201
|
Park HS, Matsuoka Y, Luongo C, Yang L, Santos C, Liu X, Ahlers LRH, Moore IN, Afroz S, Johnson RF, Lafont BAP, Dorward DW, Fischer ER, Martens C, Samal SK, Munir S, Buchholz UJ, Le Nouën C. Intranasal immunization with avian paramyxovirus type 3 expressing SARS-CoV-2 spike protein protects hamsters against SARS-CoV-2. NPJ Vaccines 2022; 7:72. [PMID: 35764659 PMCID: PMC9240059 DOI: 10.1038/s41541-022-00493-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 05/11/2022] [Indexed: 12/13/2022] Open
Abstract
Current vaccines against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) are administered parenterally and appear to be more protective in the lower versus the upper respiratory tract. Vaccines are needed that directly stimulate immunity in the respiratory tract, as well as systemic immunity. We used avian paramyxovirus type 3 (APMV3) as an intranasal vaccine vector to express the SARS-CoV-2 spike (S) protein. A lack of pre-existing immunity in humans and attenuation by host-range restriction make APMV3 a vector of interest. The SARS-CoV-2 S protein was stabilized in its prefusion conformation by six proline substitutions (S-6P) rather than the two that are used in most vaccine candidates, providing increased stability. APMV3 expressing S-6P (APMV3/S-6P) replicated to high titers in embryonated chicken eggs and was genetically stable, whereas APMV3 expressing non-stabilized S or S-2P were unstable. In hamsters, a single intranasal dose of APMV3/S-6P induced strong serum IgG and IgA responses to the S protein and its receptor-binding domain, and strong serum neutralizing antibody responses to SARS-CoV-2 isolate WA1/2020 (lineage A). Sera from APMV3/S-6P-immunized hamsters also efficiently neutralized Alpha and Beta variants of concern. Immunized hamsters challenged with WA1/2020 did not exhibit the weight loss and lung inflammation observed in empty-vector-immunized controls; SARS-CoV-2 replication in the upper and lower respiratory tract of immunized animals was low or undetectable compared to the substantial replication in controls. Thus, a single intranasal dose of APMV3/S-6P was highly immunogenic and protective against SARS-CoV-2 challenge, suggesting that APMV3/S-6P is suitable for clinical development.
Collapse
Affiliation(s)
- Hong-Su Park
- RNA Viruses Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Yumiko Matsuoka
- RNA Viruses Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Cindy Luongo
- RNA Viruses Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Lijuan Yang
- RNA Viruses Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Celia Santos
- RNA Viruses Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Xueqiao Liu
- RNA Viruses Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Laura R H Ahlers
- RNA Viruses Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Ian N Moore
- Infectious Disease Pathogenesis Section, Comparative Medicine Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Sharmin Afroz
- RNA Viruses Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Reed F Johnson
- SARS-CoV-2 Virology Core, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Bernard A P Lafont
- SARS-CoV-2 Virology Core, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - David W Dorward
- Research Technologies Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, 59840, USA
| | - Elizabeth R Fischer
- Research Technologies Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, 59840, USA
| | - Craig Martens
- Research Technologies Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, 59840, USA
| | - Siba K Samal
- Virginia-Maryland College of Veterinary Medicine, University of Maryland, College Park, MD, 20742, USA
| | - Shirin Munir
- RNA Viruses Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Ursula J Buchholz
- RNA Viruses Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA.
| | - Cyril Le Nouën
- RNA Viruses Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
202
|
Seyran M. Artificial intelligence and clinical data suggest the T cell-mediated SARS-CoV-2 nonstructural protein intranasal vaccines for global COVID-19 immunity. Vaccine 2022; 40:4296-4300. [PMID: 35778279 PMCID: PMC9226295 DOI: 10.1016/j.vaccine.2022.06.052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 06/13/2022] [Accepted: 06/20/2022] [Indexed: 11/11/2022]
Abstract
Advanced computational methodologies suggested SARS-CoV-2, nonstructural proteins ORF1AB, ORF3a, as the source of immunodominant peptides for T cell presentation. T cell immunity is long-lasting and compatible with COVID-19 pathology. Based on the supporting clinical data, nonstructural SARS-CoV-2 protein vaccines could provide global immunity against COVID-19.
Collapse
Affiliation(s)
- Murat Seyran
- The University of Vienna, Doctoral Studies in Natural and Technical Sciences (SPL 44), Währinger Straße, A-1090 Vienna, Austria.
| |
Collapse
|
203
|
Intranasal inoculation of an MVA-based vaccine induces IgA and protects the respiratory tract of hACE2 mice from SARS-CoV-2 infection. Proc Natl Acad Sci U S A 2022; 119:e2202069119. [PMID: 35679343 PMCID: PMC9214525 DOI: 10.1073/pnas.2202069119] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Despite the ability of current vaccines to significantly prevent severe disease due to Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), vaccinated individuals are still susceptible to infection and contribute to the spread of the virus. The present study demonstrates that a live, replication-deficient recombinant virus vaccine induces greater immunity and a greater level of protection in the respiratory tract of susceptible transgenic mice when inoculated intranasally compared with intramuscularly. Second-generation vaccines administered via the upper respiratory tract have the potential to limit the spread of SARS-CoV-2 more effectively than current vaccines. Current vaccines have greatly diminished the severity of the COVID-19 pandemic, even though they do not entirely prevent infection and transmission, likely due to insufficient immunity in the upper respiratory tract. Here, we compare intramuscular and intranasal administration of a live, replication-deficient modified vaccinia virus Ankara (MVA)–based Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) spike (S) vaccine to raise protective immune responses in the K18-hACE2 mouse model. Using a recombinant MVA expressing firefly luciferase for tracking, live imaging revealed luminescence of the respiratory tract of mice within 6 h and persisting for 3 d following intranasal inoculation, whereas luminescence remained at the site of intramuscular vaccination. Intramuscular vaccination induced S-binding–Immunoglobulin G (IgG) and neutralizing antibodies in the lungs, whereas intranasal vaccination also induced Immunoglobulin A (IgA) and higher levels of antigen-specific CD3+CD8+IFN-γ+ T cells. Similarly, IgG and neutralizing antibodies were present in the blood of mice immunized intranasally and intramuscularly, but IgA was detected only after intranasal inoculation. Intranasal boosting increased IgA after intranasal or intramuscular priming. While intramuscular vaccination prevented morbidity and cleared SARS-CoV-2 from the respiratory tract within several days after challenge, intranasal vaccination was more effective as neither infectious virus nor viral messenger (m)RNAs were detected in the nasal turbinates or lungs as early as 2 d after challenge, indicating prevention or rapid elimination of SARS-CoV-2 infection. Additionally, we determined that neutralizing antibody persisted for more than 6 mo and that serum induced to the Wuhan S protein neutralized pseudoviruses expressing the S proteins of variants, although with less potency, particularly for Beta and Omicron.
Collapse
|
204
|
Bhattacharya D. Instructing durable humoral immunity for COVID-19 and other vaccinable diseases. Immunity 2022; 55:945-964. [PMID: 35637104 PMCID: PMC9085459 DOI: 10.1016/j.immuni.2022.05.004] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 05/02/2022] [Accepted: 05/05/2022] [Indexed: 11/29/2022]
Abstract
Many aspects of SARS-CoV-2 have fully conformed with the principles established by decades of viral immunology research, ultimately leading to the crowning achievement of highly effective COVID-19 vaccines. Nonetheless, the pandemic has also exposed areas where our fundamental knowledge is thinner. Some key unknowns are the duration of humoral immunity post-primary infection or vaccination and how long booster shots confer protection. As a corollary, if protection does not last as long as desired, what are some ways it can be improved? Here, I discuss lessons from other infections and vaccines that point to several key features that influence durable antibody production and the perseverance of immunity. These include (1) the specific innate sensors that are initially triggered, (2) the kinetics of antigen delivery and persistence, (3) the starting B cell receptor (BCR) avidity and antigen valency, and (4) the memory B cell subsets that are recalled by boosters. I further highlight the fundamental B cell-intrinsic and B cell-extrinsic pathways that, if understood better, would provide a rational framework for vaccines to reliably provide durable immunity.
Collapse
Affiliation(s)
- Deepta Bhattacharya
- Department of Immunobiology, University of Arizona College of Medicine, Tucson, AZ 85724, USA.
| |
Collapse
|
205
|
Mittal D, Ali SA. Use of Nanomaterials for Diagnosis and Treatment: The Advancement of Next-Generation Antiviral Therapy. Microb Drug Resist 2022; 28:670-697. [PMID: 35696335 DOI: 10.1089/mdr.2021.0281] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Globally, viral illness propagation is the leading cause of morbidity and death, causing wreaking havoc on socioeconomic development and health care systems. The rise of infected individuals has outpaced the existing critical care facilities. Early and sophisticated methods are desperately required in this respect to halt the spread of the infection. Therefore, early detection of infectious agents and an early treatment approach may help minimize viral outbreaks. Conventional point-of-care diagnostic techniques such as computed tomography scan, quantitative real time polymerase chain reaction (qRT-PCR), X-ray, and immunoassay are still deemed valuable. However, the labor demanding, low sensitivity, and complex infrastructure needed for these methods preclude their use in distant areas. Nanotechnology has emerged as a potentially transformative technology due to its promise as an effective theranostic platform for diagnosing and treating viral infection, circumventing the limits of traditional techniques. Their unique physical and chemical characteristics make nanoparticles (NPs) advantageous for drug delivery platforms due to their size, encapsulation efficiency, improved bioavailability, effectiveness, immunogenicity, and antiviral response. This study discusses the recent research on nanotechnology-based treatments designed to combat new viruses.
Collapse
Affiliation(s)
- Deepti Mittal
- Nanosafety Lab, Division of Biochemistry, ICAR-NDRI, Karnal, Haryana, India
| | - Syed Azmal Ali
- Cell Biology and Proteomics Lab, Animal Biotechnology Center, ICAR-NDRI, Karnal, Haryana, India
| |
Collapse
|
206
|
Newly Emerged Antiviral Strategies for SARS-CoV-2: From Deciphering Viral Protein Structural Function to the Development of Vaccines, Antibodies, and Small Molecules. Int J Mol Sci 2022; 23:ijms23116083. [PMID: 35682761 PMCID: PMC9181103 DOI: 10.3390/ijms23116083] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 05/20/2022] [Accepted: 05/27/2022] [Indexed: 01/09/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) caused by the infection of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has become the most severe health crisis, causing extraordinary economic disruption worldwide. SARS-CoV-2 is a single-stranded RNA-enveloped virus. The process of viral replication and particle packaging is finished in host cells. Viral proteins, including both structural and nonstructural proteins, play important roles in the viral life cycle, which also provides the targets of treatment. Therefore, a better understanding of the structural function of virus proteins is crucial to speed up the development of vaccines and therapeutic strategies. Currently, the structure and function of proteins encoded by the SARS-CoV-2 genome are reviewed by several studies. However, most of them are based on the analysis of SARS-CoV-1 particles, lacking a systematic review update for SARS-CoV-2. Here, we specifically focus on the structure and function of proteins encoded by SARS-CoV-2. Viral proteins that contribute to COVID-19 infection and disease pathogenesis are reviewed according to the most recent research findings. The structure-function correlation of viral proteins provides a fundamental rationale for vaccine development and targeted therapy. Then, current antiviral vaccines are updated, such as inactive viral vaccines and protein-based vaccines and DNA, mRNA, and circular RNA vaccines. A summary of other therapeutic options is also reviewed, including monoclonal antibodies such as a cross-neutralizer antibody, a constructed cobinding antibody, a dual functional monoclonal antibody, an antibody cocktail, and an engineered bispecific antibody, as well as peptide-based inhibitors, chemical compounds, and clustered regularly interspaced short palindromic repeats (CRISPR) exploration. Overall, viral proteins and their functions provide the basis for targeted therapy and vaccine development.
Collapse
|
207
|
Intranasal administration of a recombinant RBD vaccine induces long-term immunity against Omicron-included SARS-CoV-2 variants. Signal Transduct Target Ther 2022; 7:159. [PMID: 35581200 PMCID: PMC9112270 DOI: 10.1038/s41392-022-01002-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 03/23/2022] [Accepted: 04/05/2022] [Indexed: 02/05/2023] Open
Abstract
The outbreak of coronavirus disease 2019 (COVID-19) has posed great threats to global health and economy. Several effective vaccines are available now, but additional booster immunization is required to retain or increase the immune responses owing to waning immunity and the emergency of new variant strains. The deficiency of intramuscularly delivered vaccines to induce mucosal immunity urged the development of mucosal vaccines. Here, we developed an adjuvanted intranasal RBD vaccine and monitored its long-term immunogenicity against both wild-type and mutant strains of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), including Omicron variants, in mice. Three-dose intranasal immunization with this vaccine induced and maintained high levels of neutralizing IgG antibodies in the sera for at least 1 year. Strong mucosal immunity was also provoked, including mucosal secretory IgA and lung-resident memory T cells (TRM). We also demonstrated that the long-term persistence of lung TRM cells is a consequence of local T-cell proliferation, rather than T-cell migration from lymph nodes. Our data suggested that the adjuvanted intranasal RBD vaccine is a promising vaccine candidate to establish robust, long-lasting, and broad protective immunity against SARS-CoV-2 both systemically and locally.
Collapse
|
208
|
The STING Ligand and Delivery System Synergistically Enhance the Immunogenicity of an Intranasal Spike SARS-CoV-2 Vaccine Candidate. Biomedicines 2022; 10:biomedicines10051142. [PMID: 35625879 PMCID: PMC9138454 DOI: 10.3390/biomedicines10051142] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 05/02/2022] [Accepted: 05/06/2022] [Indexed: 11/23/2022] Open
Abstract
The respiratory organ serves as a primary target site for SARS-CoV-2. Thus, the vaccine-stimulating immune response of the respiratory tract is significant in controlling SARS-CoV-2 transmission and disease development. In this study, mucoadhesive nanoparticles were used to deliver SARS-CoV-2 spike proteins (S-NPs) into the nasal tracts of mice. The responses in the respiratory organ and the systemic responses were monitored. The administration of S-NPs along with cGAMP conferred a robust stimulation of antibody responses in the respiratory tract, as demonstrated by an increase of IgA and IgG antibodies toward the spike proteins in bronchoalveolar lavages (BALs) and the lungs. Interestingly, the elicited antibodies were able to neutralize both the wild-type and Delta variant strains of SARS-CoV-2. Significantly, the intranasal immunization also stimulated systemic responses. This is evidenced by the increased production of circulating IgG and IgA, which were able to neutralize and bind specifically to the SARS-CoV-2 virion and spike protein. Additionally, this intranasal administration potently activated a splenic T cell response and the production of Th-1 cytokines, suggesting that this vaccine may well activate a cellular response in the respiratory tract. The results demonstrate that STING agonist strongly acts as an adjuvant to the immunogenicity of S-NPs. This platform may be an ideal vaccine against SARS-CoV-2.
Collapse
|
209
|
Mettelman RC, Allen EK, Thomas PG. Mucosal immune responses to infection and vaccination in the respiratory tract. Immunity 2022; 55:749-780. [PMID: 35545027 PMCID: PMC9087965 DOI: 10.1016/j.immuni.2022.04.013] [Citation(s) in RCA: 89] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 04/12/2022] [Accepted: 04/15/2022] [Indexed: 01/25/2023]
Abstract
The lungs are constantly exposed to inhaled debris, allergens, pollutants, commensal or pathogenic microorganisms, and respiratory viruses. As a result, innate and adaptive immune responses in the respiratory tract are tightly regulated and are in continual flux between states of enhanced pathogen clearance, immune-modulation, and tissue repair. New single-cell-sequencing techniques are expanding our knowledge of airway cellular complexity and the nuanced connections between structural and immune cell compartments. Understanding these varied interactions is critical in treatment of human pulmonary disease and infections and in next-generation vaccine design. Here, we review the innate and adaptive immune responses in the lung and airways following infection and vaccination, with particular focus on influenza virus and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The ongoing SARS-CoV-2 pandemic has put pulmonary research firmly into the global spotlight, challenging previously held notions of respiratory immunity and helping identify new populations at high risk for respiratory distress.
