City 07360, Mexico;
| | - Beatriz Xoconostle-Cázares
- Department of Biotechnology and Bioengineering, Centro de Investigación y de Estudios Avanzados (CINVESTAV), Av. Instituto Politécnico Nacional 2508, México City 07360, Mexico; (L.N.-M.); (G.M.-P.); (B.C.-P.); (B.V.-H.); (A.O.-M.); (R.R.-M.); (D.T.-S.); (J.A.R.-P.)
| |
Collapse
|
31
|
Yeast-produced RBD-based recombinant protein vaccines elicit broadly neutralizing antibodies and durable protective immunity against SARS-CoV-2 infection. Cell Discov 2021; 7:71. [PMID: 34408130 PMCID: PMC8372230 DOI: 10.1038/s41421-021-00315-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 07/21/2021] [Indexed: 12/15/2022] Open
Abstract
Massive production of efficacious SARS-CoV-2 vaccines is essential for controlling the ongoing COVID-19 pandemic. We report here the preclinical development of yeast-produced receptor-binding domain (RBD)-based recombinant protein SARS-CoV-2 vaccines. We found that monomeric RBD of SARS-CoV-2 could be efficiently produced as a secreted protein from transformed Pichia pastoris (P. pastoris) yeast. Yeast-derived RBD-monomer possessed functional conformation and was able to elicit protective level of neutralizing antibodies in mice. We further designed and expressed a genetically linked dimeric RBD protein in yeast. The engineered dimeric RBD was more potent than the monomeric RBD in inducing long-lasting neutralizing antibodies. Mice immunized with either monomeric RBD or dimeric RBD were effectively protected from live SARS-CoV-2 virus challenge even at 18 weeks after the last vaccine dose. Importantly, we found that the antisera raised against the RBD of a single SARS-CoV-2 prototype strain could effectively neutralize the two predominant circulating variants B.1.1.7 and B.1.351, implying broad-spectrum protective potential of the RBD-based vaccines. Our data demonstrate that yeast-derived RBD-based recombinant SARS-CoV-2 vaccines are feasible and efficacious, opening up a new avenue for rapid and cost-effective production of SARS-CoV-2 vaccines to achieve global immunization.
Collapse
|
32
|
Yang Y, Zang J, Xu S, Zhang X, Yuan S, Wang H, Lavillette D, Zhang C, Huang Z. Elicitation of Broadly Neutralizing Antibodies against B.1.1.7, B.1.351, and B.1.617.1 SARS-CoV-2 Variants by Three Prototype Strain-Derived Recombinant Protein Vaccines. Viruses 2021; 13:v13081421. [PMID: 34452287 PMCID: PMC8402859 DOI: 10.3390/v13081421] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 07/12/2021] [Accepted: 07/14/2021] [Indexed: 11/16/2022] Open
Abstract
The ongoing coronavirus disease 2019 (COVID-19) pandemic is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Most of the currently approved SARS-CoV-2 vaccines use the prototype strain-derived spike (S) protein or its receptor-binding domain (RBD) as the vaccine antigen. The emergence of several novel SARS-CoV-2 variants has raised concerns about potential immune escape. In this study, we performed an immunogenicity comparison of prototype strain-derived RBD, S1, and S ectodomain trimer (S-trimer) antigens and evaluated their induction of neutralizing antibodies against three circulating SARS-CoV-2 variants, including B.1.1.7, B.1.351, and B.1.617.1. We found that, at the same antigen dose, the RBD and S-trimer vaccines were more potent than the S1 vaccine in eliciting long-lasting, high-titer broadly neutralizing antibodies in mice. The RBD immune sera remained highly effective against the B.1.1.7, B.1.351, and B.1.617.1 variants despite the corresponding neutralizing titers decreasing by 1.2-, 2.8-, and 3.5-fold relative to that against the wild-type strain. Significantly, the S-trimer immune sera exhibited comparable neutralization potency (less than twofold variation in neutralizing GMTs) towards the prototype strain and all three variants tested. These findings provide valuable information for further development of recombinant protein-based SARS-CoV-2 vaccines and support the continued use of currently approved SARS-CoV-2 vaccines in the regions/countries where variant viruses circulate.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Chao Zhang
- Correspondence: (C.Z.); (Z.H.); Tel.: +86-21-54923066 (C.Z.); +86-21-54923067 (Z.H.)
| | - Zhong Huang
- Correspondence: (C.Z.); (Z.H.); Tel.: +86-21-54923066 (C.Z.); +86-21-54923067 (Z.H.)
| |
Collapse
|
33
|
Valdes-Balbin Y, Santana-Mederos D, Quintero L, Fernández S, Rodriguez L, Sanchez Ramirez B, Perez-Nicado R, Acosta C, Méndez Y, Ricardo MG, Hernandez T, Bergado G, Pi F, Valdes A, Carmenate T, Ramirez U, Oliva R, Soubal JP, Garrido R, Cardoso F, Landys M, Gonzalez H, Farinas M, Enriquez J, Noa E, Suarez A, Fang C, Espinosa LA, Ramos Y, González LJ, Climent Y, Rojas G, Relova-Hernández E, Cabrera Infante Y, Losada SL, Boggiano T, Ojito E, León K, Chiodo F, Paquet F, Chen GW, Rivera DG, Garcia-Rivera D, Verez Bencomo V. SARS-CoV-2 RBD-Tetanus Toxoid Conjugate Vaccine Induces a Strong Neutralizing Immunity in Preclinical Studies. ACS Chem Biol 2021; 16:1223-1233. [PMID: 34219448 DOI: 10.1021/acschembio.1c00272] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Controlling the global COVID-19 pandemic depends, among other measures, on developing preventive vaccines at an unprecedented pace. Vaccines approved for use and those in development intend to elicit neutralizing antibodies to block viral sites binding to the host's cellular receptors. Virus infection is mediated by the spike glycoprotein trimer on the virion surface via its receptor binding domain (RBD). Antibody response to this domain is an important outcome of immunization and correlates well with viral neutralization. Here, we show that macromolecular constructs with recombinant RBD conjugated to tetanus toxoid (TT) induce a potent immune response in laboratory animals. Some advantages of immunization with RBD-TT conjugates include a predominant IgG immune response due to affinity maturation and long-term specific B-memory cells. These result demonstrate the potential of the conjugate COVID-19 vaccine candidates and enable their advance to clinical evaluation under the name SOBERANA02, paving the way for other antiviral conjugate vaccines.
