1
|
Deng Y, Cheng H, Zhu J, Jiang Y, Sun H, Li G, Wei J, Xue R, Feng R, Cao J, Yu W, Wang Y, Xu M, Zou Q, Li H. Tamibarotene-Loaded Nanoemulsion Incorporating Toll-like Receptor 2/6 Agonist as an Intramuscular Adjuvant System Enhances Gastrointestinal Mucosal Immunity. ACS NANO 2025; 19:19149-19167. [PMID: 40376869 DOI: 10.1021/acsnano.5c00563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2025]
Abstract
Parenteral subunit vaccines typically elicit systemic humoral immune responses but often struggle to induce mucosal immunity. Herein, we developed a promising adjuvant system, TB/P2C-NE, a tamibarotene-loaded nanoemulsion incorporating the TLR2/6 agonist Pam2CSK4. Upon intramuscular vaccination, TB/P2C-NE promoted antigen-specific mucosal immune responses in the gastrointestinal tract, accompanied by systemic humoral and cellular response. Mechanistically, tamibarotene upregulated the intestinal homing molecule CCR9 on lymphocytes through dendritic cell modulation, while Pam2CSK4 increased IL-6 secretion at the injection sites, further amplifying CCR9 expression and lymphocyte activation and leading to enhanced lymphocyte homing to the intestinal mucosa and a subsequent boost in mucosal immunity. Notably, TB/P2C-NE induced long-term gastrointestinal mucosal responses, maintaining elevated sIgA levels for up to three months post-immunization, and also induced gastrointestinal mucosal immunity in combination with a polysaccharide conjugate antigen. Immunization with recombinant intimin using TB/P2C-NE as the adjuvant resulted in a robust protective effect against the EHEC O157:H7 challenge. In summary, TB/P2C-NE offers an adjuvant strategy potentially accelerating the development of vaccines targeting gastrointestinal infections.
Collapse
Affiliation(s)
- Yan Deng
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Hao Cheng
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Ji Zhu
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Yue Jiang
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Hongwu Sun
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Guocheng Li
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Jing Wei
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, China
- School of Clinical Medicine, Jiamusi University, Jiamusi, Heilongjiang 154007, China
| | - Ruoyi Xue
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Rang Feng
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Jingwen Cao
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, China
- School of Clinical Medicine, Jiamusi University, Jiamusi, Heilongjiang 154007, China
| | - Wenkang Yu
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Yalan Wang
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Mingqi Xu
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, China
- School of Clinical Medicine, Jiamusi University, Jiamusi, Heilongjiang 154007, China
| | - Quanming Zou
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Haibo Li
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, China
| |
Collapse
|
2
|
de Oliveira B, Goes WM, Nascimento FC, Carnielli JBT, Ferreira ER, de Carvalho AF, Dos Reis PVM, Pereira M, Ricotta TQN, Dos Santos LM, de Souza RP, Cargnelutti DE, Mottram JC, Teixeira SR, Fernandes AP, Gazzinelli RT. Characterization of a novel Leishmania antigen containing a repetitive domain and its potential use as a prophylactic and therapeutic vaccine. mSphere 2025; 10:e0009725. [PMID: 40261025 DOI: 10.1128/msphere.00097-25] [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: 03/07/2025] [Accepted: 03/21/2025] [Indexed: 04/24/2025] Open
Abstract
Human visceral leishmaniasis (HVL) is the second most lethal tropical parasitic disease. Currently, no prophylactic or therapeutic vaccines exist for HVL. Thus, the development of an efficacious vaccine is still needed. We previously performed an immunoproteomics analysis on Leishmania amazonensis parasite extracts to identify immunodominant antigens recognized by the sera of vaccinated and protected mice. Among the identified antigens, we discovered a novel, previously unstudied repetitive protein, initially annotated in Leishmania genomes as a kinetoplast-associated protein-like protein from Leishmania infantum (LinKAP), containing conserved domains (trichohyalin-plectin-homology [TPH] and TolA) that are associated with other mitochondrial proteins. LinKAP sequences are conserved across trypanosomatids, including Endotrypanum, Leishmania, and Trypanosoma species. Using differential centrifugation of Leishmania subcellular structures, we showed that LinKAP was enriched in fractions colocalizing with other mitochondrial proteins. mNeonGreen labeling at the endogenous locus using CRISPR-Cas9 and confocal microscopy confirmed that LinKAP is a mitochondrial-associated protein in Leishmania but not specifically colocalized with kDNA. We cloned and expressed a truncated version of LinKAP (rLinKAP), containing part (15) of the several LinKAP amino acid repeats, demonstrating over 85% homology across L. infantum, L. amazonensis, L. braziliensis, and L. mexicana species. An adjuvanted formulation of LinKAP with Poly ICLC, a polyinosinic-polycytidylic acid (Poly I:C) stabilized with carboxymethylcellulose and polylysine, was used to vaccinate mice and hamsters as a prophylactic vaccine for visceral leishmaniasis. Animals immunized with rLinKAP showed a potent cellular and humoral response and a significant decrease in tissue parasitism when challenged with L. infantum. We also tested rLinKAP as a therapeutic vaccine in mice. Following therapeutic vaccination, antibody responses were enhanced, and cellular responses became apparent. Our treatment protocol inhibited splenic parasite burden by 75% in treated mice. In conclusion, our antigen discovery strategy and the observed protective effect highlight rLinKAP as a promising vaccine candidate for leishmaniasis. IMPORTANCE A previous reverse vaccinology study identified kinetoplast-associated protein-like protein from Leishmania infantum (LinKAP) as a potential new vaccine target, as this protein was recognized by the sera of protected mice in extracts of Leishmania promastigotes. Interestingly, LinKAP is a repetitive protein containing trichohyalin-plectin-homology (TPH) and TolA domains and was initially annotated as a kinetoplast-associated protein. We further characterized LinKAP as a mitochondrial-associated protein highly conserved among trypanosomatids. We also validated LinKAP as a promising vaccine antigen by using a truncated version of LinKAP (rLinKAP) as both a prophylactic and therapeutic vaccine, adjuvanted with Poly ICLC, to immunize animals against visceral leishmaniasis (VL). This disease, caused by the Leishmania parasite, affects several populations globally and still lacks highly effective vaccines. Identifying LinKAP and its preliminary characterization also provides new perspectives for studying its role in the parasite's biology.
Collapse
Affiliation(s)
- Bianca de Oliveira
- Centro de Tecnologia de Vacinas (CTVacinas), Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Instituto René Rachou, Fundação Osvaldo Cruz-Minas, Belo Horizonte, Minas Gerais, Brazil
| | - Wanessa M Goes
- Centro de Tecnologia de Vacinas (CTVacinas), Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Frederico C Nascimento
- Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Juliana B T Carnielli
- York Biomedical Research Institute, Department of Biology, University of York, York, United Kingdom
| | - Eden R Ferreira
- York Biomedical Research Institute, Department of Biology, University of York, York, United Kingdom
| | - Alex Fiorini de Carvalho
- Centro de Tecnologia de Vacinas (CTVacinas), Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Pablo Victor Mendes Dos Reis
- Centro de Tecnologia de Vacinas (CTVacinas), Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Milton Pereira
- Centro de Tecnologia de Vacinas (CTVacinas), Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | | | - Renan Pedra de Souza
- Centro de Tecnologia de Vacinas (CTVacinas), Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Diego Esteban Cargnelutti
- Instituto de Medicina y Biología Experimental de Cuyo (IMBECU), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Cuyo (UNCuyo), Mendoza, Argentina
| | - Jeremy C Mottram
- York Biomedical Research Institute, Department of Biology, University of York, York, United Kingdom
| | - Santuza R Teixeira
- Centro de Tecnologia de Vacinas (CTVacinas), Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Ana Paula Fernandes
- Centro de Tecnologia de Vacinas (CTVacinas), Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Ricardo T Gazzinelli
- Centro de Tecnologia de Vacinas (CTVacinas), Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Instituto René Rachou, Fundação Osvaldo Cruz-Minas, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
3
|
Reis EVDS, Ferreira LL, Clarindo FA, Marques-Ferreira G, de Oliveira LC, Moraes TDFS, de Carvalho LD, da Fonseca FG, Sabino ADP, Eapen MS, Teixeira MM, Valle SJ, Morris DL, Coelho-Dos-Reis JGA. Antiviral effect of Bromelain combined with acetylcysteine against SARS-CoV-2 Omicron variant. Sci Rep 2025; 15:11882. [PMID: 40195407 PMCID: PMC11976912 DOI: 10.1038/s41598-025-92242-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 02/26/2025] [Indexed: 04/09/2025] Open
Abstract
The recent pandemic represented one of the biggest challenges of modern civilization. SARS-CoV-2 remains an imminent public health threat and currently, there is no effective and greatly affordable treatment for severe COVID-19. Although standard management with dexamethasone, and physical management including physiotherapy, prone positioning and mechanical ventilation are used, severe disease patients may still succumb to infection. In this regard, BromAc® is a combination therapy of a refined protein derived from Bromelain and acetylcysteine, that shows significant mucolytic and anti-inflammatory properties. In the present study, we performed in vitro, and ex vivo analyses to assess the effect of BromAc® in inhibiting Omicron variant of SARS-CoV-2 at different levels. Here, we provide evidence of the in vitro virucidal activity of BromAc® in Vero-ACE2/TMPRSS2 cell line infected with the Omicron variant. BromAc® can also abrogate SARS-CoV-2 RNA genomic copies in tracheal aspirate (TA) samples from critically ill COVID-19 patients after long term exposure. These results were confirmed by lower spike expression observed in EpCAM+PanCKneg epithelial cells from tracheal aspirate samples after BromAc® treatment. Furthermore, atomized BromAc® promoted cleavage of the S1 Spike subunit in TA samples, demonstrating the mechanism of the antiviral activity displayed by BromAc® in human samples. These results bring novel evidence of antiviral activity in cell lines in vitro as well as in tracheal aspirate samples from critically ill COVID-19 patients, which support its potential use as an adjunct to COVID-19 management in future waves of Omicron subvariants.