Collapse
Affiliation(s)
- Robert C Mettelman
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - E Kaitlynn Allen
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Paul G Thomas
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| |
Collapse
|
210
|
Molino D, Durier C, Radenne A, Desaint C, Ropers J, Courcier S, Vieillard LV, Rekacewicz C, Parfait B, Appay V, Batteux F, Barillot E, Cogné M, Combadière B, Eberhardt CS, Gorochov G, Hupé P, Ninove L, Paul S, Pellegrin I, van der Werf S, Lefebvre M, Botelho-Nevers E, Ortega-Perez I, Jaspard M, Sow S, Lelièvre JD, de Lamballerie X, Kieny MP, Tartour E, Launay O. A comparison of Sars-Cov-2 vaccine platforms: the CoviCompare project. Nat Med 2022; 28:882-884. [PMID: 35513532 DOI: 10.1038/s41591-022-01785-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Diana Molino
- Université Paris Cité, National Institute for Health and Medical Research (INSERM) CIC 1417 Cochin Pasteur, Innovative Clinical Research Network in Vaccinology (I-REIVAC), Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Cochin, Paris, France
| | | | - Anne Radenne
- AP-HP, Hôpitaux Universitaires Pitié Salpêtrière-Charles Foix, Unité de Recherche Clinique des Hôpitaux Universitaires Pitié Salpêtrière, Paris, France
| | | | - Jacques Ropers
- AP-HP, Hôpitaux Universitaires Pitié Salpêtrière-Charles Foix, Unité de Recherche Clinique des Hôpitaux Universitaires Pitié Salpêtrière, Paris, France
| | - Soizic Courcier
- Université Paris Cité, National Institute for Health and Medical Research (INSERM) CIC 1417 Cochin Pasteur, Innovative Clinical Research Network in Vaccinology (I-REIVAC), Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Cochin, Paris, France
| | - Louis Victorien Vieillard
- Université Paris Cité, National Institute for Health and Medical Research (INSERM) CIC 1417 Cochin Pasteur, Innovative Clinical Research Network in Vaccinology (I-REIVAC), Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Cochin, Paris, France
| | - Claire Rekacewicz
- Université Paris Cité, National Institute for Health and Medical Research (INSERM) CIC 1417 Cochin Pasteur, Innovative Clinical Research Network in Vaccinology (I-REIVAC), Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Cochin, Paris, France
| | - Beatrice Parfait
- AP-HP, Hôpital Cochin, Fédération des Centres de Ressources Biologiques-Plateforme de Ressources Biologiques Centre de Ressources Biologique Cochin, Paris, France
| | - Victor Appay
- Centre Hospitalier Universitaire (CHU) Bordeaux, Laboratory of Immunology and Immunogenetics, Université de Bordeaux, Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR) 5164, INSERM ERL 1303, ImmunoConcEpT, Bordeaux, France
| | - Frédéric Batteux
- AP-HP, Hôpital Cochin, Service d'Immunologie Biologique et Plateforme d'Immunomonitoring Vaccinal, Paris, France
| | - Emmanuel Barillot
- Institut Curie, PSL Research University-INSERM U900-MINES ParisTech, PSL, Paris, France
| | - Michel Cogné
- Laboratory of Immunology-Research Unit INSERM U 1236, B cell Ig Remodelling Singularities (BIGRES), Faculty of Medicine, French Blood Center (EFS Bretagne) & University Hospital, Rennes, France
| | - Béhazine Combadière
- Centre d'Immunologie et des Maladies Infectieuses-Paris (Cimi-Paris), INSERM U1135, Sorbonne Université, Paris, France
| | - Christiane S Eberhardt
- University of Geneva, Faculty of Medicine, Division of General Pediatrics, Department of Woman, Child and Adolescent Medicine and Center for Vaccinology, Geneva, Switzerland
| | - Guy Gorochov
- Sorbonne Université, INSERM, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Département d'Immunologie, AP-HP, Hôpital Pitié-Salpêtrière, Paris, France
| | - Philippe Hupé
- Institut Curie, PSL Research University-INSERM U900-MINES ParisTech, PSL, Paris, France.,CNRS, UMR 144, Paris, France
| | - Laetitia Ninove
- Aix Marseille Université, Research Institute for Sustainable Development (IRD) 190, INSERM 1207, IHU Méditerranée Infection, Unité des Virus Émergents, Marseille, France
| | - Stéphane Paul
- INSERM, U1111, CNRS, UMR 530, Immunology and Immunomonitoring Laboratory, iBiothera, CIRI-GIMAP, UCBL 1, UJM, CIC 1408, Saint-Etienne, France
| | - Isabelle Pellegrin
- Centre Hospitalier Universitaire (CHU) Bordeaux, Laboratory of Immunology and Immunogenetics, Université de Bordeaux, Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR) 5164, INSERM ERL 1303, ImmunoConcEpT, Bordeaux, France
| | - Sylvie van der Werf
- Université Paris Cité, Institut Pasteur, Unité Génétique Moléculaire Virus à ARN UMR 3569 CNRS, Paris, France
| | - Maeva Lefebvre
- CHU de Nantes, INSERM CIC1413, Maladies Infectieuses et Tropicales, Centre de Prévention des Maladies Infectieuses et Transmissibles, Nantes, France
| | - Elisabeth Botelho-Nevers
- INSERM CIC 1408, Axe Vaccinologie, CHU de Saint-Etienne, Service d'Infectiologie, Saint-Etienne, France
| | | | - Marie Jaspard
- The Alliance for International Medical Action (ALIMA), Paris, France.,University of Bordeaux, INSERM, IRD, Bordeaux Population Health Center, UMR 1219, Bordeaux, France
| | - Samba Sow
- The Center for Vaccine Development, Bamako, Mali
| | | | - Xavier de Lamballerie
- Aix Marseille Université, Research Institute for Sustainable Development (IRD) 190, INSERM 1207, IHU Méditerranée Infection, Unité des Virus Émergents, Marseille, France
| | | | - Eric Tartour
- AP-HP, Hôpital Européen Georges Pompidou, INSERM U970, PARCC, Paris, France
| | - Odile Launay
- Université Paris Cité, National Institute for Health and Medical Research (INSERM) CIC 1417 Cochin Pasteur, Innovative Clinical Research Network in Vaccinology (I-REIVAC), Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Cochin, Paris, France.
| |
Collapse
|
211
|
Jawalagatti V, Kirthika P, Hewawaduge C, Yang MS, Park JY, Oh B, Lee JH. Bacteria-enabled oral delivery of a replicon-based mRNA vaccine candidate protects against ancestral and delta variant SARS-CoV-2. Mol Ther 2022; 30:1926-1940. [PMID: 35123065 PMCID: PMC8810265 DOI: 10.1016/j.ymthe.2022.01.042] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 01/12/2022] [Accepted: 01/30/2022] [Indexed: 11/17/2022] Open
Abstract
The ongoing severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) evolution has resulted in many variants, contributing to the striking drop in vaccine efficacy and necessitating the development of next-generation vaccines to tackle antigenic diversity. Herein we developed a multivalent Semliki Forest virus replicon-based mRNA vaccine targeting the receptor binding domain (RBD), heptad repeat domain (HR), membrane protein (M), and epitopes of non-structural protein 13 (nsp13) of SARS-CoV-2. The bacteria-mediated gene delivery offers the rapid production of large quantities of vaccine at a highly economical scale and notably allows needle-free mass vaccination. Favorable T-helper (Th) 1-dominated potent antibody and cellular immune responses were detected in the immunized mice. Further, immunization induced strong cross-protective neutralizing antibodies (NAbs) against the B.1.617.2 delta variant (clade G). We recorded a difference in induction of immunoglobulin (Ig) A response by the immunization route, with the oral route eliciting a strong mucosal secretory IgA (sIgA) response, which possibly has contributed to the enhanced protection conferred by oral immunization. Hamsters immunized orally were completely protected against viral replication in the lungs and the nasal cavity. Importantly, the vaccine protected the hamsters against SARS-CoV-2-induced pneumonia. The study provides proof-of-principle findings for the development of a feasible and efficacious oral mRNA vaccine against SARS-CoV-2 and its variants.
Collapse
Affiliation(s)
- Vijayakumar Jawalagatti
- Department of Veterinary Public Health, College of Veterinary Medicine, Jeonbuk National University, Iksan 54596, South Korea
| | - Perumalraja Kirthika
- Department of Veterinary Public Health, College of Veterinary Medicine, Jeonbuk National University, Iksan 54596, South Korea
| | - Chamith Hewawaduge
- Department of Veterinary Public Health, College of Veterinary Medicine, Jeonbuk National University, Iksan 54596, South Korea
| | - Myeon-Sik Yang
- Department of Veterinary Pathology, College of Veterinary Medicine, Jeonbuk National University, Iksan 54596, South Korea
| | - Ji-Young Park
- Department of Veterinary Public Health, College of Veterinary Medicine, Jeonbuk National University, Iksan 54596, South Korea
| | - Byungkwan Oh
- Department of Veterinary Pathology, College of Veterinary Medicine, Jeonbuk National University, Iksan 54596, South Korea
| | - John Hwa Lee
- Department of Veterinary Public Health, College of Veterinary Medicine, Jeonbuk National University, Iksan 54596, South Korea.
| |
Collapse
|
212
|
Jawalagatti V, Kirthika P, Lee JH. Oral mRNA Vaccines Against Infectious Diseases- A Bacterial Perspective [Invited]. Front Immunol 2022; 13:884862. [PMID: 35592330 PMCID: PMC9110646 DOI: 10.3389/fimmu.2022.884862] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 04/11/2022] [Indexed: 01/04/2023] Open
Abstract
The mRNA vaccines from Pfizer/BioNTech and Moderna were granted emergency approval in record time in the history of vaccinology and played an instrumental role in limiting the pandemic caused by SARS-CoV-2. The success of these vaccines resulted from over 3 decades of research from many scientists. However, the development of orally administrable mRNA vaccine development is surprisingly underexplored. Our group specializing in Salmonella-based vaccines explored the possibility of oral mRNA vaccine development. Oral delivery was made possible by the exploitation of the Semliki Forest viral replicon and Salmonella vehicle for transgene amplification and gene delivery, respectively. Herein we highlight the prospect of developing oral replicon-based mRNA vaccines against infectious diseases based on our recent primary studies on SARS-CoV-2. Further, we discuss the potential advantages and limitations of bacterial gene delivery.
Collapse
Affiliation(s)
| | | | - John Hwa Lee
- Department of Veterinary Public Health, College of Veterinary Medicine, Jeonbuk National University, Iksan, South Korea
| |
Collapse
|
213
|
Peter AS, Roth E, Schulz SR, Fraedrich K, Steinmetz T, Damm D, Hauke M, Richel E, Mueller‐Schmucker S, Habenicht K, Eberlein V, Issmail L, Uhlig N, Dolles S, Grüner E, Peterhoff D, Ciesek S, Hoffmann M, Pöhlmann S, McKay PF, Shattock RJ, Wölfel R, Socher E, Wagner R, Eichler J, Sticht H, Schuh W, Neipel F, Ensser A, Mielenz D, Tenbusch M, Winkler TH, Grunwald T, Überla K, Jäck H. A pair of noncompeting neutralizing human monoclonal antibodies protecting from disease in a SARS-CoV-2 infection model. Eur J Immunol 2022; 52:770-783. [PMID: 34355795 PMCID: PMC8420377 DOI: 10.1002/eji.202149374] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 07/05/2021] [Accepted: 08/03/2021] [Indexed: 11/18/2022]
Abstract
TRIANNI mice carry an entire set of human immunoglobulin V region gene segments and are a powerful tool to rapidly isolate human monoclonal antibodies. After immunizing these mice with DNA encoding the spike protein of SARS-CoV-2 and boosting with spike protein, we identified 29 hybridoma antibodies that reacted with the SARS-CoV-2 spike protein. Nine antibodies neutralize SARS-CoV-2 infection at IC50 values in the subnanomolar range. ELISA-binding studies and DNA sequence analyses revealed one cluster of three clonally related neutralizing antibodies that target the receptor-binding domain and compete with the cellular receptor hACE2. A second cluster of six clonally related neutralizing antibodies bind to the N-terminal domain of the spike protein without competing with the binding of hACE2 or cluster 1 antibodies. SARS-CoV-2 mutants selected for resistance to an antibody from one cluster are still neutralized by an antibody from the other cluster. Antibodies from both clusters markedly reduced viral spread in mice transgenic for human ACE2 and protected the animals from SARS-CoV-2-induced weight loss. The two clusters of potent noncompeting SARS-CoV-2 neutralizing antibodies provide potential candidates for therapy and prophylaxis of COVID-19. The study further supports transgenic animals with a human immunoglobulin gene repertoire as a powerful platform in pandemic preparedness initiatives.
Collapse
|
214
|
Chen J, Wang P, Yuan L, Zhang L, Zhang L, Zhao H, Chen C, Wang X, Han J, Chen Y, Jia J, Lu Z, Hong J, Lu Z, Wang Q, Chen R, Qi R, Ma J, Zhou M, Yu H, Zhuang C, Liu X, Han Q, Wang G, Su Y, Yuan Q, Cheng T, Wu T, Ye X, Zhang T, Li C, Zhang J, Zhu H, Chen Y, Chen H, Xia N. A live attenuated virus-based intranasal COVID-19 vaccine provides rapid, prolonged, and broad protection against SARS-CoV-2. Sci Bull (Beijing) 2022; 67:1372-1387. [PMID: 35637645 PMCID: PMC9134758 DOI: 10.1016/j.scib.2022.05.018] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 04/15/2022] [Accepted: 05/25/2022] [Indexed: 12/11/2022]
Abstract
Remarkable progress has been made in developing intramuscular vaccines against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2); however, they are limited with respect to eliciting local immunity in the respiratory tract, which is the primary infection site for SARS-CoV-2. To overcome the limitations of intramuscular vaccines, we constructed a nasal vaccine candidate based on an influenza vector by inserting a gene encoding the receptor-binding domain (RBD) of the spike protein of SARS-CoV-2, named CA4-dNS1-nCoV-RBD (dNS1-RBD). A preclinical study showed that in hamsters challenged 1 d after single-dose vaccination or 9 months after booster vaccination, dNS1-RBD largely mitigated lung pathology, with no loss of body weight. Moreover, such cellular immunity is relatively unimpaired for the most concerning SARS-CoV-2 variants, especially for the latest Omicron variant. In addition, this vaccine also provides cross-protection against H1N1 and H5N1 influenza viruses. The protective immune mechanism of dNS1-RBD could be attributed to the innate immune response in the nasal epithelium, local RBD-specific T cell response in the lung, and RBD-specific IgA and IgG response. Thus, this study demonstrates that the intranasally delivered dNS1-RBD vaccine candidate may offer an important addition to the fight against the ongoing coronavirus disease 2019 pandemic and influenza infection, compensating limitations of current intramuscular vaccines.
