1
|
Kumar S, Nan L, Kalodimou G, Jany S, Freudenstein A, Brandmüller C, Müller K, Girl P, Ehmann R, Guggemos W, Seilmaier M, Wendtner CM, Volz A, Sutter G, Fux R, Tscherne A. Implementation of an Immunoassay Based on the MVA-T7pol-Expression System for Rapid Identification of Immunogenic SARS-CoV-2 Antigens: A Proof-of-Concept Study. Int J Mol Sci 2024; 25:10898. [PMID: 39456680 PMCID: PMC11508112 DOI: 10.3390/ijms252010898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 10/01/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
The emergence of hitherto unknown viral pathogens presents a great challenge for researchers to develop effective therapeutics and vaccines within a short time to avoid an uncontrolled global spread, as seen during the coronavirus disease 2019 (COVID-19) pandemic. Therefore, rapid and simple methods to identify immunogenic antigens as potential therapeutical targets are urgently needed for a better pandemic preparedness. To address this problem, we chose the well-characterized Modified Vaccinia virus Ankara (MVA)-T7pol expression system to establish a workflow to identify immunogens when a new pathogen emerges, generate candidate vaccines, and test their immunogenicity in an animal model. By using this system, we detected severe acute respiratory syndrome (SARS) coronavirus 2 (SARS-CoV-2) nucleoprotein (N)-, and spike (S)-specific antibodies in COVID-19 patient sera, which is in line with the current literature and our observations from previous immunogenicity studies. Furthermore, we detected antibodies directed against the SARS-CoV-2-membrane (M) and -ORF3a proteins in COVID-19 patient sera and aimed to generate recombinant MVA candidate vaccines expressing either the M or ORF3a protein. When testing our candidate vaccines in a prime-boost immunization regimen in humanized HLA-A2.1-/HLA-DR1-transgenic H-2 class I-/class II-knockout mice, we were able to demonstrate M- and ORF3a-specific cellular and humoral immune responses. Hence, the established workflow using the MVA-T7pol expression system represents a rapid and efficient tool to identify potential immunogenic antigens and provides a basis for future development of candidate vaccines.
Collapse
Affiliation(s)
- Satendra Kumar
- Division of Virology, Department of Veterinary Sciences, Ludwig Maximilians University Munich (LMU Munich), 85764 Oberschleißheim, Germany; (S.K.); (L.N.); (G.K.)
| | - Liangliang Nan
- Division of Virology, Department of Veterinary Sciences, Ludwig Maximilians University Munich (LMU Munich), 85764 Oberschleißheim, Germany; (S.K.); (L.N.); (G.K.)
| | - Georgia Kalodimou
- Division of Virology, Department of Veterinary Sciences, Ludwig Maximilians University Munich (LMU Munich), 85764 Oberschleißheim, Germany; (S.K.); (L.N.); (G.K.)
- German Center for Infection Research, Partner Site Munich, 85764 Oberschleißheim, Germany (R.E.)
| | - Sylvia Jany
- Division of Virology, Department of Veterinary Sciences, Ludwig Maximilians University Munich (LMU Munich), 85764 Oberschleißheim, Germany; (S.K.); (L.N.); (G.K.)
| | - Astrid Freudenstein
- Division of Virology, Department of Veterinary Sciences, Ludwig Maximilians University Munich (LMU Munich), 85764 Oberschleißheim, Germany; (S.K.); (L.N.); (G.K.)
| | - Christine Brandmüller
- Division of Virology, Department of Veterinary Sciences, Ludwig Maximilians University Munich (LMU Munich), 85764 Oberschleißheim, Germany; (S.K.); (L.N.); (G.K.)
| | - Katharina Müller
- German Center for Infection Research, Partner Site Munich, 85764 Oberschleißheim, Germany (R.E.)
- Bundeswehr Institute of Microbiology, 80937 Munich, Germany
| | - Philipp Girl
- German Center for Infection Research, Partner Site Munich, 85764 Oberschleißheim, Germany (R.E.)
- Bundeswehr Institute of Microbiology, 80937 Munich, Germany
- Chair of Bacteriology and Mycology, Department of Veterinary Sciences, Ludwig Maximilians University Munich (LMU Munich), 85764 Oberschleißheim, Germany
| | - Rosina Ehmann
- German Center for Infection Research, Partner Site Munich, 85764 Oberschleißheim, Germany (R.E.)
- Bundeswehr Institute of Microbiology, 80937 Munich, Germany
| | - Wolfgang Guggemos
- Munich Clinic Schwabing, Academic Teaching Hospital, Ludwig Maximilians University Munich (LMU Munich), 80804 Munich, Germany; (W.G.); (M.S.)
| | - Michael Seilmaier
- Munich Clinic Schwabing, Academic Teaching Hospital, Ludwig Maximilians University Munich (LMU Munich), 80804 Munich, Germany; (W.G.); (M.S.)
| | - Clemens-Martin Wendtner
- Medical Clinic III, University Hospital, Ludwig Maximilians University Munich (LMU Munich), 80336 Munich, Germany;
| | - Asisa Volz
- Institute of Virology, University of Veterinary Medicine Hannover, 30559 Hannover, Germany;
- German Center for Infection Research, Partner Site Hannover-Braunschweig, 30559 Hannover, Germany
| | - Gerd Sutter
- Division of Virology, Department of Veterinary Sciences, Ludwig Maximilians University Munich (LMU Munich), 85764 Oberschleißheim, Germany; (S.K.); (L.N.); (G.K.)
- German Center for Infection Research, Partner Site Munich, 85764 Oberschleißheim, Germany (R.E.)
| | - Robert Fux
- Division of Virology, Department of Veterinary Sciences, Ludwig Maximilians University Munich (LMU Munich), 85764 Oberschleißheim, Germany; (S.K.); (L.N.); (G.K.)
| | - Alina Tscherne
- Division of Virology, Department of Veterinary Sciences, Ludwig Maximilians University Munich (LMU Munich), 85764 Oberschleißheim, Germany; (S.K.); (L.N.); (G.K.)
- German Center for Infection Research, Partner Site Munich, 85764 Oberschleißheim, Germany (R.E.)
| |
Collapse
|
2
|
Chakravarty N, Hemani D, Paravastu R, Ahmad Z, Palani SN, Arumugaswami V, Kumar A. Mpox Virus and its ocular surface manifestations. Ocul Surf 2024; 34:108-121. [PMID: 38972544 DOI: 10.1016/j.jtos.2024.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 06/28/2024] [Accepted: 07/04/2024] [Indexed: 07/09/2024]
Abstract
The Mpox virus (MPXV) is the causative agent of human Mpox disease - a debilitating rash illness similar to smallpox. Although Clade I MPXV has remained endemic to West and Central Africa, Clade II MPXV has been responsible for many outbreaks worldwide. The most recent outbreak in 2022 resulted from the rapid spread of a new clade of MPXV, classified into Clade IIb - a distinct lineage from the previously circulating viral strains. The rapid spread and increased severity of Mpox disease by the Clade IIb strain have raised the serious public health imperative of better understanding the host and viral determinants during MPXV infection. In addition to typical skin rashes, including in the periorbital area, MPXV causes moderate to severe ophthalmic manifestations - most commonly, ocular surface complications (e.g., keratitis, conjunctivitis, blepharitis). While ocular manifestations of Clade I Mpox within the Congo basin have been well-reported, global incidence trends of ocular Mpox cases by Clade IIb are still emerging. Given the demonstrated ability of all MPXV strains to auto-inoculate ocular tissue, alongside the enhanced transmissibility of the Clade IIb virus, there is an urgent need to elucidate the mechanisms by which MPXV causes ocular anomalies. In this review, we discuss the viral and genomic structures of MPXV, the epidemiology, and pathology of systemic and ocular Mpox, as well as potential prophylactic and therapeutic interventions.
Collapse
Affiliation(s)
- Nikhil Chakravarty
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, USA; School of Medicine, California University of Science and Medicine, Colton, CA, USA
| | - Darshi Hemani
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University, Detroit, MI, USA
| | - Ramya Paravastu
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Zeeshan Ahmad
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University, Detroit, MI, USA
| | - Sankara Naynar Palani
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Vaithilingaraja Arumugaswami
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, USA; California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA, USA.
| | - Ashok Kumar
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University, Detroit, MI, USA.
| |
Collapse
|
3
|
Nimer NA, Nimer SN. Immunization against Medically Important Human Coronaviruses of Public Health Concern. THE CANADIAN JOURNAL OF INFECTIOUS DISEASES & MEDICAL MICROBIOLOGY = JOURNAL CANADIEN DES MALADIES INFECTIEUSES ET DE LA MICROBIOLOGIE MEDICALE 2024; 2024:9952803. [PMID: 38938549 PMCID: PMC11208815 DOI: 10.1155/2024/9952803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 01/14/2024] [Accepted: 05/20/2024] [Indexed: 06/29/2024]
Abstract
SARS-CoV-2 is a virus that affects the human immune system. It was observed to be on the rise since the beginning of 2020 and turned into a life-threatening pandemic. Scientists have tried to develop a possible preventive and therapeutic drug against severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) and other related coronaviruses by assessing COVID-19-recovered persons' immunity. This study aims to review immunization against SARS-CoV-2, along with exploring the interventions that have been developed for the prevention of SARS-CoV-2. This study also highlighted the role of phototherapy in treating SARS-CoV infection. The study adopted a review approach to gathering the information available and the progress that has been made in the treatment and prevention of COVID-19. Various vaccinations, including nucleotide, subunit, and vector-based vaccines, as well as attenuated and inactivated forms that have already been shown to have prophylactic efficacy against the Middle East respiratory syndrome coronavirus (MERS-CoV) and SARS-CoV, have been summarized. Neutralizing and non-neutralizing antibodies are all associated with viral infections. Because there is no specific antiviral vaccine or therapies for coronaviruses, the main treatment strategy is supportive care, which is reinforced by combining broad-spectrum antivirals, convalescent plasma, and corticosteroids. COVID-19 has been a challenge to keep reconsidering the usual approaches to regulatory evaluation as a result of getting mixed and complicated findings on the vaccines, as well as licensing procedures. However, it is observed that medicinal herbs also play an important role in treating infection of the upper respiratory tract, the principal symptom of SARS-CoV due to their natural bioactive composite. However, some Traditional Chinese Medicines contain mutagens and nephrotoxins and the toxicological properties of the majority of Chinese herbal remedies are unknown. Therefore, to treat the COVID-19 infection along with conventional treatment, it is recommended that herb-drug interaction be examined thoroughly.
Collapse
Affiliation(s)
- Nabil A. Nimer
- Faculty of Pharmacy, Philadelphia University, Amman, Jordan
| | - Seema N. Nimer
- School of Medicine, The University of Jordan, Amman, Jordan
| |
Collapse
|
4
|
López D, García-Peydró M. Could SARS-CoV-1 Vaccines in the Pipeline Have Contributed to Fighting the COVID-19 Pandemic? Lessons for the Next Coronavirus Plague. Biomedicines 2023; 12:62. [PMID: 38255169 PMCID: PMC10813159 DOI: 10.3390/biomedicines12010062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 12/20/2023] [Accepted: 12/22/2023] [Indexed: 01/24/2024] Open
Abstract
SARS-CoV-2 caused the devastating COVID-19 pandemic, which, to date, has resulted in more than 800 million confirmed cases and 7 million deaths worldwide. The rapid development and distribution (at least in high-income countries) of various vaccines prevented these overwhelming numbers of infections and deaths from being much higher. But would it have been possible to develop a prophylaxis against this pandemic more quickly? Since SARS-CoV-2 belongs to the subgenus sarbecovirus, with its highly homologous SARS-CoV-1, we propose here that while SARS-CoV-2-specific vaccines are being developed, phase II clinical trials of specific SARS-CoV-1 vaccines, which have been in the pipeline since the early 20th century, could have been conducted to test a highly probable cross-protection between SARS-CoV-1 and SARS-CoV-2.