Collapse
Affiliation(s)
- Erik Vinicius de Sousa Reis
- Laboratório de Virologia Básica e Aplicada (LVBA), Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Linziane Lopes Ferreira
- Laboratório de Virologia Básica e Aplicada (LVBA), Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Felipe Alves Clarindo
- Laboratório de Virologia Básica e Aplicada (LVBA), Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Geovane Marques-Ferreira
- Laboratório de Virologia Básica e Aplicada (LVBA), Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Leonardo Camilo de Oliveira
- CT Terapias Avançadas e Inovadoras, (CT-Terapias), Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Thaís de Fátima Silva Moraes
- Laboratório de Virologia Básica e Aplicada (LVBA), Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Luciana Debortoli de Carvalho
- Laboratório de Microbiologia, Departamento de ciências biológicas, universidade estadual de Santa Cruz, Ilhéus, Bahia, Brazil
| | - Flávio Guimarães da Fonseca
- Laboratório de Virologia Básica e Aplicada (LVBA), Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
- Centro de Tecnologia em Vacinas (CT-Vacinas), Parque Tecnológico de Belo Horizonte, Belo Horizonte, MG, Brazil
| | - Adriano de Paula Sabino
- Laboratório de Hematologia Clínica, Experimental e Molecular, Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia/UFMG, Belo Horizonte, Brazil
| | - Mathew Suji Eapen
- Laboratório de Virologia Básica e Aplicada (LVBA), Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Mauro Martins Teixeira
- CT Terapias Avançadas e Inovadoras, (CT-Terapias), Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Sarah J Valle
- Mucpharm, Sydney, Australia.
- Intensive Care Unit, St George Hospital, Sydney, Australia.
- St George Hospital Clinical School, University of New South Wales, Sydney, Australia.
| | - David L Morris
- Mucpharm, Sydney, Australia.
- Intensive Care Unit, St George Hospital, Sydney, Australia.
- St George Hospital Clinical School, University of New South Wales, Sydney, Australia.
- Department of Surgery, St George Hospital, Sydney, Australia.
| | - Jordana Grazziela Alves Coelho-Dos-Reis
- Laboratório de Virologia Básica e Aplicada (LVBA), Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil.
- CT Terapias Avançadas e Inovadoras, (CT-Terapias), Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil.
| |
Collapse
|
4
|
Gattinger P, Kozlovskaya LI, Lunin AS, Gancharova OS, Sirazova DI, Apolokhov VD, Chekina ES, Gordeychuk IV, Karaulov AV, Valenta R, Ishmukhametov AA. Fusion protein-based COVID-19 vaccines exemplified by a chimeric vaccine based on a single fusion protein (W-PreS-O). Front Immunol 2025; 16:1452814. [PMID: 39935478 PMCID: PMC11811753 DOI: 10.3389/fimmu.2025.1452814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 01/03/2025] [Indexed: 02/13/2025] Open
Abstract
In this article we discuss characteristics of fusion protein-based SARS-CoV-2 vaccines. We focus on recombinant vaccine antigens comprising fusion proteins consisting of combinations of SARS-CoV-2-derived antigens or peptides or combinations of SARS-CoV-2 antigens/peptides with SARS-CoV-2-unrelated proteins/peptides. These fusion proteins are made to increase the immunogenicity of the vaccine antigens and/or to enable special targeting of the immune system. The protein-based vaccine approach is exemplified solely in a proof of concept study by using W-PreS-O, a chimeric vaccine based on a single fusion protein (W-PreS-O), combining RBDs from Wuhan hu-1 wild-type and Omicron BA.1 with the hepatitis B virus (HBV)-derived PreS surface antigen adsorbed to aluminum hydroxide. The W-PreS-O vaccine was evaluated in Syrian hamsters which were immunized three times at three-week intervals with W-PreS-O or with aluminum hydroxide (placebo) before they were infected with Omicron BA.1. Neutralizing antibody (nAB) titers, weight, lung symptoms, and viral loads, as measured using RT-PCR in the upper and lower respiratory tracts, were determined. In addition, infectious virus titers from the lungs were measured using a plaque-forming assay. We found that W-PreS-O-vaccinated hamsters developed robust nABs against Omicron BA.1, showed almost no development of pneumonia, and had significantly reduced infectious virus titers in the lungs. Importantly, the viral loads in the nasal cavities of W-PreS-O-vaccinated hamsters were close to or above the PCR cycle threshold considered to be non-infectious. The data of our proof-of-concept study provides compelling evidence that the W-PreS-O vaccine has protective effect against Omicron BA.1 in a Syrian hamster in vivo infection model and thus support the promising results obtained also for other fusion protein-based SARS-CoV-2 vaccines.
Collapse
Affiliation(s)
- Pia Gattinger
- Center for Pathophysiology, Infectiology and Immunology, Department of Pathophysiology and Allergy Research, Division of Immunopathology, Medical University of Vienna, Vienna, Austria
| | - Luibov I. Kozlovskaya
- Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences (Institute of Poliomyelitis), Moscow, Russia
| | - Alexander S. Lunin
- Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences (Institute of Poliomyelitis), Moscow, Russia
| | - Olga S. Gancharova
- Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences (Institute of Poliomyelitis), Moscow, Russia
| | - Dina I. Sirazova
- Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences (Institute of Poliomyelitis), Moscow, Russia
- Institute for Translational Medicine and Biotechnology, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Vasiliy D. Apolokhov
- Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences (Institute of Poliomyelitis), Moscow, Russia
| | - Egor S. Chekina
- Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences (Institute of Poliomyelitis), Moscow, Russia
| | - Ilya V. Gordeychuk
- Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences (Institute of Poliomyelitis), Moscow, Russia
- Institute for Translational Medicine and Biotechnology, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Alexander V. Karaulov
- Laboratory for Immunopathology, Department of Clinical Immunology and Allergology, Sechenov First Moscow State Medical University, Moscow, Russia
- Life Improvement by Future Technologies (LIFT) Center, Moscow, Russia
| | - Rudolf Valenta
- Center for Pathophysiology, Infectiology and Immunology, Department of Pathophysiology and Allergy Research, Division of Immunopathology, Medical University of Vienna, Vienna, Austria
- Laboratory for Immunopathology, Department of Clinical Immunology and Allergology, Sechenov First Moscow State Medical University, Moscow, Russia
- Life Improvement by Future Technologies (LIFT) Center, Moscow, Russia
- Center for Molecular Allergology, Karl Landsteiner University of Health Sciences, Krems, Austria
| | - Aydar A. Ishmukhametov
- Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences (Institute of Poliomyelitis), Moscow, Russia
- Institute for Translational Medicine and Biotechnology, Sechenov First Moscow State Medical University, Moscow, Russia
| |
Collapse
|
5
|
Mendoza-Ramírez NJ, García-Cordero J, Hernández-Galicia G, Moreno-Licona NJ, Hernandez J, Cabello-Gutierrez C, Zúñiga-Ramos JA, Morales-Rios E, Pérez-Tapia SM, Ortiz-Navarrete V, Espinosa-Cantellano M, Fernández-Benavides DA, Cedillo-Barrón L. Vaccination with Plasmids Encoding the Fusion Proteins D-S1, D-S1N and O-SN from SARS-CoV-2 Induces an Effective Humoral and Cellular Immune Response in Mice. Vaccines (Basel) 2025; 13:134. [PMID: 40006682 PMCID: PMC11860763 DOI: 10.3390/vaccines13020134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 01/08/2025] [Accepted: 01/22/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Next-generation vaccines against coronavirus disease 2019 (COVID-19) focus on inducing a long-lasting immune response against severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) and its emerging variants. To achieve this, antigens other than spike proteins have been proposed, and different platforms have been evaluated. Nucleic acid-based vaccines are fundamental for this process. Preclinical data have shown that the SARS-CoV-2 nucleocapsid protein induces a protective cellular immune response, and when combined with the spike protein, the resulting humoral and cellular immune responses are effective against some SARS-CoV-2 variants. METHODS We designed a DNA vaccine against the spike and nucleocapsid proteins of SARS-CoV-2 to generate fusion proteins based on the Delta and Omicron B.5 strains. The most immunogenic regions of the spike and nucleocapsid proteins of the Delta and Omicron B strains were selected using bioinformatics. The nucleotide sequences were cloned into pcDNA3.1, and named pcDNA3.1/D-S1, pcDNA3.1/D-S1N, and pcDNA3.1/O-SN. The immunogenicity of the generated fusion proteins was evaluated by analyzing the humoral and cellular responses elicited after the immunization of BALB/c mice. RESULTS DNA immunization induced antibody production, neutralization activity, and IFN-γ production. The inclusion of the nucleocapsid regions in the plasmid greatly enhanced the immune response. Moreover, cross-reactions with the variants of interest were confirmed. CONCLUSIONS Plasmids-encoding fusion proteins combining the most immunogenic regions of the spike and nucleocapsid proteins present a promising strategy for designing new and effective vaccines against SARS-CoV-2.
Collapse
Affiliation(s)
- Noe Juvenal Mendoza-Ramírez
- Departamento de Biomedicina Molecular CINVESTAV, Av. IPN # 2508 Col, San Pedro Zacatenco 07360, Mexico; (N.J.M.-R.); (J.G.-C.); (G.H.-G.)
| | - Julio García-Cordero
- Departamento de Biomedicina Molecular CINVESTAV, Av. IPN # 2508 Col, San Pedro Zacatenco 07360, Mexico; (N.J.M.-R.); (J.G.-C.); (G.H.-G.)
| | - Gabriela Hernández-Galicia
- Departamento de Biomedicina Molecular CINVESTAV, Av. IPN # 2508 Col, San Pedro Zacatenco 07360, Mexico; (N.J.M.-R.); (J.G.-C.); (G.H.-G.)
| | - Nicole Justine Moreno-Licona
- Departamento de Bioquímica Cinvestav, Av. IPN # 2508 Col, San Pedro Zacatenco 07360, Mexico; (N.J.M.-L.); (E.M.-R.)
| | - Jesus Hernandez
- Laboratorio de Inmunología, Centro de Investigación en Alimentación y Desarrollo A. C (CIAD) Carretera a la Victoria km 0.6, Hermosillo Sonora 83304, Mexico
| | - Carlos Cabello-Gutierrez
- Departamento de Investigación en Virología y Micología, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas (INER), Calzada de Tlalpan 4502, Belisario Domínguez, Tlalpan 14080, Mexico; (C.C.-G.); (J.A.Z.-R.)
| | - Joaquín Alejandro Zúñiga-Ramos
- Departamento de Investigación en Virología y Micología, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas (INER), Calzada de Tlalpan 4502, Belisario Domínguez, Tlalpan 14080, Mexico; (C.C.-G.); (J.A.Z.-R.)