Collapse
Affiliation(s)
- Junyu Chen
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Pui Wang
- State Key Laboratory of Emerging Infectious Diseases, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China
| | - Lunzhi Yuan
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Liang Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Limin Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Hui Zhao
- National Institute for Food and Drug Control, Beijing 102629, China
| | - Congjie Chen
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Xijing Wang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Jinle Han
- Beijing Wantai Biological Pharmacy Enterprise Co., Ltd., Beijing 102206, China
| | - Yaode Chen
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Jizong Jia
- Beijing Wantai Biological Pharmacy Enterprise Co., Ltd., Beijing 102206, China
| | - Zhen Lu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Junping Hong
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Zicen Lu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Qian Wang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Rirong Chen
- State Key Laboratory of Emerging Infectious Diseases, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China
- Guangdong-Hong Kong Joint Laboratory of Emerging Infectious Diseases/Joint Laboratory for International Collaboration in Virology and Emerging Infectious Diseases, Joint Institute of Virology (STU/HKU), Shantou University, Shantou 515063, China
- EKIH Pathogen Research Institute, Shenzhen 518067, China
| | - Ruoyao Qi
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Jian Ma
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Min Zhou
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Huan Yu
- State Key Laboratory of Emerging Infectious Diseases, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China
- Guangdong-Hong Kong Joint Laboratory of Emerging Infectious Diseases/Joint Laboratory for International Collaboration in Virology and Emerging Infectious Diseases, Joint Institute of Virology (STU/HKU), Shantou University, Shantou 515063, China
- EKIH Pathogen Research Institute, Shenzhen 518067, China
| | - Chunlan Zhuang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Xiaohui Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Qiangyuan Han
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Guosong Wang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Yingying Su
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Quan Yuan
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Tong Cheng
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Ting Wu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Xiangzhong Ye
- Beijing Wantai Biological Pharmacy Enterprise Co., Ltd., Beijing 102206, China
| | - Tianying Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Changgui Li
- National Institute for Food and Drug Control, Beijing 102629, China
| | - Jun Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Huachen Zhu
- State Key Laboratory of Emerging Infectious Diseases, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China
- Guangdong-Hong Kong Joint Laboratory of Emerging Infectious Diseases/Joint Laboratory for International Collaboration in Virology and Emerging Infectious Diseases, Joint Institute of Virology (STU/HKU), Shantou University, Shantou 515063, China
- EKIH Pathogen Research Institute, Shenzhen 518067, China
| | - Yixin Chen
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Honglin Chen
- State Key Laboratory of Emerging Infectious Diseases, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China
| | - Ningshao Xia
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiamen 361102, China
| |
Collapse
|
215
|
Innate and Adaptive Immune Responses in the Upper Respiratory Tract and the Infectivity of SARS-CoV-2. Viruses 2022; 14:v14050933. [PMID: 35632675 PMCID: PMC9143801 DOI: 10.3390/v14050933] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 04/20/2022] [Accepted: 04/26/2022] [Indexed: 02/01/2023] Open
Abstract
Increasing evidence shows the nasal epithelium to be the initial site of SARS-CoV-2 infection, and that early and effective immune responses in the upper respiratory tract (URT) limit and eliminate the infection in the URT, thereby preventing infection of the lower respiratory tract and the development of severe COVID-19. SARS-CoV-2 interferes with innate immunity signaling and evolves mutants that can reduce antibody-mediated immunity in the URT. Recent genetic and immunological advances in understanding innate immunity to SARS-CoV-2 in the URT, and the ability of prior infections as well as currently available injectable and potential intranasal COVID-19 vaccines to generate anamnestic adaptive immunity in the URT, are reviewed. It is suggested that the more detailed investigation of URT immune responses to all types of COVID-19 vaccines, and the development of safe and effective COVID-19 vaccines for intranasal administration, are important needs.
Collapse
|
216
|
A single intranasal dose of human parainfluenza virus type 3-vectored vaccine induces effective antibody and memory T cell response in the lungs and protects hamsters against SARS-CoV-2. NPJ Vaccines 2022; 7:47. [PMID: 35468973 PMCID: PMC9038905 DOI: 10.1038/s41541-022-00471-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 03/21/2022] [Indexed: 12/31/2022] Open
Abstract
Respiratory tract vaccination has an advantage of needle-free delivery and induction of mucosal immune response in the portal of SARS-CoV-2 entry. We utilized human parainfluenza virus type 3 vector to generate constructs expressing the full spike (S) protein of SARS-CoV-2, its S1 subunit, or the receptor-binding domain, and tested them in hamsters as single-dose intranasal vaccines. The construct bearing full-length S induced high titers of neutralizing antibodies specific to S protein domains critical to the protein functions. Robust memory T cell responses in the lungs were also induced, which represent an additional barrier to infection and should be less sensitive than the antibody responses to mutations present in SARS-CoV-2 variants. Following SARS-CoV-2 challenge, animals were protected from the disease and detectable viral replication. Vaccination prevented induction of gene pathways associated with inflammation. These results indicate advantages of respiratory vaccination against COVID-19 and inform the design of mucosal SARS-CoV-2 vaccines.
Collapse
|
217
|
Nwagwu CS, Ugwu CN, Ogbonna JDN, Onugwu AL, Agbo CP, Echezona AC, Ezeibe EN, Uzondu S, Kenechukwu FC, Akpa PA, Momoh MA, Nnamani PO, Tarirai C, Ofokansi KC, Attama AA. Recent and advanced nano-technological strategies for COVID-19 vaccine development. METHODS IN MICROBIOLOGY 2022; 50:151-188. [PMID: 38620863 PMCID: PMC9015106 DOI: 10.1016/bs.mim.2022.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The outbreak of the COVID-19 pandemic in 2019 has been one of the greatest challenges modern medicine and science has ever faced. It has affected millions of people around the world and altered human life and activities as we once knew. The high prevalence as well as an extended period of incubations which usually does not present with symptoms have played a formidable role in the transmission and infection of millions. A lot of research has been carried out on developing suitable treatment and effective preventive measures for the control of the pandemic. Preventive strategies which include social distancing, use of masks, washing of hands, and contact tracing have been effective in slowing the spread of the virus; however, the infectious nature of the SARS-COV-2 has made these strategies unable to eradicate its spread. In addition, the continuous increase in the number of cases and death, as well as the appearance of several variants of the virus, has necessitated the development of effective and safe vaccines in a bid to ensure that human activities can return to normalcy. Nanotechnology has been of great benefit in the design of vaccines as nano-sized materials have been known to aid the safe and effective delivery of antigens as well as serve as suitable adjuvants to potentiate responses to vaccines. There are only four vaccine candidates currently approved for use in humans while many other candidates are at various levels of development. This review seeks to provide updated information on the current nano-technological strategies employed in the development of COVID-19 vaccines.
Collapse
Affiliation(s)
- Chinekwu Sherridan Nwagwu
- Drug Delivery & Nanomedicines Research Laboratory, Department of Pharmaceutics, University of Nigeria, Nsukka, Enugu State, Nigeria
| | - Chinenye Nnenna Ugwu
- Department of Pharmaceutical Microbiology and Biotechnology, University of Nigeria, Nsukka, Enugu state, Nigeria
| | - John Dike Nwabueze Ogbonna
- Drug Delivery & Nanomedicines Research Laboratory, Department of Pharmaceutics, University of Nigeria, Nsukka, Enugu State, Nigeria
| | - Adaeze Linda Onugwu
- Drug Delivery & Nanomedicines Research Laboratory, Department of Pharmaceutics, University of Nigeria, Nsukka, Enugu State, Nigeria
| | - Chinazom Precious Agbo
- Drug Delivery & Nanomedicines Research Laboratory, Department of Pharmaceutics, University of Nigeria, Nsukka, Enugu State, Nigeria
| | - Adaeze Chidiebere Echezona
- Drug Delivery & Nanomedicines Research Laboratory, Department of Pharmaceutics, University of Nigeria, Nsukka, Enugu State, Nigeria
| | - Ezinwanne Nneoma Ezeibe
- Department of Pharmaceutical Microbiology and Biotechnology, University of Nigeria, Nsukka, Enugu state, Nigeria
| | - Samuel Uzondu
- Drug Delivery & Nanomedicines Research Laboratory, Department of Pharmaceutics, University of Nigeria, Nsukka, Enugu State, Nigeria
| | - Frankline Chimaobi Kenechukwu
- Drug Delivery & Nanomedicines Research Laboratory, Department of Pharmaceutics, University of Nigeria, Nsukka, Enugu State, Nigeria
| | - Paul Achile Akpa
- Drug Delivery & Nanomedicines Research Laboratory, Department of Pharmaceutics, University of Nigeria, Nsukka, Enugu State, Nigeria
| | - Mumuni Audu Momoh
- Drug Delivery & Nanomedicines Research Laboratory, Department of Pharmaceutics, University of Nigeria, Nsukka, Enugu State, Nigeria
| | - Petra Obioma Nnamani
- Drug Delivery & Nanomedicines Research Laboratory, Department of Pharmaceutics, University of Nigeria, Nsukka, Enugu State, Nigeria
| | - Clemence Tarirai
- Department of Pharmaceutical Sciences, Tshwane University of Technology, Pretoria, South Africa
| | - Kenneth Chibuzor Ofokansi
- Drug Delivery & Nanomedicines Research Laboratory, Department of Pharmaceutics, University of Nigeria, Nsukka, Enugu State, Nigeria
| | - Anthony Amaechi Attama
- Drug Delivery & Nanomedicines Research Laboratory, Department of Pharmaceutics, University of Nigeria, Nsukka, Enugu State, Nigeria
| |
Collapse
|
218
|
Oja AE, van Lier RAW, Hombrink P. Two sides of the same coin: Protective versus pathogenic CD4 + resident memory T cells. Sci Immunol 2022; 7:eabf9393. [PMID: 35394815 DOI: 10.1126/sciimmunol.abf9393] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
The ability of the adaptive immune system to form memory is key to providing protection against secondary infections. Resident memory T cells (TRM) are specialized T cell populations that reside within tissue sites where they await reencounter with their cognate antigen. TRM are distinct from circulating memory cells, including central and effector memory T cells, both functionally and transcriptionally. Since the discovery of TRM, most research has focused on CD8+ TRM, despite that CD4+ TRM are also abundant in most tissues. In the past few years, more evidence has emerged that CD4+ TRM can contribute both protective and pathogenic roles in disease. A complexity inherent to the CD4+ TRM field is the ability of CD4+ T cells to polarize into a multitude of distinct subsets and recognize not only viruses and intracellular bacteria but also extracellular bacteria, fungi, and parasites. In this review, we outline the key features of CD4+ TRM in health and disease, including their contributions to protection against SARS-CoV-2 and potential contributions to immunopathology associated with COVID-19.
Collapse
Affiliation(s)
- Anna E Oja
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - René A W van Lier
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Pleun Hombrink
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
219
|
Williams CM, Roy S, Sun W, Furuya AM, Wijesundara DK, Furuya Y. LUNG group 2 innate lymphoid cells as a new adjuvant target to enhance intranasal vaccine efficacy against influenza. Clin Transl Immunology 2022; 11:e1381. [PMID: 35356066 PMCID: PMC8958247 DOI: 10.1002/cti2.1381] [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: 09/17/2021] [Revised: 03/05/2022] [Accepted: 03/07/2022] [Indexed: 11/12/2022] Open
Abstract
Group 2 innate lymphoid cells (ILC2) are a relatively new class of innate immune cells. Lung ILC2 are early responders that secrete type 2 cytokines in response to danger ‘alarmin’ signals such as interleukin (IL)‐33 and thymic stromal lymphopoietin. Being an early source of type 2 cytokines, ILC2 are a critical regulator of type 2 immune cells of both innate and adaptive immune responses. The immune regulatory functions of ILC2 were mostly investigated in diseases where T helper 2 inflammation predominates. However, in recent years, it has been appreciated that the role of ILC2 extends to other pathological conditions such as cancer and viral infections. In this review, we will focus on the potential role of lung ILC2 in the induction of mucosal immunity against influenza virus infection and discuss the potential utility of ILC2 as a target for mucosal vaccination.
Collapse
Affiliation(s)
- Clare M Williams
- Department of Immunology and Microbial Disease Albany Medical College Albany NY USA
| | - Sreeja Roy
- Department of Immunology and Microbial Disease Albany Medical College Albany NY USA
| | - Wei Sun
- Department of Immunology and Microbial Disease Albany Medical College Albany NY USA
| | | | - Danushka K Wijesundara
- The School of Chemistry and Molecular Biosciences The Australian Institute for Bioengineering and Nanotechnology The University of Queensland Brisbane QLD Australia
| | - Yoichi Furuya
- Department of Immunology and Microbial Disease Albany Medical College Albany NY USA
| |
Collapse
|
220
|
Mitsi E, Reiné J, Urban BC, Solórzano C, Nikolaou E, Hyder-Wright AD, Pojar S, Howard A, Hitchins L, Glynn S, Farrar MC, Liatsikos K, Collins AM, Walker NF, Hill HC, German EL, Cheliotis KS, Byrne RL, Williams CT, Cubas-Atienzar AI, Fletcher TE, Adams ER, Draper SJ, Pulido D, Beavon R, Theilacker C, Begier E, Jodar L, Gessner BD, Ferreira DM. Streptococcus pneumoniae colonization associates with impaired adaptive immune responses against SARS-CoV-2. J Clin Invest 2022; 132:e157124. [PMID: 35139037 PMCID: PMC8970672 DOI: 10.1172/jci157124] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 02/08/2022] [Indexed: 12/24/2022] Open
Abstract
BackgroundAlthough recent epidemiological data suggest that pneumococci may contribute to the risk of SARS-CoV-2 disease, cases of coinfection with Streptococcus pneumoniae in patients with coronavirus disease 2019 (COVID-19) during hospitalization have been reported infrequently. This apparent contradiction may be explained by interactions of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and pneumococci in the upper airway, resulting in the escape of SARS-CoV-2 from protective host immune responses.MethodsHere, we investigated the relationship of these 2 respiratory pathogens in 2 distinct cohorts of health care workers with asymptomatic or mildly symptomatic SARS-CoV-2 infection identified by systematic screening and patients with moderate to severe disease who presented to the hospital. We assessed the effect of coinfection on host antibody, cellular, and inflammatory responses to the virus.ResultsIn both cohorts, pneumococcal colonization was associated with diminished antiviral immune responses, which primarily affected mucosal IgA levels among individuals with mild or asymptomatic infection and cellular memory responses in infected patients.ConclusionOur findings suggest that S. pneumoniae impair host immunity to SARS-CoV-2 and raise the question of whether pneumococcal carriage also enables immune escape of other respiratory viruses and facilitates reinfection.Trial registrationISRCTN89159899 (FASTER study) and ClinicalTrials.gov NCT03502291 (LAIV study).
Collapse
Affiliation(s)
- Elena Mitsi
- Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Jesús Reiné
- Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Britta C. Urban
- Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Carla Solórzano
- Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Elissavet Nikolaou
- Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | | | - Sherin Pojar
- Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Ashleigh Howard
- Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Lisa Hitchins
- Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Sharon Glynn
- Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Madlen C. Farrar
- Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | | | - Andrea M. Collins
- Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
- Liverpool University Hospitals National Health Service (NHS) Foundation Trust, Liverpool, United Kingdom
| | - Naomi F. Walker
- Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
- Liverpool University Hospitals National Health Service (NHS) Foundation Trust, Liverpool, United Kingdom
| | - Helen C. Hill
- Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Esther L. German
- Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | | | - Rachel L. Byrne
- Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | | | - Ana I. Cubas-Atienzar
- Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
- Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Tom E. Fletcher
- Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Emily R. Adams
- Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Simon J. Draper
- Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - David Pulido
- Jenner Institute, University of Oxford, Oxford, United Kingdom
| | | | | | | | - Luis Jodar
- Pfizer Vaccines, Collegeville, Pennsylvania, USA
| | | | - Daniela M. Ferreira
- Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| |
Collapse
|
221
|
Mouro V, Fischer A. Dealing with a mucosal viral pandemic: lessons from COVID-19 vaccines. Mucosal Immunol 2022; 15:584-594. [PMID: 35505121 PMCID: PMC9062288 DOI: 10.1038/s41385-022-00517-8] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 04/13/2022] [Accepted: 04/19/2022] [Indexed: 02/04/2023]
Abstract
The development and deployment of vaccines against COVID-19 demonstrated major successes in providing immunity and preventing severe disease and death. Yet SARS-CoV-2 evolves and vaccine-induced protection wanes, meaning progress in vaccination strategies is of upmost importance. New vaccines directed at emerging viral strains are being developed while vaccination schemes with booster doses and combinations of different platform-based vaccines are being tested in trials and real-world settings. Despite these diverse approaches, COVID-19 vaccines are only delivered intramuscularly, whereas the nasal mucosa is the primary site of infection with SARS-CoV-2. Preclinical mucosal vaccines with intranasal or oral administration demonstrate promising results regarding mucosal IgA generation and tissue-resident lymphocyte responses against SARS-CoV-2. By mounting an improved local humoral and cell-mediated response, mucosal vaccination could be a safe and effective way to prevent infection, block transmission and contribute to reduce SARS-CoV-2 spread. However, questions and limitations remain: how effectively and reproducibly will vaccines penetrate mucosal barriers? Will vaccine-induced mucosal IgA responses provide sustained protection against infection?
Collapse
Affiliation(s)
- Violette Mouro
- Université Paris Cité, Paris, France.
