1
|
Ramos-Duarte VA, Orlowski A, Jaquenod de Giusti C, Corigliano MG, Legarralde A, Mendoza-Morales LF, Atela A, Sánchez MA, Sander VA, Angel SO, Clemente M. Safe plant Hsp90 adjuvants elicit an effective immune response against SARS-CoV2-derived RBD antigen. Vaccine 2024; 42:3355-3364. [PMID: 38631949 DOI: 10.1016/j.vaccine.2024.04.036] [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: 01/08/2024] [Revised: 04/10/2024] [Accepted: 04/10/2024] [Indexed: 04/19/2024]
Abstract
To better understand the role of pHsp90 adjuvant in immune response modulation, we proposed the use of the Receptor Binding Domain (RBD) of the Spike protein of SARS-CoV2, the principal candidate in the design of subunit vaccines. We evaluated the humoral and cellular immune responses against RBD through the strategy "protein mixture" (Adjuvant + Antigen). The rRBD adjuvanted with rAtHsp81.2 group showed a higher increase of the anti-rRBD IgG1, while the rRBD adjuvanted with rNbHsp90.3 group showed a significant increase in anti-rRBD IgG2b/2a. These results were consistent with the cellular immune response analysis. Spleen cell cultures from rRBD + rNbHsp90.3-immunized mice showed significantly increased IFN-γ production. In contrast, spleen cell cultures from rRBD + rAtHsp81.2-immunized mice showed significantly increased IL-4 levels. Finally, vaccines adjuvanted with rNbHsp90.3 induced higher neutralizing antibody responses compared to those adjuvanted with rAtHsp81.2. To know whether both chaperones must form complexes to generate an effective immune response, we performed co-immunoprecipitation (co-IP) assays. The results indicated that the greater neutralizing capacity observed in the rRBD adjuvanted with rNbHsp90.3 group would be given by the rRBD-rNbHsp90.3 interaction rather than by the quality of the immune response triggered by the adjuvants. These results, together with our previous results, provide a comparative benchmark of these two novel and safe vaccine adjuvants for their capacity to stimulate immunity to a subunit vaccine, demonstrating the capacity of adjuvanted SARS-CoV2 subunit vaccines. Furthermore, these results revealed differences in the ability to modulate the immune response between these two pHsp90s, highlighting the importance of adjuvant selection for future rational vaccine and adjuvant design.
Collapse
MESH Headings
- Animals
- Female
- Humans
- Mice
- Adjuvants, Immunologic/administration & dosage
- Adjuvants, Vaccine
- Antibodies, Neutralizing/immunology
- Antibodies, Neutralizing/blood
- Antibodies, Viral/immunology
- Antibodies, Viral/blood
- COVID-19/prevention & control
- COVID-19/immunology
- COVID-19 Vaccines/immunology
- HSP90 Heat-Shock Proteins/immunology
- Immunity, Cellular
- Immunity, Humoral
- Immunoglobulin G/blood
- Immunoglobulin G/immunology
- Mice, Inbred BALB C
- SARS-CoV-2/immunology
- Spike Glycoprotein, Coronavirus/immunology
- Vaccines, Subunit/immunology
- Vaccines, Subunit/administration & dosage
Collapse
Affiliation(s)
- Victor A Ramos-Duarte
- Laboratorio de Molecular Farming y Vacunas-UB6, Instituto Tecnológico de Chascomús (CONICET-UNSAM), Av. Intendente Marino Km 8.2, Chascomús, Provincia de Buenos Aires 7030, Argentina; Escuela de Bio y Nanotecnologías, Campus Miguelete, 25 de Mayo y Francia, San Martín, Provincia de Buenos Aires 1650, Argentina
| | - Alejandro Orlowski
- Centro de Investigaciones Cardiovasculares "Dr. Horacio E. Cingolani" (CONICET), Universidad Nacional de La Plata, Facultad de Ciencias Médicas, La Plata, Argentina
| | - Carolina Jaquenod de Giusti
- Centro de Investigaciones Cardiovasculares "Dr. Horacio E. Cingolani" (CONICET), Universidad Nacional de La Plata, Facultad de Ciencias Médicas, La Plata, Argentina
| | - Mariana G Corigliano
- Laboratorio de Molecular Farming y Vacunas-UB6, Instituto Tecnológico de Chascomús (CONICET-UNSAM), Av. Intendente Marino Km 8.2, Chascomús, Provincia de Buenos Aires 7030, Argentina; Escuela de Bio y Nanotecnologías, Campus Miguelete, 25 de Mayo y Francia, San Martín, Provincia de Buenos Aires 1650, Argentina
| | - Ariel Legarralde
- Laboratorio de Molecular Farming y Vacunas-UB6, Instituto Tecnológico de Chascomús (CONICET-UNSAM), Av. Intendente Marino Km 8.2, Chascomús, Provincia de Buenos Aires 7030, Argentina; Escuela de Bio y Nanotecnologías, Campus Miguelete, 25 de Mayo y Francia, San Martín, Provincia de Buenos Aires 1650, Argentina
| | - Luisa F Mendoza-Morales
- Escuela de Bio y Nanotecnologías, Campus Miguelete, 25 de Mayo y Francia, San Martín, Provincia de Buenos Aires 1650, Argentina; Laboratorio de Biotecnologías en Bovinos y Ovinos, Instituto Tecnológico de Chascomús (CONICET-UNSAM), Av. Intendente Marino Km 8.2, Provincia de Buenos Aires, Argentina
| | - Agustín Atela
- Laboratorio de Molecular Farming y Vacunas-UB6, Instituto Tecnológico de Chascomús (CONICET-UNSAM), Av. Intendente Marino Km 8.2, Chascomús, Provincia de Buenos Aires 7030, Argentina; Escuela de Bio y Nanotecnologías, Campus Miguelete, 25 de Mayo y Francia, San Martín, Provincia de Buenos Aires 1650, Argentina