- Escuela de Medicina y Ciencias de la Salud, Tecnológico de Monterrey, Monterrey 64710, Mexico
| | - Edgar Morales-Rios
- Departamento de Bioquímica Cinvestav, Av. IPN # 2508 Col, San Pedro Zacatenco 07360, Mexico; (N.J.M.-L.); (E.M.-R.)
| | - Sonia Mayra Pérez-Tapia
- Unidad de Desarrollo e Investigación en Bioterapéuticos (UDIBI), Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, México City 11340, Mexico;
| | - Vianney Ortiz-Navarrete
- Departamento de Biomedicina Molecular CINVESTAV, Av. IPN # 2508 Col, San Pedro Zacatenco 07360, Mexico; (N.J.M.-R.); (J.G.-C.); (G.H.-G.)
| | - Martha Espinosa-Cantellano
- Departamento de Infectómica y Patogénesis Molecular, CINVESTAV, Av. IPN # 2508 Col, San Pedro Zacatenco 07360, Mexico;
| | - David Andrés Fernández-Benavides
- Centro de Ingeniería y Desarrollo Industrial (CIDESI), Av. Playa Pie de la Cuesta No. 702, Desarrollo San Pablo, Querétaro 76125, Mexico;
| | - Leticia Cedillo-Barrón
- Departamento de Biomedicina Molecular CINVESTAV, Av. IPN # 2508 Col, San Pedro Zacatenco 07360, Mexico; (N.J.M.-R.); (J.G.-C.); (G.H.-G.)
| |
Collapse
|
6
|
Mambelli F, de Araujo ACVSC, Farias JP, de Andrade KQ, Ferreira LCS, Minoprio P, Leite LCC, Oliveira SC. An Update on Anti-COVID-19 Vaccines and the Challenges to Protect Against New SARS-CoV-2 Variants. Pathogens 2025; 14:23. [PMID: 39860984 PMCID: PMC11768231 DOI: 10.3390/pathogens14010023] [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: 10/04/2024] [Revised: 12/11/2024] [Accepted: 12/20/2024] [Indexed: 01/27/2025] Open
Abstract
The COVID-19 pandemic has posed a significant threat to global health systems, with extensive impacts across many sectors of society. The pandemic has been responsible for millions of deaths worldwide since its first identification in late 2019. Several actions have been taken to prevent the disease, including the unprecedented fast development and global vaccination campaigns, which were pivotal in reducing symptoms and deaths. Given the impact of the pandemic, the continuous changes of the virus, and present vaccine technologies, this review analyzes how, so far, we have met the challenge posed by the emergence of new variants and discusses how next-generation pan-coronavirus vaccines, with enhanced longevity and breadth of immune responses, may be tackled with alternative administration routes and antigen delivery platforms. By addressing these critical aspects, this review aims to contribute to the ongoing efforts to achieve long-term control of COVID-19, stimulating the discussion and work on next-generation vaccines capable of facing future waves of infection.
Collapse
Affiliation(s)
- Fábio Mambelli
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo 05508-000, Brazil; (F.M.); (A.C.V.S.C.d.A.); (K.Q.d.A.)
- Institut Pasteur de São Paulo, São Paulo 05508-020, Brazil; (L.C.S.F.); (P.M.)
| | - Ana Carolina V. S. C. de Araujo
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo 05508-000, Brazil; (F.M.); (A.C.V.S.C.d.A.); (K.Q.d.A.)
| | - Jéssica P. Farias
- Departamento de Microbiologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo 05508-000, Brazil;
| | - Kivia Q. de Andrade
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo 05508-000, Brazil; (F.M.); (A.C.V.S.C.d.A.); (K.Q.d.A.)
| | - Luis C. S. Ferreira
- Institut Pasteur de São Paulo, São Paulo 05508-020, Brazil; (L.C.S.F.); (P.M.)
- Departamento de Microbiologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo 05508-000, Brazil;
| | - Paola Minoprio
- Institut Pasteur de São Paulo, São Paulo 05508-020, Brazil; (L.C.S.F.); (P.M.)
| | - Luciana C. C. Leite
- Laboratório de Desenvolvimento de Vacinas, Instituto Butantan, São Paulo 05503-900, Brazil;
| | - Sergio C. Oliveira
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo 05508-000, Brazil; (F.M.); (A.C.V.S.C.d.A.); (K.Q.d.A.)
- Institut Pasteur de São Paulo, São Paulo 05508-020, Brazil; (L.C.S.F.); (P.M.)
| |
Collapse
|
7
|
Pinto PBA, Timis J, Chuensirikulchai K, Li QH, Lu HH, Maule E, Nguyen M, Alves RPDS, Verma SK, Ana-Sosa-Batiz F, Valentine K, Landeras-Bueno S, Kim K, Hastie K, Saphire EO, Alves A, Elong Ngono A, Shresta S. Co-immunization with spike and nucleocapsid based DNA vaccines for long-term protective immunity against SARS-CoV-2 Omicron. NPJ Vaccines 2024; 9:252. [PMID: 39702529 PMCID: PMC11659323 DOI: 10.1038/s41541-024-01043-3] [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: 03/06/2024] [Accepted: 12/01/2024] [Indexed: 12/21/2024] Open
Abstract
The continuing evolution of SARS-CoV-2 variants challenges the durability of existing spike (S)-based COVID-19 vaccines. We hypothesized that vaccines composed of both S and nucleocapsid (N) antigens would increase the durability of protection by strengthening and broadening cellular immunity compared with S-based vaccines. To test this, we examined the immunogenicity and efficacy of wild-type SARS-CoV-2 S- and N-based DNA vaccines administered individually or together to K18-hACE2 mice. S, N, and S + N vaccines all elicited polyfunctional CD4+ and CD8+ T cell responses and provided short-term cross-protection against Beta and Omicron BA.2 variants, but only co-immunization with S + N vaccines provided long-term protection against Omicron BA.2. Depletion of CD4+ and CD8+ T cells reduced the long-term efficacy, demonstrating a crucial role for T cells in the durability of protection. These findings underscore the potential to enhance long-lived protection against SARS-CoV-2 variants by combining S and N antigens in next-generation COVID-19 vaccines.
Collapse
Affiliation(s)
- Paolla Beatriz Almeida Pinto
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, 92037, USA
- Laboratory of Biotechnology and Physiology of Viral Infections, Oswaldo Cruz Institute, Fiocruz, Rio de Janeiro, 21040-900, Brazil
| | - Julia Timis
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, 92037, USA
| | - Kantinan Chuensirikulchai
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, 92037, USA
- Department of Microbiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Qin Hui Li
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, 92037, USA
| | - Hsueh Han Lu
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, 92037, USA
| | - Erin Maule
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, 92037, USA
| | - Michael Nguyen
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, 92037, USA
| | | | | | | | - Kristen Valentine
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, 92037, USA
| | - Sara Landeras-Bueno
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, 92037, USA
- University Cardenal Herrera-CEU, CEU Universities, Valencia, 46113, Spain
| | - Kenneth Kim
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, 92037, USA
- Microscopy and Histology Core Facility, La Jolla Institute for Immunology, La Jolla, 92037, USA
| | - Kathryn Hastie
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, 92037, USA
| | - Erica Ollmann Saphire
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, 92037, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California San Diego, La Jolla, 92093, USA
| | - Ada Alves
- Laboratory of Biotechnology and Physiology of Viral Infections, Oswaldo Cruz Institute, Fiocruz, Rio de Janeiro, 21040-900, Brazil
| | - Annie Elong Ngono
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, 92037, USA.
| | - Sujan Shresta
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, 92037, USA.
- Department of Pediatrics, Division of Host-Microbe Systems and Therapeutics, University of California San Diego, La Jolla, 92093, USA.
| |
Collapse
|
8
|
Hojo-Souza NS, de Castro JT, Rivelli GG, Azevedo PO, Oliveira ER, Faustino LP, Salazar N, Bagno FF, Carvalho AF, Rattis B, Lourenço KL, Gomes IP, Assis BRD, Piccin M, Fonseca FG, Durigon E, Silva JS, de Souza RP, Goulart GAC, Santiago H, Fernandes APS, Teixeira SR, Gazzinelli RT. SpiN-Tec: A T cell-based recombinant vaccine that is safe, immunogenic, and shows high efficacy in experimental models challenged with SARS-CoV-2 variants of concern. Vaccine 2024; 42:126394. [PMID: 39368129 DOI: 10.1016/j.vaccine.2024.126394] [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: 12/12/2023] [Revised: 09/11/2024] [Accepted: 09/21/2024] [Indexed: 10/07/2024]
Abstract
The emergence of new SARS-CoV-2 variants of concern associated with waning immunity induced by natural infection or vaccines currently in use suggests that the COVID-19 pandemic will become endemic. Investing in new booster vaccines using different platforms is a promising way to enhance protection and keep the disease under control. Here, we evaluated the immunogenicity, efficacy, and safety of the SpiN-Tec vaccine, based on a chimeric recombinant protein (SpiN) adjuvanted with CTVad1 (MF59-based adjuvant), aiming at boosting immunity against variants of concern of SARS-CoV-2. Immunization of K18-hACE-2 transgenic mice and hamsters induced high antibody titers and cellular immune response to the SpiN protein as well as to its components, RBD and N proteins. Importantly in a heterologous prime/boost protocol with a COVID-19 vaccine approved for emergency use (ChAdOx1), SpiN-Tec enhanced the level of circulation neutralizing antibodies (nAb). In addition to protection against the Wuhan isolate, protection against the Delta and Omicron variants was also observed as shown by reduced viral load and lung pathology. Toxicity and safety tests performed in rats demonstrated that the SpiN-Tec vaccine was safe and, based on these results, the SpiN-Tec phase I/II clinical trial was approved.