Collapse
Affiliation(s)
| | | | - Lauren Quintero
- Finlay Vaccine Institute, 200 and 21 Street, Havana 11600, Cuba
| | | | - Laura Rodriguez
- Finlay Vaccine Institute, 200 and 21 Street, Havana 11600, Cuba
| | | | | | - Claudia Acosta
- Finlay Vaccine Institute, 200 and 21 Street, Havana 11600, Cuba
| | - Yanira Méndez
- Laboratory of Synthetic and Biomolecular Chemistry, Faculty of Chemistry, University of Havana, Zapata y G, Havana 10400, Cuba
| | - Manuel G. Ricardo
- Laboratory of Synthetic and Biomolecular Chemistry, Faculty of Chemistry, University of Havana, Zapata y G, Havana 10400, Cuba
| | - Tays Hernandez
- Center of Molecular Immunology, P.O. Box 16040, 216 St., Havana, Cuba
| | - Gretchen Bergado
- Center of Molecular Immunology, P.O. Box 16040, 216 St., Havana, Cuba
| | - Franciscary Pi
- Center of Molecular Immunology, P.O. Box 16040, 216 St., Havana, Cuba
| | - Annet Valdes
- Center of Molecular Immunology, P.O. Box 16040, 216 St., Havana, Cuba
| | - Tania Carmenate
- Center of Molecular Immunology, P.O. Box 16040, 216 St., Havana, Cuba
| | - Ubel Ramirez
- Finlay Vaccine Institute, 200 and 21 Street, Havana 11600, Cuba
| | - Reinaldo Oliva
- Finlay Vaccine Institute, 200 and 21 Street, Havana 11600, Cuba
| | | | - Raine Garrido
- Finlay Vaccine Institute, 200 and 21 Street, Havana 11600, Cuba
| | - Felix Cardoso
- Finlay Vaccine Institute, 200 and 21 Street, Havana 11600, Cuba
| | - Mario Landys
- Finlay Vaccine Institute, 200 and 21 Street, Havana 11600, Cuba
| | | | - Mildrey Farinas
- Finlay Vaccine Institute, 200 and 21 Street, Havana 11600, Cuba
| | - Juliet Enriquez
- National Civil Defense Research Laboratory, Mayabeque 32700, Cuba
| | - Enrique Noa
- National Civil Defense Research Laboratory, Mayabeque 32700, Cuba
| | - Anamary Suarez
- National Civil Defense Research Laboratory, Mayabeque 32700, Cuba
| | - Cheng Fang
- Shanghai Fenglin Glycodrug Promotion Center, Shanghai 200032, China
| | - Luis A. Espinosa
- Center for Genetic Engineering and Biotechnology, Ave 31 e/158 y 190, Havana 10600, Cuba
| | - Yassel Ramos
- Center for Genetic Engineering and Biotechnology, Ave 31 e/158 y 190, Havana 10600, Cuba
| | - Luis Javier González
- Center for Genetic Engineering and Biotechnology, Ave 31 e/158 y 190, Havana 10600, Cuba
| | - Yanet Climent
- Finlay Vaccine Institute, 200 and 21 Street, Havana 11600, Cuba
| | - Gertrudis Rojas
- Center of Molecular Immunology, P.O. Box 16040, 216 St., Havana, Cuba
| | | | | | - Sum Lai Losada
- Center of Molecular Immunology, P.O. Box 16040, 216 St., Havana, Cuba
| | - Tammy Boggiano
- Center of Molecular Immunology, P.O. Box 16040, 216 St., Havana, Cuba
| | - Eduardo Ojito
- Center of Molecular Immunology, P.O. Box 16040, 216 St., Havana, Cuba
| | - Kalet León
- Center of Molecular Immunology, P.O. Box 16040, 216 St., Havana, Cuba
| | - Fabrizio Chiodo
- Finlay Vaccine Institute, 200 and 21 Street, Havana 11600, Cuba
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam 1081HV, The Netherlands
- Institute of Biomolecular Chemistry, National Research Council (CNR), Pozzuoli 80078, Napoli, Italy
| | - Françoise Paquet
- Centre de Biophysique Moléculaire, CNRS UPR 4301, rue Charles Sadron F-45071, Orléans, Cedex 2, France
| | - Guang-Wu Chen
- Chengdu Olisynn Biotech. Co. Ltd., and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, People’s Republic of China
| | - Daniel G. Rivera
- Laboratory of Synthetic and Biomolecular Chemistry, Faculty of Chemistry, University of Havana, Zapata y G, Havana 10400, Cuba
| | | | | |
Collapse
|
34
|
Smith CC, Olsen KS, Gentry KM, Sambade M, Beck W, Garness J, Entwistle S, Willis C, Vensko S, Woods A, Fini M, Carpenter B, Routh E, Kodysh J, O'Donnell T, Haber C, Heiss K, Stadler V, Garrison E, Sandor AM, Ting JPY, Weiss J, Krajewski K, Grant OC, Woods RJ, Heise M, Vincent BG, Rubinsteyn A. Landscape and selection of vaccine epitopes in SARS-CoV-2. Genome Med 2021; 13:101. [PMID: 34127050 PMCID: PMC8201469 DOI: 10.1186/s13073-021-00910-1] [Citation(s) in RCA: 28] [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: 07/15/2020] [Accepted: 05/14/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Early in the pandemic, we designed a SARS-CoV-2 peptide vaccine containing epitope regions optimized for concurrent B cell, CD4+ T cell, and CD8+ T cell stimulation. The rationale for this design was to drive both humoral and cellular immunity with high specificity while avoiding undesired effects such as antibody-dependent enhancement (ADE). METHODS We explored the set of computationally predicted SARS-CoV-2 HLA-I and HLA-II ligands, examining protein source, concurrent human/murine coverage, and population coverage. Beyond MHC affinity, T cell vaccine candidates were further refined by predicted immunogenicity, sequence conservation, source protein abundance, and coverage of high frequency HLA alleles. B cell epitope regions were chosen from linear epitope mapping studies of convalescent patient serum, followed by filtering for surface accessibility, sequence conservation, spatial localization near functional domains of the spike glycoprotein, and avoidance of glycosylation sites. RESULTS From 58 initial candidates, three B cell epitope regions were identified. From 3730 (MHC-I) and 5045 (MHC-II) candidate ligands, 292 CD8+ and 284 CD4+ T cell epitopes were identified. By combining these B cell and T cell analyses, as well as a manufacturability heuristic, we proposed a set of 22 SARS-CoV-2 vaccine peptides for use in subsequent murine studies. We curated a dataset of ~ 1000 observed T cell epitopes from convalescent COVID-19 patients across eight studies, showing 8/15 recurrent epitope regions to overlap with at least one of our candidate peptides. Of the 22 candidate vaccine peptides, 16 (n = 10 T cell epitope optimized; n = 6 B cell epitope optimized) were manually selected to decrease their degree of sequence overlap and then synthesized. The immunogenicity of the synthesized vaccine peptides was validated using ELISpot and ELISA following murine vaccination. Strong T cell responses were observed in 7/10 T cell epitope optimized peptides following vaccination. Humoral responses were deficient, likely due to the unrestricted conformational space inhabited by linear vaccine peptides. CONCLUSIONS Overall, we find our selection process and vaccine formulation to be appropriate for identifying T cell epitopes and eliciting T cell responses against those epitopes. Further studies are needed to optimize prediction and induction of B cell responses, as well as study the protective capacity of predicted T and B cell epitopes.