Collapse
Affiliation(s)
- Daniel López
- Presentation and Immune Regulation Unit, Centro Nacional de Microbiología, Instituto de Salud Carlos III, 28220 Majadahonda, Spain
| | | |
Collapse
|
5
|
Daian E Silva DSO, Cox LJ, Rocha AS, Lopes-Ribeiro Á, Souza JPC, Franco GM, Prado JLC, Pereira-Santos TA, Martins ML, Coelho-Dos-Reis JGA, Gomes-de-Pinho TM, Da Fonseca FG, Barbosa-Stancioli EF. Preclinical assessment of an anti-HTLV-1 heterologous DNA/MVA vaccine protocol expressing a multiepitope HBZ protein. Virol J 2023; 20:304. [PMID: 38115107 PMCID: PMC10731796 DOI: 10.1186/s12985-023-02264-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 12/07/2023] [Indexed: 12/21/2023] Open
Abstract
BACKGROUND Human T-lymphotropic virus 1 (HTLV-1) is associated with the development of several pathologies and chronic infection in humans. The inefficiency of the available treatments and the challenge in developing a protective vaccine highlight the need to produce effective immunotherapeutic tools. The HTLV-1 basic leucine zipper (bZIP) factor (HBZ) plays an important role in the HTLV-1 persistence, conferring a survival advantage to infected cells by reducing the HTLV-1 proteins expression, allowing infected cells to evade immune surveillance, and enhancing cell proliferation leading to increased proviral load. METHODS We have generated a recombinant Modified Virus Vaccinia Ankara (MVA-HBZ) and a plasmid DNA (pcDNA3.1(+)-HBZ) expressing a multiepitope protein based on peptides of HBZ to study the immunogenic potential of this viral-derived protein in BALB/c mice model. Mice were immunized in a prime-boost heterologous protocol and their splenocytes (T CD4+ and T CD8+) were immunophenotyped by flow cytometry and the humoral response was evaluated by ELISA using HBZ protein produced in prokaryotic vector as antigen. RESULTS T CD4+ and T CD8+ lymphocytes cells stimulated by HBZ-peptides (HBZ42-50 and HBZ157-176) showed polyfunctional double positive responses for TNF-α/IFN-γ, and TNF-α/IL-2. Moreover, T CD8+ cells presented a tendency in the activation of effector memory cells producing granzyme B (CD44+High/CD62L-Low), and the activation of Cytotoxic T Lymphocytes (CTLs) and cytotoxic responses in immunized mice were inferred through the production of granzyme B by effector memory T cells and the expression of CD107a by CD8+ T cells. The overall data is consistent with a directive and effector recall response, which may be able to operate actively in the elimination of HTLV-1-infected cells and, consequently, in the reduction of the proviral load. Sera from immunized mice, differently from those of control animals, showed IgG-anti-HBZ production by ELISA. CONCLUSIONS Our results highlight the potential of the HBZ multiepitope protein expressed from plasmid DNA and a poxviral vector as candidates for therapeutic vaccine.
Collapse
Affiliation(s)
- D S O Daian E Silva
- Laboratório de Virologia Básica e Aplicada, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, Campus Pampulha, Belo Horizonte, MG, CEP 31270-901, Brazil
- GIPH - Grupo Interdisciplinar de Pesquisas em HTLV, Interdisciplinary HTLV Research Group, Belo Horizonte, Brazil
| | - L J Cox
- Laboratório de Virologia Básica e Aplicada, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, Campus Pampulha, Belo Horizonte, MG, CEP 31270-901, Brazil
- GIPH - Grupo Interdisciplinar de Pesquisas em HTLV, Interdisciplinary HTLV Research Group, Belo Horizonte, Brazil
| | - A S Rocha
- Laboratório de Virologia Básica e Aplicada, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, Campus Pampulha, Belo Horizonte, MG, CEP 31270-901, Brazil
- GIPH - Grupo Interdisciplinar de Pesquisas em HTLV, Interdisciplinary HTLV Research Group, Belo Horizonte, Brazil
| | - Á Lopes-Ribeiro
- Laboratório de Virologia Básica e Aplicada, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, Campus Pampulha, Belo Horizonte, MG, CEP 31270-901, Brazil
| | - J P C Souza
- Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, 31270-901, Brazil
| | - G M Franco
- Laboratório de Virologia Básica e Aplicada, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, Campus Pampulha, Belo Horizonte, MG, CEP 31270-901, Brazil
- GIPH - Grupo Interdisciplinar de Pesquisas em HTLV, Interdisciplinary HTLV Research Group, Belo Horizonte, Brazil
| | - J L C Prado
- Laboratório de Virologia Básica e Aplicada, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, Campus Pampulha, Belo Horizonte, MG, CEP 31270-901, Brazil
| | - T A Pereira-Santos
- Laboratório de Virologia Básica e Aplicada, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, Campus Pampulha, Belo Horizonte, MG, CEP 31270-901, Brazil
- GIPH - Grupo Interdisciplinar de Pesquisas em HTLV, Interdisciplinary HTLV Research Group, Belo Horizonte, Brazil
| | - M L Martins
- GIPH - Grupo Interdisciplinar de Pesquisas em HTLV, Interdisciplinary HTLV Research Group, Belo Horizonte, Brazil
- Gerência de Desenvolvimento Técnico Científico, Fundação Centro de Hematologia e Hemoterapia do Estado de Minas Gerais - Hemominas, Belo Horizonte, Brazil
| | - J G A Coelho-Dos-Reis
- Laboratório de Virologia Básica e Aplicada, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, Campus Pampulha, Belo Horizonte, MG, CEP 31270-901, Brazil
- GIPH - Grupo Interdisciplinar de Pesquisas em HTLV, Interdisciplinary HTLV Research Group, Belo Horizonte, Brazil
| | - T M Gomes-de-Pinho
- Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, 31270-901, Brazil
| | - F G Da Fonseca
- Laboratório de Virologia Básica e Aplicada, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, Campus Pampulha, Belo Horizonte, MG, CEP 31270-901, Brazil
- Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, 31270-901, Brazil
| | - E F Barbosa-Stancioli
- Laboratório de Virologia Básica e Aplicada, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, Campus Pampulha, Belo Horizonte, MG, CEP 31270-901, Brazil.
- GIPH - Grupo Interdisciplinar de Pesquisas em HTLV, Interdisciplinary HTLV Research Group, Belo Horizonte, Brazil.
| |
Collapse
|
6
|
López D, García-Peydró M. Immunoinformatics lessons on the current COVID-19 pandemic and future coronavirus zoonoses. Front Immunol 2023; 14:1118267. [PMID: 38149247 PMCID: PMC10749959 DOI: 10.3389/fimmu.2023.1118267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 11/29/2023] [Indexed: 12/28/2023] Open
Affiliation(s)
- Daniel López
- Presentation and Immune Regulation Unit, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, Spain
| | - Marina García-Peydró
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas (CSIC) - Universidad Autónoma de Madrid (UAM), Madrid, Spain
| |
Collapse
|
7
|
Kaushik N, Patel P, Bhartiya P, Shin Y, Kim JH, Choi EH, Kaushik NK. Glycolytic stress deteriorates 229E virulence to improve host defense response. Microbes Infect 2023; 25:105150. [PMID: 37178787 PMCID: PMC10174727 DOI: 10.1016/j.micinf.2023.105150] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 04/28/2023] [Accepted: 05/09/2023] [Indexed: 05/15/2023]
Abstract
Viral infection treatment is a difficult task due to its complex structure and metabolism. Additionally, viruses can alter the metabolism of host cells, mutate, and readily adjust to harsh environments. Coronavirus stimulates glycolysis, weakens mitochondrial activity, and impairs infected cells. In this study, we investigated the efficacy of 2-DG in inhibiting coronavirus-induced metabolic processes and antiviral host defense systems, which have not been explored so far. 2-Deoxy-d-glucose (2-DG), a molecule restricting substrate availability, has recently gained attention as a potential antiviral drug. The results revealed that 229E human coronavirus promoted glycolysis, producing a significant increase in the concentration of fluorescent 2-NBDG, a glucose analog, particularly in the infected host cells. The addition of 2-DG decreased its viral replication and suppressed infection-induced cell death and cytopathic effects, thereby improving the antiviral host defense response. It was also observed that administration of low doses of 2-DG inhibited glucose uptake, indicating that 2-DG consumption in virus-infected host cells was mediated by high-affinity glucose transporters, whose levels were amplified upon coronavirus infection. Our findings indicated that 2-DG could be a potential drug to improve the host defense system in coronavirus-infected cells.
Collapse
Affiliation(s)
- Neha Kaushik
- Department of Biotechnology, College of Engineering, The University of Suwon, Hwaseong, 18323, Republic of Korea
| | - Paritosh Patel
- Department of Electrical and Biological Physics, Plasma Bioscience Research Center, Kwangwoon University, Seoul, 01897, Republic of Korea
| | - Pradeep Bhartiya
- Department of Electrical and Biological Physics, Plasma Bioscience Research Center, Kwangwoon University, Seoul, 01897, Republic of Korea
| | - Yungoh Shin
- Department of Electrical and Biological Physics, Plasma Bioscience Research Center, Kwangwoon University, Seoul, 01897, Republic of Korea
| | - June Hyun Kim
- Department of Biotechnology, College of Engineering, The University of Suwon, Hwaseong, 18323, Republic of Korea
| | - Eun Ha Choi
- Department of Electrical and Biological Physics, Plasma Bioscience Research Center, Kwangwoon University, Seoul, 01897, Republic of Korea.
| | - Nagendra Kumar Kaushik
- Department of Electrical and Biological Physics, Plasma Bioscience Research Center, Kwangwoon University, Seoul, 01897, Republic of Korea.
| |
Collapse
|
8
|
Perdiguero B, Pérez P, Marcos-Villar L, Albericio G, Astorgano D, Álvarez E, Sin L, Elena Gómez C, García-Arriaza J, Esteban M. Highly attenuated poxvirus-based vaccines against emerging viral diseases. J Mol Biol 2023:168173. [PMID: 37301278 DOI: 10.1016/j.jmb.2023.168173] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 06/04/2023] [Accepted: 06/05/2023] [Indexed: 06/12/2023]
Abstract
Although one member of the poxvirus family, variola virus, has caused one of the most devastating human infections worldwide, smallpox, the knowledge gained over the last 30 years on the molecular, virological and immunological mechanisms of these viruses has allowed the use of members of this family as vectors for the generation of recombinant vaccines against numerous pathogens. In this review, we cover different aspects of the history and biology of poxviruses with emphasis on their application as vaccines, from first- to fourth-generation, against smallpox, monkeypox, emerging viral diseases highlighted by the World Health Organization (COVID-19, Crimean-Congo haemorrhagic fever, Ebola and Marburg virus diseases, Lassa fever, Middle East respiratory syndrome and severe acute respiratory syndrome, Nipah and other henipaviral diseases, Rift Valley fever and Zika), as well as against one of the most concerning prevalent virus, the Human Immunodeficiency Virus, the causative agent of AcquiredImmunodeficiency Syndrome. We discuss the implications in human health of the 2022 monkeypox epidemic affecting many countries, and the rapid prophylactic and therapeutic measures adopted to control virus dissemination within the human population. We also describe the preclinical and clinical evaluation of the Modified Vaccinia virus Ankara and New York vaccinia virus poxviral strains expressing heterologous antigens from the viral diseases listed above. Finally, we report different approaches to improve the immunogenicity and efficacy of poxvirus-based vaccine candidates, such as deletion of immunomodulatory genes, insertion of host-range genes and enhanced transcription of foreign genes through modified viral promoters. Some future prospects are also highlighted.
Collapse
Affiliation(s)
- Beatriz Perdiguero
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain.
| | - Patricia Pérez
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain.
| | - Laura Marcos-Villar
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Guillermo Albericio
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - David Astorgano
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Enrique Álvarez
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Laura Sin
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Carmen Elena Gómez
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Juan García-Arriaza
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Mariano Esteban
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain.
| |
Collapse
|
9
|
Lorenzo MM, Marín-López A, Chiem K, Jimenez-Cabello L, Ullah I, Utrilla-Trigo S, Calvo-Pinilla E, Lorenzo G, Moreno S, Ye C, Park JG, Matía A, Brun A, Sánchez-Puig JM, Nogales A, Mothes W, Uchil PD, Kumar P, Ortego J, Fikrig E, Martinez-Sobrido L, Blasco R. Vaccinia Virus Strain MVA Expressing a Prefusion-Stabilized SARS-CoV-2 Spike Glycoprotein Induces Robust Protection and Prevents Brain Infection in Mouse and Hamster Models. Vaccines (Basel) 2023; 11:1006. [PMID: 37243110 PMCID: PMC10220993 DOI: 10.3390/vaccines11051006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/11/2023] [Accepted: 05/15/2023] [Indexed: 05/28/2023] Open
Abstract
The COVID-19 pandemic has underscored the importance of swift responses and the necessity of dependable technologies for vaccine development. Our team previously developed a fast cloning system for the modified vaccinia virus Ankara (MVA) vaccine platform. In this study, we reported on the construction and preclinical testing of a recombinant MVA vaccine obtained using this system. We obtained recombinant MVA expressing the unmodified full-length SARS-CoV-2 spike (S) protein containing the D614G amino-acid substitution (MVA-Sdg) and a version expressing a modified S protein containing amino-acid substitutions designed to stabilize the protein a in a pre-fusion conformation (MVA-Spf). S protein expressed by MVA-Sdg was found to be expressed and was correctly processed and transported to the cell surface, where it efficiently produced cell-cell fusion. Version Spf, however, was not proteolytically processed, and despite being transported to the plasma membrane, it failed to induce cell-cell fusion. We assessed both vaccine candidates in prime-boost regimens in the susceptible transgenic K18-human angiotensin-converting enzyme 2 (K18-hACE2) in mice and in golden Syrian hamsters. Robust immunity and protection from disease was induced with either vaccine in both animal models. Remarkably, the MVA-Spf vaccine candidate produced higher levels of antibodies, a stronger T cell response, and a higher degree of protection from challenge. In addition, the level of SARS-CoV-2 in the brain of MVA-Spf inoculated mice was decreased to undetectable levels. Those results add to our current experience and range of vaccine vectors and technologies for developing a safe and effective COVID-19 vaccine.