Collapse
MESH Headings
- Animals
- COVID-19 Vaccines/immunology
- SARS-CoV-2/immunology
- COVID-19/prevention & control
- COVID-19/immunology
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- Antibodies, Neutralizing/blood
- Antibodies, Neutralizing/immunology
- Mice
- Vaccines, Synthetic/immunology
- Vaccines, Synthetic/administration & dosage
- Cricetinae
- Mice, Transgenic
- Spike Glycoprotein, Coronavirus/immunology
- Spike Glycoprotein, Coronavirus/genetics
- T-Lymphocytes/immunology
- Female
- Immunogenicity, Vaccine
- Humans
- Rats
- Disease Models, Animal
- Adjuvants, Vaccine
- Immunization, Secondary
- Vaccine Efficacy
Collapse
Affiliation(s)
- Natália S Hojo-Souza
- Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais, Brazil; Instituto René Rachou, Fundação Oswaldo Cruz-Minas, Brazil
| | - Júlia T de Castro
- Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais, Brazil; Instituto René Rachou, Fundação Oswaldo Cruz-Minas, Brazil; Plataforma Bi-Institucional de Pesquisa em Medicina Translacional, Fundação Oswaldo Cruz, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Brazil
| | - Graziella G Rivelli
- Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais, Brazil
| | - Patrick O Azevedo
- Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais, Brazil; Instituto René Rachou, Fundação Oswaldo Cruz-Minas, Brazil
| | | | - Lídia P Faustino
- Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais, Brazil; Instituto René Rachou, Fundação Oswaldo Cruz-Minas, Brazil
| | - Natália Salazar
- Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais, Brazil
| | - Flávia F Bagno
- Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais, Brazil
| | - Alex F Carvalho
- Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais, Brazil
| | - Bruna Rattis
- Plataforma Bi-Institucional de Pesquisa em Medicina Translacional, Fundação Oswaldo Cruz, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Brazil
| | - Karine L Lourenço
- Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais, Brazil
| | - Isabela P Gomes
- Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais, Brazil
| | - Bruna R D Assis
- Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais, Brazil; Faculdade de Farmácia, Universidade Federal de Minas Gerais, Brazil
| | - Mariela Piccin
- Plataforma Bi-Institucional de Pesquisa em Medicina Translacional, Fundação Oswaldo Cruz, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Brazil
| | - Flávio G Fonseca
- Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais, Brazil; Departamento de Microbiologia, Universidade Federal de Minas Gerais, Brazil
| | - Edison Durigon
- Instituto de Ciências Biológicas, Universidade de São Paulo, Brazil
| | - João S Silva
- Plataforma Bi-Institucional de Pesquisa em Medicina Translacional, Fundação Oswaldo Cruz, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Brazil
| | - Renan P de Souza
- Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais, Brazil; Departamento de Genética, Ecologia e Evolução, Universidade Federal de Minas Gerais, Brazil
| | - Gisele A C Goulart
- Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais, Brazil; Faculdade de Farmácia, Universidade Federal de Minas Gerais, Brazil
| | - Helton Santiago
- Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais, Brazil; Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Brazil
| | - Ana Paula S Fernandes
- Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais, Brazil; Faculdade de Farmácia, Universidade Federal de Minas Gerais, Brazil
| | - Santuza R Teixeira
- Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais, Brazil; Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Brazil
| | - Ricardo T Gazzinelli
- Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais, Brazil; Instituto René Rachou, Fundação Oswaldo Cruz-Minas, Brazil; Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Brazil.
| |
Collapse
|
9
|
Lee P, Kim J, Oh H, Kim CU, Jeong AY, Lee MS, Jang MS, Hong JJ, Park JE, Kim DJ. Coronavirus nucleocapsid-based vaccine provides partial protection against hetero-species coronavirus in murine models. Antiviral Res 2024; 231:105991. [PMID: 39181216 DOI: 10.1016/j.antiviral.2024.105991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 08/06/2024] [Accepted: 08/21/2024] [Indexed: 08/27/2024]
Abstract
Most coronavirus vaccines focus on the spike (S) antigen, but the frequent mutations in S raise concerns about the vaccine efficacy against new variants. Although additional antigens with conserved sequences are have been tested, the extent to which these vaccines can provide immunity against different coronavirus species remains unclear. In this study, we assessed the potential of nucleocapsid (N) as a coronavirus vaccine antigen. Immunization with MERS-CoV N induced robust immune responses, providing significant protection against MERS-CoV. Notably, MERS-CoV N elicited cross-reactive T cell responses to SARS-CoV-2 N and significantly reduced lung inflammation following a SARS-CoV-2 challenge in the transient hACE2 mouse model. However, in K18-hACE transgenic mice, the vaccine showed limited protection. Collectively, our findings suggest that coronavirus N can be an effective vaccine antigen against homologous viruses, but its efficacy may vary across different coronaviruses, highlighting the need for further research on pan-coronavirus vaccines using conserved antigens.
Collapse
Affiliation(s)
- Pureum Lee
- Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea; University of Science and Technology (UST), Daejeon, South Korea
| | - Jihee Kim
- Chungnam National University College of Veterinary Medicine, Daejeon, South Korea
| | - Hanseul Oh
- Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea; Chungbuk National University College of Veterinary Medicine, Cheongju, South Korea
| | - Chang-Ung Kim
- Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea
| | - Ahn Young Jeong
- Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea; Princeton University, Princeton, NJ, USA
| | - Moo-Seung Lee
- Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea; University of Science and Technology (UST), Daejeon, South Korea
| | | | - Jung Joo Hong
- Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea.
| | - Jung-Eun Park
- Chungnam National University College of Veterinary Medicine, Daejeon, South Korea.
| | - Doo-Jin Kim
- Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea; Chungbuk National University College of Medicine, Cheongju, South Korea; Biomedical Research Institute, Chungbuk National University Hospital, Cheongju, South Korea.
| |
Collapse
|
10
|
Xu H, Zhao X, Zhang P, Zhang Y, Zhou Q, Wu H, Fan B, Zhang S, Wu H. Absence of platelet overactivation and thrombosis formation among patients with coronary atherosclerosis disease after vaccination against SARS-CoV-2. Heliyon 2024; 10:e38336. [PMID: 39640769 PMCID: PMC11619953 DOI: 10.1016/j.heliyon.2024.e38336] [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: 04/22/2024] [Revised: 09/21/2024] [Accepted: 09/23/2024] [Indexed: 12/07/2024] Open
Abstract
Background Association of Coronavirus disease 2019 vaccines with thrombosis has raised concerns among patients with coronary atherosclerosis disease (CAD). Objectives After vaccination against SARS-CoV-2, to detect thrombosis formation in atherosclerosis ApoE-/- mice, and platelet activation, coagulation, the profile of prothrombotic antibodies, and the production of platelet factor 4 (PF4) antibodies in patients with CAD. Methods Atherosclerotic ApoE-/- mice were immunized with saline or inactivated SARS-CoV vaccines. We investigated FeCl3-induced thrombus formation in vivo, and thrombus formation under flow conditions ex vivo. Inpatients undergoing percutaneous coronary intervention (PCI) were consecutively enrolled and defined according to vaccination status. We evaluated coagulation by thrombelastograph (TEG), platelet activation makers by flow cytometry, PF4 antibody and antiphospholipid antibodies by ELISA, and SARS-CoV-2 neutralizing antibody. Results In atherosclerotic ApoE-/- mice, FeCl3-induced thrombus formation and thrombus formation under flow conditions were similar between saline-treated and inactivated SARS-CoV-2 vaccines-treated groups. A total of 182 patients undergoing PCI were included in the final analysis, of whom 92 had been vaccinated. Baseline characteristics were well balanced between unvaccinated and vaccinated groups. The expression of PAC-1 and P-selectin, the prevalence of positivity for PF4 antibodies and antiphospholipid antibodies were similar between these two groups. Conclusions Inactivated SARS-CoV-2 vaccines did not potentiate thrombosis formation in atherosclerotic mice. Inactivated SARS-CoV-2 vaccines did not enhance platelet activation, or trigger the production of PF4 and antiphospholipid antibodies in patients with CAD. In light of the observed thrombotic risks associated with adenovirus-based COVID-19 vaccines, inactivated vaccines may offer a potentially safer option for individuals with CAD.
Collapse
Affiliation(s)
- Huajie Xu
- Department of Infectious Disease, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xin Zhao
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, National Clinical Research Center for Interventional Medicine, China
| | - Peng Zhang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, National Clinical Research Center for Interventional Medicine, China
| | - Yunjie Zhang
- Department of Biostatistics, School of Public Health, Fudan University, Shanghai, China
| | - Qi Zhou
- Department of Clinical Medicine, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Huibin Wu
- Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Bing Fan
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, National Clinical Research Center for Interventional Medicine, China
| | - Si Zhang
- NHC Key Laboratory of Glycoconjugate Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Hongyi Wu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, National Clinical Research Center for Interventional Medicine, China
| |
Collapse
|
11
|
Valiate BVS, Castro JTD, Marçal TG, Andrade LAF, Oliveira LID, Maia GBF, Faustino LP, Hojo-Souza NS, Reis MAAD, Bagno FF, Salazar N, Teixeira SR, Almeida GG, Gazzinelli RT. Evaluation of an RBD-nucleocapsid fusion protein as a booster candidate for COVID-19 vaccine. iScience 2024; 27:110177. [PMID: 38993669 PMCID: PMC11238127 DOI: 10.1016/j.isci.2024.110177] [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: 08/17/2023] [Revised: 04/30/2024] [Accepted: 05/31/2024] [Indexed: 07/13/2024] Open
Abstract
Despite successful vaccines and updates, constant mutations of SARS-CoV-2 makes necessary the search for new vaccines. We generated a chimeric protein that comprises the receptor-binding domain from spike and the nucleocapsid antigens (SpiN) from SARS-CoV-2. Once SpiN elicits a protective immune response in rodents, here we show that convalescent and previously vaccinated individuals respond to SpiN. CD4+ and CD8+ T cells from these individuals produced greater amounts of IFN-γ when stimulated with SpiN, compared to SARS-CoV-2 antigens. Also, B cells from these individuals were able to secrete antibodies that recognize SpiN. When administered as a boost dose in mice previously immunized with CoronaVac, ChAdOx1-S or BNT162b2, SpiN was able to induce a greater or equivalent immune response to homologous prime/boost. Our data reveal the ability of SpiN to induce cellular and humoral responses in vaccinated human donors, rendering it a promising candidate.