- Sorbonne Université, Paris, France.
| | - Alain Fischer
- Imagine Institute, Paris, France
- Immunology and Pediatric Hematology Department, Assistance Publique-Hôpitaux de Paris, Paris, France
- Institut National de la Santé et de la Recherche Médicale UMR 1163, Paris, France
- Collège de France, Paris, France
| |
Collapse
|
222
|
Fragoso-Saavedra M, Ramírez-Estudillo C, Peláez-González DL, Ramos-Flores JO, Torres-Franco G, Núñez-Muñoz L, Marcelino-Pérez G, Segura-Covarrubias MG, González-González R, Ruiz-Medrano R, Xoconostle-Cázares B, Gayosso-Vázquez A, Reyes-Maya S, Ramírez-Andoney V, Alonso-Morales RA, Vega-López MA. Combined Subcutaneous-Intranasal Immunization With Epitope-Based Antigens Elicits Binding and Neutralizing Antibody Responses in Serum and Mucosae Against PRRSV-2 and SARS-CoV-2. Front Immunol 2022; 13:848054. [PMID: 35432364 PMCID: PMC9008747 DOI: 10.3389/fimmu.2022.848054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 03/08/2022] [Indexed: 11/23/2022] Open
Abstract
New vaccine design approaches, platforms, and immunization strategies might foster antiviral mucosal effector and memory responses to reduce asymptomatic infection and transmission in vaccinated individuals. Here, we investigated a combined parenteral and mucosal immunization scheme to induce local and serum antibody responses, employing the epitope-based antigens 3BT and NG19m. These antigens target the important emerging and re-emerging viruses PRRSV-2 and SARS-CoV-2, respectively. We assessed two versions of the 3BT protein, which contains conserved epitopes from the GP5 envelope protein of PRRSV-2: soluble and expressed by the recombinant baculovirus BacDual-3BT. On the other hand, NG19m, comprising the receptor-binding motif of the S protein of SARS-CoV-2, was evaluated as a soluble recombinant protein only. Vietnamese mini-pigs were immunized employing different inoculation routes: subcutaneous, intranasal, or a combination of both (s.c.-i.n.). Animals produced antigen-binding and neut1ralizing antibodies in serum and mucosal fluids, with varying patterns of concentration and activity, depending on the antigen and the immunization schedule. Soluble 3BT was a potent immunogen to elicit binding and neutralizing antibodies in serum, nasal mucus, and vaginal swabs. The vectored immunogen BacDual-3BT induced binding antibodies in serum and mucosae, but PRRSV-2 neutralizing activity was found in nasal mucus exclusively when administered intranasally. NG19m promoted serum and mucosal binding antibodies, which showed differing neutralizing activity. Only serum samples from subcutaneously immunized animals inhibited RBD-ACE2 interaction, while mini-pigs inoculated intranasally or via the combined s.c.-i.n. scheme produced subtle neutralizing humoral responses in the upper and lower respiratory mucosae. Our results show that intranasal immunization, alone or combined with subcutaneous delivery of epitope-based antigens, generates local and systemic binding and neutralizing antibodies. Further investigation is needed to evaluate the capability of the induced responses to prevent infection and reduce transmission.
Collapse
Affiliation(s)
- Mario Fragoso-Saavedra
- Laboratorio de Inmunobiología de las Mucosas, Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México, Mexico
| | - Carmen Ramírez-Estudillo
- Laboratorio de Inmunobiología de las Mucosas, Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México, Mexico
| | - Diana L. Peláez-González
- Unidad de Producción y Experimentación de Animales de Laboratorio, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México, Mexico
| | - Jorge O. Ramos-Flores
- Unidad de Producción y Experimentación de Animales de Laboratorio, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México, Mexico
| | - Gustavo Torres-Franco
- Unidad de Producción y Experimentación de Animales de Laboratorio, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México, Mexico
| | - Leandro Núñez-Muñoz
- Laboratorio de Biología Molecular de Plantas, Departamento de Biotecnología y Bioingeniería, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México, Mexico
| | - Gabriel Marcelino-Pérez
- Laboratorio de Biología Molecular de Plantas, Departamento de Biotecnología y Bioingeniería, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México, Mexico
| | - María G. Segura-Covarrubias
- Laboratorio de Biología Molecular de Plantas, Departamento de Biotecnología y Bioingeniería, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México, Mexico
| | - Rogelio González-González
- Laboratorio de Biología Molecular de Plantas, Departamento de Biotecnología y Bioingeniería, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México, Mexico
| | - Roberto Ruiz-Medrano
- Laboratorio de Biología Molecular de Plantas, Departamento de Biotecnología y Bioingeniería, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México, Mexico
| | - Beatriz Xoconostle-Cázares
- Laboratorio de Biología Molecular de Plantas, Departamento de Biotecnología y Bioingeniería, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México, Mexico
| | - Amanda Gayosso-Vázquez
- Laboratorio de Genética Molecular, Departamento de Genética y Bioestadística, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Silvia Reyes-Maya
- Laboratorio de Genética Molecular, Departamento de Genética y Bioestadística, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Vianey Ramírez-Andoney
- Laboratorio de Genética Molecular, Departamento de Genética y Bioestadística, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Rogelio A. Alonso-Morales
- Laboratorio de Genética Molecular, Departamento de Genética y Bioestadística, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Marco A. Vega-López
- Laboratorio de Inmunobiología de las Mucosas, Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México, Mexico
| |
Collapse
|
223
|
Li XF, Cui Z, Fan H, Chen Q, Cao L, Qiu HY, Zhang NN, Xu YP, Zhang RR, Zhou C, Ye Q, Deng YQ, Guo Y, Qin S, Fan K, Wang L, Jia Z, Cui Y, Wang X, Qin CF. A highly immunogenic live-attenuated vaccine candidate prevents SARS-CoV-2 infection and transmission in hamsters. Innovation (N Y) 2022; 3:100221. [PMID: 35252935 PMCID: PMC8888354 DOI: 10.1016/j.xinn.2022.100221] [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: 11/11/2021] [Accepted: 02/28/2022] [Indexed: 01/08/2023] Open
Abstract
The highly pathogenic and readily transmissible SARS-CoV-2 has caused a global coronavirus pandemic, urgently requiring effective countermeasures against its rapid expansion. All available vaccine platforms are being used to generate safe and effective COVID-19 vaccines. Here, we generated a live-attenuated candidate vaccine strain by serial passaging of a SARS-CoV-2 clinical isolate in Vero cells. Deep sequencing revealed the dynamic adaptation of SARS-CoV-2 in Vero cells, resulting in a stable clone with a deletion of seven amino acids (N679SPRRAR685) at the S1/S2 junction of the S protein (named VAS5). VAS5 showed significant attenuation of replication in multiple human cell lines, human airway epithelium organoids, and hACE2 mice. Viral fitness competition assays demonstrated that VAS5 showed specific tropism to Vero cells but decreased fitness in human cells compared with the parental virus. More importantly, a single intranasal injection of VAS5 elicited a high level of neutralizing antibodies and prevented SARS-CoV-2 infection in mice as well as close-contact transmission in golden Syrian hamsters. Structural and biochemical analysis revealed a stable and locked prefusion conformation of the S trimer of VAS5, which most resembles SARS-CoV-2-3Q-2P, an advanced vaccine immunogen (NVAX-CoV2373). Further systematic antigenic profiling and immunogenicity validation confirmed that the VAS5 S trimer presents an enhanced antigenic mimic of the wild-type S trimer. Our results not only provide a potent live-attenuated vaccine candidate against COVID-19 but also clarify the molecular and structural basis for the highly attenuated and super immunogenic phenotype of VAS5.
Collapse
Affiliation(s)
- Xiao-Feng Li
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing 100071, China
| | - Zhen Cui
- CAS Key Laboratory of Infection and Immunity, National Laboratory of Macromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Hang Fan
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing 100071, China
| | - Qi Chen
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing 100071, China
| | - Lei Cao
- CAS Key Laboratory of Infection and Immunity, National Laboratory of Macromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Hong-Ying Qiu
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing 100071, China
| | - Na-Na Zhang
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing 100071, China
- School of Medicine, Tsinghua University, Beijing 100084, China
| | - Yan-Peng Xu
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing 100071, China
| | - Rong-Rong Zhang
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing 100071, China
| | - Chao Zhou
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing 100071, China
| | - Qing Ye
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing 100071, China
| | - Yong-Qiang Deng
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing 100071, China
| | - Yan Guo
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing 100071, China
| | - Si Qin
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing 100071, China
| | - Kaiyue Fan
- CAS Key Laboratory of Infection and Immunity, National Laboratory of Macromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Lei Wang
- CAS Key Laboratory of Infection and Immunity, National Laboratory of Macromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Zijing Jia
- CAS Key Laboratory of Infection and Immunity, National Laboratory of Macromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Yujun Cui
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing 100071, China
| | - Xiangxi Wang
- CAS Key Laboratory of Infection and Immunity, National Laboratory of Macromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Cheng-Feng Qin
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing 100071, China
- Research Unit of Discovery and Tracing of Natural Focus Diseases, Chinese Academy of Medical Sciences, Beijing 100071, China
| |
Collapse
|
224
|
Wong TY, Horspool AM, Russ BP, Ye C, Lee KS, Winters MT, Bevere JR, Miller OA, Rader NA, Cooper M, Kieffer T, Sourimant J, Greninger AL, Plemper RK, Denvir J, Cyphert HA, Barbier M, Torrelles JB, Martinez I, Martinez-Sobrido L, Damron FH. Evaluating Antibody Mediated Protection against Alpha, Beta, and Delta SARS-CoV-2 Variants of Concern in K18-hACE2 Transgenic Mice. J Virol 2022; 96:e0218421. [PMID: 35080423 PMCID: PMC8941865 DOI: 10.1128/jvi.02184-21] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 01/02/2022] [Indexed: 12/04/2022] Open
Abstract
SARS-CoV-2 variants of concern (VoC) are impacting responses to the COVID-19 pandemic. Here, we utilized passive immunization using human convalescent plasma (HCP) obtained from a critically ill COVID-19 patient in the early pandemic to study the efficacy of polyclonal antibodies generated to ancestral SARS-CoV-2 against the Alpha, Beta, and Delta VoC in the K18 human angiotensin converting enzyme 2 (hACE2) transgenic mouse model. HCP protected mice from challenge with the original WA-1 SARS-CoV-2 strain; however, only partially protected mice challenged with the Alpha VoC (60% survival) and failed to save Beta challenged mice from succumbing to disease. HCP treatment groups had elevated receptor binding domain (RBD) and nucleocapsid IgG titers in the serum; however, Beta VoC viral RNA burden in the lung and brain was not decreased due to HCP treatment. While mice could be protected from WA-1 or Alpha challenge with a single dose of HCP, six doses of HCP could not decrease mortality of Delta challenged mice. Overall, these data demonstrate that VoC have enhanced immune evasion and this work underscores the need for in vivo models to evaluate future emerging strains. IMPORTANCE Emerging SARS-CoV-2 VoC are posing new problems regarding vaccine and monoclonal antibody efficacy. To better understand immune evasion tactics of the VoC, we utilized passive immunization to study the effect of early-pandemic SARS-CoV-2 HCP against, Alpha, Beta, and Delta VoC. We observed that HCP from a human infected with the original SARS-CoV-2 was unable to control lethality of Alpha, Beta, or Delta VoC in the K18-hACE2 transgenic mouse model of SARS-CoV-2 infection. Our findings demonstrate that passive immunization can be used as a model to evaluate immune evasion of emerging VoC strains.
Collapse
Affiliation(s)
- Ting Y. Wong
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center at West, Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - Alexander M. Horspool
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center at West, Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - Brynnan P. Russ
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center at West, Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - Chengjin Ye
- Host-Pathogen Interactions and Population Health Programs, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Katherine S. Lee
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center at West, Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - Michael T. Winters
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
| | - Justin R. Bevere
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center at West, Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - Olivia A. Miller
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center at West, Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - Nathaniel A. Rader
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center at West, Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - Melissa Cooper
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center at West, Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - Theodore Kieffer
- Department of Pathology, Anatomy and Laboratory Medicine, West Virginia University School of Medicine, Morgantown, West Virginia, USA
| | - Julien Sourimant
- Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia, USA
| | - Alexander L. Greninger
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Richard K. Plemper
- Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia, USA
| | - James Denvir
- Department of Biomedical Sciences, Marshall University, Huntington, West Virginia, USA
| | - Holly A. Cyphert
- Department of Biological Sciences, Marshall University, Huntington, West Virginia, USA
| | - Mariette Barbier
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center at West, Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - Jordi B. Torrelles
- Host-Pathogen Interactions and Population Health Programs, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Ivan Martinez
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- West Virginia University Cancer Institute, Morgantown, West Virginia, USA
| | - Luis Martinez-Sobrido
- Host-Pathogen Interactions and Population Health Programs, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - F. Heath Damron
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center at West, Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| |
Collapse
|
225
|
Suberi A, Grun MK, Mao T, Israelow B, Reschke M, Grundler J, Akhtar L, Lee T, Shin K, Piotrowski-Daspit AS, Homer RJ, Iwasaki A, Suh HW, Saltzman WM. Inhalable polymer nanoparticles for versatile mRNA delivery and mucosal vaccination. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022:2022.03.22.485401. [PMID: 35350207 PMCID: PMC8963702 DOI: 10.1101/2022.03.22.485401] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
An inhalable platform for mRNA therapeutics would enable minimally invasive and lung targeted delivery for a host of pulmonary diseases. Development of lung targeted mRNA therapeutics has been limited by poor transfection efficiency and risk of vehicle-induced pathology. Here we report an inhalable polymer-based vehicle for delivery of therapeutic mRNAs to the lung. We optimized biodegradable poly(amine-co-ester) polyplexes for mRNA delivery using end group modifications and polyethylene glycol. Our polyplexes achieved high transfection of mRNA throughout the lung, particularly in epithelial and antigen-presenting cells. We applied this technology to develop a mucosal vaccine for SARS-CoV-2. Intranasal vaccination with spike protein mRNA polyplexes induced potent cellular and humoral adaptive immunity and protected K18-hACE2 mice from lethal viral challenge. One-sentence summary Inhaled polymer nanoparticles (NPs) achieve high mRNA expression in the lung and induce protective immunity against SARS-CoV-2.
Collapse
|
226
|
Banerjee S, Banerjee D, Singh A, Saharan VA. A Comprehensive Investigation Regarding the Differentiation of the Procurable COVID-19 Vaccines. AAPS PharmSciTech 2022; 23:95. [PMID: 35314902 PMCID: PMC8936379 DOI: 10.1208/s12249-022-02247-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 03/06/2022] [Indexed: 11/30/2022] Open
Abstract
COVID-19 caused by coronavirus SARS-CoV-2 became a serious threat to humankind for the past couple of years. The development of vaccine and its immediate application might be the only to escape from the grasp of this demoniac pandemic. Approximately 343 clinical trials on COVID-19 vaccines are ongoing currently, and almost all countries are motivating ongoing researches at warp speed for the development of vaccines against COVID-19. This review explores the progress in the development of the vaccines, their current status of ongoing clinical research, mechanisms, and regulatory approvals. Many pharmaceutical companies are already in the endgame for manufacturing various vaccines of which some are already being marketed across the globe, while others are yet to get approval for marketing. The primary aim of this review is to compare regulatory accepted vaccines in terms of their composition, doses, regulatory status, and efficacy. The study is conducted by grouping into approved and unapproved vaccines for marketing. Different routes of administration of vaccines along with the efficacy of the routes are also presented in the review. A wide range of database and clinical trial data is reviewed for sorting out the information on different vaccines. Unfortunately, many mutations (alpha, beta, gamma, delta, kappa, omicron etc.) of SARS-CoV-2 have attacked people in very short time, which is the great challenge for investigational vaccines. Moreover, some vaccines like Pfizer's BNT162, Oxford's ChAdOx1, Moderna's mRNA-1273, and Bharat Biotech's Covaxin have got regulatory approval in some countries for its distribution which may prove to stand tall against the pandemic.