Collapse
Affiliation(s)
- Christof C Smith
- Department of Microbiology and Immunology, UNC School of Medicine, Chapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, CB# 7295, Chapel Hill, NC, 27599-7295, USA
| | - Kelly S Olsen
- Department of Microbiology and Immunology, UNC School of Medicine, Chapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, CB# 7295, Chapel Hill, NC, 27599-7295, USA
| | - Kaylee M Gentry
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, CB# 7295, Chapel Hill, NC, 27599-7295, USA
| | - Maria Sambade
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, CB# 7295, Chapel Hill, NC, 27599-7295, USA
| | - Wolfgang Beck
- Department of Microbiology and Immunology, UNC School of Medicine, Chapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, CB# 7295, Chapel Hill, NC, 27599-7295, USA
| | - Jason Garness
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, CB# 7295, Chapel Hill, NC, 27599-7295, USA
| | - Sarah Entwistle
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, CB# 7295, Chapel Hill, NC, 27599-7295, USA
| | - Caryn Willis
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, CB# 7295, Chapel Hill, NC, 27599-7295, USA
| | - Steven Vensko
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, CB# 7295, Chapel Hill, NC, 27599-7295, USA
| | - Allison Woods
- Department of Microbiology and Immunology, UNC School of Medicine, Chapel Hill, NC, USA
| | - Misha Fini
- Department of Microbiology and Immunology, UNC School of Medicine, Chapel Hill, NC, USA
| | - Brandon Carpenter
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, CB# 7295, Chapel Hill, NC, 27599-7295, USA
| | - Eric Routh
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, CB# 7295, Chapel Hill, NC, 27599-7295, USA
| | - Julia Kodysh
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Timothy O'Donnell
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | | | | | - Erik Garrison
- Genomics Institute, University of California, Santa Cruz, CA, USA
| | - Adam M Sandor
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, CB# 7295, Chapel Hill, NC, 27599-7295, USA
| | - Jenny P Y Ting
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, CB# 7295, Chapel Hill, NC, 27599-7295, USA
- Department of Genetics, UNC School of Medicine, Chapel Hill, NC, USA
- Institute for Inflammatory Diseases, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Center for Translational Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jared Weiss
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, CB# 7295, Chapel Hill, NC, 27599-7295, USA
- Division of Medical Oncology, Department of Medicine, UNC School of Medicine, Chapel Hill, NC, USA
| | - Krzysztof Krajewski
- Department of Biochemistry and Biophysics, UNC School of Medicine, Chapel Hill, NC, USA
| | - Oliver C Grant
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, USA
| | - Robert J Woods
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, USA
| | - Mark Heise
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, CB# 7295, Chapel Hill, NC, 27599-7295, USA
- Department of Genetics, UNC School of Medicine, Chapel Hill, NC, USA
| | - Benjamin G Vincent
- Department of Microbiology and Immunology, UNC School of Medicine, Chapel Hill, NC, USA.
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, CB# 7295, Chapel Hill, NC, 27599-7295, USA.
- Computational Medicine Program, UNC School of Medicine, Chapel Hill, NC, USA.
- Curriculum in Bioinformatics and Computational Biology, UNC School of Medicine, Chapel Hill, NC, USA.
- Division of Hematology, Department of Medicine, UNC School of Medicine, Chapel Hill, NC, USA.
| | - Alex Rubinsteyn
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, CB# 7295, Chapel Hill, NC, 27599-7295, USA.
- Department of Genetics, UNC School of Medicine, Chapel Hill, NC, USA.
- Computational Medicine Program, UNC School of Medicine, Chapel Hill, NC, USA.
| |
Collapse
|
35
|
Zhang Q, Xiang R, Huo S, Zhou Y, Jiang S, Wang Q, Yu F. Molecular mechanism of interaction between SARS-CoV-2 and host cells and interventional therapy. Signal Transduct Target Ther 2021; 6:233. [PMID: 34117216 PMCID: PMC8193598 DOI: 10.1038/s41392-021-00653-w] [Citation(s) in RCA: 182] [Impact Index Per Article: 45.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 04/30/2021] [Accepted: 05/10/2021] [Indexed: 02/05/2023] Open
Abstract
The pandemic of coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection has resulted in an unprecedented setback for global economy and health. SARS-CoV-2 has an exceptionally high level of transmissibility and extremely broad tissue tropism. However, the underlying molecular mechanism responsible for sustaining this degree of virulence remains largely unexplored. In this article, we review the current knowledge and crucial information about how SARS-CoV-2 attaches on the surface of host cells through a variety of receptors, such as ACE2, neuropilin-1, AXL, and antibody-FcγR complexes. We further explain how its spike (S) protein undergoes conformational transition from prefusion to postfusion with the help of proteases like furin, TMPRSS2, and cathepsins. We then review the ongoing experimental studies and clinical trials of antibodies, peptides, or small-molecule compounds with anti-SARS-CoV-2 activity, and discuss how these antiviral therapies targeting host-pathogen interaction could potentially suppress viral attachment, reduce the exposure of fusion peptide to curtail membrane fusion and block the formation of six-helix bundle (6-HB) fusion core. Finally, the specter of rapidly emerging SARS-CoV-2 variants deserves a serious review of broad-spectrum drugs or vaccines for long-term prevention and control of COVID-19 in the future.
Collapse
Affiliation(s)
- Qianqian Zhang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
| | - Rong Xiang
- College of Life Sciences, Hebei Agricultural University, Baoding, China
| | - Shanshan Huo
- College of Life Sciences, Hebei Agricultural University, Baoding, China
| | - Yunjiao Zhou
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
| | - Shibo Jiang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China.
| | - Qiao Wang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China.
| | - Fei Yu
- College of Life Sciences, Hebei Agricultural University, Baoding, China.
| |
Collapse
|
36
|
van Dülmen M, Muthmann N, Rentmeister A. Chemo-Enzymatic Modification of the 5' Cap Maintains Translation and Increases Immunogenic Properties of mRNA. Angew Chem Int Ed Engl 2021; 60:13280-13286. [PMID: 33751748 PMCID: PMC8250829 DOI: 10.1002/anie.202100352] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Indexed: 12/19/2022]
Abstract
Eukaryotic mRNAs are emerging modalities for protein replacement therapy and vaccination. Their 5' cap is important for mRNA translation and immune response and can be naturally methylated at different positions by S-adenosyl-l-methionine (AdoMet)-dependent methyltransferases (MTases). We report on the cosubstrate scope of the MTase CAPAM responsible for methylation at the N6 -position of adenosine start nucleotides using synthetic AdoMet analogs. The chemo-enzymatic propargylation enabled production of site-specifically modified reporter-mRNAs. These cap-propargylated mRNAs were efficiently translated and showed ≈3-fold increased immune response in human cells. The same effects were observed when the receptor binding domain (RBD) of SARS-CoV-2-a currently tested epitope for mRNA vaccination-was used. Site-specific chemo-enzymatic modification of eukaryotic mRNA may thus be a suitable strategy to modulate translation and immune response of mRNAs for future therapeutic applications.
Collapse
Affiliation(s)
- Melissa van Dülmen
- Department of Chemistry and PharmacyInstitute of BiochemistryCorrensstrasse 3648149MünsterGermany
| | - Nils Muthmann
- Department of Chemistry and PharmacyInstitute of BiochemistryCorrensstrasse 3648149MünsterGermany
| | - Andrea Rentmeister
- Department of Chemistry and PharmacyInstitute of BiochemistryCorrensstrasse 3648149MünsterGermany
- Cells in Motion Interfaculty CenterUniversity of MünsterGermany
| |
Collapse
|
37
|
Dülmen M, Muthmann N, Rentmeister A. Eine chemo‐enzymatische Modifizierung der 5′‐Kappe erhält die Translation und erhöht die Immunogenität der mRNA. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202100352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Melissa Dülmen
- Fachbereich Chemie und Pharmazie Institut für Biochemie Corrensstrasse 36 48149 Münster Deutschland
| | - Nils Muthmann
- Fachbereich Chemie und Pharmazie Institut für Biochemie Corrensstrasse 36 48149 Münster Deutschland
| | - Andrea Rentmeister
- Fachbereich Chemie und Pharmazie Institut für Biochemie Corrensstrasse 36 48149 Münster Deutschland
- Cells in Motion Interfaculty Center Westfälische Wilhelms-Universität Münster Deutschland
| |
Collapse
|
38
|
Valdes-Balbin Y, Santana-Mederos D, Paquet F, Fernandez S, Climent Y, Chiodo F, Rodríguez L, Sanchez Ramirez B, Leon K, Hernandez T, Castellanos-Serra L, Garrido R, Chen GW, Garcia-Rivera D, Rivera DG, Verez-Bencomo V. Molecular Aspects Concerning the Use of the SARS-CoV-2 Receptor Binding Domain as a Target for Preventive Vaccines. ACS CENTRAL SCIENCE 2021; 7:757-767. [PMID: 34075345 PMCID: PMC8084267 DOI: 10.1021/acscentsci.1c00216] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Indexed: 02/08/2023]
Abstract
The development of recombinant COVID-19 vaccines has resulted from scientific progress made at an unprecedented speed during 2020. The recombinant spike glycoprotein monomer, its trimer, and its recombinant receptor-binding domain (RBD) induce a potent anti-RBD neutralizing antibody response in animals. In COVID-19 convalescent sera, there is a good correlation between the antibody response and potent neutralization. In this review, we summarize with a critical view the molecular aspects associated with the interaction of SARS-CoV-2 RBD with its receptor in human cells, the angiotensin-converting enzyme 2 (ACE2), the epitopes involved in the neutralizing activity, and the impact of virus mutations thereof. Recent trends in RBD-based vaccines are analyzed, providing detailed insights into the role of antigen display and multivalence in the immune response of vaccines under development.