Collapse
Affiliation(s)
- Bruno Vinicius Santos Valiate
- Fundação Oswaldo Cruz-Minas, Belo Horizonte 30.190-002, MG, Brazil
- Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais, Parque Tecnológico de Belo Horizonte, Belo Horizonte 31.310-260, MG, Brazil
| | - Julia Teixeira de Castro
- Fundação Oswaldo Cruz-Minas, Belo Horizonte 30.190-002, MG, Brazil
- Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais, Parque Tecnológico de Belo Horizonte, Belo Horizonte 31.310-260, MG, Brazil
| | | | - Luis Adan Flores Andrade
- Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais, Parque Tecnológico de Belo Horizonte, Belo Horizonte 31.310-260, MG, Brazil
| | - Livia Isabela de Oliveira
- Fundação Oswaldo Cruz-Minas, Belo Horizonte 30.190-002, MG, Brazil
- Fundação Hospitalar do Estado de Minas Gerais, Belo Horizonte 31.630-901, MG, Brazil
| | | | | | - Natalia S Hojo-Souza
- Fundação Oswaldo Cruz-Minas, Belo Horizonte 30.190-002, MG, Brazil
- Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais, Parque Tecnológico de Belo Horizonte, Belo Horizonte 31.310-260, MG, Brazil
| | | | - Flávia Fonseca Bagno
- Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais, Parque Tecnológico de Belo Horizonte, Belo Horizonte 31.310-260, MG, Brazil
| | - Natalia Salazar
- Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais, Parque Tecnológico de Belo Horizonte, Belo Horizonte 31.310-260, MG, Brazil
| | - Santuza R Teixeira
- Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais, Parque Tecnológico de Belo Horizonte, Belo Horizonte 31.310-260, MG, Brazil
| | - Gregório Guilherme Almeida
- Fundação Oswaldo Cruz-Minas, Belo Horizonte 30.190-002, MG, Brazil
- Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais, Parque Tecnológico de Belo Horizonte, Belo Horizonte 31.310-260, MG, Brazil
| | - Ricardo Tostes Gazzinelli
- Fundação Oswaldo Cruz-Minas, Belo Horizonte 30.190-002, MG, Brazil
- Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais, Parque Tecnológico de Belo Horizonte, Belo Horizonte 31.310-260, MG, Brazil
| |
Collapse
|
12
|
Mendoza-Ramírez NJ, García-Cordero J, Shrivastava G, Cedillo-Barrón L. The Key to Increase Immunogenicity of Next-Generation COVID-19 Vaccines Lies in the Inclusion of the SARS-CoV-2 Nucleocapsid Protein. J Immunol Res 2024; 2024:9313267. [PMID: 38939745 PMCID: PMC11208798 DOI: 10.1155/2024/9313267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 05/06/2024] [Accepted: 05/09/2024] [Indexed: 06/29/2024] Open
Abstract
Vaccination is one of the most effective prophylactic public health interventions for the prevention of infectious diseases such as coronavirus disease (COVID-19). Considering the ongoing need for new COVID-19 vaccines, it is crucial to modify our approach and incorporate more conserved regions of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) to effectively address emerging viral variants. The nucleocapsid protein is a structural protein of SARS-CoV-2 that is involved in replication and immune responses. Furthermore, this protein offers significant advantages owing to the minimal accumulation of mutations over time and the inclusion of key T-cell epitopes critical for SARS-CoV-2 immunity. A novel strategy that may be suitable for the new generation of vaccines against COVID-19 is to use a combination of antigens, including the spike and nucleocapsid proteins, to elicit robust humoral and potent cellular immune responses, along with long-lasting immunity. The strategic use of multiple antigens aims to enhance vaccine efficacy and broaden protection against viruses, including their variants. The immune response against the nucleocapsid protein from other coronavirus is long-lasting, and it can persist up to 11 years post-infection. Thus, the incorporation of nucleocapsids (N) into vaccine design adds an important dimension to vaccination efforts and holds promise for bolstering the ability to combat COVID-19 effectively. In this review, we summarize the preclinical studies that evaluated the use of the nucleocapsid protein as antigen. This study discusses the use of nucleocapsid alone and its combination with spike protein or other proteins of SARS-CoV-2.
Collapse
Affiliation(s)
- Noe Juvenal Mendoza-Ramírez
- Departamento de Biomedicina MolecularCINVESTAV IPN, Av. IPN # 2508 Col, San Pedro Zacatenco, Mexico City 07360, Mexico
| | - Julio García-Cordero
- Departamento de Biomedicina MolecularCINVESTAV IPN, Av. IPN # 2508 Col, San Pedro Zacatenco, Mexico City 07360, Mexico
| | - Gaurav Shrivastava
- Laboratory of Malaria and Vector ResearchNational Institute of Allergy and Infectious DiseasesNational Institutes of Health, Rockville, MD, USA
| | - Leticia Cedillo-Barrón
- Departamento de Biomedicina MolecularCINVESTAV IPN, Av. IPN # 2508 Col, San Pedro Zacatenco, Mexico City 07360, Mexico
| |
Collapse
|
13
|
Huang Y, Chen J, Chen S, Huang C, Li B, Li J, Jin Z, Zhang Q, Pan P, Du W, Liu L, Liu Z. Molecular characterization of SARS-CoV-2 nucleocapsid protein. Front Cell Infect Microbiol 2024; 14:1415885. [PMID: 38846351 PMCID: PMC11153676 DOI: 10.3389/fcimb.2024.1415885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 05/07/2024] [Indexed: 06/09/2024] Open
Abstract
Corona Virus Disease 2019 (COVID-19) is a highly prevalent and potent infectious disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Until now, the world is still endeavoring to develop new ways to diagnose and treat COVID-19. At present, the clinical prevention and treatment of COVID-19 mainly targets the spike protein on the surface of SRAS-CoV-2. However, with the continuous emergence of SARS-CoV-2 Variants of concern (VOC), targeting the spike protein therapy shows a high degree of limitation. The Nucleocapsid Protein (N protein) of SARS-CoV-2 is highly conserved in virus evolution and is involved in the key process of viral infection and assembly. It is the most expressed viral structural protein after SARS-CoV-2 infection in humans and has high immunogenicity. Therefore, N protein as the key factor of virus infection and replication in basic research and clinical application has great potential research value. This article reviews the research progress on the structure and biological function of SARS-CoV-2 N protein, the diagnosis and drug research of targeting N protein, in order to promote researchers' further understanding of SARS-CoV-2 N protein, and lay a theoretical foundation for the possible outbreak of new and sudden coronavirus infectious diseases in the future.
Collapse
Affiliation(s)
- Yanping Huang
- Department of Infectious Diseases, Renmin Hospital, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China
- Shiyan Key Laboratory of Virology, Hubei University of Medicine, Shiyan, China
| | - Junkai Chen
- Department of Infectious Diseases, Renmin Hospital, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China
- Shiyan Key Laboratory of Virology, Hubei University of Medicine, Shiyan, China
| | - Siwei Chen
- Department of Infectious Diseases, Renmin Hospital, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China
- Shiyan Key Laboratory of Virology, Hubei University of Medicine, Shiyan, China
| | - Congcong Huang
- Department of Infectious Diseases, Renmin Hospital, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China
- Shiyan Key Laboratory of Virology, Hubei University of Medicine, Shiyan, China
| | - Bei Li
- Department of Infectious Diseases, Renmin Hospital, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China
- Shiyan Key Laboratory of Virology, Hubei University of Medicine, Shiyan, China
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, China
| | - Jian Li
- Department of Infectious Diseases, Renmin Hospital, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China
- Shiyan Key Laboratory of Virology, Hubei University of Medicine, Shiyan, China
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, China
| | - Zhixiong Jin
- Department of Infectious Diseases, Renmin Hospital, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China
- Shiyan Key Laboratory of Virology, Hubei University of Medicine, Shiyan, China
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, China
| | - Qiwei Zhang
- Central Laboratory, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Pan Pan
- Guangdong Provincial Key Laboratory of Virology, Institute of Medical Microbiology, Jinan University, Guangzhou, China
| | - Weixing Du
- Department of Infectious Diseases, Renmin Hospital, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China
| | - Long Liu
- Department of Infectious Diseases, Renmin Hospital, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China
- Shiyan Key Laboratory of Virology, Hubei University of Medicine, Shiyan, China
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, China
| | - Zhixin Liu
- Department of Infectious Diseases, Renmin Hospital, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China
- Shiyan Key Laboratory of Virology, Hubei University of Medicine, Shiyan, China
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, China
| |
Collapse
|
14
|
Liu C, Xue RY, Li GC, Zhang Y, Wu WY, Liu JY, Feng R, Jin Z, Deng Y, Jin ZL, Cheng H, Mao L, Zou QM, Li HB. pGM-CSF as an adjuvant in DNA vaccination against SARS-CoV-2. Int J Biol Macromol 2024; 264:130660. [PMID: 38460634 DOI: 10.1016/j.ijbiomac.2024.130660] [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: 01/16/2024] [Revised: 02/19/2024] [Accepted: 03/04/2024] [Indexed: 03/11/2024]
Abstract
The emergence of SARS-CoV-2 presents a significant global public health dilemma. Vaccination has long been recognized as the most effective means of preventing the spread of infectious diseases. DNA vaccines have attracted attention due to their safety profile, cost-effectiveness, and ease of production. This study aims to assess the efficacy of plasmid-encoding GM-CSF (pGM-CSF) as an adjuvant to augment the specific humoral and cellular immune response elicited by DNA vaccines based on the receptor-binding domain (RBD) antigen. Compared to the use of plasmid-encoded RBD (pRBD) alone, mice that were immunized with a combination of pRBD and pGM-CSF exhibited significantly elevated levels of RBD-specific antibody titers in serum, BALF, and nasal wash. Furthermore, these mice generated more potent neutralization antibodies against both the wild-type and Omicron pseudovirus, as well as the ancestral virus. In addition, pGM-CSF enhanced pRBD-induced CD4+ and CD8+ T cell responses and promoted central memory T cells storage in the spleen. At the same time, tissue-resident memory T (Trm) cells in the lung also increased significantly, and higher levels of specific responses were maintained 60 days post the final immunization. pGM-CSF may play an adjuvant role by promoting antigen expression, immune cells recruitment and GC B cell responses. In conclusion, pGM-CSF may be an effective adjuvant candidate for the DNA vaccines against SARS-CoV-2.
Collapse
Affiliation(s)
- Chang Liu
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, PR China; Department of Pharmacy, Chinese People's Liberation Army Unit 32265, Guangzhou 510310, PR China
| | - Ruo-Yi Xue
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, PR China
| | - Guo-Cheng Li
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, PR China
| | - Yi Zhang
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, PR China
| | - Wei-Yi Wu
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, PR China
| | - Jing-Yi Liu
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, PR China
| | - Rang Feng
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, PR China
| | - Zhe Jin
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, PR China
| | - Yan Deng
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, PR China
| | - Zi-Li Jin
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, PR China
| | - Hao Cheng
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, PR China
| | - Ling Mao
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, PR China
| | - Quan-Ming Zou
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, PR China.
| | - Hai-Bo Li
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, PR China.
| |
Collapse
|
15
|
Yu H, Guan F, Miller H, Lei J, Liu C. The role of SARS-CoV-2 nucleocapsid protein in antiviral immunity and vaccine development. Emerg Microbes Infect 2023; 12:e2164219. [PMID: 36583642 PMCID: PMC9980416 DOI: 10.1080/22221751.2022.2164219] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
ABSTRACTThe coronavirus disease 2019 (COVID-19) has caused enormous health risks and global economic disruption. This disease is caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The SARS-CoV-2 nucleocapsid protein is a structural protein involved in viral replication and assembly. There is accumulating evidence indicating that the nucleocapsid protein is multi-functional, playing a key role in the pathogenesis of COVID-19 and antiviral immunity against SARS-CoV-2. Here, we summarize its potential application in the prevention of COVID-19, which is based on its role in inflammation, cell death, antiviral innate immunity, and antiviral adaptive immunity.