Collapse
Affiliation(s)
- Surojit Banerjee
- School of Pharmaceutical Sciences and Technology, Sardar Bhagwan Singh University, Balawala, Dehradun, 248001, Uttarakhand, India.
| | - Debadri Banerjee
- School of Pharmaceutical Sciences and Technology, Sardar Bhagwan Singh University, Balawala, Dehradun, 248001, Uttarakhand, India
| | - Anupama Singh
- School of Pharmaceutical Sciences and Technology, Sardar Bhagwan Singh University, Balawala, Dehradun, 248001, Uttarakhand, India
| | - Vikas Anand Saharan
- School of Pharmaceutical Sciences and Technology, Sardar Bhagwan Singh University, Balawala, Dehradun, 248001, Uttarakhand, India
| |
Collapse
|
227
|
Brdovčak MC, Materljan J, Šustić M, Ravlić S, Ružić T, Lisnić B, Miklić K, Brizić I, Matešić MP, Lisnić VJ, Halassy B, Rončević D, Knežević Z, Štefan L, Bertoglio F, Schubert M, Čičin-Šain L, Markotić A, Jonjić S, Krmpotić A. ChAdOx1-S adenoviral vector vaccine applied intranasally elicits superior mucosal immunity compared to the intramuscular route of vaccination. Eur J Immunol 2022; 52:936-945. [PMID: 35304741 PMCID: PMC9087383 DOI: 10.1002/eji.202249823] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 03/11/2022] [Accepted: 03/14/2022] [Indexed: 11/09/2022]
Abstract
COVID-19 vaccines prevent severe forms of the disease, but do not warrant complete protection against breakthrough infections. This could be due to suboptimal mucosal immunity at the site of virus entry, given that all currently approved vaccines are administered via the intramuscular route. In this study we assessed humoral and cellular immune responses in BALB/c mice after intranasal and intramuscular immunization with adenoviral vector ChAdOx1-S expressing full-length Spike protein of SARS-CoV-2. We showed that both routes of vaccination induced a potent IgG antibody response, as well as robust neutralizing capacity, but intranasal vaccination elicited a superior IgA antibody titer in the sera and in the respiratory mucosa. Bronchoalveolar lavage from intranasally immunized mice efficiently neutralized SARS-CoV-2, which has not been the case in intramuscularly immunized group. Moreover, substantially higher percentages of epitope-specific CD8 T cells exhibiting a tissue resident phenotype were found in the lungs of intranasally immunized animals. Finally, both intranasal and intramuscular vaccination with ChAdOx1-S efficiently protected the mice after the challenge with recombinant herpesvirus expressing the Spike protein. Our results demonstrate that intranasal application of adenoviral vector ChAdOx1-S induces superior mucosal immunity and therefore could be a promising strategy for putting the COVID-19 pandemic under control. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Maja Cokarić Brdovčak
- Center for Proteomics, University of Rijeka, Faculty of Medicine, Rijeka, 51000, Croatia
| | - Jelena Materljan
- Center for Proteomics, University of Rijeka, Faculty of Medicine, Rijeka, 51000, Croatia.,Department of Histology and Embryology, University of Rijeka, Rijeka, 51000, Croatia
| | - Marko Šustić
- Center for Proteomics, University of Rijeka, Faculty of Medicine, Rijeka, 51000, Croatia
| | - Sanda Ravlić
- Centre for Research and Knowledge Transfer in Biotechnology, University of Zagreb, Zagreb, 10000, Croatia
| | - Tina Ružić
- Center for Proteomics, University of Rijeka, Faculty of Medicine, Rijeka, 51000, Croatia
| | - Berislav Lisnić
- Center for Proteomics, University of Rijeka, Faculty of Medicine, Rijeka, 51000, Croatia
| | - Karmela Miklić
- Center for Proteomics, University of Rijeka, Faculty of Medicine, Rijeka, 51000, Croatia
| | - Ilija Brizić
- Center for Proteomics, University of Rijeka, Faculty of Medicine, Rijeka, 51000, Croatia
| | | | - Vanda Juranić Lisnić
- Center for Proteomics, University of Rijeka, Faculty of Medicine, Rijeka, 51000, Croatia
| | - Beata Halassy
- Centre for Research and Knowledge Transfer in Biotechnology, University of Zagreb, Zagreb, 10000, Croatia
| | | | | | - Leo Štefan
- JGL d.d. Jadran Galenski Laboratorij, Rijeka, 51 000, Croatia
| | - Federico Bertoglio
- Institute for Biochemistry, Biotechnology and Bioinformatics, Technische Universität Braunschweig, Braunschweig, 38106, Germany
| | - Maren Schubert
- Institute for Biochemistry, Biotechnology and Bioinformatics, Technische Universität Braunschweig, Braunschweig, 38106, Germany
| | - Luka Čičin-Šain
- Helmholtz Center for Infection Research, Department of Viral Immunology, Braunschweig, 38124, Germany
| | - Alemka Markotić
- University Hospital for Infectious Diseases "Fran Mihaljević", Zagreb, 10000, Croatia
| | - Stipan Jonjić
- Center for Proteomics, University of Rijeka, Faculty of Medicine, Rijeka, 51000, Croatia
| | - Astrid Krmpotić
- Department of Histology and Embryology, University of Rijeka, Rijeka, 51000, Croatia
| |
Collapse
|
228
|
Pi-Estopiñan F, Pérez MT, Fraga A, Bergado G, Díaz GD, Orosa I, Díaz M, Solozábal JA, Rodríguez LM, Garcia-Rivera D, Macías C, Jerez Y, Casadesús AV, Fernández-Marrero B, Bermúdez E, Plasencia CA, Sánchez B, Hernández T. A cell-based ELISA as surrogate of virus neutralization assay for RBD SARS-CoV-2 specific antibodies. Vaccine 2022; 40:1958-1967. [PMID: 35193792 PMCID: PMC8856731 DOI: 10.1016/j.vaccine.2022.02.044] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 12/20/2021] [Accepted: 02/09/2022] [Indexed: 12/23/2022]
Abstract
SARS-CoV-2, the cause of the COVID-19 pandemic, has provoked a global crisis and death of millions of people. Several serological assays to determine the quality of the immune response against SARS-CoV-2 and the efficacy of vaccines have been developed, among them the gold standard conventional virus neutralization assays. However, these tests are time consuming, require biosafety level 3 (BSL3), and are low throughput and expensive. This has motivated the development of alternative methods, including molecular inhibition assays. Herein, we present a safe cell-based ELISA-virus neutralization test (cbE-VNT) as a surrogate for the conventional viral neutralization assays that detects the inhibition of SARS-CoV-2 RBD binding to ACE2-bearing cells independently of species. Our test shows a very good correlation with the conventional and molecular neutralization assays and achieves 100% specificity and 95% sensitivity. cbE-VNT is cost-effective, fast and enables a large-scale serological evaluation that can be performed in a BSL2 laboratory, allowing its use in pre-clinical and clinical investigations.
Collapse
Affiliation(s)
- Franciscary Pi-Estopiñan
- Immunology and Immunotherapy Division, Center of Molecular Immunology (CIM), P.O. Box 16040, 216 St., Havana, Cuba
| | - María Teresa Pérez
- National Laboratory of Civil Defense (NLCD), Jamaica Highway and National Highway, San José of Lajas, Mayabeque, Cuba
| | - Anitza Fraga
- National Laboratory of Civil Defense (NLCD), Jamaica Highway and National Highway, San José of Lajas, Mayabeque, Cuba
| | - Gretchen Bergado
- Immunology and Immunotherapy Division, Center of Molecular Immunology (CIM), P.O. Box 16040, 216 St., Havana, Cuba
| | - Geidy D Díaz
- Immunology and Immunotherapy Division, Center of Molecular Immunology (CIM), P.O. Box 16040, 216 St., Havana, Cuba
| | - Ivette Orosa
- Immunology and Immunotherapy Division, Center of Molecular Immunology (CIM), P.O. Box 16040, 216 St., Havana, Cuba
| | - Marianniz Díaz
- Immunology and Immunotherapy Division, Center of Molecular Immunology (CIM), P.O. Box 16040, 216 St., Havana, Cuba
| | - Joaquín Antonio Solozábal
- Quality Control Department, Center of Molecular Immunology (CIM), P.O. Box 16040, 216 St., Havana, Cuba
| | | | | | | | - Yanet Jerez
- Institute of Hematology and Immunology (IHI), Havana, Cuba
| | - Ana V Casadesús
- Immunology and Immunotherapy Division, Center of Molecular Immunology (CIM), P.O. Box 16040, 216 St., Havana, Cuba
| | - Briandy Fernández-Marrero
- Immunology and Immunotherapy Division, Center of Molecular Immunology (CIM), P.O. Box 16040, 216 St., Havana, Cuba
| | - Ernesto Bermúdez
- Immunology and Immunotherapy Division, Center of Molecular Immunology (CIM), P.O. Box 16040, 216 St., Havana, Cuba
| | - Claudia A Plasencia
- Immunology and Immunotherapy Division, Center of Molecular Immunology (CIM), P.O. Box 16040, 216 St., Havana, Cuba
| | - Belinda Sánchez
- Immunology and Immunotherapy Division, Center of Molecular Immunology (CIM), P.O. Box 16040, 216 St., Havana, Cuba
| | - Tays Hernández
- Immunology and Immunotherapy Division, Center of Molecular Immunology (CIM), P.O. Box 16040, 216 St., Havana, Cuba.
| |
Collapse
|
229
|
Wang H, Li P, Zhang M, Bi J, He Y, Li F, Yu R, Gao F, Kong W, Yu B, Chen L, Yu X. Vaccine with bacterium-like particles displaying HIV-1 gp120 trimer elicits specific mucosal responses and neutralizing antibodies in rhesus macaques. Microb Biotechnol 2022; 15:2022-2039. [PMID: 35290714 PMCID: PMC9249329 DOI: 10.1111/1751-7915.14022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 02/06/2022] [Indexed: 11/27/2022] Open
Abstract
Preclinical studies have shown that the induction of secretory IgA (sIgA) in mucosa and neutralizing antibodies (NAbs) in sera is essential for designing vaccines that can effectively block the transmission of HIV-1. We previously showed that a vaccine consisting of bacterium-like particles (BLPs) displaying Protan-gp120AE-MTQ (PAM) could induce mucosal immune responses through intranasal (IN) immunization in mice and NAbs through intramuscular (IM) immunization in guinea pigs. Here, we evaluated the ability of this vaccine BLP-PAM to elicit HIV-1-specific mucosal and systemic immune responses through IN and IM immunization combination strategies in rhesus macaques. First, the morphology, antigenicity and epitope accessibility of the vaccine were analysed by transmission electron microscopy, bio-layer interferometry and ELISA. In BLP-PAM-immunized macaques, HIV-1-specific sIgA were rapidly induced through IN immunization in situ and distant mucosal sites, although the immune responses are relatively weak. Furthermore, the HIV-1-specific IgG and IgA antibody levels in mucosal secretions were enhanced and maintained, while production of serum NAbs against heterologous HIV-1 tier 1 and 2 pseudoviruses was elicited after IM boost. Additionally, situ mucosal responses and systemic T cell immune responses were improved by rAd2-gp120AE boost immunization via the IN and IM routes. These results suggested that BLP-based delivery in combination with the IN and IM immunization approach represents a potential vaccine strategy against HIV-1.
Collapse
Affiliation(s)
- Huaiyu Wang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, 130012, China
| | - Pingchao Li
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Mo Zhang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, 130012, China
| | - Jinpeng Bi
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, 130012, China
| | - Yizi He
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences, Guangzhou, 510530, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Fangshen Li
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, 130012, China
| | - Rongzhen Yu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, 130012, China
| | - Feng Gao
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, 130012, China.,Department of Medicine, Duke University Medical Center, Durham, NC, 27710, USA
| | - Wei Kong
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, 130012, China.,Key Laboratory for Molecular Enzymology and Engineering, The Ministry of Education, School of Life Sciences, Jilin University, Changchun, 130012, China
| | - Bin Yu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, 130012, China
| | - Ling Chen
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences, Guangzhou, 510530, China.,University of Chinese Academy of Sciences, Beijing, 100049, China.,The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510060, China
| | - Xianghui Yu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, 130012, China.,Key Laboratory for Molecular Enzymology and Engineering, The Ministry of Education, School of Life Sciences, Jilin University, Changchun, 130012, China
| |
Collapse
|
230
|
Intranasal administration of BReC-CoV-2 COVID-19 vaccine protects K18-hACE2 mice against lethal SARS-CoV-2 challenge. NPJ Vaccines 2022; 7:36. [PMID: 35288576 PMCID: PMC8921182 DOI: 10.1038/s41541-022-00451-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 01/20/2022] [Indexed: 12/21/2022] Open
Abstract
SARS-CoV-2 is a viral respiratory pathogen responsible for the current global pandemic and the disease that causes COVID-19. All current WHO approved COVID-19 vaccines are administered through the muscular route. We have developed a prototype two-dose vaccine (BReC-CoV-2) by combining the Receptor Binding Domain (RBD) antigen, via conjugation to Diphtheria toxoid (EcoCRM®). The vaccine is adjuvanted with Bacterial Enzymatic Combinatorial Chemistry (BECC), BECC470. Intranasal (IN) administration of BreC-CoV-2 in K18-hACE2 mice induced a strong systemic and localized immune response in the respiratory tissues which provided protection against the Washington strain of SARS-CoV-2. Protection provided after IN administration of BReC-CoV-2 was associated with decreased viral RNA copies in the lung, robust RBD IgA titers in the lung and nasal wash, and induction of broadly neutralizing antibodies in the serum. We also observed that BReC-CoV-2 vaccination administered using an intramuscular (IM) prime and IN boost protected mice from a lethal challenge dose of the Delta variant of SARS-CoV-2. IN administration of BReC-CoV-2 provided better protection than IM only administration to mice against lethal challenge dose of SARS-CoV-2. These data suggest that the IN route of vaccination induces localized immune responses that can better protect against SARS-CoV-2 than the IM route in the upper respiratory tract.
Collapse
|
231
|
Zhang B, Jin Y, Zhang L, Wang H, Wang X. Pentamidine Ninety Years on: the Development and Applications of Pentamidine and its Analogs. Curr Med Chem 2022; 29:4602-4609. [PMID: 35289252 DOI: 10.2174/0929867329666220314121446] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 12/25/2021] [Accepted: 01/07/2022] [Indexed: 11/22/2022]
Abstract
Pentamidine, an FDA-approved human drug for many protozoal infections, was initially synthesized in the late 1930s and first reported to be curative for parasitosis in the 1940s. After ninety years of sometimes quiet growth, pentamidine and its derivatives have gone far beyond antibacterial agents, including but not limited to the ligands of DNA minor groove, modulators of PPIs (protein-protein interactions) of the transmembrane domain 5 of lateral membrane protein 1, and the blockers of the SARS-CoV-2 3a channel. This mini review highlights the development and applications of pentamidine and its analogs, aiming to provide insights for further developing pentamidine derivatives in the following decades.
Collapse
Affiliation(s)
- Bo Zhang
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, China;
- Department of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui, China
| | - Yushan Jin
- Department of Immunology and Department of Cell & Systems Biology, University of Toronto, Toronto, M5S 3G3, Canada
| | - Lei Zhang
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, China
| | - Hongshuang Wang
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, China
| | - Xiaohui Wang
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, China;
- Department of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui, China
| |
Collapse
|
232
|
O’Donnell KL, Gourdine T, Fletcher P, Shifflett K, Furuyama W, Clancy CS, Marzi A. VSV-Based Vaccines Reduce Virus Shedding and Viral Load in Hamsters Infected with SARS-CoV-2 Variants of Concern. Vaccines (Basel) 2022; 10:435. [PMID: 35335067 PMCID: PMC8951568 DOI: 10.3390/vaccines10030435] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 03/07/2022] [Accepted: 03/11/2022] [Indexed: 12/11/2022] Open
Abstract
The continued progression of the COVID-19 pandemic can partly be attributed to the ability of SARS-CoV-2 to mutate and introduce new viral variants. Some of these variants with the potential to spread quickly and conquer the globe are termed variants of concern (VOC). The existing vaccines implemented on a global scale are based on the ancestral strain, which has resulted in increased numbers of breakthrough infections as these VOC have emerged. It is imperative to show protection against VOC infection with newly developed vaccines. Previously, we evaluated two vesicular stomatitis virus (VSV)-based vaccines expressing the SARS-CoV-2 spike protein alone (VSV-SARS2) or in combination with the Ebola virus glycoprotein (VSV-SARS2-EBOV) and demonstrated their fast-acting potential. Here, we prolonged the time to challenge; we vaccinated hamsters intranasally (IN) or intramuscularly 28 days prior to infection with three SARS-CoV-2 VOC-the Alpha, Beta, and Delta variants. IN vaccination with either the VSV-SARS2 or VSV-SARS2-EBOV resulted in the highest protective efficacy as demonstrated by decreased virus shedding and lung viral load of vaccinated hamsters. Histopathologic analysis of the lungs revealed the least amount of lung damage in the IN-vaccinated animals regardless of the challenge virus. This data demonstrates the ability of a VSV-based vaccine to not only protect from disease caused by SARS-CoV-2 VOC but also reduce viral shedding.