| | - Manuel A Sánchez
- Laboratorio de Molecular Farming y Vacunas-UB6, Instituto Tecnológico de Chascomús (CONICET-UNSAM), Av. Intendente Marino Km 8.2, Chascomús, Provincia de Buenos Aires 7030, Argentina; Escuela de Bio y Nanotecnologías, Campus Miguelete, 25 de Mayo y Francia, San Martín, Provincia de Buenos Aires 1650, Argentina
| | - Valeria A Sander
- Escuela de Bio y Nanotecnologías, Campus Miguelete, 25 de Mayo y Francia, San Martín, Provincia de Buenos Aires 1650, Argentina; Laboratorio de Biotecnologías en Bovinos y Ovinos, Instituto Tecnológico de Chascomús (CONICET-UNSAM), Av. Intendente Marino Km 8.2, Provincia de Buenos Aires, Argentina
| | - Sergio O Angel
- Escuela de Bio y Nanotecnologías, Campus Miguelete, 25 de Mayo y Francia, San Martín, Provincia de Buenos Aires 1650, Argentina; Laboratorio de Parasitología Molecular-UB2, Instituto Tecnológico de Chascomús (CONICET-UNSAM), Av. Intendente Marino Km 8.2, Chascomús, Provincia de Buenos Aires, Argentina
| | - Marina Clemente
- Laboratorio de Molecular Farming y Vacunas-UB6, Instituto Tecnológico de Chascomús (CONICET-UNSAM), Av. Intendente Marino Km 8.2, Chascomús, Provincia de Buenos Aires 7030, Argentina; Escuela de Bio y Nanotecnologías, Campus Miguelete, 25 de Mayo y Francia, San Martín, Provincia de Buenos Aires 1650, Argentina.
| |
Collapse
|
2
|
Hromić-Jahjefendić A, Lundstrom K, Adilović M, Aljabali AAA, Tambuwala MM, Serrano-Aroca Á, Uversky VN. Autoimmune response after SARS-CoV-2 infection and SARS-CoV-2 vaccines. Autoimmun Rev 2024; 23:103508. [PMID: 38160960 DOI: 10.1016/j.autrev.2023.103508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 12/19/2023] [Indexed: 01/03/2024]
Abstract
The complicated relationships between autoimmunity, COVID-19, and COVID-19 vaccinations are described, giving insight into their intricacies. Antinuclear antibodies (ANA), anti-Ro/SSA, rheumatoid factor, lupus anticoagulant, and antibodies against interferon (IFN)-I have all been consistently found in COVID-19 patients, indicating a high prevalence of autoimmune reactions following viral exposure. Furthermore, the discovery of human proteins with structural similarities to SARS-CoV-2 peptides as possible autoantigens highlights the complex interplay between the virus and the immune system in initiating autoimmunity. An updated summary of the current status of COVID-19 vaccines is presented. We present probable pathways underpinning the genesis of COVID-19 autoimmunity, such as bystander activation caused by hyperinflammatory conditions, viral persistence, and the creation of neutrophil extracellular traps. These pathways provide important insights into the development of autoimmune-related symptoms ranging from organ-specific to systemic autoimmune and inflammatory illnesses, demonstrating the wide influence of COVID-19 on the immune system.
Collapse
Affiliation(s)
- Altijana Hromić-Jahjefendić
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Hrasnicka cesta 15, 71000 Sarajevo, Bosnia and Herzegovina.
| | | | - Muhamed Adilović
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Hrasnicka cesta 15, 71000 Sarajevo, Bosnia and Herzegovina.
| | - Alaa A A Aljabali
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Yarmouk University, P.O. Box 566, Irbid 21163, Jordan.
| | - Murtaza M Tambuwala
- Lincoln Medical School, Brayford Pool Campus, University of Lincoln, Lincoln LN6 7TS, UK.
| | - Ángel Serrano-Aroca
- Biomaterials and Bioengineering Laboratory, Centro de Investigación Traslacional San Alberto Magno, Universidad Católica de Valencia San Vicente Mártir, c/Guillem de Castro 94, 46001, Valencia, Spain.
| | - Vladimir N Uversky
- Department of Molecular Medicine and USF Health Byrd Alzheimer's Institute, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA.
| |
Collapse
|
3
|
Lundstrom K. COVID-19 Vaccines: Where Did We Stand at the End of 2023? Viruses 2024; 16:203. [PMID: 38399979 PMCID: PMC10893040 DOI: 10.3390/v16020203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 01/23/2024] [Accepted: 01/25/2024] [Indexed: 02/25/2024] Open
Abstract
Vaccine development against SARS-CoV-2 has been highly successful in slowing down the COVID-19 pandemic. A wide spectrum of approaches including vaccines based on whole viruses, protein subunits and peptides, viral vectors, and nucleic acids has been developed in parallel. For all types of COVID-19 vaccines, good safety and efficacy have been obtained in both preclinical animal studies and in clinical trials in humans. Moreover, emergency use authorization has been granted for the major types of COVID-19 vaccines. Although high safety has been demonstrated, rare cases of severe adverse events have been detected after global mass vaccinations. Emerging SARS-CoV-2 variants possessing enhanced infectivity have affected vaccine protection efficacy requiring re-design and re-engineering of novel COVID-19 vaccine candidates. Furthermore, insight is given into preparedness against emerging SARS-CoV-2 variants.