Collapse
Affiliation(s)
- Haiyun Yu
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Fei Guan
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Heather Miller
- Laboratory of Intracellular Parasites, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Jiahui Lei
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Chaohong Liu
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China, Chaohong Liu
| |
Collapse
|
16
|
Russo M, Mendes-Corrêa MC, Lins BB, Kersten V, Pernambuco Filho PCA, Martins TR, Tozetto-Mendoza TR, Vilas Boas LS, Gomes BM, Dati LMM, Duarte-Neto AN, Reigado GR, Frederico ABT, de Brito e Cunha DRDA, de Paula AV, da Silva JIG, Vasconcelos CFM, Chambergo FS, Nunes VA, Ano Bom APD, Castilho LR, Martins RAP, Hirata MH, Mirotti L. Intranasal Liposomal Formulation of Spike Protein Adjuvanted with CpG Protects and Boosts Heterologous Immunity of hACE2 Transgenic Mice to SARS-CoV-2 Infection. Vaccines (Basel) 2023; 11:1732. [PMID: 38006064 PMCID: PMC10675295 DOI: 10.3390/vaccines11111732] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 11/05/2023] [Accepted: 11/08/2023] [Indexed: 11/26/2023] Open
Abstract
Mucosal vaccination appears to be suitable to protect against SARS-CoV-2 infection. In this study, we tested an intranasal mucosal vaccine candidate for COVID-19 that consisted of a cationic liposome containing a trimeric SARS-CoV-2 spike protein and CpG-ODNs, a Toll-like receptor 9 agonist, as an adjuvant. In vitro and in vivo experiments indicated the absence of toxicity following the intranasal administration of this vaccine formulation. First, we found that subcutaneous or intranasal vaccination protected hACE-2 transgenic mice from infection with the wild-type (Wuhan) SARS-CoV-2 strain, as shown by weight loss and mortality indicators. However, when compared with subcutaneous administration, the intranasal route was more effective in the pulmonary clearance of the virus and induced higher neutralizing antibodies and anti-S IgA titers. In addition, the intranasal vaccination afforded protection against gamma, delta, and omicron virus variants of concern. Furthermore, the intranasal vaccine formulation was superior to intramuscular vaccination with a recombinant, replication-deficient chimpanzee adenovirus vector encoding the SARS-CoV-2 spike glycoprotein (Oxford/AstraZeneca) in terms of virus lung clearance and production of neutralizing antibodies in serum and bronchial alveolar lavage (BAL). Finally, the intranasal liposomal formulation boosted heterologous immunity induced by previous intramuscular vaccination with the Oxford/AstraZeneca vaccine, which was more robust than homologous immunity.
Collapse
Affiliation(s)
- Momtchilo Russo
- Department of Immunology, Institute of Biomedical Science, University of São Paulo (ICB-USP), São Paulo 05508-000, Brazil
| | - Maria Cássia Mendes-Corrêa
- Laboratório de Virologia (LIM52), Instituto de Medicina Tropical de São Paulo, Faculdade de Medicina da Universidade de São Paulo (FM-USP), São Paulo 05403-000, Brazil; (M.C.M.-C.); (T.R.M.)
| | - Bruna B. Lins
- Department of Immunology, Institute of Biomedical Science, University of São Paulo (ICB-USP), São Paulo 05508-000, Brazil
| | - Victor Kersten
- Department of Immunology, Institute of Biomedical Science, University of São Paulo (ICB-USP), São Paulo 05508-000, Brazil
| | - Paulo C. A. Pernambuco Filho
- Department of Immunology, Institute of Biomedical Science, University of São Paulo (ICB-USP), São Paulo 05508-000, Brazil
| | - Toni Ricardo Martins
- Laboratório de Virologia (LIM52), Instituto de Medicina Tropical de São Paulo, Faculdade de Medicina da Universidade de São Paulo (FM-USP), São Paulo 05403-000, Brazil; (M.C.M.-C.); (T.R.M.)
- Faculdade de Ciências Farmacêuticas, Universidade Federal do Amazonas (UFAM), Manaus 69080-900, Brazil
| | - Tânia Regina Tozetto-Mendoza
- Laboratório de Virologia (LIM52), Instituto de Medicina Tropical de São Paulo, Faculdade de Medicina da Universidade de São Paulo (FM-USP), São Paulo 05403-000, Brazil; (M.C.M.-C.); (T.R.M.)
| | - Lucy Santos Vilas Boas
- Laboratório de Virologia (LIM52), Instituto de Medicina Tropical de São Paulo, Faculdade de Medicina da Universidade de São Paulo (FM-USP), São Paulo 05403-000, Brazil; (M.C.M.-C.); (T.R.M.)
| | - Brisa Moreira Gomes
- Department of Immunology, Institute of Biomedical Science, University of São Paulo (ICB-USP), São Paulo 05508-000, Brazil
| | - Livia Mendonça Munhoz Dati
- Departamento de Analises Clinicas e Toxicologicas, Faculdade de Ciências Farmacêuticas da Universidade de Sao Paulo (FCF-USP), São Paulo 05508-000, Brazil (M.H.H.)
| | - Amaro Nunes Duarte-Neto
- Departamento de Patologia, Faculdade de Medicina da Universidade de São Paulo (FM-USP), São Paulo 05403-000, Brazil
| | - Gustavo Roncoli Reigado
- Laboratório de Biotecnologia, Escola de Artes, Ciências e Humanidades, Universidade de São Paulo (EACH-USP), São Paulo 03828-000, Brazil (F.S.C.); (V.A.N.)
| | - Ana Beatriz T. Frederico
- Immunological Technology Laboratory, Institute of Immunobiological Technology (Bio-Manguinhos), Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro 21040-900, Brazil (A.P.D.A.B.)
| | - Danielle R. de A. de Brito e Cunha
- Immunological Technology Laboratory, Institute of Immunobiological Technology (Bio-Manguinhos), Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro 21040-900, Brazil (A.P.D.A.B.)
| | - Anderson Vicente de Paula
- Laboratório de Virologia (LIM52), Instituto de Medicina Tropical de São Paulo, Faculdade de Medicina da Universidade de São Paulo (FM-USP), São Paulo 05403-000, Brazil; (M.C.M.-C.); (T.R.M.)
| | - José Igor G. da Silva
- Programa de Biologia Celular e do Desenvolvimento, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, Brazil (R.A.P.M.)
| | - Carlos F. Moreira Vasconcelos
- Programa de Biologia Celular e do Desenvolvimento, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, Brazil (R.A.P.M.)
| | - Felipe S. Chambergo
- Laboratório de Biotecnologia, Escola de Artes, Ciências e Humanidades, Universidade de São Paulo (EACH-USP), São Paulo 03828-000, Brazil (F.S.C.); (V.A.N.)
| | - Viviane Abreu Nunes
- Laboratório de Biotecnologia, Escola de Artes, Ciências e Humanidades, Universidade de São Paulo (EACH-USP), São Paulo 03828-000, Brazil (F.S.C.); (V.A.N.)
| | - Ana Paula Dinis Ano Bom
- Immunological Technology Laboratory, Institute of Immunobiological Technology (Bio-Manguinhos), Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro 21040-900, Brazil (A.P.D.A.B.)
| | - Leda R. Castilho
- Cell Culture Engineering Laboratory, COPPE, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-598, Brazil;
| | - Rodrigo A. P. Martins
- Programa de Biologia Celular e do Desenvolvimento, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, Brazil (R.A.P.M.)
| | - Mario Hiroyuki Hirata
- Departamento de Analises Clinicas e Toxicologicas, Faculdade de Ciências Farmacêuticas da Universidade de Sao Paulo (FCF-USP), São Paulo 05508-000, Brazil (M.H.H.)
| | - Luciana Mirotti
- Institute of Science and Technology in Biomodels (ICTB), Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro 21040-900, Brazil
| |
Collapse
|
17
|
Cao J, Gu H, Zhang X, Yun H, Li J, Si CY, Zhang J, Wang H. Intranasal inoculation of female BALB/c mice with replication-deficient human adenovirus type 5 expressing SARS-CoV-2 nucleocapsid protein aggravates lung pathology upon re-encountering the antigen. Virus Res 2023; 335:199201. [PMID: 37595663 PMCID: PMC10470087 DOI: 10.1016/j.virusres.2023.199201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 07/27/2023] [Accepted: 08/15/2023] [Indexed: 08/20/2023]
Abstract
Preclinical studies indicate that SARS-CoV-2 nucleocapsid (N)-based vaccines, along with other viral protein(s), confer protection in various animal models against infection by SARS-CoV-2 ancestral virus and variants of concern. However, the optimal vaccination procedure and the role of N-specific host adaptive immune responses remain elusive. Here, we report that intranasal inoculation with replication-deficient human adenovirus type 5 expressing SARS-CoV-2 N protein (Ad5-N) conferred no protection in the lung of female BALB/c mice upon re-encountering the antigen, either by 10-fold Ad5-N re-exposure or sublethal infection of mouse-adapted SARS-CoV-2. By contrast, this procedure led to aggravated lung pathology with more necroptotic CD3+ T cells and Ly6G+ granulocytes, which was associated with the accumulation of IFN-γ-expressing antigen-experienced CD4+ and CD8+ T cells. These findings pre-caution the clinical application of this vaccination procedure. Furthermore, our data suggest that excessive host adaptive immune responses against N protein contributes to COVID-19 pathogenesis.
Collapse
Affiliation(s)
- Junxia Cao
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Hongjing Gu
- Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Xueting Zhang
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Hongfang Yun
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Jiarong Li
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China; University of South China, Hengyang Medical School, Hengyang 421001, China
| | - Chuan-Yimu Si
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Jiyan Zhang
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China; University of South China, Hengyang Medical School, Hengyang 421001, China; Anhui Medical University, Hefei 230032, China; Chinese Institute for Brain Research, Beijing 102206, China.
| | - Hui Wang
- Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China.
| |
Collapse
|
18
|
Song W, Fang Z, Ma F, Li J, Huang Z, Zhang Y, Li J, Chen K. The role of SARS-CoV-2 N protein in diagnosis and vaccination in the context of emerging variants: present status and prospects. Front Microbiol 2023; 14:1217567. [PMID: 37675423 PMCID: PMC10478715 DOI: 10.3389/fmicb.2023.1217567] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 07/31/2023] [Indexed: 09/08/2023] Open
Abstract
Despite many countries rapidly revising their strategies to prevent contagions, the number of people infected with Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) continues to surge. The emergent variants that can evade the immune response significantly affect the effectiveness of mainstream vaccines and diagnostic products based on the original spike protein. Therefore, it is essential to focus on the highly conserved nature of the nucleocapsid protein as a potential target in the field of vaccines and diagnostics. In this regard, our review initially discusses the structure, function, and mechanism of action of N protein. Based on this discussion, we summarize the relevant research on the in-depth development and application of diagnostic methods and vaccines based on N protein, such as serology and nucleic acid detection. Such valuable information can aid in designing more efficient diagnostic and vaccine tools that could help end the SARS-CoV-2 pandemic.