Collapse
Affiliation(s)
- Kyle L. O’Donnell
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA (T.G.); (P.F.); (K.S.); (W.F.)
| | - Tylisha Gourdine
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA (T.G.); (P.F.); (K.S.); (W.F.)
| | - Paige Fletcher
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA (T.G.); (P.F.); (K.S.); (W.F.)
| | - Kyle Shifflett
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA (T.G.); (P.F.); (K.S.); (W.F.)
| | - Wakako Furuyama
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA (T.G.); (P.F.); (K.S.); (W.F.)
| | - Chad S. Clancy
- Rocky Mountain Veterinary Branch, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA;
| | - Andrea Marzi
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA (T.G.); (P.F.); (K.S.); (W.F.)
| |
Collapse
|
233
|
STxB as an Antigen Delivery Tool for Mucosal Vaccination. Toxins (Basel) 2022; 14:toxins14030202. [PMID: 35324699 PMCID: PMC8948715 DOI: 10.3390/toxins14030202] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 03/03/2022] [Accepted: 03/07/2022] [Indexed: 12/31/2022] Open
Abstract
Immunotherapy against cancer and infectious disease holds the promise of high efficacy with minor side effects. Mucosal vaccines to protect against tumors or infections disease agents that affect the upper airways or the lung are still lacking, however. One mucosal vaccine candidate is the B-subunit of Shiga toxin, STxB. In this review, we compare STxB to other immunotherapy vectors. STxB is a non-toxic protein that binds to a glycosylated lipid, termed globotriaosylceramide (Gb3), which is preferentially expressed by dendritic cells. We review the use of STxB for the cross-presentation of tumor or viral antigens in a MHC class I-restricted manner to induce humoral immunity against these antigens in addition to polyfunctional and persistent CD4+ and CD8+ T lymphocytes capable of protecting against viral infection or tumor growth. Other literature will be summarized that documents a powerful induction of mucosal IgA and resident memory CD8+ T cells against mucosal tumors specifically when STxB-antigen conjugates are administered via the nasal route. It will also be pointed out how STxB-based vaccines have been shown in preclinical cancer models to synergize with other therapeutic modalities (immune checkpoint inhibitors, anti-angiogenic therapy, radiotherapy). Finally, we will discuss how molecular aspects such as low immunogenicity, cross-species conservation of Gb3 expression, and lack of toxicity contribute to the competitive positioning of STxB among the different DC targeting approaches. STxB thereby appears as an original and innovative tool for the development of mucosal vaccines in infectious diseases and cancer.
Collapse
|
234
|
Huang J, Ding Y, Yao J, Zhang M, Zhang Y, Xie Z, Zuo J. Nasal Nanovaccines for SARS-CoV-2 to Address COVID-19. Vaccines (Basel) 2022; 10:vaccines10030405. [PMID: 35335037 PMCID: PMC8952855 DOI: 10.3390/vaccines10030405] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 03/04/2022] [Accepted: 03/04/2022] [Indexed: 02/06/2023] Open
Abstract
COVID-19 is still prevalent around the globe. Although some SARS-CoV-2 vaccines have been distributed to the population, the shortcomings of vaccines and the continuous emergence of SARS-CoV-2 mutant virus strains are a cause for concern. Thus, it is vital to continue to improve vaccines and vaccine delivery methods. One option is nasal vaccination, which is more convenient than injections and does not require a syringe. Additionally, stronger mucosal immunity is produced under nasal vaccination. The easy accessibility of the intranasal route is more advantageous than injection in the context of the COVID-19 pandemic. Nanoparticles have been proven to be suitable delivery vehicles and adjuvants, and different NPs have different advantages. The shortcomings of the SARS-CoV-2 vaccine may be compensated by selecting or modifying different nanoparticles. It travels along the digestive tract to the intestine, where it is presented by GALT, tissue-resident immune cells, and gastrointestinal lymph nodes. Nasal nanovaccines are easy to use, safe, multifunctional, and can be distributed quickly, demonstrating strong prospects as a vaccination method for SARS-CoV-2, SARS-CoV-2 variants, or SARS-CoV-n.
Collapse
Affiliation(s)
- Jialu Huang
- The Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang 421001, China; (J.H.); (M.Z.); (Y.Z.); (Z.X.)
| | - Yubo Ding
- Nanhua Hospital Affiliated to University of South China, Hengyang Medical School, University of South China, Hengyang 421002, China; (Y.D.); (J.Y.)
| | - Jingwei Yao
- Nanhua Hospital Affiliated to University of South China, Hengyang Medical School, University of South China, Hengyang 421002, China; (Y.D.); (J.Y.)
| | - Minghui Zhang
- The Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang 421001, China; (J.H.); (M.Z.); (Y.Z.); (Z.X.)
| | - Yu Zhang
- The Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang 421001, China; (J.H.); (M.Z.); (Y.Z.); (Z.X.)
| | - Zhuoyi Xie
- The Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang 421001, China; (J.H.); (M.Z.); (Y.Z.); (Z.X.)
| | - Jianhong Zuo
- The Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang 421001, China; (J.H.); (M.Z.); (Y.Z.); (Z.X.)
- Nanhua Hospital Affiliated to University of South China, Hengyang Medical School, University of South China, Hengyang 421002, China; (Y.D.); (J.Y.)
- The Third Affiliated Hospital of University of South China, Hengyang Medical School, University of South China, Hengyang 421900, China
- Correspondence: ; Tel.: +86-7345-675219
| |
Collapse
|
235
|
Feng F, Wen Z, Chen J, Yuan Y, Wang C, Sun C. Strategies to Develop a Mucosa-Targeting Vaccine against Emerging Infectious Diseases. Viruses 2022; 14:v14030520. [PMID: 35336927 PMCID: PMC8952777 DOI: 10.3390/v14030520] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 02/27/2022] [Accepted: 03/01/2022] [Indexed: 02/06/2023] Open
Abstract
Numerous pathogenic microbes, including viruses, bacteria, and fungi, usually infect the host through the mucosal surfaces of the respiratory tract, gastrointestinal tract, and reproductive tract. The mucosa is well known to provide the first line of host defense against pathogen entry by physical, chemical, biological, and immunological barriers, and therefore, mucosa-targeting vaccination is emerging as a promising strategy for conferring superior protection. However, there are still many challenges to be solved to develop an effective mucosal vaccine, such as poor adhesion to the mucosal surface, insufficient uptake to break through the mucus, and the difficulty in avoiding strong degradation through the gastrointestinal tract. Recently, increasing efforts to overcome these issues have been made, and we herein summarize the latest findings on these strategies to develop mucosa-targeting vaccines, including a novel needle-free mucosa-targeting route, the development of mucosa-targeting vectors, the administration of mucosal adjuvants, encapsulating vaccines into nanoparticle formulations, and antigen design to conjugate with mucosa-targeting ligands. Our work will highlight the importance of further developing mucosal vaccine technology to combat the frequent outbreaks of infectious diseases.
Collapse
Affiliation(s)
- Fengling Feng
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China; (F.F.); (Z.W.); (J.C.); (Y.Y.); (C.W.)
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou 510080, China
| | - Ziyu Wen
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China; (F.F.); (Z.W.); (J.C.); (Y.Y.); (C.W.)
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou 510080, China
| | - Jiaoshan Chen
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China; (F.F.); (Z.W.); (J.C.); (Y.Y.); (C.W.)
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou 510080, China
| | - Yue Yuan
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China; (F.F.); (Z.W.); (J.C.); (Y.Y.); (C.W.)
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou 510080, China
| | - Congcong Wang
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China; (F.F.); (Z.W.); (J.C.); (Y.Y.); (C.W.)
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou 510080, China
| | - Caijun Sun
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China; (F.F.); (Z.W.); (J.C.); (Y.Y.); (C.W.)
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou 510080, China
- Correspondence:
| |
Collapse
|
236
|
Afkhami S, D'Agostino MR, Zhang A, Stacey HD, Marzok A, Kang A, Singh R, Bavananthasivam J, Ye G, Luo X, Wang F, Ang JC, Zganiacz A, Sankar U, Kazhdan N, Koenig JFE, Phelps A, Gameiro SF, Tang S, Jordana M, Wan Y, Mossman KL, Jeyanathan M, Gillgrass A, Medina MFC, Smaill F, Lichty BD, Miller MS, Xing Z. Respiratory mucosal delivery of next-generation COVID-19 vaccine provides robust protection against both ancestral and variant strains of SARS-CoV-2. Cell 2022; 185:896-915.e19. [PMID: 35180381 PMCID: PMC8825346 DOI: 10.1016/j.cell.2022.02.005] [Citation(s) in RCA: 172] [Impact Index Per Article: 57.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 12/16/2021] [Accepted: 02/02/2022] [Indexed: 12/28/2022]
Abstract
The emerging SARS-CoV-2 variants of concern (VOCs) threaten the effectiveness of current COVID-19 vaccines administered intramuscularly and designed to only target the spike protein. There is a pressing need to develop next-generation vaccine strategies for broader and long-lasting protection. Using adenoviral vectors (Ad) of human and chimpanzee origin, we evaluated Ad-vectored trivalent COVID-19 vaccines expressing spike-1, nucleocapsid, and RdRp antigens in murine models. We show that single-dose intranasal immunization, particularly with chimpanzee Ad-vectored vaccine, is superior to intramuscular immunization in induction of the tripartite protective immunity consisting of local and systemic antibody responses, mucosal tissue-resident memory T cells and mucosal trained innate immunity. We further show that intranasal immunization provides protection against both the ancestral SARS-CoV-2 and two VOC, B.1.1.7 and B.1.351. Our findings indicate that respiratory mucosal delivery of Ad-vectored multivalent vaccine represents an effective next-generation COVID-19 vaccine strategy to induce all-around mucosal immunity against current and future VOC.
Collapse
Affiliation(s)
- Sam Afkhami
- McMaster Immunology Research Centre, M.G. DeGroote Institute for Infectious Disease Research, Department of Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Michael R D'Agostino
- McMaster Immunology Research Centre, M. G. DeGroote Institute for Infectious Disease Research, Department of Biochemistry & Biomedical Sciences, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Ali Zhang
- McMaster Immunology Research Centre, M. G. DeGroote Institute for Infectious Disease Research, Department of Biochemistry & Biomedical Sciences, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Hannah D Stacey
- McMaster Immunology Research Centre, M. G. DeGroote Institute for Infectious Disease Research, Department of Biochemistry & Biomedical Sciences, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Art Marzok
- McMaster Immunology Research Centre, M. G. DeGroote Institute for Infectious Disease Research, Department of Biochemistry & Biomedical Sciences, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Alisha Kang
- McMaster Immunology Research Centre, M.G. DeGroote Institute for Infectious Disease Research, Department of Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Ramandeep Singh
- McMaster Immunology Research Centre, M.G. DeGroote Institute for Infectious Disease Research, Department of Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Jegarubee Bavananthasivam
- McMaster Immunology Research Centre, M.G. DeGroote Institute for Infectious Disease Research, Department of Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Gluke Ye
- McMaster Immunology Research Centre, M.G. DeGroote Institute for Infectious Disease Research, Department of Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Xiangqian Luo
- McMaster Immunology Research Centre, M.G. DeGroote Institute for Infectious Disease Research, Department of Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada; Department of Pediatric Otolaryngology, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Fuan Wang
- McMaster Immunology Research Centre, M.G. DeGroote Institute for Infectious Disease Research, Department of Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Jann C Ang
- McMaster Immunology Research Centre, M. G. DeGroote Institute for Infectious Disease Research, Department of Biochemistry & Biomedical Sciences, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Anna Zganiacz
- McMaster Immunology Research Centre, M.G. DeGroote Institute for Infectious Disease Research, Department of Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Uma Sankar
- McMaster Immunology Research Centre, M.G. DeGroote Institute for Infectious Disease Research, Department of Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Natallia Kazhdan
- McMaster Immunology Research Centre, M.G. DeGroote Institute for Infectious Disease Research, Department of Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Joshua F E Koenig
- McMaster Immunology Research Centre, M.G. DeGroote Institute for Infectious Disease Research, Department of Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Allyssa Phelps
- McMaster Immunology Research Centre, M.G. DeGroote Institute for Infectious Disease Research, Department of Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Steven F Gameiro
- McMaster Immunology Research Centre, M.G. DeGroote Institute for Infectious Disease Research, Department of Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Shangguo Tang
- Department of Pathology and Molecular Medicine, M.G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Manel Jordana
- McMaster Immunology Research Centre, M.G. DeGroote Institute for Infectious Disease Research, Department of Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Yonghong Wan
- McMaster Immunology Research Centre, M.G. DeGroote Institute for Infectious Disease Research, Department of Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Karen L Mossman
- McMaster Immunology Research Centre, M.G. DeGroote Institute for Infectious Disease Research, Department of Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Mangalakumari Jeyanathan
- McMaster Immunology Research Centre, M.G. DeGroote Institute for Infectious Disease Research, Department of Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Amy Gillgrass
- McMaster Immunology Research Centre, M.G. DeGroote Institute for Infectious Disease Research, Department of Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Maria Fe C Medina
- McMaster Immunology Research Centre, M.G. DeGroote Institute for Infectious Disease Research, Department of Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Fiona Smaill
- Department of Pathology and Molecular Medicine, M.G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Brian D Lichty
- McMaster Immunology Research Centre, M.G. DeGroote Institute for Infectious Disease Research, Department of Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada.
| | - Matthew S Miller
- McMaster Immunology Research Centre, M. G. DeGroote Institute for Infectious Disease Research, Department of Biochemistry & Biomedical Sciences, McMaster University, Hamilton, ON L8S 4K1, Canada.
| | - Zhou Xing
- McMaster Immunology Research Centre, M.G. DeGroote Institute for Infectious Disease Research, Department of Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada.
| |
Collapse
|
237
|
Mabrouk MT, Huang W, Martinez‐Sobrido L, Lovell JF. Advanced Materials for SARS-CoV-2 Vaccines. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2107781. [PMID: 34894000 PMCID: PMC8957524 DOI: 10.1002/adma.202107781] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 11/28/2021] [Indexed: 05/09/2023]
Abstract
The ongoing coronavirus disease 2019 (COVID-19) pandemic, caused by severe acute respiratory coronavirus 2 (SARS-CoV-2), has killed untold millions worldwide and has hurtled vaccines into the spotlight as a go-to approach to mitigate it. Advances in virology, genomics, structural biology, and vaccine technologies have enabled a rapid and unprecedented rollout of COVID-19 vaccines, although much of the developing world remains unvaccinated. Several new vaccine platforms have been developed or deployed against SARS-CoV-2, with most targeting the large viral Spike immunogen. Those that safely induce strong and durable antibody responses at low dosages are advantageous, as well are those that can be rapidly produced at a large scale. Virtually all COVID-19 vaccines and adjuvants possess nanoscale or microscale dimensions and represent diverse and unique biomaterials. Viral vector vaccine platforms, lipid nanoparticle mRNA vaccines and multimeric display technologies for subunit vaccines have received much attention. Nanoscale vaccine adjuvants have also been used in combination with other vaccines. To deal with the ongoing pandemic, and to be ready for potential future ones, advanced vaccine technologies will continue to be developed in the near future. Herein, the recent use of advanced materials used for developing COVID-19 vaccines is summarized.
Collapse
Affiliation(s)
- Moustafa T. Mabrouk
- Department of Biomedical EngineeringUniversity at BuffaloState University of New YorkBuffaloNY14260USA
| | - Wei‐Chiao Huang
- Department of Biomedical EngineeringUniversity at BuffaloState University of New YorkBuffaloNY14260USA
| | - Luis Martinez‐Sobrido
- Division of Disease Intervention and PreventionTexas Biomedical Research InstituteSan AntonioTX78227USA
| | - Jonathan F. Lovell
- Department of Biomedical EngineeringUniversity at BuffaloState University of New YorkBuffaloNY14260USA
| |
Collapse
|
238
|
Nanoparticle and virus-like particle vaccine approaches against SARS-CoV-2. J Microbiol 2022; 60:335-346. [PMID: 35089583 PMCID: PMC8795728 DOI: 10.1007/s12275-022-1608-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 12/16/2021] [Accepted: 12/16/2021] [Indexed: 02/06/2023]
Abstract
The global spread of coronavirus disease 2019 caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection has provoked an urgent need for prophylactic measures. Several innovative vaccine platforms have been introduced and billions of vaccine doses have been administered worldwide. To enable the creation of safer and more effective vaccines, additional platforms are under development. These include the use of nanoparticle (NP) and virus-like particle (VLP) technology. NP vaccines utilize self-assembling scaffold structures designed to load the entire spike protein or receptor-binding domain of SARS-CoV-2 in a trimeric configuration. In contrast, VLP vaccines are genetically modified recombinant viruses that are considered safe, as they are generally replication-defective. Furthermore, VLPs have indigenous immunogenic potential due to their microbial origin. Importantly, NP and VLP vaccines have shown stronger immunogenicity with greater protection by mimicking the physicochemical characteristics of SARS-CoV-2. The study of NP- and VLP-based coronavirus vaccines will help ensure the development of rapid-response technology against SARS-CoV-2 variants and future coronavirus pandemics.