Collapse
|
4
|
Kachko A, Selvaraj P, Liu S, Kim J, Rotstein D, Stauft CB, Chabot S, Rajasagi N, Zhao Y, Wang T, Major M. Vaccine-associated respiratory pathology correlates with viral clearance and protective immunity after immunization with self-amplifying RNA expressing the spike (S) protein of SARS-CoV-2 in mouse models. Vaccine 2024; 42:608-619. [PMID: 38142216 DOI: 10.1016/j.vaccine.2023.12.052] [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: 07/10/2023] [Revised: 12/18/2023] [Accepted: 12/18/2023] [Indexed: 12/25/2023]
Abstract
In this study, we evaluated the immunogenicity and protective immunity of in vitro transcribed Venezuelan equine encephalitis virus (VEEV TC-83 strain) self-amplifying RNA (saRNA) encoding the SARS-CoV-2 spike (S) protein in wild type (S-WT) and stabilized pre-fusion conformations (S-PP). Immunization with S-WT and S-PP saRNA induced specific neutralizing antibody responses in both K18-Tg hACE2 (K18) and BALB/c mice, as assessed using SARS-CoV-2 pseudotyped viruses. Protective immunity was assessed in challenge experiments. Two immunizations with S-WT and S-PP induced protective immunity, evidenced by lower mortality, lower weight loss and more than one log10 lower subgenomic virus RNA titers in the upper and lower respiratory tracts in both K18 and BALB/c mice. Histopathologic examination of lungs post-challenge showed that immunization with S-WT and S-PP resulted in a higher degree of immune cell infiltration and inflammatory changes, compared with control mice, characterized by high levels of T- and B-cell infiltration. No substantial differences were found in the presence and localization of eosinophils, macrophages, neutrophils, and natural killer cells. CD4 and CD8 T-cell depletion post immunization resulted in reduced lung inflammation post challenge but also prolonged virus clearance. These data indicate that immunization with saRNA encoding the SARS-CoV-2 S protein induces immune responses that are protective following challenge, that virus clearance is associated with pulmonary changes caused by T-cell and B-cell infiltration in the lungs, but that this T and B-cell infiltration plays an important role in viral clearance.
Collapse
Affiliation(s)
- Alla Kachko
- Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA.
| | - Prabhuanand Selvaraj
- Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | - Shufeng Liu
- Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | - Jaekwan Kim
- Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | - David Rotstein
- Division of Food Compliance, Center for Veterinary Medicine, Food and Drug Administration, Rockville, MD, USA
| | - Charles B Stauft
- Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | - Sylvie Chabot
- Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | - Naveen Rajasagi
- Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | - Yangqing Zhao
- Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | - Tony Wang
- Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | - Marian Major
- Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| |
Collapse
|
5
|
Meyers G, Tews BA. Self-Replicating RNA Derived from the Genomes of Positive-Strand RNA Viruses. Methods Mol Biol 2024; 2786:25-49. [PMID: 38814389 DOI: 10.1007/978-1-0716-3770-8_2] [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: 05/31/2024]
Abstract
Self-replicating RNA derived from the genomes of positive-strand RNA viruses represents a powerful tool for both molecular studies on virus biology and approaches to novel safe and effective vaccines. The following chapter summarizes the principles how such RNAs can be established and used for design of vaccines. Due to the large variety of strategies needed to circumvent specific pitfalls in the design of such constructs the technical details of the experiments are not described here but can be found in the cited literature.
Collapse
Affiliation(s)
- Gregor Meyers
- Institut für Immunologie, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Greifswald-Insel Riems, Germany
| | - Birke Andrea Tews
- Institut für Infektionsmedizin, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Greifswald-Insel Riems, Germany.
| |
Collapse
|
6
|
Koseki T, Teramachi M, Koga M, Ko MSH, Amano T, Yu H, Amano M, Leyder E, Badiola M, Ray P, Kim J, Ko AC, Achour A, Weng NP, Imai T, Yoshida H, Taniuchi S, Shintani A, Fujigaki H, Kondo M, Doi Y. A Phase I/II Clinical Trial of Intradermal, Controllable Self-Replicating Ribonucleic Acid Vaccine EXG-5003 against SARS-CoV-2. Vaccines (Basel) 2023; 11:1767. [PMID: 38140172 PMCID: PMC10747308 DOI: 10.3390/vaccines11121767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 11/11/2023] [Accepted: 11/22/2023] [Indexed: 12/24/2023] Open
Abstract
mRNA vaccines against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) have played a key role in reducing morbidity and mortality from coronavirus disease 2019 (COVID-19). We conducted a double-blind, placebo-controlled phase I/II trial to evaluate the safety, tolerability, and immunogenicity of EXG-5003, a two-dose, controllable self-replicating RNA vaccine against SARS-CoV-2. EXG-5003 encodes the receptor binding domain (RBD) of SARS-CoV-2 and was administered intradermally without lipid nanoparticles (LNPs). The participants were followed for 12 months. Forty healthy participants were enrolled in Cohort 1 (5 µg per dose, n = 16; placebo, n = 4) and Cohort 2 (25 µg per dose, n = 16; placebo, n = 4). No safety concerns were observed with EXG-5003 administration. SARS-CoV-2 RBD antibody titers and neutralizing antibody titers were not elevated in either cohort. Elicitation of antigen-specific cellular immunity was observed in the EXG-5003 recipients in Cohort 2. At the 12-month follow-up, participants who had received an approved mRNA vaccine (BNT162b2 or mRNA-1273) >1 month after receiving the second dose of EXG-5003 showed higher cellular responses compared with equivalently vaccinated participants in the placebo group. The findings suggest a priming effect of EXG-5003 on the long-term cellular immunity of approved SARS-CoV-2 mRNA vaccines.