Collapse
Affiliation(s)
- Wanchen Song
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhongbiao Fang
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Feike Ma
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Jiaxuan Li
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Zhiwei Huang
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Yanjun Zhang
- Key Laboratory of Public Health Detection and Etiological Research of Zhejiang Province, Department of Microbiology, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, China
| | - Jianhua Li
- Key Laboratory of Public Health Detection and Etiological Research of Zhejiang Province, Department of Microbiology, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, China
| | - Keda Chen
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| |
Collapse
|
19
|
Dias Assis BR, Gomes IP, de Castro JT, Rivelli GG, de Castro NS, Gomez-Mendoza DP, Bagno FF, Hojo-Souza NS, Chaves Maia AL, Lages EB, da Fonseca FG, Ribeiro Teixeira SM, Fernandes AP, Gazzinelli RT, Castro Goulart GA. Quality attributes of CTVad1, a nanoemulsified adjuvant for phase I clinical trial of SpiN COVID-19 vaccine. Nanomedicine (Lond) 2023; 18:1175-1194. [PMID: 37712604 DOI: 10.2217/nnm-2023-0122] [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: 09/16/2023] Open
Abstract
Aim: To develop, characterize and evaluate an oil/water nanoemulsion with squalene (CTVad1) to be approved as an adjuvant for the SpiN COVID-19 vaccine clinical trials. Materials & methods: Critical process parameters (CPPs) of CTVad1 were standardized to meet the critical quality attributes (CQAs) of an adjuvant for human use. CTVad1 and the SpiN-CTVad1 vaccine were submitted to physicochemical, stability, in vitro and in vivo studies. Results & conclusion: All CQAs were met in the CTVad1 production process. SpiN- CTVad1 met CQAs and induced high levels of antibodies and specific cellular responses in in vivo studies. These results represented a critical step in the process developed to meet regulatory requirements for the SpiN COVID-19 vaccine clinical trial.
Collapse
Affiliation(s)
- Bruna Rodrigues Dias Assis
- Department of Pharmaceuticals, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, MG, 31270-901, Brazil
- Centro de Tecnologia de Vacinas da Universidade Federal de Minas Gerais, Belo Horizonte, Belo Horizonte, MG, 31310-260, Brazil
| | - Isabela Pereira Gomes
- Centro de Tecnologia de Vacinas da Universidade Federal de Minas Gerais, Belo Horizonte, Belo Horizonte, MG, 31310-260, Brazil
| | - Júlia Teixeira de Castro
- Centro de Tecnologia de Vacinas da Universidade Federal de Minas Gerais, Belo Horizonte, Belo Horizonte, MG, 31310-260, Brazil
| | - Graziella Gomes Rivelli
- Centro de Tecnologia de Vacinas da Universidade Federal de Minas Gerais, Belo Horizonte, Belo Horizonte, MG, 31310-260, Brazil
| | - Natália Salazar de Castro
- Centro de Tecnologia de Vacinas da Universidade Federal de Minas Gerais, Belo Horizonte, Belo Horizonte, MG, 31310-260, Brazil
| | - Diana Paola Gomez-Mendoza
- Centro de Tecnologia de Vacinas da Universidade Federal de Minas Gerais, Belo Horizonte, Belo Horizonte, MG, 31310-260, Brazil
| | - Flávia Fonseca Bagno
- Centro de Tecnologia de Vacinas da Universidade Federal de Minas Gerais, Belo Horizonte, Belo Horizonte, MG, 31310-260, Brazil
| | - Natália Satchiko Hojo-Souza
- Centro de Tecnologia de Vacinas da Universidade Federal de Minas Gerais, Belo Horizonte, Belo Horizonte, MG, 31310-260, Brazil
- Instituto René Rachou, Fundação Oswaldo Cruz-Minas, Belo Horizonte, MG, 30190-002, Brazil
| | - Ana Luiza Chaves Maia
- Centro de Tecnologia de Vacinas da Universidade Federal de Minas Gerais, Belo Horizonte, Belo Horizonte, MG, 31310-260, Brazil
| | - Eduardo Burgarelli Lages
- Centro de Tecnologia de Vacinas da Universidade Federal de Minas Gerais, Belo Horizonte, Belo Horizonte, MG, 31310-260, Brazil
| | - Flávio Guimaraes da Fonseca
- Centro de Tecnologia de Vacinas da Universidade Federal de Minas Gerais, Belo Horizonte, Belo Horizonte, MG, 31310-260, Brazil
- Department of Microbiology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, 31270-901, Brazil
| | - Santuza Maria Ribeiro Teixeira
- Centro de Tecnologia de Vacinas da Universidade Federal de Minas Gerais, Belo Horizonte, Belo Horizonte, MG, 31310-260, Brazil
- Department of Biochemistry & Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, 31270-901, Brazil
| | - Ana Paula Fernandes
- Centro de Tecnologia de Vacinas da Universidade Federal de Minas Gerais, Belo Horizonte, Belo Horizonte, MG, 31310-260, Brazil
- Department of Clinical & Toxicological Analysis, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, MG, 31270-901, Brazil
| | - Ricardo Tostes Gazzinelli
- Centro de Tecnologia de Vacinas da Universidade Federal de Minas Gerais, Belo Horizonte, Belo Horizonte, MG, 31310-260, Brazil
- Instituto René Rachou, Fundação Oswaldo Cruz-Minas, Belo Horizonte, MG, 30190-002, Brazil
- Department of Biochemistry & Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, 31270-901, Brazil
| | - Gisele Assis Castro Goulart
- Department of Pharmaceuticals, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, MG, 31270-901, Brazil
- Centro de Tecnologia de Vacinas da Universidade Federal de Minas Gerais, Belo Horizonte, Belo Horizonte, MG, 31310-260, Brazil
| |
Collapse
|
20
|
Chan BCL, Li P, Tsang MSM, Sung JCC, Kwong KWY, Zheng T, Hon SSM, Lau CP, Cheng W, Chen F, Lau CBS, Leung PC, Wong CK. Creating a Vaccine-like Supplement against Respiratory Infection Using Recombinant Bacillus subtilis Spores Expressing SARS-CoV-2 Spike Protein with Natural Products. Molecules 2023; 28:4996. [PMID: 37446658 DOI: 10.3390/molecules28134996] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 06/19/2023] [Accepted: 06/24/2023] [Indexed: 07/15/2023] Open
Abstract
Vaccination is the most effective method of combating COVID-19 infection, but people with a psychological fear of needles and side effects are hesitant to receive the current vaccination, and alternative delivery methods may help. Bacillus subtilis, a harmless intestinal commensal, has recently earned a strong reputation as a vaccine production host and delivery vector, with advantages such as low cost, safety for human consumption, and straightforward oral administration. In this study, we have succeeded generating "S spores" by engineering B. subtilis with spore coat proteins resembling the spike (S) protein of the ancestral SARS-CoV-2 coronavirus. With the addition of two immunostimulating natural products as adjuvants, namely Astragalus membranaceus (Fisch.) Bge (AM) and Coriolus versicolor (CV), oral administration of S spores could elicit mild immune responses against COVID-19 infection without toxicity. Mucosal IgA against the S protein was enhanced by co-feeding with AM and CV in an S spores-inoculated mouse model. Faster and stronger IgG responses against the S protein were observed when the mice were fed with S spores prior to vaccination with the commercial COVID-19 vaccine CoronaVac. In vitro studies demonstrated that AM, CV, and B. subtilis spores could dose-dependently activate both macrophages and dendritic cells by secreting innate immunity-related IL-1β, IL-6, and TNF-α, and some other proinflammatory chemokines and cytokines. In conclusion, the combination of S spores with AM and CV may be helpful in developing a vaccine-like supplement against respiratory infection.
Collapse
Affiliation(s)
- Ben Chung-Lap Chan
- Institute of Chinese Medicine and State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Hong Kong, China
| | - Peiting Li
- Institute of Chinese Medicine and State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Hong Kong, China
| | - Miranda Sin-Man Tsang
- Institute of Chinese Medicine and State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Hong Kong, China
- China-Australia International Research Centre for Chinese Medicine, School of Health and Biomedical Sciences, STEM College, RMIT University, Bundoora, VIC 3083, Australia
| | | | | | - Tao Zheng
- Institute of Chinese Medicine and State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Hong Kong, China
| | - Sharon Sze-Man Hon
- Institute of Chinese Medicine and State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Hong Kong, China
- Department of Chemical Pathology, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, NT, Hong Kong, China
| | - Ching-Po Lau
- Institute of Chinese Medicine and State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Hong Kong, China
| | - Wen Cheng
- Institute of Chinese Medicine and State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Hong Kong, China
| | - Fang Chen
- Institute of Chinese Medicine and State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Hong Kong, China
| | - Clara Bik-San Lau
- Institute of Chinese Medicine and State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Hong Kong, China
| | - Ping-Chung Leung
- Institute of Chinese Medicine and State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Hong Kong, China
| | - Chun-Kwok Wong
- Institute of Chinese Medicine and State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Hong Kong, China
- Department of Chemical Pathology, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, NT, Hong Kong, China
- Li Dak Sum Yip Yio Chin R & D Centre for Chinese Medicine, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
21
|
Elder E, Bangalore Revanna C, Johansson C, Wallin RPA, Sjödahl J, Winqvist O, Mirazimi A. Protective immunity induced by an inhaled SARS-CoV-2 subunit vaccine. Vaccine 2023:S0264-410X(23)00684-9. [PMID: 37353452 PMCID: PMC10242152 DOI: 10.1016/j.vaccine.2023.06.015] [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: 10/07/2022] [Revised: 05/31/2023] [Accepted: 06/02/2023] [Indexed: 06/25/2023]
Abstract
Targeting the site of infection is a promising strategy for improving vaccine effectivity. To date, licensed COVID-19 vaccines have been administered intramuscularly despite the fact that SARS-CoV-2 is a respiratory virus. Here, we aim to induce local protective mucosal immune responses with an inhaled subunit vaccine candidate, ISR52, based on the SARS-CoV-2 Spike S1 protein. When tested in a lethal challenge hACE2 transgenic SARS-CoV-2 mouse model, intranasal and intratracheal administration of ISR52 provided superior protection against severe infection, compared to the subcutaneous injection of the vaccine. Interestingly for a protein-based vaccine, inhaled ISR52 elicited both CD4 and CD8 T-cell Spike-specific responses that were maintained for at least 6 months in wild-type mice. Induced IgG and IgA responses cross-reacting with several SARS- CoV-2 variants of concern were detected in the lung and in serum and protected animals displayed neutralizing antibodies. Based on our results, we are developing ISR52 as a dry powder formulation for inhalation, that does not require cold-chain distribution or the use of needle administration, for evaluation in a Phase I/II clinical trial.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Ali Mirazimi
- National Veterinary Institute, Uppsala, Sweden; Clinical Microbiology, LABMED, Karolinska Institute, Sweden.