Collapse
Affiliation(s)
- María M. Lorenzo
- Departamento de Biotecnología, INIA CSIC, Carretera La Coruña km 7.5, E-28040 Madrid, Spain; (M.M.L.); (S.M.); (A.M.); (J.M.S.-P.)
| | - Alejandro Marín-López
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06519, USA; (A.M.-L.); (I.U.); (E.F.)
| | - Kevin Chiem
- Texas Biomedical Research Institute, San Antonio, TX 78227, USA; (K.C.); (C.Y.); (J.-G.P.); (P.K.)
| | - Luis Jimenez-Cabello
- Centro de Investigación en Sanidad Animal, INIA CSIC, Carretera Valdeolmos a El Casar, Valdeolmos, E-28130 Madrid, Spain; (L.J.-C.); (S.U.-T.); (E.C.-P.); (G.L.); (A.B.); (A.N.); (J.O.)
| | - Irfan Ullah
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06519, USA; (A.M.-L.); (I.U.); (E.F.)
| | - Sergio Utrilla-Trigo
- Centro de Investigación en Sanidad Animal, INIA CSIC, Carretera Valdeolmos a El Casar, Valdeolmos, E-28130 Madrid, Spain; (L.J.-C.); (S.U.-T.); (E.C.-P.); (G.L.); (A.B.); (A.N.); (J.O.)
| | - Eva Calvo-Pinilla
- Centro de Investigación en Sanidad Animal, INIA CSIC, Carretera Valdeolmos a El Casar, Valdeolmos, E-28130 Madrid, Spain; (L.J.-C.); (S.U.-T.); (E.C.-P.); (G.L.); (A.B.); (A.N.); (J.O.)
| | - Gema Lorenzo
- Centro de Investigación en Sanidad Animal, INIA CSIC, Carretera Valdeolmos a El Casar, Valdeolmos, E-28130 Madrid, Spain; (L.J.-C.); (S.U.-T.); (E.C.-P.); (G.L.); (A.B.); (A.N.); (J.O.)
| | - Sandra Moreno
- Departamento de Biotecnología, INIA CSIC, Carretera La Coruña km 7.5, E-28040 Madrid, Spain; (M.M.L.); (S.M.); (A.M.); (J.M.S.-P.)
- Centro de Investigación en Sanidad Animal, INIA CSIC, Carretera Valdeolmos a El Casar, Valdeolmos, E-28130 Madrid, Spain; (L.J.-C.); (S.U.-T.); (E.C.-P.); (G.L.); (A.B.); (A.N.); (J.O.)
| | - Chengjin Ye
- Texas Biomedical Research Institute, San Antonio, TX 78227, USA; (K.C.); (C.Y.); (J.-G.P.); (P.K.)
| | - Jun-Gyu Park
- Texas Biomedical Research Institute, San Antonio, TX 78227, USA; (K.C.); (C.Y.); (J.-G.P.); (P.K.)
| | - Alejandro Matía
- Departamento de Biotecnología, INIA CSIC, Carretera La Coruña km 7.5, E-28040 Madrid, Spain; (M.M.L.); (S.M.); (A.M.); (J.M.S.-P.)
| | - Alejandro Brun
- Centro de Investigación en Sanidad Animal, INIA CSIC, Carretera Valdeolmos a El Casar, Valdeolmos, E-28130 Madrid, Spain; (L.J.-C.); (S.U.-T.); (E.C.-P.); (G.L.); (A.B.); (A.N.); (J.O.)
| | - Juana M. Sánchez-Puig
- Departamento de Biotecnología, INIA CSIC, Carretera La Coruña km 7.5, E-28040 Madrid, Spain; (M.M.L.); (S.M.); (A.M.); (J.M.S.-P.)
| | - Aitor Nogales
- Centro de Investigación en Sanidad Animal, INIA CSIC, Carretera Valdeolmos a El Casar, Valdeolmos, E-28130 Madrid, Spain; (L.J.-C.); (S.U.-T.); (E.C.-P.); (G.L.); (A.B.); (A.N.); (J.O.)
| | - Walther Mothes
- Department of Microbial Pathogenesis, Yale School of Medicine, New Haven, CT 06510, USA; (W.M.); (P.D.U.)
| | - Pradeep D. Uchil
- Department of Microbial Pathogenesis, Yale School of Medicine, New Haven, CT 06510, USA; (W.M.); (P.D.U.)
| | - Priti Kumar
- Texas Biomedical Research Institute, San Antonio, TX 78227, USA; (K.C.); (C.Y.); (J.-G.P.); (P.K.)
| | - Javier Ortego
- Centro de Investigación en Sanidad Animal, INIA CSIC, Carretera Valdeolmos a El Casar, Valdeolmos, E-28130 Madrid, Spain; (L.J.-C.); (S.U.-T.); (E.C.-P.); (G.L.); (A.B.); (A.N.); (J.O.)
| | - Erol Fikrig
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06519, USA; (A.M.-L.); (I.U.); (E.F.)
| | - Luis Martinez-Sobrido
- Texas Biomedical Research Institute, San Antonio, TX 78227, USA; (K.C.); (C.Y.); (J.-G.P.); (P.K.)
| | - Rafael Blasco
- Departamento de Biotecnología, INIA CSIC, Carretera La Coruña km 7.5, E-28040 Madrid, Spain; (M.M.L.); (S.M.); (A.M.); (J.M.S.-P.)
| |
Collapse
|
10
|
Evaluation and comparison of four quantitative SARS-CoV-2 serological assays in COVID-19 patients and immunized healthy individuals, cancer patients, and patients with immunosuppressive therapy. Clin Biochem 2023; 116:1-6. [PMID: 36849050 PMCID: PMC9968448 DOI: 10.1016/j.clinbiochem.2023.02.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 02/20/2023] [Indexed: 02/27/2023]
Abstract
BACKGROUND Semi-quantitative and quantitative immunoassays are the most commonly used methodology to evaluate immunity post immunization. OBJECTIVES To compare four quantitative SARS-CoV-2 serological assays in COVID-19 patients and immunized healthy individuals, cancer patients, and patients with immunosuppressive therapy. STUDY DESIGN 210 serological samples from COVID-19 infection and vaccination cohorts were used to create a serological sample repository. Serological methods from four manufacturers, namely Euroimmun, Roche, Abbott, and DiaSorin, were evaluated for quantitative, semi-quantitative, and qualitative antibody measurements. All four methods measure IgG antibodies against the SARS-CoV-2 spike receptor-binding domain and report the results in Binding Antibody Unit/mL (BAU/mL). A Total Error Allowable (TEa) of ±25% was chosen as the criteria to determine whether two methods are clinically equivalent quantitatively. Semi-quantitative results (titers) were derived using numeric antibody concentration divided by the cut-off value for each method. RESULTS All paired quantitative comparisons demonstrated unacceptable performance. With ±25% as TEa, the best agreement was 74 (35.2% out of 210 samples) between Euroimmun and DiaSorin, whereas the lowest agreement was 11 (5.2% out of 210 samples) between Euroimmun and Roche. Antibody titers amongst all four methods were significantly different (p < 0.001). The highest titer difference from the same sample is between Roche and DiaSorin with a 1392-fold difference. On qualitative comparison, none of the paired comparison showed acceptable comparison (p < 0.001). CONCLUSIONS Poor correlation exists between four evaluated assays, quantitatively, semi-quantitatively, and qualitatively. Further harmonization of assays is required to achieve comparable measurements.
Collapse
|
11
|
Shishido AA, Barnes AH, Narayanan S, Chua JV. COVID-19 Vaccines-All You Want to Know. Semin Respir Crit Care Med 2023; 44:143-172. [PMID: 36646092 DOI: 10.1055/s-0042-1759779] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2) pandemic has led to an unprecedented public health crisis. The collective global response has led to production of multiple safe and effective vaccines utilizing novel platforms to combat the virus that have propelled the field of vaccinology forward. Significant challenges to universal vaccine effectiveness remain, including immune evasion by SARS-CoV-2 variants, waning of immune response, inadequate knowledge of correlates of protection, and dosing in special populations. This review serves as a detailed evaluation of the development of the current SARS-CoV-2 vaccines, their effectiveness, and challenges to their deployment as a preventive tool.
Collapse
Affiliation(s)
- Akira A Shishido
- Division of Clinical Care and Research, Institute of Human Virology, University of Maryland School of Medicine, Baltimore, Maryland.,Division of Infectious Diseases, Virginia Commonwealth University, Richmond, Virginia
| | - Ashley H Barnes
- Division of Clinical Care and Research, Institute of Human Virology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Shivakumar Narayanan
- Division of Clinical Care and Research, Institute of Human Virology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Joel V Chua
- Division of Clinical Care and Research, Institute of Human Virology, University of Maryland School of Medicine, Baltimore, Maryland
| |
Collapse
|
12
|
Kaushik NK, Bhartiya P, Kaushik N, Shin Y, Nguyen LN, Park JS, Kim D, Choi EH. Nitric-oxide enriched plasma-activated water inactivates 229E coronavirus and alters antiviral response genes in human lung host cells. Bioact Mater 2023; 19:569-580. [PMID: 35574062 PMCID: PMC9080223 DOI: 10.1016/j.bioactmat.2022.05.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 04/20/2022] [Accepted: 05/03/2022] [Indexed: 12/18/2022] Open
Abstract
The ongoing pandemic caused by the novel coronavirus, SARS-CoV-2, is influencing global health. Moreover, there is a major threat of future coronaviruses affecting the entire world in a similar, or even more dreadful, manner. Therefore, effective and biocompatible therapeutic options against coronaviruses are urgently needed. To address this challenge, medical specialists require a well-informed and safe approach to treating human coronaviruses (HCoVs). Herein, an environmental friendly approach for viral inactivation, based on plasma technology, was considered. A microwave plasma system was employed for the generation of the high amount of gaseous nitric oxide to prepare nitric oxide enriched plasma-activated water (NO-PAW), the effects of which on coronaviruses, have not been reported to date. To determine these effects, alpha-HCoV-229E was used in an experimental model. We found that NO-PAW treatment effectively inhibited coronavirus infection in host lung cells, visualized by evaluating the cytopathic effect and expression level of spike proteins. Interestingly, NO-PAW showed minimal toxicity towards lung host cells, suggesting its potential for therapeutic application. Moreover, this new approach resulted in viral inactivation and greatly improved the gene levels involved in host antiviral responses. Together, our findings provide evidence of an initiation point for further progress toward the clinical development of antiviral treatments, including such coronaviruses.