Collapse
Affiliation(s)
- Takenao Koseki
- Department of Pharmacotherapeutics and Informatics, School of Medicine, Fujita Health University, Toyoake 470-1192, Japan;
| | - Mayumi Teramachi
- Center for Clinical Trial and Research Support, School of Medicine, Fujita Health University, Toyoake 470-1192, Japan; (M.T.); (M.K.)
| | - Minako Koga
- KM Pharmaceutical Consulting, Washington, DC 20006, USA;
| | - Minoru S. H. Ko
- Elixirgen Therapeutics, Inc., Baltimore, MD 21205, USA; (M.S.H.K.); (T.A.); (H.Y.); (M.A.); (E.L.); (M.B.); (P.R.); (J.K.); (A.C.K.)
| | - Tomokazu Amano
- Elixirgen Therapeutics, Inc., Baltimore, MD 21205, USA; (M.S.H.K.); (T.A.); (H.Y.); (M.A.); (E.L.); (M.B.); (P.R.); (J.K.); (A.C.K.)
| | - Hong Yu
- Elixirgen Therapeutics, Inc., Baltimore, MD 21205, USA; (M.S.H.K.); (T.A.); (H.Y.); (M.A.); (E.L.); (M.B.); (P.R.); (J.K.); (A.C.K.)
| | - Misa Amano
- Elixirgen Therapeutics, Inc., Baltimore, MD 21205, USA; (M.S.H.K.); (T.A.); (H.Y.); (M.A.); (E.L.); (M.B.); (P.R.); (J.K.); (A.C.K.)
| | - Erica Leyder
- Elixirgen Therapeutics, Inc., Baltimore, MD 21205, USA; (M.S.H.K.); (T.A.); (H.Y.); (M.A.); (E.L.); (M.B.); (P.R.); (J.K.); (A.C.K.)
| | - Maria Badiola
- Elixirgen Therapeutics, Inc., Baltimore, MD 21205, USA; (M.S.H.K.); (T.A.); (H.Y.); (M.A.); (E.L.); (M.B.); (P.R.); (J.K.); (A.C.K.)
| | - Priyanka Ray
- Elixirgen Therapeutics, Inc., Baltimore, MD 21205, USA; (M.S.H.K.); (T.A.); (H.Y.); (M.A.); (E.L.); (M.B.); (P.R.); (J.K.); (A.C.K.)
| | - Jiyoung Kim
- Elixirgen Therapeutics, Inc., Baltimore, MD 21205, USA; (M.S.H.K.); (T.A.); (H.Y.); (M.A.); (E.L.); (M.B.); (P.R.); (J.K.); (A.C.K.)
| | - Akihiro C. Ko
- Elixirgen Therapeutics, Inc., Baltimore, MD 21205, USA; (M.S.H.K.); (T.A.); (H.Y.); (M.A.); (E.L.); (M.B.); (P.R.); (J.K.); (A.C.K.)
| | - Achouak Achour
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, National Institutes of Health, Baltimore, MD 20892, USA; (A.A.); (N.-p.W.)
| | - Nan-ping Weng
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, National Institutes of Health, Baltimore, MD 20892, USA; (A.A.); (N.-p.W.)
| | - Takumi Imai
- Department of Medical Statistics, Graduate School of Medicine, Osaka Metropolitan University, Osaka 545-8585, Japan; (T.I.); (H.Y.); (S.T.); (A.S.)
| | - Hisako Yoshida
- Department of Medical Statistics, Graduate School of Medicine, Osaka Metropolitan University, Osaka 545-8585, Japan; (T.I.); (H.Y.); (S.T.); (A.S.)
| | - Satsuki Taniuchi
- Department of Medical Statistics, Graduate School of Medicine, Osaka Metropolitan University, Osaka 545-8585, Japan; (T.I.); (H.Y.); (S.T.); (A.S.)
| | - Ayumi Shintani
- Department of Medical Statistics, Graduate School of Medicine, Osaka Metropolitan University, Osaka 545-8585, Japan; (T.I.); (H.Y.); (S.T.); (A.S.)
| | - Hidetsugu Fujigaki
- Department of Advanced Diagnostic System Development, Graduate School of Health Sciences, Fujita Health University, Toyoake 470-1192, Japan
| | - Masashi Kondo
- Center for Clinical Trial and Research Support, School of Medicine, Fujita Health University, Toyoake 470-1192, Japan; (M.T.); (M.K.)
- Department of Respiratory Medicine, School of Medicine, Fujita Health University, Toyoake 470-1192, Japan
| | - Yohei Doi
- Departments of Microbiology and Infectious Diseases, School of Medicine, Fujita Health University, Toyoake 470-1192, Japan
- Center for Infectious Disease Research, Fujita Health University, Toyoake 470-1192, Japan
- Division of Infectious Diseases, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
| |
Collapse
|
7
|
Lundstrom K. Application of DNA Replicons in Gene Therapy and Vaccine Development. Pharmaceutics 2023; 15:pharmaceutics15030947. [PMID: 36986808 PMCID: PMC10054396 DOI: 10.3390/pharmaceutics15030947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 03/04/2023] [Accepted: 03/13/2023] [Indexed: 03/17/2023] Open
Abstract
DNA-based gene therapy and vaccine development has received plenty of attention lately. DNA replicons based on self-replicating RNA viruses such as alphaviruses and flaviviruses have been of particular interest due to the amplification of RNA transcripts leading to enhanced transgene expression in transfected host cells. Moreover, significantly reduced doses of DNA replicons compared to conventional DNA plasmids can elicit equivalent immune responses. DNA replicons have been evaluated in preclinical animal models for cancer immunotherapy and for vaccines against infectious diseases and various cancers. Strong immune responses and tumor regression have been obtained in rodent tumor models. Immunization with DNA replicons has provided robust immune responses and protection against challenges with pathogens and tumor cells. DNA replicon-based COVID-19 vaccines have shown positive results in preclinical animal models.