| |
Collapse
|
22
|
Azevedo PO, Hojo-Souza NS, Faustino LP, Fumagalli MJ, Hirako IC, Oliveira ER, Figueiredo MM, Carvalho AF, Doro D, Benevides L, Durigon E, Fonseca F, Machado AM, Fernandes AP, Teixeira SR, Silva JS, Gazzinelli RT. Differential requirement of neutralizing antibodies and T cells on protective immunity to SARS-CoV-2 variants of concern. NPJ Vaccines 2023; 8:15. [PMID: 36781862 PMCID: PMC9923671 DOI: 10.1038/s41541-023-00616-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 01/27/2023] [Indexed: 02/15/2023] Open
Abstract
The current COVID-19 vaccines protect against severe disease, but are not effective in controlling replication of the Variants of Concern (VOCs). Here, we used the existing pre-clinical models of severe and moderate COVID-19 to evaluate the efficacy of a Spike-based DNA vaccine (pCTV-WS) for protection against different VOCs. Immunization of transgenic (K18-hACE2) mice and hamsters induced significant levels of neutralizing antibodies (nAbs) to Wuhan and Delta isolates, but not to the Gamma and Omicron variants. Nevertheless, the pCTV-WS vaccine offered significant protection to all VOCs. Consistently, protection against lung pathology and viral load to Wuhan or Delta was mediated by nAbs, whereas in the absence of nAbs, T cells controlled viral replication, disease and lethality in mice infected with either the Gamma or Omicron variants. Hence, considering the conserved nature of CD4 and CD8 T cell epitopes, we corroborate the hypothesis that induction of effector T-cells should be a main goal for new vaccines against the emergent SARS-CoV-2 VOCs.
Collapse
Affiliation(s)
- Patrick O. Azevedo
- grid.8430.f0000 0001 2181 4888Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil ,grid.418068.30000 0001 0723 0931Instituto René Rachou, Fundação Oswaldo Cruz-Minas, Belo Horizonte, Brazil
| | - Natália S. Hojo-Souza
- grid.8430.f0000 0001 2181 4888Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil ,grid.418068.30000 0001 0723 0931Instituto René Rachou, Fundação Oswaldo Cruz-Minas, Belo Horizonte, Brazil
| | - Lídia P. Faustino
- grid.8430.f0000 0001 2181 4888Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil ,grid.418068.30000 0001 0723 0931Instituto René Rachou, Fundação Oswaldo Cruz-Minas, Belo Horizonte, Brazil
| | - Marcílio J. Fumagalli
- grid.11899.380000 0004 1937 0722Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil
| | - Isabella C. Hirako
- grid.418068.30000 0001 0723 0931Instituto René Rachou, Fundação Oswaldo Cruz-Minas, Belo Horizonte, Brazil
| | - Emiliano R. Oliveira
- grid.418068.30000 0001 0723 0931Instituto René Rachou, Fundação Oswaldo Cruz-Minas, Belo Horizonte, Brazil
| | - Maria M. Figueiredo
- grid.8430.f0000 0001 2181 4888Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Alex F. Carvalho
- grid.8430.f0000 0001 2181 4888Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Daniel Doro
- grid.8430.f0000 0001 2181 4888Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil ,grid.418068.30000 0001 0723 0931Instituto René Rachou, Fundação Oswaldo Cruz-Minas, Belo Horizonte, Brazil
| | - Luciana Benevides
- Plataforma Bi-Institucional de Pesquisa em Medicina Translacional - Fiocruz/SP, São Paulo, Brazil
| | - Edison Durigon
- grid.11899.380000 0004 1937 0722Instituto de Ciências Biológicas, Universidade de São Paulo, São Paulo, Brazil
| | - Flávio Fonseca
- grid.8430.f0000 0001 2181 4888Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil ,grid.8430.f0000 0001 2181 4888Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Alexandre M. Machado
- grid.8430.f0000 0001 2181 4888Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil ,grid.418068.30000 0001 0723 0931Instituto René Rachou, Fundação Oswaldo Cruz-Minas, Belo Horizonte, Brazil
| | - Ana P. Fernandes
- grid.8430.f0000 0001 2181 4888Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil ,grid.8430.f0000 0001 2181 4888Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Santuza R. Teixeira
- grid.8430.f0000 0001 2181 4888Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil ,grid.8430.f0000 0001 2181 4888Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - João S. Silva
- Plataforma Bi-Institucional de Pesquisa em Medicina Translacional - Fiocruz/SP, São Paulo, Brazil
| | - Ricardo T. Gazzinelli
- grid.8430.f0000 0001 2181 4888Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil ,grid.418068.30000 0001 0723 0931Instituto René Rachou, Fundação Oswaldo Cruz-Minas, Belo Horizonte, Brazil ,Plataforma Bi-Institucional de Pesquisa em Medicina Translacional - Fiocruz/SP, São Paulo, Brazil ,grid.8430.f0000 0001 2181 4888Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil ,grid.168645.80000 0001 0742 0364University of Massachusetts Medical School, Worcester, Massachusetts, USA
| |
Collapse
|
23
|
Okada Y, Kayano T, Anzai A, Zhang T, Nishiura H. Protection against SARS-CoV-2 BA.4 and BA.5 subvariants via vaccination and natural infection: A modeling study. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2023; 20:2530-2543. [PMID: 36899545 DOI: 10.3934/mbe.2023118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
With continuing emergence of new SARS-CoV-2 variants, understanding the proportion of the population protected against infection is crucial for public health risk assessment and decision-making and so that the general public can take preventive measures. We aimed to estimate the protection against symptomatic illness caused by SARS-CoV-2 Omicron variants BA.4 and BA.5 elicited by vaccination against and natural infection with other SARS-CoV-2 Omicron subvariants. We used a logistic model to define the protection rate against symptomatic infection caused by BA.1 and BA.2 as a function of neutralizing antibody titer values. Applying the quantified relationships to BA.4 and BA.5 using two different methods, the estimated protection rate against BA.4 and BA.5 was 11.3% (95% confidence interval [CI]: 0.01-25.4) (method 1) and 12.9% (95% CI: 8.8-18.0) (method 2) at 6 months after a second dose of BNT162b2 vaccine, 44.3% (95% CI: 20.0-59.3) (method 1) and 47.3% (95% CI: 34.1-60.6) (method 2) at 2 weeks after a third BNT162b2 dose, and 52.3% (95% CI: 25.1-69.2) (method 1) and 54.9% (95% CI: 37.6-71.4) (method 2) during the convalescent phase after infection with BA.1 and BA.2, respectively. Our study indicates that the protection rate against BA.4 and BA.5 are significantly lower compared with those against previous variants and may lead to substantial morbidity, and overall estimates were consistent with empirical reports. Our simple yet practical models enable prompt assessment of public health impacts posed by new SARS-CoV-2 variants using small sample-size neutralization titer data to support public health decisions in urgent situations.
Collapse
Affiliation(s)
- Yuta Okada
- Kyoto University School of Public Health, Yoshida-Konoe, Sakyo-ku, Kyoto 606-8601, Japan
| | - Taishi Kayano
- Kyoto University School of Public Health, Yoshida-Konoe, Sakyo-ku, Kyoto 606-8601, Japan
| | - Asami Anzai
- Kyoto University School of Public Health, Yoshida-Konoe, Sakyo-ku, Kyoto 606-8601, Japan
| | - Tong Zhang
- Kyoto University School of Public Health, Yoshida-Konoe, Sakyo-ku, Kyoto 606-8601, Japan
| | - Hiroshi Nishiura
- Kyoto University School of Public Health, Yoshida-Konoe, Sakyo-ku, Kyoto 606-8601, Japan
| |
Collapse
|
24
|
Natalini A, Simonetti S, Sher C, D’Oro U, Hayday AC, Di Rosa F. Durable CD8 T Cell Memory against SARS-CoV-2 by Prime/Boost and Multi-Dose Vaccination: Considerations on Inter-Dose Time Intervals. Int J Mol Sci 2022; 23:14367. [PMID: 36430845 PMCID: PMC9698736 DOI: 10.3390/ijms232214367] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 11/08/2022] [Accepted: 11/14/2022] [Indexed: 11/22/2022] Open
Abstract
Facing the COVID-19 pandemic, anti-SARS-CoV-2 vaccines were developed at unprecedented pace, productively exploiting contemporary fundamental research and prior art. Large-scale use of anti-SARS-CoV-2 vaccines has greatly limited severe morbidity and mortality. Protection has been correlated with high serum titres of neutralizing antibodies capable of blocking the interaction between the viral surface protein spike and the host SARS-CoV-2 receptor, ACE-2. Yet, vaccine-induced protection subsides over time, and breakthrough infections are commonly observed, mostly reflecting the decay of neutralizing antibodies and the emergence of variant viruses with mutant spike proteins. Memory CD8 T cells are a potent weapon against viruses, as they are against tumour cells. Anti-SARS-CoV-2 memory CD8 T cells are induced by either natural infection or vaccination and can be potentially exploited against spike-mutated viruses. We offer here an overview of current research about the induction of anti-SARS-CoV-2 memory CD8 T cells by vaccination, in the context of prior knowledge on vaccines and on fundamental mechanisms of immunological memory. We focus particularly on how vaccination by two doses (prime/boost) or more (boosters) promotes differentiation of memory CD8 T cells, and on how the time-length of inter-dose intervals may influence the magnitude and persistence of CD8 T cell memory.
Collapse
Affiliation(s)
- Ambra Natalini
- Institute of Molecular Biology and Pathology, National Research Council of Italy (CNR), 00161 Rome, Italy
- Immunosurveillance Laboratory, The Francis Crick Institute, London NW1 1AT, UK
| | - Sonia Simonetti
- Institute of Molecular Biology and Pathology, National Research Council of Italy (CNR), 00161 Rome, Italy
- Medical Oncology Department, Campus Bio-Medico University, 00128 Rome, Italy
| | - Carmel Sher
- Institute of Molecular Biology and Pathology, National Research Council of Italy (CNR), 00161 Rome, Italy
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | | | - Adrian C. Hayday
- Immunosurveillance Laboratory, The Francis Crick Institute, London NW1 1AT, UK
- Peter Gorer Department of Immunobiology, King’s College London, London WC2R 2LS, UK
- National Institute for Health and Research (NIHR) Biomedical Research Center (BRC), Guy’s and St Thomas’ NHS Foundation Trust, King’s College London, London WC2R 2LS, UK
| | - Francesca Di Rosa
- Institute of Molecular Biology and Pathology, National Research Council of Italy (CNR), 00161 Rome, Italy
| |
Collapse
|