Collapse
Affiliation(s)
- Nagendra Kumar Kaushik
- Department of Electrical and Biological Physics, Plasma Bioscience Research Center, Kwangwoon University, Seoul, 01897, Republic of Korea
| | - Pradeep Bhartiya
- Department of Electrical and Biological Physics, Plasma Bioscience Research Center, Kwangwoon University, Seoul, 01897, Republic of Korea
| | - Neha Kaushik
- Department of Biotechnology, College of Engineering, The University of Suwon, Hwaseong-si, 18323, Republic of Korea
| | - Yungoh Shin
- Department of Electrical and Biological Physics, Plasma Bioscience Research Center, Kwangwoon University, Seoul, 01897, Republic of Korea
| | - Linh Nhat Nguyen
- Department of Electrical and Biological Physics, Plasma Bioscience Research Center, Kwangwoon University, Seoul, 01897, Republic of Korea
| | - Jang Sick Park
- Department of Electrical and Biological Physics, Plasma Bioscience Research Center, Kwangwoon University, Seoul, 01897, Republic of Korea
| | - Doyoung Kim
- Department of Electrical and Biological Physics, Plasma Bioscience Research Center, Kwangwoon University, Seoul, 01897, Republic of Korea
| | - Eun Ha Choi
- Department of Electrical and Biological Physics, Plasma Bioscience Research Center, Kwangwoon University, Seoul, 01897, Republic of Korea
| |
Collapse
|
13
|
Shariatifar H, Farasat A. Affinity enhancement of CR3022 binding to RBD; in silico site directed mutagenesis using molecular dynamics simulation approaches. J Biomol Struct Dyn 2023; 41:81-90. [PMID: 34796779 DOI: 10.1080/07391102.2021.2004230] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a disease which caused by a novel beta coronavirus. Structural and non-structural proteins are expressed by the virus gene fragments. The RBD of the S1 protein of the virus has the ability to interact with potent antibodies including CR3022, which was characterized to target the S protein of the virus which can efficiently neutralize the SARS-CoV in vitro and in vivo. In current study, we aimed to design CR3022 based antibody with high affinity compared with wild-type CR3022 using MD simulation method. Two variants were designed based on the amino acid binding conformation and the free binding energy of the critical amino acids which involved in CR3022-RBD interactions were evaluated. In this study three complexes were evaluated; CR3022-RBD, V1-RBD and V2-RBD using molecular dynamics simulations carried out for 100 ns in each case. Then, all the complexes were simulated for 100 ns. In the next step, to calculate the free binding affinity of the wild CR3022 and mutant antibody (V1 and V2) with RBD, the PMF method was performed. The RMSD profile demonstrated that all three complexes were equilibrated after 85 ns. Furthermore, the free binding energy results indicated that the V2-RBD complex has the higher binding affinity than V1-RBD and CR3022-RBD complexes. It should be noted that in above variants, the electrostatic energy and the number of H-bonds between the antibody and RBD increased. Thus, it is suggested that both designed antibodies could be considered as appropriate candidates for covid-19 disease treatment.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Hanifeh Shariatifar
- Health Products Safety Research Center, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Alireza Farasat
- Cellular and Molecular Research Center, Research Institute for Prevention of Non-Communicable Diseases, Qazvin University of Medical Sciences, Qazvin, Iran
| |
Collapse
|
14
|
Zolfaghari MA, Ghadiri Moghaddam F, Rajput S, Karimi A, Naghi Vishteh M, Mahmoodpoor A, Dolati S, Yousefi M. SARS-CoV-2 vaccines: A double-edged sword throughout rapid evolution of COVID-19. Cell Biol Int 2022; 46:2009-2017. [PMID: 36047303 PMCID: PMC9539123 DOI: 10.1002/cbin.11903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 07/12/2022] [Accepted: 08/24/2022] [Indexed: 11/11/2022]
Abstract
After more than 2 years of the coronavirus disease 2019 pandemic caused by severe acute respiratory syndrome coronavirus 2, several questions have remained unanswered that affected our daily lives. Although substantial vaccine development could resist this challenge, emerging new variants in different countries could be considered as potent concerns regarding the adverse effects of reinfection or postvaccination. Precisely, these concerns address some significant and probable outcomes in vaccinated or reinfected models, followed by some virus challenges, such as antibody-dependent enhancement and cytokine storm. Therefore, the importance of evaluating the effectiveness of neutralizing antibodies (nAbs) elicited by vaccination and the rise of new variants must be addressed.
Collapse
Affiliation(s)
- Mohammad Ali Zolfaghari
- Student Research CommitteeTabriz University of Medical SciencesTabrizIran
- Department of Molecular Medicine, Faculty of Advanced Medical SciencesTabriz University of Medical SciencesTabrizIran
| | | | - Shabnam Rajput
- Department of Pediatrics, School of MedicineJahrom University of Medical SciencesJahromIran
| | - Abbas Karimi
- Department of Molecular Medicine, Faculty of Advanced Medical SciencesTabriz University of Medical SciencesTabrizIran
- Biotechnology Research CenterTabriz University of Medical SciencesTabrizIran
| | - Mohadeseh Naghi Vishteh
- Department of Genetics and Molecular Biology, School of MedicineIsfahan University of Medical SciencesIsfahanIran
| | - Ata Mahmoodpoor
- Department of AnesthesiologyTabriz University of Medical SciencesTabrizIran
| | - Sanam Dolati
- Physical Medicine and Rehabilitation Research Center, Aging Research InstituteTabriz University of Medical SciencesTabrizIran
| | - Mehdi Yousefi
- Stem Cell Research CenterTabriz University of Medical SciencesTabrizIran
- Department of Immunology, Faculty of MedicineTabriz University of Medical SciencesTabrizIran
| |
Collapse
|
15
|
Ali A, Zhang GF, Hu C, Yuan B, Jahan S, Kitsios GD, Morris A, Gao SJ, Panat R. Ultrarapid and ultrasensitive detection of SARS-CoV-2 antibodies in COVID-19 patients via a 3D-printed nanomaterial-based biosensing platform. J Med Virol 2022; 94:5808-5826. [PMID: 35981973 PMCID: PMC9538259 DOI: 10.1002/jmv.28075] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 08/07/2022] [Accepted: 08/17/2022] [Indexed: 01/06/2023]
Abstract
Rapid detection of antibodies during infection and after vaccination is critical for the control of infectious outbreaks, understanding immune response, and evaluating vaccine efficacy. In this manuscript, we evaluate a simple ultrarapid test for SARS-CoV-2 antibodies in COVID-19 patients, which gives quantitative results (i.e., antibody concentration) in 10-12 s using a previously reported nanomaterial-based three-dimensional (3D)-printed biosensing platform. This platform consists of a micropillar array electrode fabricated via 3D printing of aerosolized gold nanoparticles and coated with nanoflakes of graphene and specific SARS-CoV-2 antigens, including spike S1, S1 receptor-binding domain (RBD) and nucleocapsid (N). The sensor works on the principle of electrochemical transduction, where the change of sensor impedance is realized by the interactions between the viral proteins attached to the sensor electrode surface and the antibodies. The three sensors were used to test samples from 17 COVID-19 patients and 3 patients without COVID-19. Unlike other serological tests, the 3D sensors quantitatively detected antibodies at a concentration as low as picomole within 10-12 s in human plasma samples. We found that the studied COVID-19 patients had higher concentrations of antibodies to spike proteins (RBD and S1) than to the N protein. These results demonstrate the enormous potential of the rapid antibody test platform for understanding patients' immunity, disease epidemiology and vaccine efficacy, and facilitating the control and prevention of infectious epidemics.
Collapse
Affiliation(s)
- Azahar Ali
- Department of Mechanical Engineering, Carnegie Mellon
University, Pittsburgh, PA, 15213 USA
- Current address: Department of Animal and Poultry Sciences,
Virginia Tech, Blacksburg, VA, 24061 USA
| | - George Fei Zhang
- Cancer Virology Program, UPMC Hillman Cancer Center and
Department of Microbiology and Molecular Genetics, University of Pittsburgh School
of Medicine, Pittsburgh, PA, 15213 USA
| | - Chunshan Hu
- Department of Mechanical Engineering, Carnegie Mellon
University, Pittsburgh, PA, 15213 USA
| | - Bin Yuan
- Department of Mechanical Engineering, Carnegie Mellon
University, Pittsburgh, PA, 15213 USA
| | - Sanjida Jahan
- Department of Mechanical Engineering, Carnegie Mellon
University, Pittsburgh, PA, 15213 USA
| | - Georgios D. Kitsios
- Division of Pulmonary, Allergy and Critical Care Medicine,
Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA,
15213 USA
| | - Alison Morris
- Division of Pulmonary, Allergy and Critical Care Medicine,
Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA,
15213 USA
| | - Shou-Jiang Gao
- Cancer Virology Program, UPMC Hillman Cancer Center and
Department of Microbiology and Molecular Genetics, University of Pittsburgh School
of Medicine, Pittsburgh, PA, 15213 USA
| | - Rahul Panat
- Department of Mechanical Engineering, Carnegie Mellon
University, Pittsburgh, PA, 15213 USA
| |
Collapse
|
16
|
Babaeimarzangou SS, Zaker H, Soleimannezhadbari E, Gamchi NS, Kazeminia M, Tarighi S, Seyedian H, Tsatsakis A, Spandidos DA, Margina D. Vaccine development for zoonotic viral diseases caused by positive‑sense single‑stranded RNA viruses belonging to the Coronaviridae and Togaviridae families (Review). Exp Ther Med 2022; 25:42. [PMID: 36569444 PMCID: PMC9768462 DOI: 10.3892/etm.2022.11741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 11/10/2022] [Indexed: 12/02/2022] Open
Abstract
Outbreaks of zoonotic viral diseases pose a severe threat to public health and economies worldwide, with this currently being more prominent than it previously was human history. These emergency zoonotic diseases that originated and transmitted from vertebrates to humans have been estimated to account for approximately one billion cases of illness and have caused millions of deaths worldwide annually. The recent emergence of severe acute respiratory syndrome coronavirus-2 (coronavirus disease 2019) is an excellent example of the unpredictable public health threat causing a pandemic. The present review summarizes the literature data regarding the main vaccine developments in human clinical phase I, II and III trials against the zoonotic positive-sense single-stranded RNA viruses belonging to the Coronavirus and Alphavirus genera, including severe acute respiratory syndrome, Middle east respiratory syndrome, Venezuelan equine encephalitis virus, Semliki Forest virus, Ross River virus, Chikungunya virus and O'nyong-nyong virus. That there are neither vaccines nor effective antiviral drugs available against most of these viruses is undeniable. Therefore, new explosive outbreaks of these zoonotic viruses may surely be expected. The present comprehensive review provides an update on the status of vaccine development in different clinical trials against these viruses, as well as an overview of the present results of these trials.
Collapse
Affiliation(s)
- Seyed Sajjad Babaeimarzangou
- Division of Poultry Health and Diseases, Department of Clinical Sciences, Faculty of Veterinary Medicine, Urmia University, Urmia 5756151818, Iran
| | - Himasadat Zaker
- Histology and Microscopic Analysis Division, RASTA Specialized Research Institute (RSRI), West Azerbaijan Science and Technology Park (WASTP), Urmia 5756115322, Iran
| | | | - Naeimeh Shamsi Gamchi
- Histology and Microscopic Analysis Division, RASTA Specialized Research Institute (RSRI), West Azerbaijan Science and Technology Park (WASTP), Urmia 5756115322, Iran
| | - Masoud Kazeminia
- Department of Food Hygiene and Quality Control, Faculty of Veterinary Medicine, University of Tehran, Tehran 1417935840, Iran
| | - Shima Tarighi
- Veterinary Office of West Azerbaijan Province, Urmia 5717617695, Iran
| | - Homayon Seyedian
- Faculty of Veterinary Medicine, Urmia University, Urmia 5756151818, Iran
| | - Aristidis Tsatsakis
- Laboratory of Toxicology, Department of Medicine, University of Crete, 71307 Heraklion, Greece,Correspondence to: Professor Denisa Margina, Department of Biochemistry, Faculty of Pharmacy, ‘Carol Davila’ University of Medicine and Pharmacy, 6 Traian Vuia Street, 020956 Bucharest, Romania
| | - Demetrios A. Spandidos
- Laboratory of Clinical Virology, School of Medicine, University of Crete, 71003 Heraklion, Greece
| | - Denisa Margina
- Department of Biochemistry, Faculty of Pharmacy, ‘Carol Davila’ University of Medicine and Pharmacy, 020956 Bucharest, Romania,Correspondence to: Professor Denisa Margina, Department of Biochemistry, Faculty of Pharmacy, ‘Carol Davila’ University of Medicine and Pharmacy, 6 Traian Vuia Street, 020956 Bucharest, Romania
| |
Collapse
|
17
|
Yang N, Garcia A, Meyer C, Tuschl T, Merghoub T, Wolchok JD, Deng L. Heat-inactivated modified vaccinia virus Ankara boosts Th1 cellular and humoral immunity as a vaccine adjuvant. NPJ Vaccines 2022; 7:120. [PMID: 36261460 PMCID: PMC9580433 DOI: 10.1038/s41541-022-00542-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 09/26/2022] [Indexed: 12/02/2022] Open
Abstract
Protein or peptide-based subunit vaccines have generated excitement and renewed interest in combating human cancer or COVID-19 outbreak. One major concern for subunit vaccine application is the weak immune responses induced by protein or peptides. Developing novel and effective vaccine adjuvants are critical for the success of subunit vaccines. Here we explored the potential of heat-inactivated MVA (heat-iMVA) as a vaccine adjuvant. Heat-iMVA dramatically enhances T cell responses and antibodies responses, mainly toward Th1 immune responses when combined with protein or peptide-based immunogen. The adjuvant effect of Heat-iMVA is stronger than live MVA and is dependent on the cGAS/STING-mediated cytosolic DNA-sensing pathway. In a therapeutic vaccination model based on tumor neoantigen peptide vaccine, Heat-iMVA significantly extended the survival and delayed tumor growth. When combined with SARS-CoV-2 spike protein, Heat-iMVA induced more robust spike-specific antibody production and more potent neutralization antibodies. Our results support that Heat-iMVA can be developed as a safe and potent vaccine adjuvant for subunit vaccines against cancer or SARS-CoV-2.
Collapse
Affiliation(s)
- Ning Yang
- Dermatology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Aitor Garcia
- Laboratory of RNA Molecular Biology, The Rockefeller University, New York, NY, 10065, USA
| | - Cindy Meyer
- Laboratory of RNA Molecular Biology, The Rockefeller University, New York, NY, 10065, USA
| | - Thomas Tuschl
- Laboratory of RNA Molecular Biology, The Rockefeller University, New York, NY, 10065, USA
| | - Taha Merghoub
- Immuno-oncology service, Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
- Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - Jedd D Wolchok
- Immuno-oncology service, Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
- Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - Liang Deng
- Dermatology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA.
- Immuno-oncology service, Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA.
- Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Weill Cornell Medical College, New York, NY, USA.
| |
Collapse
|
18
|
Masoomi Nomandan SZ, Azimzadeh Irani M, Hosseini SM. In silico design of refined ferritin-SARS-CoV-2 glyco-RBD nanoparticle vaccine. Front Mol Biosci 2022; 9:976490. [PMID: 36148012 PMCID: PMC9486171 DOI: 10.3389/fmolb.2022.976490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 08/11/2022] [Indexed: 12/04/2022] Open
Abstract
With the onset of Coronavirus disease 2019 (COVID-19) pandemic, all attention was drawn to finding solutions to cure the coronavirus disease. Among all vaccination strategies, the nanoparticle vaccine has been shown to stimulate the immune system and provide optimal immunity to the virus in a single dose. Ferritin is a reliable self-assembled nanoparticle platform for vaccine production that has already been used in experimental studies. Furthermore, glycosylation plays a crucial role in the design of antibodies and vaccines and is an essential element in developing effective subunit vaccines. In this computational study, ferritin nanoparticles and glycosylation, which are two unique facets of vaccine design, were used to model improved nanoparticle vaccines for the first time. In this regard, molecular modeling and molecular dynamics simulation were carried out to construct three atomistic models of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) receptor binding domain (RBD)-ferritin nanoparticle vaccine, including unglycosylated, glycosylated, and modified with additional O-glycans at the ferritin–RBD interface. It was shown that the ferritin–RBD complex becomes more stable when glycans are added to the ferritin–RBD interface and optimal performance of this nanoparticle can be achieved. If validated experimentally, these findings could improve the design of nanoparticles against all microbial infections.
Collapse
|
19
|
Nurpeisova A, Khairullin B, Abitaev R, Shorayeva K, Jekebekov K, Kalimolda E, Kerimbayev A, Akylbayeva K, Abay Z, Myrzakhmetova B, Nakhanov A, Absatova Z, Nurabayev S, Orynbayev M, Assanzhanova N, Abeuov K, Kutumbetov L, Kassenov M, Abduraimov Y, Zakarya K. Safety and immunogenicity of the first Kazakh inactivated vaccine for COVID-19. Hum Vaccin Immunother 2022; 18:2087412. [PMID: 35960911 DOI: 10.1080/21645515.2022.2087412] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
This article describes the results of a preclinical safety and immunogenicity study of QazCovid-in®, the first COVID-19 vaccine developed in Kazakhstan, on BALB/c mice, rats, ferrets, Syrian hamsters and rhesus macaques (Macaca mulatta). The study's safety data suggests that this immunobiological preparation can be technically considered a Class 5 nontoxic vaccine. The series of injections that were made did not produce any adverse effect or any change in the general condition of the model animals' health, while macroscopy and histology studies identified no changes in the internal organs of the BALB/c mice and rats. This study has demonstrated that a double immunization enhances the growth of antibody titers as assessed by the microneutralization assay (MNA) and the enzyme-linked immunosorbent assay (ELISA) in a pre-clinical immunogenicity test on animal models. The best GMT results were assessed in MNA and ELISA 7 days after re-vaccination; however, we noted that GMT antibody results in ELISA were lower than in MNA. A comparative GMT assessment after the first immunization and the re-immunization identified significant differences between model animal groups and a growth of GMT antibodies in all of them; also, differences between the gender groups were statistically significant. Moreover, the most marked MNA immune response to the QazCovid-in® vaccine was seen in the Syrian hamsters, while their SARS-CoV-2-specific antibody activity as assessed with ELISA was the lowest.
Collapse
Affiliation(s)
- Ainur Nurpeisova
- Research Institute for Biological Safety Problems (RIBSP), Guardeyskiy, Kazakhstan
| | - Berik Khairullin
- Research Institute for Biological Safety Problems (RIBSP), Guardeyskiy, Kazakhstan
| | - Ruslan Abitaev
- Research Institute for Biological Safety Problems (RIBSP), Guardeyskiy, Kazakhstan
| | - Kamshat Shorayeva
- Research Institute for Biological Safety Problems (RIBSP), Guardeyskiy, Kazakhstan
| | - Kuanish Jekebekov
- Research Institute for Biological Safety Problems (RIBSP), Guardeyskiy, Kazakhstan
| | - Elina Kalimolda
- Research Institute for Biological Safety Problems (RIBSP), Guardeyskiy, Kazakhstan
| | - Aslan Kerimbayev
- Research Institute for Biological Safety Problems (RIBSP), Guardeyskiy, Kazakhstan
| | - Karligash Akylbayeva
- Research Institute for Biological Safety Problems (RIBSP), Guardeyskiy, Kazakhstan
| | - Zhandos Abay
- Research Institute for Biological Safety Problems (RIBSP), Guardeyskiy, Kazakhstan
| | | | - Aziz Nakhanov
- Research Institute for Biological Safety Problems (RIBSP), Guardeyskiy, Kazakhstan
| | - Zharkinay Absatova
- Research Institute for Biological Safety Problems (RIBSP), Guardeyskiy, Kazakhstan
| | - Sergazy Nurabayev
- Research Institute for Biological Safety Problems (RIBSP), Guardeyskiy, Kazakhstan
| | - Mukhit Orynbayev
- Research Institute for Biological Safety Problems (RIBSP), Guardeyskiy, Kazakhstan
| | - Nurika Assanzhanova
- Research Institute for Biological Safety Problems (RIBSP), Guardeyskiy, Kazakhstan
| | - Khairulla Abeuov
- Research Institute for Biological Safety Problems (RIBSP), Guardeyskiy, Kazakhstan
| | - Lespek Kutumbetov
- Research Institute for Biological Safety Problems (RIBSP), Guardeyskiy, Kazakhstan
| | - Markhabat Kassenov
- Research Institute for Biological Safety Problems (RIBSP), Guardeyskiy, Kazakhstan
| | - Yergaly Abduraimov
- Research Institute for Biological Safety Problems (RIBSP), Guardeyskiy, Kazakhstan
| | - Kunsulu Zakarya
- Research Institute for Biological Safety Problems (RIBSP), Guardeyskiy, Kazakhstan
| |
Collapse
|
20
|
Prediction of Conserved HLA Class I and Class II Epitopes from SARS-CoV-2 Licensed Vaccines Supports T-Cell Cross-Protection against SARS-CoV-1. Biomedicines 2022; 10:biomedicines10071622. [PMID: 35884927 PMCID: PMC9313420 DOI: 10.3390/biomedicines10071622] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 06/28/2022] [Accepted: 07/05/2022] [Indexed: 11/17/2022] Open
Abstract
Heterologous immunity-inducing vaccines against different pathogens are necessary to deal with new pandemics. In this study, the possible impact of COVID-19 licensed formulations in the cytotoxic and the helper cellular immune responses against SARS-CoV-1 is analyzed for the 567 and 41 most abundant HLA class I and II alleles, respectively. Computational prediction showed that most of these 608 alleles, which cover >90% of the human population, contain enough conserved T-cell epitopes among SARS-CoV-1 and SARS-CoV-2 spike proteins. In addition, the vast majority of these predicted peptides were defined as epitopes recognized by CD4+ or CD8+ T lymphocytes, showing a very high correlation between the bioinformatics prediction and the experimental assays. These data suggest that both cytotoxic and helper cellular immune protection elicited by the currently licensed COVID-19 vaccines should be effective against SARS-CoV-1 infection. Lastly, this study has potential implications for public health against current and future pandemics, given that the SARS-CoV-1 vaccines in pipeline since the early 20th century could generate similarly cross-protection against COVID-19.
Collapse
|
21
|
Lin Q, Lu C, Hong Y, Li R, Chen J, Chen W, Chen J. Animal models for studying coronavirus infections and developing antiviral agents and vaccines. Antiviral Res 2022; 203:105345. [PMID: 35605699 PMCID: PMC9122840 DOI: 10.1016/j.antiviral.2022.105345] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 04/30/2022] [Accepted: 05/17/2022] [Indexed: 01/17/2023]
Abstract
In addition to severe acute respiratory syndrome coronavirus (SARS-CoV) and Middle East respiratory syndrome coronavirus (MERS-CoV), SARS-CoV-2 has become the third deadly coronavirus that infects humans and causes the new coronavirus disease (COVID-19). COVID-19 has already caused more than six million deaths worldwide and it is likely the biggest pandemic of this century faced by mankind. Although many studies on SARS-CoV-2 have been conducted, a detailed understanding of SARS-CoV-2 and COVID-19 is still lacking. Animal models are indispensable for studying its pathogenesis and developing vaccines and antivirals. In this review, we analyze animal models of coronavirus infections and explore their applications on antivirals and vaccines.
Collapse
Affiliation(s)
- Qisheng Lin
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Chunni Lu
- Centre for Inflammatory Diseases, Monash University Department of Medicine, Monash Medical Centre, Monash University, Clayton, Victoria 3168, Australia
| | - Yuqi Hong
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Runfeng Li
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| | - Jinding Chen
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Weisan Chen
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, 3086, Australia.
| | - Jianxin Chen
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China.
| |
Collapse
|
22
|
Cicaloni V, Costanti F, Pasqui A, Bianchini M, Niccolai N, Bongini P. A Bioinformatics Approach to Investigate Structural and Non-Structural Proteins in Human Coronaviruses. Front Genet 2022; 13:891418. [PMID: 35774504 PMCID: PMC9237418 DOI: 10.3389/fgene.2022.891418] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 05/04/2022] [Indexed: 11/13/2022] Open
Abstract
Recent studies confirmed that people unexposed to SARS-CoV-2 have preexisting reactivity, probably due to previous exposure to widely circulating common cold coronaviruses. Such preexistent reactivity against SARS-CoV-2 comes from memory T cells that can specifically recognize a SARS-CoV-2 epitope of structural and non-structural proteins and the homologous epitopes from common cold coronaviruses. Therefore, it is important to understand the SARS-CoV-2 cross-reactivity by investigating these protein sequence similarities with those of different circulating coronaviruses. In addition, the emerging SARS-CoV-2 variants lead to an intense interest in whether mutations in proteins (especially in the spike) could potentially compromise vaccine effectiveness. Since it is not clear that the differences in clinical outcomes are caused by common cold coronaviruses, a deeper investigation on cross-reactive T-cell immunity to SARS-CoV-2 is crucial to examine the differential COVID-19 symptoms and vaccine performance. Therefore, the present study can be a starting point for further research on cross-reactive T cell recognition between circulating common cold coronaviruses and SARS-CoV-2, including the most recent variants Delta and Omicron. In the end, a deep learning approach, based on Siamese networks, is proposed to accurately and efficiently calculate a BLAST-like similarity score between protein sequences.
Collapse
Affiliation(s)
| | - Filippo Costanti
- Department of Information Engineering and Mathematics, University of Siena, Siena, Italy
| | | | - Monica Bianchini
- Department of Information Engineering and Mathematics, University of Siena, Siena, Italy
| | - Neri Niccolai
- Department of Biotechnology, Chemistry, and Pharmacy, University of Siena, Siena, Italy
| | - Pietro Bongini
- Department of Information Engineering and Mathematics, University of Siena, Siena, Italy
- Department of Information Engineering, University of Florence, Firenze, Italy
- *Correspondence: Pietro Bongini,
| |
Collapse
|
23
|
McHenry A, Iyer K, Wang J, Liu C, Harigopal M. Detection of SARS-CoV-2 in tissue: the comparative roles of RT-qPCR, in situ RNA hybridization, and immunohistochemistry. Expert Rev Mol Diagn 2022; 22:559-574. [PMID: 35658709 DOI: 10.1080/14737159.2022.2085508] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION The emergence of SARS-CoV-2, the causative agent the COVID-19 pandemic, has led to a rapidly expanding arsenal of molecular diagnostic assays for the detection of viral material in tissue specimens. AREAS COVERED We review the value and shortcomings of available tissue-based assays for SARS-CoV-2 detection in formalin-fixed paraffin-embedded (FFPE) tissue, including immunohistochemistry, in situ hybridization, and quantitative reverse transcription PCR (RT-qPCR). The validation, accuracy, and comparative utility of each method is discussed. Subsequently, we identify commercially available antibodies which render the greatest specificity and reproducibility of staining in FFPE specimens. EXPERT OPINION We offer expert opinion on the efficacy of such techniques and guidance for future implementation, both clinical and experimental.