Collapse
|
8
|
Zhang T, Wang Z, Yang J, Xu X. Immunogenicity of novel DNA vaccines encoding receptor-binding domain (RBD) dimer-Fc fusing antigens derived from different SARS-CoV-2 variants of concern. J Med Virol 2023; 95:e28563. [PMID: 36755368 DOI: 10.1002/jmv.28563] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 01/16/2023] [Accepted: 02/06/2023] [Indexed: 02/10/2023]
Abstract
The continuously emerging of severe acute respiratory syndrome coronavirus-2 variants of concern (VOCs) led to a decline in effectiveness of the first-generation vaccines. Therefore, optimized vaccines and vaccination strategies, which show advantages in protecting against VOCs, are urgently needed. Here we constructed an optimized DNA vaccine plasmid containing built-in CpG adjuvant, and designed vaccine candidates encoding five forms of antigens derived from Wuhan-Hu-1. The results showed that plasmid with receptor binding domain (RBD) dimer-Fc fusing antigen (2RBD-Fc) induced the highest level of RBD-specific IgG and neutralizing antibodies in mice. Then 2dRBD-Fc and 2omRBD-Fc vaccines, respectively derived from delta and omicron VOCs, were constructed. The 2dRBD-Fc induced potent humoral and cellular immune responses, while the immunogenicity of 2omRBD-Fc was low. We also observed that sequential immunization with 2RBD-Fc, 2dRBD-Fc and 2omRBD-Fc effectively elicited neutralizing antibodies against each immunized strain, and RBD-specific T cell responses. To be noted, the Wuhan-Hu-1, delta and omicron neutralizing antibody titers induced by sequential immunization were comparable to that induced by repetitive immunization with 2RBD-Fc, 2dRBD-Fc or 2omRBD-Fc respectively. The results suggest that sequential immunization with DNA vaccines encoding potent antigens derived from different VOCs, may be a promising strategy to elicit immune responses against multiple variants.
Collapse
Affiliation(s)
- Ting Zhang
- Department of Biophysics and Structural Biology, School of Basic Medicine Peking Union Medical College, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, Beijing, China
| | - Zhirong Wang
- Department of Biophysics and Structural Biology, School of Basic Medicine Peking Union Medical College, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, Beijing, China
| | - Jiaojiao Yang
- Department of Biophysics and Structural Biology, School of Basic Medicine Peking Union Medical College, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, Beijing, China
| | - Xuemei Xu
- Department of Biophysics and Structural Biology, School of Basic Medicine Peking Union Medical College, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
9
|
Elliott T, Cheeseman HM, Evans AB, Day S, McFarlane LR, O’Hara J, Kalyan M, Amini F, Cole T, Winston A, Fidler S, Pollock KM, Harker JA, Shattock RJ. Enhanced immune responses following heterologous vaccination with self-amplifying RNA and mRNA COVID-19 vaccines. PLoS Pathog 2022; 18:e1010885. [PMID: 36194628 PMCID: PMC9565686 DOI: 10.1371/journal.ppat.1010885] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 10/14/2022] [Accepted: 09/14/2022] [Indexed: 11/06/2022] Open
Abstract
The optimal vaccination strategy to boost responses in the context of pre-existing immune memory to the SARS-CoV-2 spike (S) glycoprotein is an important question for global public health. To address this, we explored the SARS-CoV-2-specific humoral and cellular immune responses to a novel self-amplifying RNA (saRNA) vaccine followed by a UK authorised mRNA vaccine (BNT162b2) in individuals with and without previous COVID-19, and compared these responses with those who received an authorised vaccine alone. 35 subjects receiving saRNA (saRNA group) as part of the COVAC1 clinical trial and an additional 40 participants receiving an authorised SARS-CoV-2 vaccine only (non-saRNA group) were recruited. Antibody responses were measured by ELISA and a pseudoneutralisation assay for wildtype, Delta and Omicron variants. Cellular responses were measured by IFN-ƴ ELISpot and an activation induced marker (AIM) assay. Approximately 50% in each group had previous COVID-19 prior to vaccination, confirmed by PCR or antibody positivity on ELISA. All of those who received saRNA subsequently received a full course of an authorised vaccine. The majority (83%) of those receiving saRNA who were COVID-19 naïve at baseline seroconverted following the second dose, and those with previous COVID-19 had an increase in antibody titres two weeks following saRNA vaccination (median 27-fold), however titres were lower when compared to mRNA vaccination. Two weeks following the 2nd authorised mRNA vaccine dose, binding and neutralising antibody titres were significantly higher in the saRNA participants with previous COVID-19, compared to non-saRNA, or COVID-19 naive saRNA participants. Cellular responses were again highest in this group, with a higher proportion of spike specific CD8+ than CD4+ T cells when compared to those receiving the mRNA vaccine only. These findings suggest an immunological benefit of increased antigen exposure, both from natural infection and vaccination, particularly evident in those receiving heterologous vaccination with saRNA and mRNA.