Collapse
|
239
|
Ivanova N, Sotirova Y, Gavrailov G, Nikolova K, Andonova V. Advances in the Prophylaxis of Respiratory Infections by the Nasal and the Oromucosal Route: Relevance to the Fight with the SARS-CoV-2 Pandemic. Pharmaceutics 2022; 14:530. [PMID: 35335905 PMCID: PMC8953301 DOI: 10.3390/pharmaceutics14030530] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 02/19/2022] [Accepted: 02/23/2022] [Indexed: 11/22/2022] Open
Abstract
In this time of COVID-19 pandemic, the strategies for prevention of the infection are a primary concern. Looking more globally on the subject and acknowledging the high degree of misuse of protective face masks from the population, we focused this review on alternative pharmaceutical developments eligible for self-defense against respiratory infections. In particular, the attention herein is directed to the nasal and oromucosal formulations intended to boost the local immunity, neutralize or mechanically "trap" the pathogens at the site of entry (nose or mouth). The current work presents a critical review of the contemporary methods of immune- and chemoprophylaxis and their suitability and applicability in topical mucosal dosage forms for SARS-CoV-2 prophylaxis.
Collapse
Affiliation(s)
- Nadezhda Ivanova
- Department of Pharmaceutical Technologies, Faculty of Pharmacy, Medical University of Varna, 55 Marin Drinov Str., 9000 Varna, Bulgaria; (Y.S.); (G.G.); (V.A.)
| | - Yoana Sotirova
- Department of Pharmaceutical Technologies, Faculty of Pharmacy, Medical University of Varna, 55 Marin Drinov Str., 9000 Varna, Bulgaria; (Y.S.); (G.G.); (V.A.)
| | - Georgi Gavrailov
- Department of Pharmaceutical Technologies, Faculty of Pharmacy, Medical University of Varna, 55 Marin Drinov Str., 9000 Varna, Bulgaria; (Y.S.); (G.G.); (V.A.)
| | - Krastena Nikolova
- Department of Physics and Biophysics, Faculty of Pharmacy, Medical University of Varna, 55 Marin Drinov Str., 9000 Varna, Bulgaria;
| | - Velichka Andonova
- Department of Pharmaceutical Technologies, Faculty of Pharmacy, Medical University of Varna, 55 Marin Drinov Str., 9000 Varna, Bulgaria; (Y.S.); (G.G.); (V.A.)
| |
Collapse
|
240
|
Bellier B, Saura A, Luján LA, Molina CR, Luján HD, Klatzmann D. A Thermostable Oral SARS-CoV-2 Vaccine Induces Mucosal and Protective Immunity. Front Immunol 2022; 13:837443. [PMID: 35281065 PMCID: PMC8913903 DOI: 10.3389/fimmu.2022.837443] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 02/07/2022] [Indexed: 12/04/2022] Open
Abstract
An ideal protective vaccine against SARS-CoV-2 should not only be effective in preventing disease, but also in preventing virus transmission. It should also be well accepted by the population and have a simple logistic chain. To fulfill these criteria, we developed a thermostable, orally administered vaccine that can induce a robust mucosal neutralizing immune response. We used our platform based on retrovirus-derived enveloped virus-like particles (eVLPs) harnessed with variable surface proteins (VSPs) from the intestinal parasite Giardia lamblia, affording them resistance to degradation and the triggering of robust mucosal cellular and antibody immune responses after oral administration. We made eVLPs expressing various forms of the SARS-CoV-2 Spike protein (S), with or without membrane protein (M) expression. We found that prime-boost administration of VSP-decorated eVLPs expressing a pre-fusion stabilized form of S and M triggers robust mucosal responses against SARS-CoV-2 in mice and hamsters, which translate into complete protection from a viral challenge. Moreover, they dramatically boosted the IgA mucosal response of intramuscularly injected vaccines. We conclude that our thermostable orally administered eVLP vaccine could be a valuable addition to the current arsenal against SARS-CoV-2, in a stand-alone prime-boost vaccination strategy or as a boost for existing vaccines.
Collapse
Affiliation(s)
- Bertrand Bellier
- Sorbonne Université, INSERM, UMRS 959, Immunology-Immunopathology-Immunotherapy, i3, Paris, France
| | - Alicia Saura
- Centro de Investigación y Desarrollo en Inmunología y Enfermedades Infecciosas (CIDIE), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)/Universidad Católica de Córdoba (UCC), Córdoba, Argentina
- Facultad de Ciencias de la Salud, Universidad Católica de Córdoba (UCC), Córdoba, Argentina
| | - Lucas A. Luján
- Centro de Investigación y Desarrollo en Inmunología y Enfermedades Infecciosas (CIDIE), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)/Universidad Católica de Córdoba (UCC), Córdoba, Argentina
- Facultad de Ciencias de la Salud, Universidad Católica de Córdoba (UCC), Córdoba, Argentina
| | - Cecilia R. Molina
- Centro de Investigación y Desarrollo en Inmunología y Enfermedades Infecciosas (CIDIE), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)/Universidad Católica de Córdoba (UCC), Córdoba, Argentina
- Facultad de Ciencias de la Salud, Universidad Católica de Córdoba (UCC), Córdoba, Argentina
| | - Hugo D. Luján
- Centro de Investigación y Desarrollo en Inmunología y Enfermedades Infecciosas (CIDIE), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)/Universidad Católica de Córdoba (UCC), Córdoba, Argentina
- Facultad de Ciencias de la Salud, Universidad Católica de Córdoba (UCC), Córdoba, Argentina
- *Correspondence: Hugo D. Luján, ; David Klatzmann,
| | - David Klatzmann
- Sorbonne Université, INSERM, UMRS 959, Immunology-Immunopathology-Immunotherapy, i3, Paris, France
- AP-HP, Hôpital Pitié-Salpêtrière, Clinical Investigation Center for Biotherapies (CIC-BTi) and Immunology-Inflammation-Infectiology and Dermatology Department (3iD), Paris, France
- *Correspondence: Hugo D. Luján, ; David Klatzmann,
| |
Collapse
|
241
|
Ren C, Gao Y, Zhang C, Zhou C, Hong Y, Qu M, Zhao Z, Du Y, Yang L, Liu B, Wang S, Han M, Shen Y, Liu Y. Respiratory Mucosal Immunity: Kinetics of Secretory Immunoglobulin A in Sputum and Throat Swabs From COVID-19 Patients and Vaccine Recipients. Front Microbiol 2022; 13:782421. [PMID: 35283823 PMCID: PMC8914317 DOI: 10.3389/fmicb.2022.782421] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 01/03/2022] [Indexed: 11/13/2022] Open
Abstract
While IgM and IgG response to SARS-CoV-2 has been extensively studied, relatively little is known about secretory IgA (sIgA) response in respiratory mucosa. Here we report IgA response to the SARS-CoV-2 in sputum, throat swabs, and serum with nucleocapsid protein (NP) enzyme-linked immunosorbent assays (ELISA) in a cohort of 28 COVID-19 patients and 55 vaccine recipients. The assays showed sIgA in respiratory mucosa could be detected on the first day after illness onset (AIO), and the median conversion time for sIgA in sputum, throat swabs, and serum was 3, 4, and 10 days, respectively. The positive rates of sIgA first week AIO were 100% (24/28) and 85.7% (24/28) in sputum and throat swabs, respectively, and were both 100% during the mid-onset (2–3 weeks AIO). During the recovery period, sIgA positive rates in sputum and throat swabs gradually decreased from 60.7% (17/28) and 57.1% (16/28) 1 month AIO and the sIgA antibodies were all undetectable 6 months AIO. However, serum IgA positive rate was still 100% at 4 months and 53.6% (15/28) at 6 months. Throat swabs obtained from volunteers who received inactivated SARS-CoV-2 vaccines by intramuscular delivery all showed negative results in IgA ELISA. These findings will likely improve our understanding of respiratory mucosal immunity of this emerging disease and help in containing the pandemic and developing vaccines.
Collapse
Affiliation(s)
- Cuiping Ren
- Department of Microbiology and Parasitology, Anhui Medical University, Hefei, China
- Anhui Provincial Laboratory of Pathogen Biology, Anhui Medical University, Hefei, China
- Anhui Key Laboratory of Zoonosis of High Institution, Anhui Medical University, Hefei, China
- Laboratory of Tropical and Parasitic Diseases Control, Anhui Medical University, Hefei, China
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Yong Gao
- Department of Clinical Laboratory, The Second People’s Hospital of Fuyang, Fuyang, China
| | - Cong Zhang
- Department of Microbiology and Parasitology, Anhui Medical University, Hefei, China
- Anhui Provincial Laboratory of Pathogen Biology, Anhui Medical University, Hefei, China
- Anhui Key Laboratory of Zoonosis of High Institution, Anhui Medical University, Hefei, China
- Laboratory of Tropical and Parasitic Diseases Control, Anhui Medical University, Hefei, China
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Chang Zhou
- Department of Microbiology and Parasitology, Anhui Medical University, Hefei, China
- Anhui Provincial Laboratory of Pathogen Biology, Anhui Medical University, Hefei, China
- Anhui Key Laboratory of Zoonosis of High Institution, Anhui Medical University, Hefei, China
- Laboratory of Tropical and Parasitic Diseases Control, Anhui Medical University, Hefei, China
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Ying Hong
- Maanshan Center for Disease Control and Prevention, Maanshan, China
| | - Mingsheng Qu
- Department of Microbiology and Parasitology, Anhui Medical University, Hefei, China
- Anhui Provincial Laboratory of Pathogen Biology, Anhui Medical University, Hefei, China
- Anhui Key Laboratory of Zoonosis of High Institution, Anhui Medical University, Hefei, China
- Laboratory of Tropical and Parasitic Diseases Control, Anhui Medical University, Hefei, China
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Zhirong Zhao
- Maanshan Center for Disease Control and Prevention, Maanshan, China
| | - Yinan Du
- Department of Microbiology and Parasitology, Anhui Medical University, Hefei, China
- Anhui Provincial Laboratory of Pathogen Biology, Anhui Medical University, Hefei, China
- Anhui Key Laboratory of Zoonosis of High Institution, Anhui Medical University, Hefei, China
- Laboratory of Tropical and Parasitic Diseases Control, Anhui Medical University, Hefei, China
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Li Yang
- Department of Microbiology and Parasitology, Anhui Medical University, Hefei, China
- Anhui Provincial Laboratory of Pathogen Biology, Anhui Medical University, Hefei, China
- Anhui Key Laboratory of Zoonosis of High Institution, Anhui Medical University, Hefei, China
- Laboratory of Tropical and Parasitic Diseases Control, Anhui Medical University, Hefei, China
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Boyu Liu
- Department of Microbiology and Parasitology, Anhui Medical University, Hefei, China
- Anhui Provincial Laboratory of Pathogen Biology, Anhui Medical University, Hefei, China
- Anhui Key Laboratory of Zoonosis of High Institution, Anhui Medical University, Hefei, China
- Laboratory of Tropical and Parasitic Diseases Control, Anhui Medical University, Hefei, China
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Siying Wang
- Department of Microbiology and Parasitology, Anhui Medical University, Hefei, China
- Anhui Provincial Laboratory of Pathogen Biology, Anhui Medical University, Hefei, China
- Anhui Key Laboratory of Zoonosis of High Institution, Anhui Medical University, Hefei, China
- Laboratory of Tropical and Parasitic Diseases Control, Anhui Medical University, Hefei, China
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Mingfeng Han
- Department of Clinical Laboratory, The Second People’s Hospital of Fuyang, Fuyang, China
- *Correspondence: Mingfeng Han,
| | - Yuxian Shen
- Department of Microbiology and Parasitology, Anhui Medical University, Hefei, China
- Anhui Provincial Laboratory of Pathogen Biology, Anhui Medical University, Hefei, China
- Anhui Key Laboratory of Zoonosis of High Institution, Anhui Medical University, Hefei, China
- Laboratory of Tropical and Parasitic Diseases Control, Anhui Medical University, Hefei, China
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
- Yuxian Shen,
| | - Yan Liu
- Department of Microbiology and Parasitology, Anhui Medical University, Hefei, China
- Anhui Provincial Laboratory of Pathogen Biology, Anhui Medical University, Hefei, China
- Anhui Key Laboratory of Zoonosis of High Institution, Anhui Medical University, Hefei, China
- Laboratory of Tropical and Parasitic Diseases Control, Anhui Medical University, Hefei, China
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
- Yan Liu,
| |
Collapse
|
242
|
Furuyama W, Shifflett K, Pinski AN, Griffin AJ, Feldmann F, Okumura A, Gourdine T, Jankeel A, Lovaglio J, Hanley PW, Thomas T, Clancy CS, Messaoudi I, O’Donnell KL, Marzi A. Rapid Protection from COVID-19 in Nonhuman Primates Vaccinated Intramuscularly but Not Intranasally with a Single Dose of a Vesicular Stomatitis Virus-Based Vaccine. mBio 2022; 13:e0337921. [PMID: 35012339 PMCID: PMC8749411 DOI: 10.1128/mbio.03379-21] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 11/23/2021] [Indexed: 01/08/2023] Open
Abstract
The ongoing pandemic of coronavirus (CoV) disease 2019 (COVID-19) continues to exert a significant burden on health care systems worldwide. With limited treatments available, vaccination remains an effective strategy to counter transmission of severe acute respiratory syndrome CoV 2 (SARS-CoV-2). Recent discussions concerning vaccination strategies have focused on identifying vaccine platforms, number of doses, route of administration, and time to reach peak immunity against SARS-CoV-2. Here, we generated a single-dose, fast-acting vesicular stomatitis virus (VSV)-based vaccine derived from the licensed Ebola virus (EBOV) vaccine rVSV-ZEBOV, expressing the SARS-CoV-2 spike protein and the EBOV glycoprotein (VSV-SARS2-EBOV). Rhesus macaques vaccinated intramuscularly (i.m.) with a single dose of VSV-SARS2-EBOV were protected within 10 days and did not show signs of COVID-19 pneumonia. In contrast, intranasal (i.n.) vaccination resulted in limited immunogenicity and enhanced COVID-19 pneumonia compared to results for control animals. While both i.m. and i.n. vaccination induced neutralizing antibody titers, only i.m. vaccination resulted in a significant cellular immune response. RNA sequencing data bolstered these results by revealing robust activation of the innate and adaptive immune transcriptional signatures in the lungs of i.m. vaccinated animals only. Overall, the data demonstrate that VSV-SARS2-EBOV is a potent single-dose COVID-19 vaccine candidate that offers rapid protection based on the protective efficacy observed in our study. IMPORTANCE The vesicular stomatitis virus (VSV) vaccine platform rose to fame in 2019, when a VSV-based Ebola virus (EBOV) vaccine was approved by the European Medicines Agency and the U.S. Food and Drug Administration for human use against the deadly disease. Here, we demonstrate the protective efficacy of a VSV-EBOV-based COVID-19 vaccine against challenge in nonhuman primates (NHPs). When a single dose of the VSV-SARS2-EBOV vaccine was administered intramuscularly (i.m.), the NHPs were protected from COVID-19 within 10 days. In contrast, if the vaccine was administered intranasally, there was no benefit from the vaccine and the NHPs developed pneumonia. The i.m. vaccinated NHPs quickly developed antigen-specific IgG, including neutralizing antibodies. Transcriptional analysis highlighted the development of protective innate and adaptive immune responses in the i.m. vaccination group only.