Collapse
Affiliation(s)
- Austin McHenry
- Yale University School of Medicine, Department of Pathology, New Haven, CT, 06520, United States
| | - Krishna Iyer
- Yale University School of Medicine, Department of Pathology, New Haven, CT, 06520, United States
| | - Jianhi Wang
- Yale University School of Medicine, Department of Pathology, New Haven, CT, 06520, United States
| | - Chen Liu
- Yale University School of Medicine, Department of Pathology, New Haven, CT, 06520, United States
| | - Malini Harigopal
- Yale University School of Medicine, Department of Pathology, New Haven, CT, 06520, United States
| |
Collapse
|
24
|
Ávalos I, Lao T, Rodríguez EM, Zamora Y, Rodríguez A, Ramón A, Lemos G, Cabrales A, Bequet-Romero M, Casillas D, Andújar I, Espinosa LA, González LJ, Alvarez Y, Carpio Y, Estrada MP. Chimeric Antigen by the Fusion of SARS-CoV-2 Receptor Binding Domain with the Extracellular Domain of Human CD154: A Promising Improved Vaccine Candidate. Vaccines (Basel) 2022; 10:897. [PMID: 35746505 PMCID: PMC9228316 DOI: 10.3390/vaccines10060897] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 05/26/2022] [Accepted: 05/27/2022] [Indexed: 02/04/2023] Open
Abstract
COVID-19 is a respiratory viral disease caused by a new coronavirus called SARS-CoV-2. This disease has spread rapidly worldwide with a high rate of morbidity and mortality. The receptor-binding domain (RBD) of protein spike (S) mediates the attachment of the virus to the host's cellular receptor. The RBD domain constitutes a very attractive target for subunit vaccine development due to its ability to induce a neutralizing antibody response against the virus. With the aim of boosting the immunogenicity of RBD, it was fused to the extracellular domain of CD154, an immune system modulator molecule. To obtain the chimeric protein, stable transduction of HEK-293 was carried out with recombinant lentivirus and polyclonal populations and cell clones were obtained. RBD-CD was purified from culture supernatant and further characterized by several techniques. RBD-CD immunogenicity evaluated in mice and non-human primates (NHP) indicated that recombinant protein was able to induce a specific and high IgG response after two doses. NHP sera also neutralize SARS-CoV-2 infection of Vero E6 cells. RBD-CD could improve the current vaccines against COVID-19, based in the enhancement of the host humoral and cellular response. Further experiments are necessary to confirm the utility of RBD-CD as a prophylactic vaccine and/or booster purpose.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Yamila Carpio
- Center for Genetic Engineering and Biotechnology, CIGB, Ave. 31 E/158 y 190, Havana 10600, Cuba; (I.Á.); (T.L.); (E.M.R.); (Y.Z.); (A.R.); (A.R.); (G.L.); (A.C.); (M.B.-R.); (D.C.); (I.A.); (L.A.E.); (L.J.G.); (Y.A.)
| | - Mario Pablo Estrada
- Center for Genetic Engineering and Biotechnology, CIGB, Ave. 31 E/158 y 190, Havana 10600, Cuba; (I.Á.); (T.L.); (E.M.R.); (Y.Z.); (A.R.); (A.R.); (G.L.); (A.C.); (M.B.-R.); (D.C.); (I.A.); (L.A.E.); (L.J.G.); (Y.A.)
| |
Collapse
|
25
|
Boulton S, Poutou J, Martin NT, Azad T, Singaravelu R, Crupi MJF, Jamieson T, He X, Marius R, Petryk J, Tanese de Souza C, Austin B, Taha Z, Whelan J, Khan ST, Pelin A, Rezaei R, Surendran A, Tucker S, Fekete EEF, Dave J, Diallo JS, Auer R, Angel JB, Cameron DW, Cailhier JF, Lapointe R, Potts K, Mahoney DJ, Bell JC, Ilkow CS. Single-dose replicating poxvirus vector-based RBD vaccine drives robust humoral and T cell immune response against SARS-CoV-2 infection. Mol Ther 2022; 30:1885-1896. [PMID: 34687845 PMCID: PMC8527104 DOI: 10.1016/j.ymthe.2021.10.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/24/2021] [Accepted: 10/10/2021] [Indexed: 02/01/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic requires the continued development of safe, long-lasting, and efficacious vaccines for preventive responses to major outbreaks around the world, and especially in isolated and developing countries. To combat severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), we characterize a temperature-stable vaccine candidate (TOH-Vac1) that uses a replication-competent, attenuated vaccinia virus as a vector to express a membrane-tethered spike receptor binding domain (RBD) antigen. We evaluate the effects of dose escalation and administration routes on vaccine safety, efficacy, and immunogenicity in animal models. Our vaccine induces high levels of SARS-CoV-2 neutralizing antibodies and favorable T cell responses, while maintaining an optimal safety profile in mice and cynomolgus macaques. We demonstrate robust immune responses and protective immunity against SARS-CoV-2 variants after only a single dose. Together, these findings support further development of our novel and versatile vaccine platform as an alternative or complementary approach to current vaccines.
Collapse
Affiliation(s)
- Stephen Boulton
- Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Joanna Poutou
- Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Nikolas T Martin
- Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Taha Azad
- Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Ragunath Singaravelu
- Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Mathieu J F Crupi
- Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Taylor Jamieson
- Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Xiaohong He
- Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Ricardo Marius
- Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Julia Petryk
- Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Christiano Tanese de Souza
- Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Bradley Austin
- Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Zaid Taha
- Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Jack Whelan
- Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Sarwat T Khan
- Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
| | - Adrian Pelin
- Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Reza Rezaei
- Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Abera Surendran
- Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Sarah Tucker
- Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Emily E F Fekete
- Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Jaahnavi Dave
- Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Jean-Simon Diallo
- Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Rebecca Auer
- Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Jonathan B Angel
- Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada; Department of Medicine, The Ottawa Hospital, Ottawa, ON K1H 8L6, Canada
| | - D William Cameron
- Division of Infectious Disease, Department of Medicine, University of Ottawa at The Ottawa Hospital/ Research Institute, Ottawa, ON K1H 8L6, Canada
| | | | - Réjean Lapointe
- Institut du Cancer de Montréal, Montréal, Québec H2X 0A9, Canada
| | - Kyle Potts
- Arnie Charbonneau Cancer Institute, Calgary, AB T2N 4Z6, Canada; Alberta Children's Hospital Research Institute, Calgary, AB T2N 6A8, Canada; Department of Microbiology, Immunology and Infectious Disease, Cumming School of Medicine, University of Calgary, Calgary, AB T2T 1N4, Canada
| | - Douglas J Mahoney
- Arnie Charbonneau Cancer Institute, Calgary, AB T2N 4Z6, Canada; Alberta Children's Hospital Research Institute, Calgary, AB T2N 6A8, Canada; Department of Microbiology, Immunology and Infectious Disease, Cumming School of Medicine, University of Calgary, Calgary, AB T2T 1N4, Canada
| | - John C Bell
- Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada.
| | - Carolina S Ilkow
- Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada.
| |
Collapse
|
26
|
Sadeghalvad M, Mansourabadi AH, Noori M, Nejadghaderi SA, Masoomikarimi M, Alimohammadi M, Rezaei N. Recent developments in SARS-CoV-2 vaccines: A systematic review of the current studies. Rev Med Virol 2022; 33:e2359. [PMID: 35491495 PMCID: PMC9348268 DOI: 10.1002/rmv.2359] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 04/11/2022] [Accepted: 04/14/2022] [Indexed: 01/28/2023]
Abstract
Designing and manufacturing efficient vaccines against coronavirus disease 2019 (COVID-19) is a major objective. In this systematic review, we aimed to evaluate the most important vaccines under construction worldwide, their efficiencies and clinical results in healthy individuals and in those with specific underlying diseases. We conducted a comprehensive search in PubMed, Scopus, EMBASE, and Web of Sciences by 1 December 2021 to identify published research studies. The inclusion criteria were publications that evaluated the immune responses and safety of COVID-19 vaccines in healthy individuals and in those with pre-existing diseases. We also searched the VAERS database to estimate the incidence of adverse events of special interest (AESI) post COVID-19 vaccination. Almost all investigated vaccines were well tolerated and developed good levels of both humoural and cellular responses. A protective and efficient humoural immune response develops after the second or third dose of vaccine and a longer interval (about 28 days) between the first and second injections of vaccine could induce higher antibody responses. The vaccines were less immunogenic in immunocompromised patients, particularly those with haematological malignancies. In addition, we found that venous and arterial thrombotic events, Bell's palsy, and myocarditis/pericarditis were the most common AESI. The results showed the potency of the SARS-CoV-2 vaccines to protect subjects against disease. The provision of further effective and safe vaccines is necessary in order to reach a high coverage of immunisation programs across the globe and to provide protection against infection itself.
Collapse
Affiliation(s)
- Mona Sadeghalvad
- Department of ImmunologySchool of MedicineTehran University of Medical SciencesTehranIran
| | | | - Maryam Noori
- Student Research Committee, School of MedicineIran University of Medical SciencesTehranIran,Urology Research CenterTehran University of Medical SciencesTehranIran
| | - Seyed Aria Nejadghaderi
- Systematic Review and Meta‐Analysis Expert Group (SRMEG)Universal Scientific Education and Research Network (USERN)TehranIran,School of MedicineShahid Beheshti University of Medical SciencesTehranIran
| | - Masoomeh Masoomikarimi
- Department of ImmunologySchool of MedicineTehran University of Medical SciencesTehranIran
| | - Masoumeh Alimohammadi
- Department of ImmunologySchool of MedicineTehran University of Medical SciencesTehranIran,Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA)Universal Scientific Education and Research Network (USERN)TehranIran,Research Center for ImmunodeficienciesChildren's Medical CenterTehran University of Medical SciencesTehranIran
| | - Nima Rezaei
- Department of ImmunologySchool of MedicineTehran University of Medical SciencesTehranIran,Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA)Universal Scientific Education and Research Network (USERN)TehranIran,Research Center for ImmunodeficienciesChildren's Medical CenterTehran University of Medical SciencesTehranIran
| |
Collapse
|
27
|
Advances in Targeting ACE2 for Developing COVID-19 Therapeutics. Ann Biomed Eng 2022; 50:1734-1749. [PMID: 36261668 PMCID: PMC9581451 DOI: 10.1007/s10439-022-03094-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 09/29/2022] [Indexed: 01/01/2023]
Abstract
Since the onset of the coronavirus pandemic in December 2019, the SARS-CoV-2 virus has accounted for over 6.3 million lives resulting in the demand to develop novel therapeutic approaches to target and treat SARS-CoV-2. Improved understanding of viral entry and infection mechanisms has led to identifying different target receptors to mitigate infection in the host. Researchers have been working on identifying and targeting potential therapeutic target receptors utilizing different candidate drugs. Angiotensin-converting enzyme-2 (ACE2) has been known to perform critical functions in maintaining healthy cardiorespiratory function. However, ACE2 also functions as the binding site for the spike protein of SARS-CoV-2, allowing the virus to enter the cells and ensue infection. Therefore, drugs targeting ACE2 receptors can be considered as therapeutic candidates. Strategies targeting the level of ACE2 expression have been investigated and compared to other potential therapeutic targets, such as TMPRSS2, RdRp, and DPP4. This mini review discusses the key therapeutic approaches that target the ACE2 receptor, which is critical to the cellular entry and propagation of the novel SARS-CoV-2. In addition, we summarize the main advantages of ACE2 targeting against alternative approaches for the treatment of COVID-19.
Collapse
|
28
|
Response kinetics of different classes of antibodies to SARS-CoV2 infection in the Japanese population: The IgA and IgG titers increased earlier than the IgM titers. Int Immunopharmacol 2021; 103:108491. [PMID: 34954559 PMCID: PMC8687758 DOI: 10.1016/j.intimp.2021.108491] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 12/14/2021] [Accepted: 12/17/2021] [Indexed: 12/13/2022]
Abstract
To better understand the immune responses to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection in individuals with COVID-19, it is important to investigate the kinetics of the antibody responses and their associations with the clinical course in different populations, since there seem to be considerable differences between Western and Asian populations in the clinical features and spread of COVID-19. In this study, we serially measured the serum titers of IgM, IgG and IgA antibodies generated against the nucleocapsid protein (NCP), S1 subunit of the spike protein (S1), and receptor-binding domain in the S1 subunit (RBD) of SARS-CoV-2 in Japanese individuals with COVID-19. Among the IgM, IgG, and IgA antibodies, IgA antibodies against all of the aforementioned viral proteins were the first to appear after the infection, and IgG and/or IgA seroconversion often preceded IgM seroconversion. In regard to the timeline of the antibody responses to the different viral proteins (NCP, S1 and RBD), IgA against NCP appeared than IgA against S1 or RBD, while IgM and IgG against S1 appeared earlier than IgM/IgG against NCP or RBD. The IgG responses to all three viral proteins and responses of all three antibody classes to S1 and RBD were sustained for longer durations than the IgA/IgM responses to all three viral proteins and responses of all three antibody classes to NCP, respectively. The seroconversion of IgA against NCP occurred later and less frequently in patients with mild COVID-19. These results suggest possible differences in the antibody responses to SARS-CoV-2 antigens between the Japanese and Western populations.