Collapse
Affiliation(s)
| | | | - Françoise Paquet
- Centre
de Biophysique Moléculaire, Centre
National de la Recherche Scientifique UPR 4301, rue Charles Sadron, F-45071, Orléans, Cedex 2, France
| | | | - Yanet Climent
- Finlay
Vaccine Institute, 200
and 21 Street, Havana 11600, Cuba
| | - Fabrizio Chiodo
- Department
of Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands, 1081 HV
- Institute
of Biomolecular Chemistry, National Research
Council (CNR), Pozzuoli, Napoli, Italy
| | - Laura Rodríguez
- Finlay
Vaccine Institute, 200
and 21 Street, Havana 11600, Cuba
| | | | - Kalet Leon
- Center
of Molecular Immunology, P.O. Box 16040, 216 Street, Havana, Cuba
| | - Tays Hernandez
- Center
of Molecular Immunology, P.O. Box 16040, 216 Street, Havana, Cuba
| | | | - Raine Garrido
- Finlay
Vaccine Institute, 200
and 21 Street, Havana 11600, Cuba
| | - Guang-Wu Chen
- Chengdu
Olisynn Biotech. Co. Ltd. and State Key Laboratory of Biotherapy and
Cancer Center, West China Hospital, Sichuan
University, Chengdu 610041, People’s Republic of China
| | | | - Daniel G. Rivera
- Laboratory
of Synthetic and Biomolecular Chemistry, Faculty of Chemistry, University of Havana, Zapata & G, Havana 10400, Cuba
| | | |
Collapse
|
39
|
Cao M, Su X, Jiang S. Broad-Spectrum Anti-coronavirus Vaccines and Therapeutics to Combat the Current COVID-19 Pandemic and Future Coronavirus Disease Outbreaks. Stem Cell Reports 2021; 16:398-411. [PMID: 33691145 PMCID: PMC7940144 DOI: 10.1016/j.stemcr.2020.12.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 12/18/2020] [Accepted: 12/18/2020] [Indexed: 12/14/2022] Open
Abstract
While the COVID-19 pandemic caused by SARS-CoV-2 is continuing, it may become worse in the coming winter months with a high potential for the emergence and spread of escape variants of SARS-CoV-2. SARS-related CoVs (SARSr-CoVs) from bats may also cause outbreaks of emerging coronavirus diseases in the future. These predictions call for the development of broad-spectrum anti-coronavirus vaccines and therapeutics to combat the current COVID-19 pandemic and future emerging coronavirus disease epidemics. In this review, we describe advances and challenges in the development of broad-spectrum vaccines and neutralizing antibodies against lineage B betacoronaviruses (β-CoV-Bs), including SARS-CoV-2, SARS-CoV, and SARSr-CoVs, as well as peptide-based pan-CoV fusion inhibitors and their potential in the prevention and treatment of COVID-19 and other human coronavirus infections.
Collapse
Affiliation(s)
- Miao Cao
- Key Laboratory of Medical Molecular Virology of MOE/MOH, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Xiaojie Su
- Key Laboratory of Medical Molecular Virology of MOE/MOH, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Shibo Jiang
- Key Laboratory of Medical Molecular Virology of MOE/MOH, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China.
| |
Collapse
|
40
|
Jeong H, Choi YM, Seo H, Kim BJ. A Novel DNA Vaccine Against SARS-CoV-2 Encoding a Chimeric Protein of Its Receptor-Binding Domain (RBD) Fused to the Amino-Terminal Region of Hepatitis B Virus preS1 With a W4P Mutation. Front Immunol 2021; 12:637654. [PMID: 33732258 PMCID: PMC7959807 DOI: 10.3389/fimmu.2021.637654] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 02/08/2021] [Indexed: 12/15/2022] Open
Abstract
A coronavirus SARS-CoV-2, which has caused the pandemic viral pneumonia disease COVID-19, significantly threatens global public health, highlighting the need to develop effective and safe vaccines against its infection. In this study, we developed a novel DNA vaccine candidate against SARS-CoV-2 by expressing a chimeric protein of its receptor-binding domain (RBD) fused to a 33-bp sequence (11 aa) from the hepatitis B virus (HBV) preS1 region with a W4P mutation (W4P-RBD) at the N-terminal region and evaluated its immunogenicity. In vitro transfection experiments in multiple cell lines demonstrated that W4P-RBD vs. wild-type RBD protein (W-RBD) led to enhanced production of IL-6 and TNFα at the transcription and translation levels, suggesting the adjuvant potential of N-terminal HBV preS1 sequences for DNA vaccines against SARS-CoV-2. W4P-RBD also led to enhanced production of IgG and IgA, which can neutralize and block SARS-CoV-2 infection in both blood sera and bronchoalveolar lavage (BAL) fluid from the lung in vaccinated mice. Additionally, W4P-RBD led to an enhanced T-cell-mediated cellular immune response under S1 protein stimulation. In summary, W4P-RBD led to robust humoral and cell-mediated immune responses against SARS-CoV-2 in vaccinated mice, highlighting its feasibility as a novel DNA vaccine to protect against SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Hyein Jeong
- Department of Biomedical Sciences, College of Medicine, Microbiology and Immunology and Liver Research Institute, Seoul National University, Seoul, South Korea
| | - Yu-Min Choi
- Department of Biomedical Sciences, College of Medicine, Microbiology and Immunology and Liver Research Institute, Seoul National University, Seoul, South Korea
| | - Hyejun Seo
- Department of Biomedical Sciences, College of Medicine, Microbiology and Immunology and Liver Research Institute, Seoul National University, Seoul, South Korea
| | - Bum-Joon Kim
- Department of Biomedical Sciences, College of Medicine, Microbiology and Immunology and Liver Research Institute, Seoul National University, Seoul, South Korea
| |
Collapse
|
41
|
Laing ED, Sterling SL, Richard SA, Epsi NJ, Coggins S, Samuels EC, Phogat S, Yan L, Moreno N, Coles CL, Drew M, Mehalko J, English CE, Merritt S, Mende K, Munster VJ, de Wit E, Chung KK, Millar EV, Tribble DR, Simons MP, Pollett SD, Agan BK, Esposito D, Lanteri C, Clifton GT, Mitre E, Burgess TH, Broder CC. Antigen-based multiplex strategies to discriminate SARS-CoV-2 natural and vaccine induced immunity from seasonal human coronavirus humoral responses. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2021:2021.02.10.21251518. [PMID: 33594376 PMCID: PMC7885935 DOI: 10.1101/2021.02.10.21251518] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Sensitive and specific SARS-CoV-2 antibody assays remain critical for community and hospital-based SARS-CoV-2 sero-surveillance. With the rollout of SARS-CoV-2 vaccines, such assays must be able to distinguish vaccine from natural immunity to SARS-CoV-2 and related human coronaviruses. Here, we developed and implemented multiplex microsphere-based immunoassay strategies for COVD-19 antibody studies that incorporates spike protein trimers of SARS-CoV-2 and the endemic seasonal human coronaviruses (HCoV), enabling high throughout measurement of pre-existing cross-reactive antibodies. We varied SARS-CoV-2 antigen compositions within the multiplex assay, allowing direct comparisons of the effects of spike protein, receptor-binding domain protein (RBD) and nucleocapsid protein (NP) based SARS-CoV-2 antibody detection. Multiplex immunoassay performance characteristics are antigen-dependent, and sensitivities and specificities range 92-99% and 94-100%, respectively, for human subject samples collected as early as 7-10 days from symptom onset. SARS-CoV-2 spike and RBD had a strong correlative relationship for the detection of IgG. Correlation between detectable IgG reactive with spike and NP also had strong relationship, however, several PCR-positive and spike IgG-positive serum samples were NP IgG-negative. This spike and NP multiplex immunoassay has the potential to be useful for differentiation between vaccination and natural infection induced antibody responses. We also assessed the induction of de novo SARS-CoV-2 IgG cross reactions with SARS-CoV and MERS-CoV spike proteins. Furthermore, multiplex immunoassays that incorporate spike proteins of SARS-CoV-2 and HCoVs will permit investigations into the influence of HCoV antibodies on COVID-19 clinical outcomes and SARS-CoV-2 antibody durability.