Collapse
Affiliation(s)
- Wakako Furuyama
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Kyle Shifflett
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Amanda N. Pinski
- Department of Molecular Biology and Biochemistry, University of California—Irvine, Irvine, California, USA
| | - Amanda J. Griffin
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Friederike Feldmann
- Rocky Mountain Veterinary Branch, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Atsushi Okumura
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Tylisha Gourdine
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Allen Jankeel
- Department of Molecular Biology and Biochemistry, University of California—Irvine, Irvine, California, USA
| | - Jamie Lovaglio
- Rocky Mountain Veterinary Branch, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Patrick W. Hanley
- Rocky Mountain Veterinary Branch, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Tina Thomas
- Rocky Mountain Veterinary Branch, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Chad S. Clancy
- Rocky Mountain Veterinary Branch, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Ilhem Messaoudi
- Department of Molecular Biology and Biochemistry, University of California—Irvine, Irvine, California, USA
| | - Kyle L. O’Donnell
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Andrea Marzi
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| |
Collapse
|
243
|
Intensity and Dynamics of Anti-SARS-CoV-2 Immune Responses after BNT162b2 mRNA Vaccination: Implications for Public Health Vaccination Strategies. Vaccines (Basel) 2022; 10:vaccines10020316. [PMID: 35214774 PMCID: PMC8879063 DOI: 10.3390/vaccines10020316] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 02/11/2022] [Accepted: 02/14/2022] [Indexed: 01/27/2023] Open
Abstract
The aim of our study was to investigate the immunogenicity of the BNT162b2 vaccination according to the age and medical status of vaccinated individuals. A total of 511 individuals were enrolled (median age: 54.0 years, range: 19–105); 509 of these individuals (99.6%) received two doses of BNT162b2 at an interval of 21 days. IgG and IgA responses were evaluated on days 21, 42, 90, and 180 after the first dose with chemiluminescent microparticle and ELISA assays. The cell-mediated immune responses were assessed by an automated interferon-gamma release assay. We demonstrated positive antibody responses after vaccination for the majority of enrolled participants, although waning of IgG and IgA titers was also observed over time. We further observed that the intensity of humoral responses was positively correlated with increased age and prior COVID-19 infection (either before or after the first vaccination). Moreover, we found that only a medical history of autoimmune disease could affect the intensity of IgA and IgG responses (3 weeks after the primary and secondary immunization, respectively), while development of systemic adverse reactions after the second vaccination dose was significantly associated with the height of IgG responses. Finally, we identified a clear correlation between humoral and cellular responses, suggesting that the study of cellular responses is not required as a routine laboratory test after vaccination. Our results provide useful information about the immunogenicity of COVID-19 vaccination with significant implications for public health vaccination strategies.
Collapse
|
244
|
Jawalagatti V, Kirthika P, Hewawaduge C, Park JY, Yang MS, Oh B, So MY, Kim B, Lee JH. A Simplified SARS-CoV-2 Mouse Model Demonstrates Protection by an Oral Replicon-Based mRNA Vaccine. Front Immunol 2022; 13:811802. [PMID: 35250985 PMCID: PMC8888445 DOI: 10.3389/fimmu.2022.811802] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 01/25/2022] [Indexed: 12/24/2022] Open
Abstract
A mouse model of SARS-CoV-2 that can be developed in any molecular biology lab with standard facilities will be valuable in evaluating drugs and vaccines. Here we present a simplified SARS-CoV-2 mouse model exploiting the rapid adenoviral purification method. Mice that are sensitive to SARS-CoV-2 infection were generated by transducing human angiotensin-converting enzyme 2 (hACE2) by an adenovirus. The expression kinetics of the hACE2 in transduced mice were assessed by immunohistochemistry, RT-PCR, and qPCR. Further, the ability of the hACE2 to support viral replication was determined in vitro and in vivo. The hACE2 expression in the lungs of mice was observed for at least nine days after transduction. The murine macrophages expressing hACE2 supported viral replication with detection of high viral titers. Next, in vivo studies were carried out to determine viral replication and lung disease following SARS-CoV-2 challenge. The model supported viral replication, and the challenged mouse developed lung disease characteristic of moderate interstitial pneumonia. Further, we illustrated the utility of the system by demonstrating protection using an oral mRNA vaccine. The multicistronic vaccine design enabled by the viral self-cleaving peptides targets receptor binding domain (RBD), heptad repeat domain (HR), membrane glycoprotein (M) and epitopes of nsp13 of parental SARS-CoV-2. Further, Salmonella and Semliki Forest virus replicon were exploited, respectively, for gene delivery and mRNA expression. We recorded potent cross-protective neutralizing antibodies in immunized mice against the SARS-CoV-2 delta variant. The vaccine protected the mice against viral replication and SARS-CoV-2-induced weight loss and lung pathology. The findings support the suitability of the model for preclinical evaluation of anti-SARS-CoV-2 therapies and vaccines. In addition, the findings provide novel insights into mRNA vaccine design against infectious diseases not limiting to SARS-CoV-2.
Collapse
Affiliation(s)
- Vijayakumar Jawalagatti
- Department of Veterinary Public Health, College of Veterinary Medicine, Jeonbuk National University, Iksan, South Korea
| | - Perumalraja Kirthika
- Department of Veterinary Public Health, College of Veterinary Medicine, Jeonbuk National University, Iksan, South Korea
| | - Chamith Hewawaduge
- Department of Veterinary Public Health, College of Veterinary Medicine, Jeonbuk National University, Iksan, South Korea
| | - Ji-Young Park
- Department of Veterinary Public Health, College of Veterinary Medicine, Jeonbuk National University, Iksan, South Korea
| | - Myeon-Sik Yang
- Department of Veterinary Pathology, College of Veterinary Medicine, Jeonbuk National University, Iksan, South Korea
| | - Byungkwan Oh
- Department of Veterinary Pathology, College of Veterinary Medicine, Jeonbuk National University, Iksan, South Korea
| | - Mi Young So
- Department of Veterinary Pathology, College of Veterinary Medicine, Jeonbuk National University, Iksan, South Korea
| | - Bumseok Kim
- Department of Veterinary Pathology, College of Veterinary Medicine, Jeonbuk National University, Iksan, South Korea
| | - John Hwa Lee
- Department of Veterinary Public Health, College of Veterinary Medicine, Jeonbuk National University, Iksan, South Korea
| |
Collapse
|
245
|
Chang A, Yu J. Fighting Fire with Fire: Immunogenicity of Viral Vectored Vaccines against COVID-19. Viruses 2022; 14:380. [PMID: 35215973 PMCID: PMC8874888 DOI: 10.3390/v14020380] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 02/09/2022] [Accepted: 02/09/2022] [Indexed: 11/16/2022] Open
Abstract
The persistent expansion of the coronavirus disease 2019 (COVID-19) global pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) requires the rapid development of safe and effective countermeasures to reduce transmission, morbidity, and mortality. Several highly efficacious vaccines are actively being deployed around the globe to expedite mass vaccination and control of COVID-19. Notably, viral vectored vaccines (VVVs) are among the first to be approved for global distribution and use. In this review, we examine the humoral, cellular, and innate immune responses elicited by viral vectors, and the immune correlates of protection against COVID-19 in preclinical and clinical studies. We also discuss the durability and breadth of immune response induced by VVVs and boosters. Finally, we present challenges associated with VVVs and offer solutions for overcoming certain limitations of current vaccine regimens. Collectively, this review provides the rationale for expanding the portfolio of VVVs against SARS-CoV-2.
Collapse
MESH Headings
- Animals
- Antibodies, Neutralizing/blood
- Antibodies, Neutralizing/immunology
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- COVID-19/immunology
- COVID-19/prevention & control
- COVID-19 Vaccines/genetics
- COVID-19 Vaccines/immunology
- Clinical Trials as Topic
- Disease Models, Animal
- Genetic Vectors/immunology
- Immunity, Cellular
- Immunity, Humoral
- Immunity, Innate
- Immunization, Secondary
- Immunogenicity, Vaccine
- SARS-CoV-2/immunology
- Spike Glycoprotein, Coronavirus/genetics
- Vaccination
- Viral Vaccines/classification
- Viral Vaccines/genetics
- Viral Vaccines/immunology
Collapse
Affiliation(s)
- Aiquan Chang
- Beth Israel Deaconess Medical Center, Center for Virology and Vaccine Research, Harvard Medical School, Boston, MA 02115, USA
| | - Jingyou Yu
- Beth Israel Deaconess Medical Center, Center for Virology and Vaccine Research, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
246
|
Alu A, Chen L, Lei H, Wei Y, Tian X, Wei X. Intranasal COVID-19 vaccines: From bench to bed. EBioMedicine 2022; 76:103841. [PMID: 35085851 PMCID: PMC8785603 DOI: 10.1016/j.ebiom.2022.103841] [Citation(s) in RCA: 128] [Impact Index Per Article: 42.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Revised: 01/09/2022] [Accepted: 01/12/2022] [Indexed: 02/05/2023] Open
Abstract
Currently licensed COVID-19 vaccines are all designed for intramuscular (IM) immunization. However, vaccination today failed to prevent the virus infection through the upper respiratory tract, which is partially due to the absence of mucosal immunity activation. Despite the emerging severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants, the next generation of COVID-19 vaccine is in demand and intranasal (IN) vaccination method has been demonstrated to be potent in inducing both mucosal and systemic immune responses. Presently, although not licensed, various IN vaccines against SARS-CoV-2 are under intensive investigation, with 12 candidates reaching clinical trials at different phases. In this review, we give a detailed description about current status of IN COVID-19 vaccines, including virus-vectored vaccines, recombinant subunit vaccines and live attenuated vaccines. The ongoing clinical trials for IN vaccines are highlighted. Additionally, the underlying mechanisms of mucosal immunity and potential mucosal adjuvants and nasal delivery devices are also summarized.
Collapse
Affiliation(s)
- Aqu Alu
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Li Chen
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hong Lei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yuquan Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiaohe Tian
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
247
|
Xu F, Wu S, Yi L, Peng S, Wang F, Si W, Hou L, Zhu T. Safety, mucosal and systemic immunopotency of an aerosolized adenovirus-vectored vaccine against SARS-CoV-2 in rhesus macaques. Emerg Microbes Infect 2022; 11:438-441. [PMID: 35094672 PMCID: PMC8803102 DOI: 10.1080/22221751.2022.2030199] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Mucosal immunity provides a potential for preventing initial infection and stopping subsequent transmission of SARS-CoV-2. Here, we examined the safety and immunogenicity of a replication-defective adenovirus type-5 vectored vaccine (Ad5-nCov) encoding SARS-CoV-2 spike protein delivered by nebulization inhalation in rhesus macaques. The vaccine-associated clinical pathology and toxicity were not observed in the NHP model. The extensive safety study indicated that Ad5-nCoV was mainly confined to the organs related to respiratory system and was rapidly cleared away from the system. Our results showed that Ad5-nCoV delivered by inhalation robustly elicited both systematic and mucosal immune responses against SARS-nCoV-2 and variants. Thus, Ad5-nCoV inhalation may provide an effective, safe and non-invasive vaccination strategy for the control of SARS-CoV-2.
Collapse
Affiliation(s)
- Fang Xu
- CanSino Biologics, Tianjin, People’s Republic of China
| | - Shipo Wu
- Institute of Biotechnology, Academy of Military Medical Sciences, Beijing, People’s Republic of China
| | - Linan Yi
- CanSino Biologics, Tianjin, People’s Republic of China
| | - Shaodan Peng
- CanSino Biologics, Tianjin, People’s Republic of China
| | - Fan Wang
- CanSino Biologics, Tianjin, People’s Republic of China
| | - Weixue Si
- CanSino Biologics, Tianjin, People’s Republic of China
| | - Lihua Hou
- Institute of Biotechnology, Academy of Military Medical Sciences, Beijing, People’s Republic of China
| | - Tao Zhu
- CanSino Biologics, Tianjin, People’s Republic of China
| |
Collapse
|
248
|
Mao T, Israelow B, Suberi A, Zhou L, Reschke M, Peña-Hernández MA, Dong H, Homer RJ, Saltzman WM, Iwasaki A. Unadjuvanted intranasal spike vaccine booster elicits robust protective mucosal immunity against sarbecoviruses. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022. [PMID: 35118464 DOI: 10.1101/2022.01.24.477597] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
As the SARS-CoV-2 pandemic enters its third year, vaccines that not only prevent disease, but also prevent transmission are needed to help reduce global disease burden. Currently approved parenteral vaccines induce robust systemic immunity, but poor immunity at the respiratory mucosa. Here we describe the development of a novel vaccine strategy, Prime and Spike, based on unadjuvanted intranasal spike boosting that leverages existing immunity generated by primary vaccination to elicit mucosal immune memory within the respiratory tract. We show that Prime and Spike induces robust T resident memory cells, B resident memory cells and IgA at the respiratory mucosa, boosts systemic immunity, and completely protects mice with partial immunity from lethal SARS-CoV-2 infection. Using divergent spike proteins, Prime and Spike enables induction of cross-reactive immunity against sarbecoviruses without invoking original antigenic sin. ONE-SENTENCE SUMMARY Broad sarbecovirus protective mucosal immunity is generated by unadjuvanted intranasal spike boost in preclinical model.
Collapse
|
249
|
Focosi D, Maggi F, Casadevall A. Mucosal Vaccines, Sterilizing Immunity, and the Future of SARS-CoV-2 Virulence. Viruses 2022; 14:187. [PMID: 35215783 PMCID: PMC8878800 DOI: 10.3390/v14020187] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 01/15/2022] [Accepted: 01/17/2022] [Indexed: 02/01/2023] Open
Abstract
Sterilizing immunity after vaccination is desirable to prevent the spread of infection from vaccinees, which can be especially dangerous in hospital settings while managing frail patients. Sterilizing immunity requires neutralizing antibodies at the site of infection, which for respiratory viruses such as SARS-CoV-2 implies the occurrence of neutralizing IgA in mucosal secretions. Systemic vaccination by intramuscular delivery induces no or low-titer neutralizing IgA against vaccine antigens. Mucosal priming or boosting, is needed to provide sterilizing immunity. On the other side of the coin, sterilizing immunity, by zeroing interhuman transmission, could confine SARS-CoV-2 in animal reservoirs, preventing spontaneous attenuation of virulence in humans as presumably happened with the endemic coronaviruses. We review here the pros and cons of each vaccination strategy, the current mucosal SARS-CoV-2 vaccines under development, and their implications for public health.
Collapse
Affiliation(s)
- Daniele Focosi
- North-Western Tuscany Blood Bank, Pisa University Hospital, 56124 Pisa, Italy
| | - Fabrizio Maggi
- Department of Medicine and Surgery, University of Insubria, 21100 Varese, Italy;
| | - Arturo Casadevall
- Department of Medicine, Johns Hopkins School of Public Health and School of Medicine, Baltimore, MD 21218, USA;
| |
Collapse
|
250
|
A novel STING agonist-adjuvanted pan-sarbecovirus vaccine elicits potent and durable neutralizing antibody and T cell responses in mice, rabbits and NHPs. Cell Res 2022; 32:269-287. [PMID: 35046518 PMCID: PMC8767042 DOI: 10.1038/s41422-022-00612-2] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 01/03/2022] [Indexed: 12/23/2022] Open
Abstract
The emergence of SARS-CoV-2 variants and potentially other highly pathogenic sarbecoviruses in the future highlights the need for pan-sarbecovirus vaccines. Here, we discovered a new STING agonist, CF501, and found that CF501-adjuvanted RBD-Fc vaccine (CF501/RBD-Fc) elicited significantly stronger neutralizing antibody (nAb) and T cell responses than Alum- and cGAMP-adjuvanted RBD-Fc in mice. Vaccination of rabbits and rhesus macaques (nonhuman primates, NHPs) with CF501/RBD-Fc elicited exceptionally potent nAb responses against SARS-CoV-2 and its nine variants and 41 S-mutants, SARS-CoV and bat SARSr-CoVs. CF501/RBD-Fc-immunized hACE2-transgenic mice were almost completely protected against SARS-CoV-2 challenge, even 6 months after the initial immunization. NHPs immunized with a single dose of CF501/RBD-Fc produced high titers of nAbs. The immunized macaques also exhibited durable humoral and cellular immune responses and showed remarkably reduced viral load in the upper and lower airways upon SARS-CoV-2 challenge even at 108 days post the final immunization. Thus, CF501/RBD-Fc can be further developed as a novel pan-sarbecovirus vaccine to combat current and future outbreaks of sarbecovirus diseases.
Collapse
|