Collapse
Affiliation(s)
- Tamara Elliott
- Department of Infectious Disease, Imperial College London, United Kingdom
- Imperial College Healthcare NHS Trust, London, United Kingdom
- Imperial College NIHR BRC, London, United Kingdom
| | | | - Abbey B. Evans
- Department of Infectious Disease, Imperial College London, United Kingdom
| | - Suzanne Day
- Department of Infectious Disease, Imperial College London, United Kingdom
| | - Leon R. McFarlane
- Department of Infectious Disease, Imperial College London, United Kingdom
| | - Jessica O’Hara
- Department of Infectious Disease, Imperial College London, United Kingdom
| | - Mohini Kalyan
- Department of Infectious Disease, Imperial College London, United Kingdom
| | - Fahimah Amini
- Department of Infectious Disease, Imperial College London, United Kingdom
| | - Tom Cole
- Department of Infectious Disease, Imperial College London, United Kingdom
- Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Alan Winston
- Department of Infectious Disease, Imperial College London, United Kingdom
- Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Sarah Fidler
- Department of Infectious Disease, Imperial College London, United Kingdom
- Imperial College Healthcare NHS Trust, London, United Kingdom
- Imperial College NIHR BRC, London, United Kingdom
| | - Katrina M. Pollock
- Department of Infectious Disease, Imperial College London, United Kingdom
- Imperial College Healthcare NHS Trust, London, United Kingdom
| | | | - Robin J. Shattock
- Department of Infectious Disease, Imperial College London, United Kingdom
| |
Collapse
|
10
|
Moliner-Morro A, McInerney GM, Hanke L. Nanobodies in the limelight: Multifunctional tools in the fight against viruses. J Gen Virol 2022; 103. [PMID: 35579613 DOI: 10.1099/jgv.0.001731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Antibodies are natural antivirals generated by the vertebrate immune system in response to viral infection or vaccination. Unsurprisingly, they are also key molecules in the virologist's molecular toolbox. With new developments in methods for protein engineering, protein functionalization and application, smaller antibody-derived fragments are moving in focus. Among these, camelid-derived nanobodies play a prominent role. Nanobodies can replace full-sized antibodies in most applications and enable new possible applications for which conventional antibodies are challenging to use. Here we review the versatile nature of nanobodies, discuss their promise as antiviral therapeutics, for diagnostics, and their suitability as research tools to uncover novel aspects of viral infection and disease.
Collapse
Affiliation(s)
- Ainhoa Moliner-Morro
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Gerald M McInerney
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | | |
Collapse
|
11
|
Hawman DW, Meade-White K, Archer J, Leventhal SS, Wilson D, Shaia C, Randall S, Khandhar AP, Krieger K, Hsiang TY, Gale M, Berglund P, Fuller DH, Feldmann H, Erasmus JH. SARS-CoV2 variant-specific replicating RNA vaccines protect from disease following challenge with heterologous variants of concern. eLife 2022; 11:e75537. [PMID: 35191378 PMCID: PMC8983041 DOI: 10.7554/elife.75537] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Accepted: 02/17/2022] [Indexed: 11/14/2022] Open
Abstract
Despite mass public health efforts, the SARS-CoV2 pandemic continues as of late 2021 with resurgent case numbers in many parts of the world. The emergence of SARS-CoV2 variants of concern (VoCs) and evidence that existing vaccines that were designed to protect from the original strains of SARS-CoV-2 may have reduced potency for protection from infection against these VoC is driving continued development of second-generation vaccines that can protect against multiple VoC. In this report, we evaluated an alphavirus-based replicating RNA vaccine expressing Spike proteins from the original SARS-CoV-2 Alpha strain and recent VoCs delivered in vivo via a lipid inorganic nanoparticle. Vaccination of both mice and Syrian Golden hamsters showed that vaccination induced potent neutralizing titers against each homologous VoC but reduced neutralization against heterologous challenges. Vaccinated hamsters challenged with homologous SARS-CoV2 variants exhibited complete protection from infection. In addition, vaccinated hamsters challenged with heterologous SARS-CoV-2 variants exhibited significantly reduced shedding of infectious virus. Our data demonstrate that this vaccine platform can be updated to target emergent VoCs, elicits significant protective immunity against SARS-CoV2 variants and supports continued development of this platform.
Collapse
Affiliation(s)
- David W Hawman
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain LaboratoriesHamiltonUnited States
| | - Kimberly Meade-White
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain LaboratoriesHamiltonUnited States
| | | | - Shanna S Leventhal
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain LaboratoriesHamiltonUnited States
| | - Drew Wilson
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain LaboratoriesHamiltonUnited States
| | - Carl Shaia
- Rocky Mountain Veterinary Branch, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain LaboratoriesHamiltonUnited States
| | - Samantha Randall
- Department of Microbiology, University of Washington School of MedicineSeattleUnited States
| | | | | | - Tien-Ying Hsiang
- Center for Innate Immunity and Immune Disease, Department of Immunology, University of Washington School of MedicineSeattleUnited States
| | - Michael Gale
- Center for Innate Immunity and Immune Disease, Department of Immunology, University of Washington School of MedicineSeattleUnited States
| | | | | | - Heinz Feldmann
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain LaboratoriesHamiltonUnited States
| | - Jesse H Erasmus
- HDT BioSeattleUnited States
- Department of Microbiology, University of Washington School of MedicineSeattleUnited States
| |
Collapse
|
12
|
Immunogenicity and protective efficacy of a DNA vaccine inducing optimal expression of the SARS-CoV-2 S gene in hACE2 mice. Arch Virol 2022; 167:2519-2528. [PMID: 36083350 PMCID: PMC9462073 DOI: 10.1007/s00705-022-05562-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 06/28/2022] [Indexed: 01/07/2023]
Abstract
The wide spread of coronavirus disease 2019 (COVID-19) has significantly threatened public health. Human herd immunity induced by vaccination is essential to fight the epidemic. Therefore, highly immunogenic and safe vaccines are necessary to control SARS-CoV-2, whose S protein is the antigenic determinant responsible for eliciting antibodies that prevent viral entry and fusion. In this study, we developed a SARS-CoV-2 DNA vaccine expressing the S protein, named pVAX-S-OP, which was optimized according to the human-origin codon preference and using polyinosinic-polycytidylic acid as an adjuvant. pVAX-S-OP induced specific antibodies and neutralizing antibodies in BALB/c and hACE2 transgenic mice. Furthermore, we observed 1.43-fold higher antibody titers in mice receiving pVAX-S-OP plus adjuvant than in those receiving pVAX-S-OP alone. Interferon gamma production in the pVAX-S-OP-immunized group was 1.58 times (CD3+CD4+IFN-gamma+) and 2.29 times (CD3+CD8+IFN-gamma+) lower than that in the pVAX-S-OP plus adjuvant group but higher than that in the control group. The pVAX-S-OP vaccine was also observed to stimulate a Th1-type immune response. When, hACE2 transgenic mice were challenged with SARS-CoV-2, qPCR detection of N and E genes showed that the viral RNA loads in pVAX-S-OP-immunized mice lung tissues were 104 times and 106 times lower than those of the PBS control group, which shows that the vaccine could reduce the amount of live virus in the lungs of hACE2 mice. In addition, pathological sections showed less lung damage in the pVAX-S-OP-immunized group. Taken together, our results demonstrated that pVAX-S-OP has significant immunogenicity, which provides support for developing SARS-CoV-2 DNA candidate vaccines.