Collapse
Affiliation(s)
- Eric D. Laing
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Spencer L. Sterling
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD USA
| | - Stephanie A. Richard
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD USA
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Nusrat J. Epsi
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD USA
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Si’Ana Coggins
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD USA
| | - Emily C. Samuels
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD USA
| | - Shreshta Phogat
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD USA
| | - Lianying Yan
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD USA
| | - Nicole Moreno
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD USA
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Christian L. Coles
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD USA
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Matthew Drew
- Protein Expression Laboratory, National Cancer Institute RAS Initiative, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Jennifer Mehalko
- Protein Expression Laboratory, National Cancer Institute RAS Initiative, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Caroline E. English
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD USA
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Scott Merritt
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD USA
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Brooke Army Medical Center, JBSA Fort Sam Houston, TX, USA
| | - Katrin Mende
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD USA
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Brooke Army Medical Center, JBSA Fort Sam Houston, TX, USA
| | - Vincent J. Munster
- Laboratory of Virology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Emmie de Wit
- Laboratory of Virology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Kevin K. Chung
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Eugene V. Millar
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD USA
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - David R. Tribble
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Mark P. Simons
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Simon D. Pollett
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD USA
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Brian K. Agan
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD USA
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Dominic Esposito
- Protein Expression Laboratory, National Cancer Institute RAS Initiative, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Charlotte Lanteri
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD USA
| | | | - Edward Mitre
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Timothy H. Burgess
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD USA
| | - Christopher C. Broder
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| |
Collapse
|
42
|
Carrillo J, Izquierdo-Useros N, Ávila-Nieto C, Pradenas E, Clotet B, Blanco J. Humoral immune responses and neutralizing antibodies against SARS-CoV-2; implications in pathogenesis and protective immunity. Biochem Biophys Res Commun 2021; 538:187-191. [PMID: 33187644 PMCID: PMC7648524 DOI: 10.1016/j.bbrc.2020.10.108] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 10/26/2020] [Indexed: 12/15/2022]
Abstract
The magnitude and the quality of humoral responses against SARS-CoV-2 have been associated with clinical outcome. Although the elicitation of humoral responses against different viral proteins is rapid and occurs in most infected individuals, its magnitude is highly variable among them and positively correlates with COVID-19 disease severity. This rapid response is characterized by the almost concomitant appearance of virus-specific IgG, IgA and IgM antibodies that contain neutralizing antibodies directed against different epitopes of the Spike glycoprotein. Of particularly interest, the antibodies against domain of the Spike that interacts with the cellular receptor ACE2, known as the receptor binding domain (RBD), are present in most infected individuals and are block viral entry and infectivity. Such neutralizing antibodies protect different animal species when administered before virus exposure; therefore, its elicitation is the main target of current vaccine approaches and their clinical use as recombinant monoclonal antibodies (mAbs) is being explored. Yet, little information exists on the duration of humoral responses during natural infection. This is a key issue that will impact the management of the pandemic and determine the utility of seroconversion studies and the level of herd immunity. Certainly, several cases of reinfection have been reported, suggesting that immunity could be transient, as reported for other coronaviruses. In summary, although the kinetics of the generation of antibodies against SASR-CoV-2 and their protective activity have been clearly defined, their role in COVID-19 pathogenesis and the length of these responses are still open questions.
Collapse
Affiliation(s)
- Jorge Carrillo
- IrsiCaixa AIDS Research Institute, Germans Trias I Pujol Research Institute (IGTP), Can Ruti Campus, 08916, Badalona, Catalonia, Spain,Corresponding author. Institut de Recerca de la Sida, IrsiCaixa Hospital Universitari Germans Trias i Pujol, Ctra. de Canyet s/n. 2a Planta Maternal, 08916, Badalona, Barcelona, Spain
| | - Nuria Izquierdo-Useros
- IrsiCaixa AIDS Research Institute, Germans Trias I Pujol Research Institute (IGTP), Can Ruti Campus, 08916, Badalona, Catalonia, Spain
| | - Carlos Ávila-Nieto
- IrsiCaixa AIDS Research Institute, Germans Trias I Pujol Research Institute (IGTP), Can Ruti Campus, 08916, Badalona, Catalonia, Spain
| | - Edwards Pradenas
- IrsiCaixa AIDS Research Institute, Germans Trias I Pujol Research Institute (IGTP), Can Ruti Campus, 08916, Badalona, Catalonia, Spain
| | - Bonaventura Clotet
- IrsiCaixa AIDS Research Institute, Germans Trias I Pujol Research Institute (IGTP), Can Ruti Campus, 08916, Badalona, Catalonia, Spain,Infectious Diseases Department, Germans Trias i Pujol Hospital, Badalona, Catalonia, Spain,University of Vic (UVic-UCC), Vic, Catalonia, Spain
| | - Julià Blanco
- IrsiCaixa AIDS Research Institute, Germans Trias I Pujol Research Institute (IGTP), Can Ruti Campus, 08916, Badalona, Catalonia, Spain,University of Vic (UVic-UCC), Vic, Catalonia, Spain,Corresponding author. Institut de Recerca de la Sida, IrsiCaixa Hospital Universitari Germans Trias i Pujol, Ctra. de Canyet s/n. 2a Planta Maternal, 08916, Badalona, Barcelona, Spain
| |
Collapse
|
43
|
Scourfield DO, Reed SG, Quastel M, Alderson J, Bart VMT, Teijeira Crespo A, Jones R, Pring E, Richter FC, Burnell SEA. The role and uses of antibodies in COVID-19 infections: a living review. OXFORD OPEN IMMUNOLOGY 2021; 2:iqab003. [PMID: 34192270 PMCID: PMC7928637 DOI: 10.1093/oxfimm/iqab003] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 12/18/2020] [Accepted: 01/07/2021] [Indexed: 12/19/2022] Open
Abstract
Coronavirus disease 2019 has generated a rapidly evolving field of research, with the global scientific community striving for solutions to the current pandemic. Characterizing humoral responses towards SARS-CoV-2, as well as closely related strains, will help determine whether antibodies are central to infection control, and aid the design of therapeutics and vaccine candidates. This review outlines the major aspects of SARS-CoV-2-specific antibody research to date, with a focus on the various prophylactic and therapeutic uses of antibodies to alleviate disease in addition to the potential of cross-reactive therapies and the implications of long-term immunity.