Collapse
|
29
|
Dangi T, Palacio N, Sanchez S, Park M, Class J, Visvabharathy L, Ciucci T, Koralnik IJ, Richner JM, Penaloza-MacMaster P. Cross-protective immunity following coronavirus vaccination and coronavirus infection. J Clin Invest 2021; 131:151969. [PMID: 34623973 DOI: 10.1172/jci151969] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 10/07/2021] [Indexed: 12/16/2022] Open
Abstract
Although severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccines have shown efficacy against SARS-CoV-2, it is unknown if coronavirus vaccines can also protect against other coronaviruses that may infect humans in the future. Here, we show that coronavirus vaccines elicited cross-protective immune responses against heterologous coronaviruses. In particular, we show that a severe acute respiratory syndrome coronavirus 1 (SARS-CoV-1) vaccine developed in 2004 and known to protect against SARS-CoV-1 conferred robust heterologous protection against SARS-CoV-2 in mice. Similarly, prior coronavirus infections conferred heterologous protection against distinct coronaviruses. Cross-reactive immunity was also reported in patients with coronavirus disease 2019 (COVID-19) and in individuals who received SARS-CoV-2 vaccines, and transfer of plasma from these individuals into mice improved protection against coronavirus challenges. These findings provide the first demonstration to our knowledge that coronavirus vaccines (and prior coronavirus infections) can confer broad protection against heterologous coronaviruses and establish a rationale for universal coronavirus vaccines.
Collapse
Affiliation(s)
- Tanushree Dangi
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Nicole Palacio
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Sarah Sanchez
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Mincheol Park
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Jacob Class
- Department of Microbiology and Immunology, University of Illinois at Chicago (UIC), Chicago, Illinois, USA
| | - Lavanya Visvabharathy
- Ken and Ruth Davee Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Thomas Ciucci
- David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, New York, USA.,Department of Microbiology and Immunology, Center for Vaccine Biology and Immunology, University of Rochester, Rochester, New York, USA
| | - Igor J Koralnik
- Ken and Ruth Davee Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Justin M Richner
- Department of Microbiology and Immunology, University of Illinois at Chicago (UIC), Chicago, Illinois, USA
| | - Pablo Penaloza-MacMaster
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
30
|
Zhuang Z, Liu D, Sun J, Li F, Zhao J. Immune responses to human respiratory coronaviruses infection in mouse models. Curr Opin Virol 2021; 52:102-111. [PMID: 34906757 PMCID: PMC8665230 DOI: 10.1016/j.coviro.2021.11.015] [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: 07/31/2021] [Revised: 11/20/2021] [Accepted: 11/23/2021] [Indexed: 12/23/2022]
Abstract
Human respiratory coronaviruses (HCoVs), including the recently emerged SARS-CoV-2, the causative agent of the coronavirus disease 2019 (COVID-19) pandemic, potentially cause severe lung infections and multiple organ damages, emphasizing the urgent need for antiviral therapeutics and vaccines against HCoVs. Small animal models, especially mice, are ideal tools for deciphering the pathogenesis of HCoV infections as well as virus-induced immune responses, which is critical for antiviral drug development and vaccine design. In this review, we focus on the antiviral innate immune response, antibody response and T cell response in HCoV infected mouse models, and discuss the potential implications for understanding the anti-HCoV immunity and fighting the COVID-19 pandemic.
Collapse
Affiliation(s)
- Zhen Zhuang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510182, China
| | - Donglan Liu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510182, China
| | - Jing Sun
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510182, China
| | - Fang Li
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510182, China
| | - Jincun Zhao
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510182, China; Guangzhou Laboratory, Bio-Island, Guangzhou, Guangdong 510320, China.
| |
Collapse
|
31
|
Cordeiro AS, Patil-Sen Y, Shivkumar M, Patel R, Khedr A, Elsawy MA. Nanovaccine Delivery Approaches and Advanced Delivery Systems for the Prevention of Viral Infections: From Development to Clinical Application. Pharmaceutics 2021; 13:2091. [PMID: 34959372 PMCID: PMC8707864 DOI: 10.3390/pharmaceutics13122091] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 11/30/2021] [Accepted: 12/01/2021] [Indexed: 02/07/2023] Open
Abstract
Viral infections causing pandemics and chronic diseases are the main culprits implicated in devastating global clinical and socioeconomic impacts, as clearly manifested during the current COVID-19 pandemic. Immunoprophylaxis via mass immunisation with vaccines has been shown to be an efficient strategy to control such viral infections, with the successful and recently accelerated development of different types of vaccines, thanks to the advanced biotechnological techniques involved in the upstream and downstream processing of these products. However, there is still much work to be done for the improvement of efficacy and safety when it comes to the choice of delivery systems, formulations, dosage form and route of administration, which are not only crucial for immunisation effectiveness, but also for vaccine stability, dose frequency, patient convenience and logistics for mass immunisation. In this review, we discuss the main vaccine delivery systems and associated challenges, as well as the recent success in developing nanomaterials-based and advanced delivery systems to tackle these challenges. Manufacturing and regulatory requirements for the development of these systems for successful clinical and marketing authorisation were also considered. Here, we comprehensively review nanovaccines from development to clinical application, which will be relevant to vaccine developers, regulators, and clinicians.
Collapse
Affiliation(s)
- Ana Sara Cordeiro
- Leicester Institute for Pharmaceutical Innovation, Leicester School of Pharmacy, De Montfort University, Leicester LE1 9BH, UK; (A.S.C.); (M.S.); (A.K.)
| | - Yogita Patil-Sen
- Wrightington, Wigan and Leigh Teaching Hospitals NHS Foundation Trust, National Health Service, Wigan WN6 0SZ, UK;
| | - Maitreyi Shivkumar
- Leicester Institute for Pharmaceutical Innovation, Leicester School of Pharmacy, De Montfort University, Leicester LE1 9BH, UK; (A.S.C.); (M.S.); (A.K.)
| | - Ronak Patel
- School of Pharmacy and Biomedical Sciences, University of Central Lancashire, Preston PR1 2HE, UK;
| | - Abdulwahhab Khedr
- Leicester Institute for Pharmaceutical Innovation, Leicester School of Pharmacy, De Montfort University, Leicester LE1 9BH, UK; (A.S.C.); (M.S.); (A.K.)
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
| | - Mohamed A. Elsawy
- Leicester Institute for Pharmaceutical Innovation, Leicester School of Pharmacy, De Montfort University, Leicester LE1 9BH, UK; (A.S.C.); (M.S.); (A.K.)
| |
Collapse
|
32
|
MVA vector expression of SARS-CoV-2 spike protein and protection of adult Syrian hamsters against SARS-CoV-2 challenge. NPJ Vaccines 2021; 6:145. [PMID: 34862398 PMCID: PMC8642471 DOI: 10.1038/s41541-021-00410-8] [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: 06/25/2021] [Accepted: 11/09/2021] [Indexed: 12/19/2022] Open
Abstract
Numerous vaccine candidates against SARS-CoV-2, the causative agent of the COVID-19 pandemic, are under development. The majority of vaccine candidates to date are designed to induce immune responses against the viral spike (S) protein, although different forms of S antigen have been incorporated. To evaluate the yield and immunogenicity of different forms of S, we constructed modified vaccinia virus Ankara (MVA) vectors expressing full-length S (MVA-S), the RBD, and soluble S ectodomain and tested their immunogenicity in dose-ranging studies in mice. All three MVA vectors induced spike-specific immunoglobulin G after one subcutaneous immunization and serum titers were boosted following a second immunization. The MVA-S and MVA-ssM elicited the strongest neutralizing antibody responses. In assessing protective efficacy, MVA-S-immunized adult Syrian hamsters were challenged with SARS-CoV-2 (USA/WA1/2020). MVA-S-vaccinated hamsters exhibited less severe manifestations of atypical pneumocyte hyperplasia, hemorrhage, vasculitis, and especially consolidation, compared to control animals. They also displayed significant reductions in gross pathology scores and weight loss, and a moderate reduction in virus shedding was observed post challenge in nasal washes. There was evidence of reduced viral replication by in situ hybridization, although the reduction in viral RNA levels in lungs and nasal turbinates did not reach significance. Taken together, the data indicate that immunization with two doses of an MVA vector expressing SARS-CoV-2 S provides protection against a stringent SARS-CoV-2 challenge of adult Syrian hamsters, reaffirm the utility of this animal model for evaluating candidate SARS-CoV-2 vaccines, and demonstrate the value of an MVA platform in facilitating vaccine development against SARS-CoV-2.
Collapse
|
33
|
Velusamy P, Kiruba K, Su CH, Arun V, Anbu P, Gopinath SCB, Vaseeharan B. SARS-CoV-2 spike protein: Site-specific breakpoints for the development of COVID-19 vaccines. JOURNAL OF KING SAUD UNIVERSITY. SCIENCE 2021; 33:101648. [PMID: 34690467 PMCID: PMC8523302 DOI: 10.1016/j.jksus.2021.101648] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 09/08/2021] [Accepted: 10/07/2021] [Indexed: 05/08/2023]
Abstract
SARS-CoV2 is a member of human coronaviruses and is the causative agent of the present pandemic COVID-19 virus. In order to control COVID-19, studies on viral structure and mechanism of infectivity and pathogenicity are sorely needed. The spike (S) protein is comprised of S1 & S2 subunits. These spike protein subunits enable viral attachment by binding to the host cell via ACE-2 (angiotensin converting enzyme-2) receptor, thus facilitating the infection. During viral entry, one of the key steps is the cleavage of the S1-S2 spike protein subunits via surface TMPRSS2 (transmembrane protease serine 2) and results in viral infection. Hence, the S-protein is critical for the viral attachment and penetration into the host. The rapid advancement of our knowledge on the structural and functional aspects of the spike protein could lead to development of numerous candidate vaccines against SARS-CoV2. Here the authors discuss about the structure of spike protein and explore its related functions. Our aim is to provide a better understanding that may aid in fighting against CoVID-19 and its treatment.
Collapse
Affiliation(s)
- Palaniyandi Velusamy
- Research and Development Wing, Central Research Laboratory, Sree Balaji Medical College and Hospital, Bharath Institute of Higher Education and Research (BIHER), Chennai- 600 044, TN, India
| | - Kannan Kiruba
- Department of Biotechnology, University of Madras, Guindy Campus, Chennai 600 025, TN, India
| | - Chia-Hung Su
- Department of Chemical Engineering, Ming Chi University of Technology, Taishan, Taipei 24301, Taiwan
| | - Viswanathan Arun
- Department of Biotechnology SRFBMST, Sri Ramachandra Institute of Higher Education and Research, Porur, Chennai 600 116, TN, India
| | - Periasamy Anbu
- Department of Biological Engineering, College of Engineering, Inha University, Incheon 402-751, Republic of Korea
| | - Subash C B Gopinath
- Faculty of Chemical Engineering Technology and Institute of Nano Electronic Engineering, Universiti Malaysia Perlis, Arau 02600, Perlis, Malaysia
| | - Baskaralingam Vaseeharan
- Nanobiosciences and Nanopharmacology Division, Biomaterials and Biotechnology in Animal Health Lab, Department of Animal Health and Management, Alagappa University, Science Campus 6th Floor, Karaikudi 630 004, Tamil Nadu, India
| |
Collapse
|
34
|
Kandimalla R, Chakraborty P, Vallamkondu J, Chaudhary A, Samanta S, Reddy PH, De Feo V, Dewanjee S. Counting on COVID-19 Vaccine: Insights into the Current Strategies, Progress and Future Challenges. Biomedicines 2021; 9:1740. [PMID: 34829969 PMCID: PMC8615473 DOI: 10.3390/biomedicines9111740] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 10/30/2021] [Accepted: 11/03/2021] [Indexed: 12/27/2022] Open
Abstract
The emergence of a novel coronavirus viz., severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in late 2019 and its subsequent substantial spread produced the coronavirus disease 2019 (COVID-19) pandemic worldwide. Given its unprecedented infectivity and pathogenicity, the COVID-19 pandemic had a devastating impact on human health, and its clinical management has been a great challenge, which has led to the development and speedy trials of several vaccine candidates against SARS-CoV-2 at an exceptional pace. As a result, several COVID-19 vaccines were made commercially available in the first half of 2021. Although several COVID-19 vaccines showed promising results, crucial insights into their epidemiology, protective mechanisms, and the propensities of reinfection are not largely reviewed. In the present report, we provided insights into the prospects of vaccination against COVID-19 and assessed diverse vaccination strategies including DNA, mRNA, protein subunits, vector-based, live attenuated, and inactivated whole/viral particle-based vaccines. Next, we reviewed major aspects of various available vaccines approved by the World Health Organization and by the local administrations to use against COVID-19. Moreover, we comprehensively assessed the success of these approved vaccines and also their untoward effects, including the possibility of reinfection. We also provided an update on the vaccines that are under development and could be promising candidates in the future. Conclusively, we provided insights into the COVID-19 vaccine epidemiology, their potency, and propensity for SARS-CoV-2 reinfection, while a careful review of their current status, strategies, success, and future challenges was also presented.