Collapse
|
13
|
Hawman DW, Meade-White K, Archer J, Leventhal S, Wilson D, Shaia C, Randall S, Khandhar AP, Hsiang TY, Gale M, Berglund P, Fuller DH, Feldmann H, Erasmus JH. SARS-CoV2 variant-specific replicating RNA vaccines protect from disease and pathology and reduce viral shedding following challenge with heterologous SARS-CoV2 variants of concern. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021:2021.12.10.472134. [PMID: 34931189 PMCID: PMC8687464 DOI: 10.1101/2021.12.10.472134] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Despite mass public health efforts, the SARS-CoV2 pandemic continues as of late-2021 with resurgent case numbers in many parts of the world. The emergence of SARS-CoV2 variants of concern (VoC) and evidence that existing vaccines that were designed to protect from the original strains of SARS-CoV-2 may have reduced potency for protection from infection against these VoC is driving continued development of second generation vaccines that can protect against multiple VoC. In this report, we evaluated an alphavirus-based replicating RNA vaccine expressing Spike proteins from the original SARS-CoV-2 Alpha strain and recent VoCs delivered in vivo via a lipid inorganic nanoparticle. Vaccination of both mice and Syrian Golden hamsters showed that vaccination induced potent neutralizing titers against each homologous VoC but reduced neutralization against heterologous challenges. Vaccinated hamsters challenged with homologous SARS-CoV2 variants exhibited complete protection from infection. In addition, vaccinated hamsters challenged with heterologous SARS-CoV-2 variants exhibited significantly reduced shedding of infectious virus. Our data demonstrate that this vaccine platform elicits significant protective immunity against SARS-CoV2 variants and supports continued development of this platform.
Collapse
Affiliation(s)
- David W Hawman
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT 59840, USA
| | - Kimberly Meade-White
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT 59840, USA
| | | | - Shanna Leventhal
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT 59840, USA
| | - Drew Wilson
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT 59840, USA
| | - Carl Shaia
- Rocky Mountain Veterinary Branch, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT 59840, USA
| | - Samantha Randall
- Department of Microbiology, University of Washington School of Medicine, Seattle, WA 98109, USA
| | | | - Tien-Ying Hsiang
- Center for Innate Immunity and Immune Disease, Department of Immunology, University of Washington School of Medicine, Seattle, WA 98109, USA
| | - Michael Gale
- Center for Innate Immunity and Immune Disease, Department of Immunology, University of Washington School of Medicine, Seattle, WA 98109, USA
| | | | | | - Heinz Feldmann
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT 59840, USA
| | - Jesse H Erasmus
- HDT Bio, Seattle, WA 98102, USA
- Department of Microbiology, University of Washington School of Medicine, Seattle, WA 98109, USA
| |
Collapse
|
14
|
Langereis MA, Albulescu IC, Stammen-Vogelzangs J, Lambregts M, Stachura K, Miller S, Bosco-Lauth AM, Hartwig AE, Porter SM, Allen M, Mogler M, van Kuppeveld FJM, Bosch BJ, Vermeij P, de Groof A, Bowen RA, Davis R, Xu Z, Tarpey I. An alphavirus replicon-based vaccine expressing a stabilized Spike antigen induces protective immunity and prevents transmission of SARS-CoV-2 between cats. NPJ Vaccines 2021; 6:122. [PMID: 34671047 PMCID: PMC8528862 DOI: 10.1038/s41541-021-00390-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 10/01/2021] [Indexed: 12/01/2022] Open
Abstract
Early in the SARS-CoV-2 pandemic concerns were raised regarding infection of new animal hosts and the effect on viral epidemiology. Infection of other animals could be detrimental by causing clinical disease, allowing further mutations, and bares the risk for the establishment of a non-human reservoir. Cats were the first reported animals susceptible to natural and experimental infection with SARS-CoV-2. Given the concerns these findings raised, and the close contact between humans and cats, we aimed to develop a vaccine candidate that could reduce SARS-CoV-2 infection and in addition to prevent spread among cats. Here we report that a Replicon Particle (RP) vaccine based on Venezuelan equine encephalitis virus, known to be safe and efficacious in a variety of animal species, could induce neutralizing antibody responses in guinea pigs and cats. The design of the SARS-CoV-2 spike immunogen was critical in developing a strong neutralizing antibody response. Vaccination of cats was able to induce high neutralizing antibody responses, effective also against the SARS-CoV-2 B.1.1.7 variant. Interestingly, in contrast to control animals, the infectious virus could not be detected in oropharyngeal or nasal swabs of vaccinated cats after SARS-CoV-2 challenge. Correspondingly, the challenged control cats spread the virus to in-contact cats whereas the vaccinated cats did not transmit the virus. The results show that the RP vaccine induces protective immunity preventing SARS-CoV-2 infection and transmission. These data suggest that this RP vaccine could be a multi-species vaccine useful to prevent infection and spread to and between animals should that approach be required.