Collapse
Affiliation(s)
- D Oliver Scourfield
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK
| | - Sophie G Reed
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK
| | - Max Quastel
- Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7FZ, UK
| | - Jennifer Alderson
- Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, OX3 FTY, UK
| | - Valentina M T Bart
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK
| | - Alicia Teijeira Crespo
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff, CF14 4XN UK
| | - Ruth Jones
- Dementia Research Institute, Cardiff University, Cardiff, CF24 4HQ, UK
| | - Ellie Pring
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK
| | - Felix Clemens Richter
- Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, OX3 FTY, UK
| | - Stephanie E A Burnell
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK
| |
Collapse
|
44
|
Malladi SK, Singh R, Pandey S, Gayathri S, Kanjo K, Ahmed S, Khan MS, Kalita P, Girish N, Upadhyaya A, Reddy P, Pramanick I, Bhasin M, Mani S, Bhattacharyya S, Joseph J, Thankamani K, Raj VS, Dutta S, Singh R, Nadig G, Varadarajan R. Design of a highly thermotolerant, immunogenic SARS-CoV-2 spike fragment. J Biol Chem 2021; 296:100025. [PMID: 33154165 PMCID: PMC7832000 DOI: 10.1074/jbc.ra120.016284] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 10/30/2020] [Accepted: 11/05/2020] [Indexed: 12/19/2022] Open
Abstract
Virtually all SARS-CoV-2 vaccines currently in clinical testing are stored in a refrigerated or frozen state prior to use. This is a major impediment to deployment in resource-poor settings. Furthermore, several of them use viral vectors or mRNA. In contrast to protein subunit vaccines, there is limited manufacturing expertise for these nucleic-acid-based modalities, especially in the developing world. Neutralizing antibodies, the clearest known correlate of protection against SARS-CoV-2, are primarily directed against the receptor-binding domain (RBD) of the viral spike protein, suggesting that a suitable RBD construct might serve as a more accessible vaccine ingredient. We describe a monomeric, glycan-engineered RBD protein fragment that is expressed at a purified yield of 214 mg/l in unoptimized, mammalian cell culture and, in contrast to a stabilized spike ectodomain, is tolerant of exposure to temperatures as high as 100 °C when lyophilized, up to 70 °C in solution and stable for over 4 weeks at 37 °C. In prime:boost guinea pig immunizations, when formulated with the MF59-like adjuvant AddaVax, the RBD derivative elicited neutralizing antibodies with an endpoint geometric mean titer of ∼415 against replicative virus, comparing favorably with several vaccine formulations currently in the clinic. These features of high yield, extreme thermotolerance, and satisfactory immunogenicity suggest that such RBD subunit vaccine formulations hold great promise to combat COVID-19.
Collapse
MESH Headings
- Angiotensin-Converting Enzyme 2/chemistry
- Angiotensin-Converting Enzyme 2/genetics
- Angiotensin-Converting Enzyme 2/immunology
- Animals
- Antibodies, Neutralizing/biosynthesis
- Antibodies, Viral/biosynthesis
- Binding Sites
- COVID-19/immunology
- COVID-19/prevention & control
- COVID-19/virology
- COVID-19 Vaccines/administration & dosage
- COVID-19 Vaccines/biosynthesis
- COVID-19 Vaccines/genetics
- Escherichia coli/genetics
- Escherichia coli/metabolism
- Female
- Guinea Pigs
- HEK293 Cells
- Hot Temperature
- Humans
- Immunogenicity, Vaccine
- Models, Molecular
- Protein Binding
- Protein Conformation, alpha-Helical
- Protein Conformation, beta-Strand
- Protein Domains
- Protein Interaction Domains and Motifs
- Protein Stability
- Receptors, Virus/chemistry
- Receptors, Virus/genetics
- Receptors, Virus/immunology
- Recombinant Proteins/chemistry
- Recombinant Proteins/genetics
- Recombinant Proteins/immunology
- SARS-CoV-2/chemistry
- SARS-CoV-2/immunology
- Spike Glycoprotein, Coronavirus/chemistry
- Spike Glycoprotein, Coronavirus/genetics
- Spike Glycoprotein, Coronavirus/immunology
- Vaccination
- Vaccine Potency
Collapse
Affiliation(s)
| | - Randhir Singh
- Mynvax Private Limited, ES12, Entrepreneurship Centre, SID, Indian Institute of Science, Bengaluru, India
| | - Suman Pandey
- Mynvax Private Limited, ES12, Entrepreneurship Centre, SID, Indian Institute of Science, Bengaluru, India
| | - Savitha Gayathri
- Mynvax Private Limited, ES12, Entrepreneurship Centre, SID, Indian Institute of Science, Bengaluru, India
| | - Kawkab Kanjo
- Molecular Biophysics Unit (MBU), Indian Institute of Science, Bengaluru, India
| | - Shahbaz Ahmed
- Molecular Biophysics Unit (MBU), Indian Institute of Science, Bengaluru, India
| | | | - Parismita Kalita
- Molecular Biophysics Unit (MBU), Indian Institute of Science, Bengaluru, India
| | - Nidhi Girish
- Mynvax Private Limited, ES12, Entrepreneurship Centre, SID, Indian Institute of Science, Bengaluru, India
| | - Aditya Upadhyaya
- Mynvax Private Limited, ES12, Entrepreneurship Centre, SID, Indian Institute of Science, Bengaluru, India
| | - Poorvi Reddy
- Mynvax Private Limited, ES12, Entrepreneurship Centre, SID, Indian Institute of Science, Bengaluru, India
| | - Ishika Pramanick
- Molecular Biophysics Unit (MBU), Indian Institute of Science, Bengaluru, India
| | - Munmun Bhasin
- Molecular Biophysics Unit (MBU), Indian Institute of Science, Bengaluru, India
| | - Shailendra Mani
- Translational Health Science and Technology Institute, NCR Biotech Science Cluster, Faridabad, India
| | - Sankar Bhattacharyya
- Translational Health Science and Technology Institute, NCR Biotech Science Cluster, Faridabad, India
| | - Jeswin Joseph
- Virology Scientific Research (VSR) Laboratory, School of Biology, Indian Institute of Science Education and Research Thiruvananthapuram (IISER TVM), Kerala, India
| | - Karthika Thankamani
- Virology Scientific Research (VSR) Laboratory, School of Biology, Indian Institute of Science Education and Research Thiruvananthapuram (IISER TVM), Kerala, India
| | - V Stalin Raj
- Virology Scientific Research (VSR) Laboratory, School of Biology, Indian Institute of Science Education and Research Thiruvananthapuram (IISER TVM), Kerala, India
| | - Somnath Dutta
- Molecular Biophysics Unit (MBU), Indian Institute of Science, Bengaluru, India
| | - Ramandeep Singh
- Translational Health Science and Technology Institute, NCR Biotech Science Cluster, Faridabad, India
| | - Gautham Nadig
- Mynvax Private Limited, ES12, Entrepreneurship Centre, SID, Indian Institute of Science, Bengaluru, India
| | - Raghavan Varadarajan
- Molecular Biophysics Unit (MBU), Indian Institute of Science, Bengaluru, India; Jawaharlal Nehru Centre for Advanced Scientific Research, Bengaluru, India.
| |
Collapse
|
45
|
Laing E, Sterling S, Richard S, Epsi N, Phogat S, Samuels E, Yan L, Moreno N, Coles C, Drew M, Mehalko J, English C, Merritt S, Mende K, Chung K, Clifton G, Munster V, de Wit E, Tribble D, Agan B, Esposito D, Lanteri C, Mitre E, Burgess T, Broder C. A betacoronavirus multiplex microsphere immunoassay detects early SARS-CoV-2 seroconversion and antibody cross reactions. RESEARCH SQUARE 2020:rs.3.rs-105768. [PMID: 33269345 PMCID: PMC7709164 DOI: 10.21203/rs.3.rs-105768/v1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Sensitive and specific SARS-CoV-2 antibody assays remain critical for community and hospital-based SARS-CoV-2 surveillance. Here, we developed and applied a multiplex microsphere-based immunoassay (MMIA) for COVD-19 antibody studies that incorporates spike protein trimers of SARS-CoV-2, SARS-CoV-1, MERS-CoV, and the seasonal human betacoronaviruses, HCoV-HKU1 and HCoV-OC43, that enables measurement of off-target pre-existing cross-reactive antibodies. The MMIA performances characteristics are: 98% sensitive and 100% specific for human subject samples collected as early as 10 days from symptom onset. The MMIA permitted the simultaneous identification of SARS-CoV-2 seroconversion and the induction of SARS-CoV-2 IgG antibody cross reactions to SARS-CoV-1 and MERS-CoV. Further, synchronous increases of HCoV-OC43 IgG antibody levels was detected with SARS-CoV-2 seroconversion in a subset of subjects for whom early infection sera were available prior to their SARS-CoV-2 seroconversion, suggestive of an HCoV-OC43 memory response triggered by SARS-CoV-2 infection.
Collapse
Affiliation(s)
| | | | | | - Nusrat Epsi
- Uniformed Services University/Henry M. Jackson Foundation
| | | | - Emily Samuels
- Uniformed Services University/Henry M. Jackson Foundation
| | - Lianying Yan
- Uniformed Services University of the Health Sciences
| | - Nicole Moreno
- Uniformed Services University/Henry M. Jackson Foundation
| | | | - Matthew Drew
- Frederick National Laboratory for Cancer Research
| | | | | | - Scott Merritt
- Uniformed Services University/Henry M. Jackson Foundation/Brooke Army Medical Center
| | - Katrin Mende
- Uniformed Services University/Henry M. Jackson Foundation/Brooke Army Medical Center
| | | | | | | | - Emmie de Wit
- National Institute of Allergy and Infectious Diseases
| | | | - Brian Agan
- Uniformed Services University/Henry M. Jackson Foundation
| | | | | | | | | | | |
Collapse
|
46
|
Wang L, Xiang Y. Spike Glycoprotein-Mediated Entry of SARS Coronaviruses. Viruses 2020; 12:E1289. [PMID: 33187074 PMCID: PMC7696831 DOI: 10.3390/v12111289] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 11/07/2020] [Accepted: 11/08/2020] [Indexed: 12/12/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus (SARS-CoV) and SARS-CoV-2 are enveloped, positive-sense, single-stranded RNA viruses and causes of epidemic diseases that have resulted in public health emergencies worldwide. Angiotensin-converting enzyme 2 (ACE2) is the receptor that allows the entry of these two viruses into host cells, a key step in the life cycle of the pathogens. The characterization of the interactions of ACE2 with the viral spike glycoproteins and structural studies of the ACE2-binding-induced conformational changes in the viral spike glycoproteins have furthered our understanding of the entry processes of these two viruses, and these studies provide useful information that will facilitate the development of antiviral agents and vaccines to control the diseases.
Collapse
Affiliation(s)
| | - Ye Xiang
- Center for Infectious Disease Research, Beijing Frontier Research Center for Biological Structure & Beijing Advanced Innovation Center for Structural Biology, Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing 100084, China;
| |
Collapse
|
47
|
Liu X, Drelich A, Li W, Chen C, Sun Z, Shi M, Adams C, Mellors JW, Tseng CT, Dimitrov DS. Enhanced elicitation of potent neutralizing antibodies by the SARS-CoV-2 spike receptor binding domain Fc fusion protein in mice. Vaccine 2020; 38:7205-7212. [PMID: 33010978 PMCID: PMC7508516 DOI: 10.1016/j.vaccine.2020.09.058] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 08/19/2020] [Accepted: 09/20/2020] [Indexed: 01/23/2023]
Abstract
The development of an effective vaccine against SARS-CoV-2 is urgently needed. We generated SARS-CoV-2 RBD-Fc fusion protein and evaluated its potency to elicit neutralizing antibody response in mice. RBD-Fc elicited a higher neutralizing antibodies titer than RBD as evaluated by a pseudovirus neutralization assay and a live virus based microneutralization assay. Furthermore, RBD-Fc immunized sera better inhibited cell-cell fusion, as evaluated by a quantitative cell-cell fusion assay. The cell-cell fusion assay results correlated well with the virus neutralization potency and could be used for high-throughput screening of large panels of anti-SARS-CoV-2 antibodies and vaccines without the requirement of live virus infection in BSL3 containment. Moreover, the anti-RBD sera did not enhance the pseudotyped SARS-CoV-2 infection of K562 cells. These results demonstrate that Fc fusion can significantly improve the humoral immune response to recombinant RBD immunogen, and suggest that RBD-Fc could serve as a useful component of effective vaccines against SARS-CoV-2.
Collapse
MESH Headings
- Angiotensin-Converting Enzyme 2
- Animals
- Antibodies, Neutralizing/blood
- Antibodies, Neutralizing/immunology
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- Betacoronavirus/immunology
- COVID-19
- COVID-19 Vaccines
- Cell Fusion
- Cell Line
- Coronavirus Infections/immunology
- Coronavirus Infections/prevention & control
- Enzyme-Linked Immunosorbent Assay
- Female
- HEK293 Cells
- High-Throughput Screening Assays/methods
- Humans
- Immunity, Humoral/immunology
- Immunoglobulin Fc Fragments/genetics
- Immunoglobulin Fc Fragments/immunology
- Mice
- Mice, Inbred BALB C
- Neutralization Tests
- Pandemics/prevention & control
- Peptidyl-Dipeptidase A/genetics
- Pneumonia, Viral/prevention & control
- Protein Domains/immunology
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/immunology
- SARS-CoV-2
- Spike Glycoprotein, Coronavirus/genetics
- Spike Glycoprotein, Coronavirus/immunology
- Vaccines, Subunit/immunology
- Viral Vaccines/immunology
Collapse
Affiliation(s)
- Xianglei Liu
- Center for Antibody Therapeutics, Division of Infectious Diseases, Department of Medicine, University of Pittsburgh Medical School, 3550 Terrace St, Pittsburgh, PA 15261, USA.