Collapse
Affiliation(s)
| | - Irina C Albulescu
- Division of Infectious Diseases and Immunology, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | | | | | | | | | - Angela M Bosco-Lauth
- College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, USA
| | - Airn E Hartwig
- College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, USA
| | - Stephanie M Porter
- College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, USA
| | | | | | - Frank J M van Kuppeveld
- Division of Infectious Diseases and Immunology, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Berend-Jan Bosch
- Division of Infectious Diseases and Immunology, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | | | | | - Richard A Bowen
- College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, USA
| | | | - Zach Xu
- Merck Animal Health, Elkhorn, NE, USA
| | | |
Collapse
|
15
|
Targeting SARS-CoV-2 receptor-binding domain to cells expressing CD40 improves protection to infection in convalescent macaques. Nat Commun 2021; 12:5215. [PMID: 34471122 PMCID: PMC8410935 DOI: 10.1038/s41467-021-25382-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 07/21/2021] [Indexed: 12/27/2022] Open
Abstract
Achieving sufficient worldwide vaccination coverage against SARS-CoV-2 will require additional approaches to currently approved viral vector and mRNA vaccines. Subunit vaccines may have distinct advantages when immunizing vulnerable individuals, children and pregnant women. Here, we present a new generation of subunit vaccines targeting viral antigens to CD40-expressing antigen-presenting cells. We demonstrate that targeting the receptor-binding domain (RBD) of the SARS-CoV-2 spike protein to CD40 (αCD40.RBD) induces significant levels of specific T and B cells, with long-term memory phenotypes, in a humanized mouse model. Additionally, we demonstrate that a single dose of the αCD40.RBD vaccine, injected without adjuvant, is sufficient to boost a rapid increase in neutralizing antibodies in convalescent non-human primates (NHPs) exposed six months previously to SARS-CoV-2. Vaccine-elicited antibodies cross-neutralize different SARS-CoV-2 variants, including D614G, B1.1.7 and to a lesser extent B1.351. Such vaccination significantly improves protection against a new high-dose virulent challenge versus that in non-vaccinated convalescent animals. In this study, Marlin et al. provide insights into the potential use of subunit vaccines that induce a high level of protection against SARS-CoV-2 in animal models.
Collapse
|
16
|
Scaglione A, Opp S, Hurtado A, Lin Z, Pampeno C, Noval MG, Thannickal SA, Stapleford KA, Meruelo D. Combination of a Sindbis-SARS-CoV-2 Spike Vaccine and αOX40 Antibody Elicits Protective Immunity Against SARS-CoV-2 Induced Disease and Potentiates Long-Term SARS-CoV-2-Specific Humoral and T-Cell Immunity. Front Immunol 2021; 12:719077. [PMID: 34394127 PMCID: PMC8359677 DOI: 10.3389/fimmu.2021.719077] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 07/13/2021] [Indexed: 12/17/2022] Open
Abstract
The COVID-19 pandemic caused by the coronavirus SARS-CoV-2 is a major global public threat. Currently, a worldwide effort has been mounted to generate billions of effective SARS-CoV-2 vaccine doses to immunize the world's population at record speeds. However, there is still a demand for alternative effective vaccines that rapidly confer long-term protection and rely upon cost-effective, easily scaled-up manufacturing. Here, we present a Sindbis alphavirus vector (SV), transiently expressing the SARS-CoV-2 spike protein (SV.Spike), combined with the OX40 immunostimulatory antibody (αOX40) as a novel, highly effective vaccine approach. We show that SV.Spike plus αOX40 elicits long-lasting neutralizing antibodies and a vigorous T-cell response in mice. Protein binding, immunohistochemical, and cellular infection assays all show that vaccinated mice sera inhibits spike functions. Immunophenotyping, RNA Seq transcriptome profiles, and metabolic analysis indicate a reprogramming of T cells in vaccinated mice. Activated T cells were found to mobilize to lung tissue. Most importantly, SV.Spike plus αOX40 provided robust immune protection against infection with authentic coronavirus in transgenic mice expressing the human ACE2 receptor (hACE2-Tg). Finally, our immunization strategy induced strong effector memory response, potentiating protective immunity against re-exposure to SARS-CoV-2 spike protein. Our results show the potential of a new Sindbis virus-based vaccine platform to counteract waning immune response, which can be used as a new candidate to combat SARS-CoV-2. Given the T-cell responses elicited, our vaccine is likely to be effective against variants that are proving challenging, as well as serve as a platform to develop a broader spectrum pancoronavirus vaccine. Similarly, the vaccine approach is likely to be applicable to other pathogens.
Collapse
Affiliation(s)
- Antonella Scaglione
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, United States
| | - Silvana Opp
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, United States
| | - Alicia Hurtado
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, United States
| | - Ziyan Lin
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, United States
| | - Christine Pampeno
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, United States
| | - Maria G. Noval
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, United States
| | - Sara A. Thannickal
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, United States
| | - Kenneth A. Stapleford
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, United States
| | - Daniel Meruelo
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, United States
| |
Collapse
|