| | - Aleksandra Drelich
- Department of Microbiology & Immunology, Centers for Biodefense and Emerging Diseases, Galveston National Laboratory, 301 University Blvd, Galveston, TX 77550, USA
| | - Wei Li
- Center for Antibody Therapeutics, Division of Infectious Diseases, Department of Medicine, University of Pittsburgh Medical School, 3550 Terrace St, Pittsburgh, PA 15261, USA
| | - Chuan Chen
- Center for Antibody Therapeutics, Division of Infectious Diseases, Department of Medicine, University of Pittsburgh Medical School, 3550 Terrace St, Pittsburgh, PA 15261, USA
| | - Zehua Sun
- Center for Antibody Therapeutics, Division of Infectious Diseases, Department of Medicine, University of Pittsburgh Medical School, 3550 Terrace St, Pittsburgh, PA 15261, USA
| | - Megan Shi
- Center for Antibody Therapeutics, Division of Infectious Diseases, Department of Medicine, University of Pittsburgh Medical School, 3550 Terrace St, Pittsburgh, PA 15261, USA
| | - Cynthia Adams
- Center for Antibody Therapeutics, Division of Infectious Diseases, Department of Medicine, University of Pittsburgh Medical School, 3550 Terrace St, Pittsburgh, PA 15261, USA
| | - John W Mellors
- Center for Antibody Therapeutics, Division of Infectious Diseases, Department of Medicine, University of Pittsburgh Medical School, 3550 Terrace St, Pittsburgh, PA 15261, USA; Abound Bio, 1401 Forbes Ave, Pittsburgh, PA 15219, USA
| | - Chien-Te Tseng
- Department of Microbiology & Immunology, Centers for Biodefense and Emerging Diseases, Galveston National Laboratory, 301 University Blvd, Galveston, TX 77550, USA
| | - Dimiter S Dimitrov
- Center for Antibody Therapeutics, Division of Infectious Diseases, Department of Medicine, University of Pittsburgh Medical School, 3550 Terrace St, Pittsburgh, PA 15261, USA; Abound Bio, 1401 Forbes Ave, Pittsburgh, PA 15219, USA.
| |
Collapse
|
48
|
Laing ED, Sterling SL, Richard SA, Phogat S, Samuels EC, Epsi NJ, Yan L, Moreno N, Coles C, Mehalko J, Drew M, English C, Chung KK, Clifton GT, Munster VJ, de Wit E, Tribble D, Agan BK, Esposito D, Lanteri C, Mitre E, Burgess TH, Broder CC. A betacoronavirus multiplex microsphere immunoassay detects early SARS-CoV-2 seroconversion and controls for pre-existing seasonal human coronavirus antibody cross-reactivity. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2020:2020.10.14.20207050. [PMID: 33083807 PMCID: PMC7574255 DOI: 10.1101/2020.10.14.20207050] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
With growing concern of persistent or multiple waves of SARS-CoV-2 in the United States, sensitive and specific SARS-CoV-2 antibody assays remain critical for community and hospital-based SARS-CoV-2 surveillance. Here, we describe the development and application of a multiplex microsphere-based immunoassay (MMIA) for COVD-19 antibody studies, utilizing serum samples from non-human primate SARS-CoV-2 infection models, an archived human sera bank and subjects enrolled at five U.S. military hospitals. The MMIA incorporates prefusion stabilized spike glycoprotein trimers of SARS-CoV-2, SARS-CoV-1, MERS-CoV, and the seasonal human coronaviruses HCoV-HKU1 and HCoV-OC43, into a multiplexing system that enables simultaneous measurement of off-target pre-existing cross-reactive antibodies. We report the sensitivity and specificity performances for this assay strategy at 98% sensitivity and 100% specificity for subject samples collected as early as 10 days after the onset of symptoms. In archival sera collected prior to 2019 and serum samples from subjects PCR negative for SARS-CoV-2, we detected seroprevalence of 72% and 98% for HCoV-HKU1 and HCoV-0C43, respectively. Requiring only 1.25 μL of sera, this approach permitted the simultaneous identification of SARS-CoV-2 seroconversion and polyclonal SARS-CoV-2 IgG antibody responses to SARS-CoV-1 and MERS-CoV, further demonstrating the presence of conserved epitopes in the spike glycoprotein of zoonotic betacoronaviruses. Application of this serology assay in observational studies with serum samples collected from subjects before and after SARS-CoV-2 infection will permit an investigation of the influences of HCoV-induced antibodies on COVID-19 clinical outcomes.
Collapse
Affiliation(s)
- Eric D. Laing
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Spencer L. Sterling
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD USA
| | - Stephanie A. Richard
- Infectious Diseases Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD USA
| | - Shreshta Phogat
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD USA
| | - Emily C. Samuels
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD USA
| | - Nusrat J. Epsi
- Infectious Diseases Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD USA
| | - Lianying Yan
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD USA
| | - Nicole Moreno
- Infectious Diseases Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD USA
| | - Christian Coles
- Infectious Diseases Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD USA
| | - Jennifer Mehalko
- Protein Expression Laboratory, National Cancer Institute RAS Initiative, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Matthew Drew
- Protein Expression Laboratory, National Cancer Institute RAS Initiative, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Caroline English
- Infectious Diseases Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD USA
| | - Kevin K. Chung
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | | | - Vincent J. Munster
- Laboratory of Virology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Emmie de Wit
- Laboratory of Virology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - David Tribble
- Infectious Diseases Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Brian K. Agan
- Infectious Diseases Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD USA
| | - Dominic Esposito
- Protein Expression Laboratory, National Cancer Institute RAS Initiative, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Charlotte Lanteri
- Infectious Diseases Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Edward Mitre
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Timothy H. Burgess
- Infectious Diseases Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Christopher C. Broder
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| |
Collapse
|
49
|
Laing ED, Sterling SL, Richard SA, Phogat S, Samuels EC, Epsi NJ, Yan L, Moreno N, Coles C, Mehalko J, Drew M, English C, Chung KK, Clifton GT, Munster V, de Wit E, Tribble D, Agan B, Esposito D, Lanteri C, Mitre E, Burgess TH, Broder CC. A betacoronavirus multiplex microsphere immunoassay detects early SARS-CoV-2 seroconversion and controls for pre-existing seasonal human coronavirus antibody cross-reactivity. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2020. [PMID: 33083807 DOI: 10.1101/2020.05.21.20108985v2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 03/16/2023]
Abstract
With growing concern of persistent or multiple waves of SARS-CoV-2 in the United States, sensitive and specific SARS-CoV-2 antibody assays remain critical for community and hospital-based SARS-CoV-2 surveillance. Here, we describe the development and application of a multiplex microsphere-based immunoassay (MMIA) for COVD-19 antibody studies, utilizing serum samples from non-human primate SARS-CoV-2 infection models, an archived human sera bank and subjects enrolled at five U.S. military hospitals. The MMIA incorporates prefusion stabilized spike glycoprotein trimers of SARS-CoV-2, SARS-CoV-1, MERS-CoV, and the seasonal human coronaviruses HCoV-HKU1 and HCoV-OC43, into a multiplexing system that enables simultaneous measurement of off-target pre-existing cross-reactive antibodies. We report the sensitivity and specificity performances for this assay strategy at 98% sensitivity and 100% specificity for subject samples collected as early as 10 days after the onset of symptoms. In archival sera collected prior to 2019 and serum samples from subjects PCR negative for SARS-CoV-2, we detected seroprevalence of 72% and 98% for HCoV-HKU1 and HCoV-0C43, respectively. Requiring only 1.25 uL of sera, this approach permitted the simultaneous identification of SARS-CoV-2 seroconversion and polyclonal SARS-CoV-2 IgG antibody responses to SARS-CoV-1 and MERS-CoV, further demonstrating the presence of conserved epitopes in the spike glycoprotein of zoonotic betacoronaviruses. Application of this serology assay in observational studies with serum samples collected from subjects before and after SARS-CoV-2 infection will permit an investigation of the influences of HCoV-induced antibodies on COVID-19 clinical outcomes.
Collapse
|