Collapse
Affiliation(s)
- Ramesh Kandimalla
- Applied Biology, CSIR-Indian Institute of Technology, Uppal Road, Tarnaka, Hyderabad 500007, Telangana, India
- Department of Biochemistry, Kakatiya Medical College, Warangal 506007, Telangana, India
| | - Pratik Chakraborty
- Advanced Pharmacognosy Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, West Bengal, India;
| | | | - Anupama Chaudhary
- Orinin-BioSystems, LE-52, Lotus Road 4, CHD City, Karnal 132001, Haryana, India;
| | - Sonalinandini Samanta
- Department of Dermatology (Skin & Venereology), ESIC Medical College & Hospital, Patna 801103, Bihar, India;
| | - P. Hemachandra Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA;
- Department of Neuroscience & Pharmacology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Department of Neurology, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Public Health Department of Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Department of Speech, Language and Hearing Sciences, School Health Professions, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Vincenzo De Feo
- Department of Pharmacy, University of Salerno, 84084 Fisciano, Italy
| | - Saikat Dewanjee
- Advanced Pharmacognosy Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, West Bengal, India;
| |
Collapse
|
35
|
Machado BAS, Hodel KVS, Fonseca LMDS, Mascarenhas LAB, Andrade LPCDS, Rocha VPC, Soares MBP, Berglund P, Duthie MS, Reed SG, Badaró R. The Importance of RNA-Based Vaccines in the Fight against COVID-19: An Overview. Vaccines (Basel) 2021; 9:1345. [PMID: 34835276 PMCID: PMC8623509 DOI: 10.3390/vaccines9111345] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 11/02/2021] [Accepted: 11/15/2021] [Indexed: 12/23/2022] Open
Abstract
In recent years, vaccine development using ribonucleic acid (RNA) has become the most promising and studied approach to produce safe and effective new vaccines, not only for prophylaxis but also as a treatment. The use of messenger RNA (mRNA) as an immunogenic has several advantages to vaccine development compared to other platforms, such as lower coast, the absence of cell cultures, and the possibility to combine different targets. During the COVID-19 pandemic, the use of mRNA as a vaccine became more relevant; two out of the four most widely applied vaccines against COVID-19 in the world are based on this platform. However, even though it presents advantages for vaccine application, mRNA technology faces several pivotal challenges to improve mRNA stability, delivery, and the potential to generate the related protein needed to induce a humoral- and T-cell-mediated immune response. The application of mRNA to vaccine development emerged as a powerful tool to fight against cancer and non-infectious and infectious diseases, for example, and represents a relevant research field for future decades. Based on these advantages, this review emphasizes mRNA and self-amplifying RNA (saRNA) for vaccine development, mainly to fight against COVID-19, together with the challenges related to this approach.
Collapse
Affiliation(s)
- Bruna Aparecida Souza Machado
- SENAI Institute of Innovation (ISI) in Health Advanced Systems (CIMATEC ISI SAS), University Center SENAI/CIMATEC, Salvador 41650-010, Brazil; (K.V.S.H.); (L.M.d.S.F.); (L.A.B.M.); (L.P.C.d.S.A.); (V.P.C.R.); (M.B.P.S.); (R.B.)
| | - Katharine Valéria Saraiva Hodel
- SENAI Institute of Innovation (ISI) in Health Advanced Systems (CIMATEC ISI SAS), University Center SENAI/CIMATEC, Salvador 41650-010, Brazil; (K.V.S.H.); (L.M.d.S.F.); (L.A.B.M.); (L.P.C.d.S.A.); (V.P.C.R.); (M.B.P.S.); (R.B.)
| | - Larissa Moraes dos Santos Fonseca
- SENAI Institute of Innovation (ISI) in Health Advanced Systems (CIMATEC ISI SAS), University Center SENAI/CIMATEC, Salvador 41650-010, Brazil; (K.V.S.H.); (L.M.d.S.F.); (L.A.B.M.); (L.P.C.d.S.A.); (V.P.C.R.); (M.B.P.S.); (R.B.)
| | - Luís Alberto Brêda Mascarenhas
- SENAI Institute of Innovation (ISI) in Health Advanced Systems (CIMATEC ISI SAS), University Center SENAI/CIMATEC, Salvador 41650-010, Brazil; (K.V.S.H.); (L.M.d.S.F.); (L.A.B.M.); (L.P.C.d.S.A.); (V.P.C.R.); (M.B.P.S.); (R.B.)
| | - Leone Peter Correia da Silva Andrade
- SENAI Institute of Innovation (ISI) in Health Advanced Systems (CIMATEC ISI SAS), University Center SENAI/CIMATEC, Salvador 41650-010, Brazil; (K.V.S.H.); (L.M.d.S.F.); (L.A.B.M.); (L.P.C.d.S.A.); (V.P.C.R.); (M.B.P.S.); (R.B.)
| | - Vinícius Pinto Costa Rocha
- SENAI Institute of Innovation (ISI) in Health Advanced Systems (CIMATEC ISI SAS), University Center SENAI/CIMATEC, Salvador 41650-010, Brazil; (K.V.S.H.); (L.M.d.S.F.); (L.A.B.M.); (L.P.C.d.S.A.); (V.P.C.R.); (M.B.P.S.); (R.B.)
| | - Milena Botelho Pereira Soares
- SENAI Institute of Innovation (ISI) in Health Advanced Systems (CIMATEC ISI SAS), University Center SENAI/CIMATEC, Salvador 41650-010, Brazil; (K.V.S.H.); (L.M.d.S.F.); (L.A.B.M.); (L.P.C.d.S.A.); (V.P.C.R.); (M.B.P.S.); (R.B.)
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador 40296-710, Brazil
| | - Peter Berglund
- HDT Bio, 1616 Eastlake Ave E, Seattle, WA 98102, USA; (P.B.); (M.S.D.); (S.G.R.)
| | - Malcolm S. Duthie
- HDT Bio, 1616 Eastlake Ave E, Seattle, WA 98102, USA; (P.B.); (M.S.D.); (S.G.R.)
| | - Steven G. Reed
- HDT Bio, 1616 Eastlake Ave E, Seattle, WA 98102, USA; (P.B.); (M.S.D.); (S.G.R.)
| | - Roberto Badaró
- SENAI Institute of Innovation (ISI) in Health Advanced Systems (CIMATEC ISI SAS), University Center SENAI/CIMATEC, Salvador 41650-010, Brazil; (K.V.S.H.); (L.M.d.S.F.); (L.A.B.M.); (L.P.C.d.S.A.); (V.P.C.R.); (M.B.P.S.); (R.B.)
| |
Collapse
|
36
|
Bošnjak B, Odak I, Barros-Martins J, Sandrock I, Hammerschmidt SI, Permanyer M, Patzer GE, Greorgiev H, Gutierrez Jauregui R, Tscherne A, Schwarz JH, Kalodimou G, Ssebyatika G, Ciurkiewicz M, Willenzon S, Bubke A, Ristenpart J, Ritter C, Tuchel T, Meyer zu Natrup C, Shin DL, Clever S, Limpinsel L, Baumgärtner W, Krey T, Volz A, Sutter G, Förster R. Intranasal Delivery of MVA Vector Vaccine Induces Effective Pulmonary Immunity Against SARS-CoV-2 in Rodents. Front Immunol 2021; 12:772240. [PMID: 34858430 PMCID: PMC8632543 DOI: 10.3389/fimmu.2021.772240] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 10/25/2021] [Indexed: 01/08/2023] Open
Abstract
Antigen-specific tissue-resident memory T cells (Trms) and neutralizing IgA antibodies provide the most effective protection of the lungs from viral infections. To induce those essential components of lung immunity against SARS-CoV-2, we tested various immunization protocols involving intranasal delivery of a novel Modified Vaccinia virus Ankara (MVA)-SARS-2-spike vaccine candidate. We show that a single intranasal MVA-SARS-CoV-2-S application in mice strongly induced pulmonary spike-specific CD8+ T cells, albeit restricted production of neutralizing antibodies. In prime-boost protocols, intranasal booster vaccine delivery proved to be crucial for a massive expansion of systemic and lung tissue-resident spike-specific CD8+ T cells and the development of Th1 - but not Th2 - CD4+ T cells. Likewise, very high titers of IgG and IgA anti-spike antibodies were present in serum and broncho-alveolar lavages that possessed high virus neutralization capacities to all current SARS-CoV-2 variants of concern. Importantly, the MVA-SARS-2-spike vaccine applied in intramuscular priming and intranasal boosting treatment regimen completely protected hamsters from developing SARS-CoV-2 lung infection and pathology. Together, these results identify intramuscular priming followed by respiratory tract boosting with MVA-SARS-2-S as a promising approach for the induction of local, respiratory as well as systemic immune responses suited to protect from SARS-CoV-2 infections.
Collapse
Affiliation(s)
- Berislav Bošnjak
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Ivan Odak
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | | | - Inga Sandrock
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | | | - Marc Permanyer
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | | | - Hristo Greorgiev
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | | | - Alina Tscherne
- Division of Virology, Department of Veterinary Sciences, Ludwig Maximilian University (LMU) Munich, Munich, Germany
- German Centre for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| | - Jan Hendrik Schwarz
- Division of Virology, Department of Veterinary Sciences, Ludwig Maximilian University (LMU) Munich, Munich, Germany
| | - Georgia Kalodimou
- Division of Virology, Department of Veterinary Sciences, Ludwig Maximilian University (LMU) Munich, Munich, Germany
- German Centre for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| | - George Ssebyatika
- Center of Structural and Cell Biology in Medicine, Institute of Biochemistry, University of Lübeck, Lübeck, Germany
| | | | | | - Anja Bubke
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | | | | | - Tamara Tuchel
- Institute for Virology, University of Veterinary Medicine Hannover, Hannover, Germany
| | | | - Dai-Lun Shin
- Institute for Virology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Sabrina Clever
- Institute for Virology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Leonard Limpinsel
- Division of Virology, Department of Veterinary Sciences, Ludwig Maximilian University (LMU) Munich, Munich, Germany
| | - Wolfgang Baumgärtner
- Department of Pathology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Thomas Krey
- Center of Structural and Cell Biology in Medicine, Institute of Biochemistry, University of Lübeck, Lübeck, Germany
- German Centre for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Lübeck, Germany
- Centre for Structural Systems Biology (CSSB), Hamburg, Germany
- Institute of Virology, Hannover Medical School, Hannover, Germany
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
| | - Asisa Volz
- Division of Virology, Department of Veterinary Sciences, Ludwig Maximilian University (LMU) Munich, Munich, Germany
- German Centre for Infection Research (DZIF), Partner Site Munich, Munich, Germany
- Institute for Virology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Gerd Sutter
- Division of Virology, Department of Veterinary Sciences, Ludwig Maximilian University (LMU) Munich, Munich, Germany
- German Centre for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| | - Reinhold Förster
- Institute of Immunology, Hannover Medical School, Hannover, Germany
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
- German Centre for Infection Research (DZIF), Partner Site Hannover, Hannover, Germany
| |
Collapse
|
37
|
DNA Vaccine Administered by Cationic Lipoplexes or by In Vivo Electroporation Induces Comparable Antibody Responses against SARS-CoV-2 in Mice. Vaccines (Basel) 2021; 9:vaccines9080874. [PMID: 34451998 PMCID: PMC8402479 DOI: 10.3390/vaccines9080874] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 08/02/2021] [Accepted: 08/04/2021] [Indexed: 11/17/2022] Open
Abstract
In view of addressing the global necessity of an effective vaccine in the SARS-CoV-2 pandemic, a plasmid DNA vaccine, expressing for the spike (S) protein and formulated in lipoplexes, was manufactured and tested for in vitro transfection and in vivo immunogenicity. Blank cationic liposomes of 130.9 ± 5.8 nm in size and with a zeta potential of +48 ± 12 mV were formulated using the thin-film layer rehydration method. Liposomes were complexed with pCMVkan-S at different N/P ratios. Ratios of 0.25:1 and 1:1 were selected according to their complex stability and controlled size compared to other ratios and tested in vitro for transfection studies and in vivo for immunogenicity. Both selected formulations showed enhanced neutralizing antibody responses compared to pCMVkan-S injected alone, as well as an increased T cell response. The titers observed were similar to those of intramuscular electroporation (IM-EP), which was set as an efficacy goal.
Collapse
|
38
|
Schoeman D, Fielding BC. Human Coronaviruses: Counteracting the Damage by Storm. Viruses 2021; 13:1457. [PMID: 34452323 PMCID: PMC8402835 DOI: 10.3390/v13081457] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 07/16/2021] [Accepted: 07/20/2021] [Indexed: 12/15/2022] Open
Abstract
Over the past 18 years, three highly pathogenic human (h) coronaviruses (CoVs) have caused severe outbreaks, the most recent causative agent, SARS-CoV-2, being the first to cause a pandemic. Although much progress has been made since the COVID-19 pandemic started, much about SARS-CoV-2 and its disease, COVID-19, is still poorly understood. The highly pathogenic hCoVs differ in some respects, but also share some similarities in clinical presentation, the risk factors associated with severe disease, and the characteristic immunopathology associated with the progression to severe disease. This review aims to highlight these overlapping aspects of the highly pathogenic hCoVs-SARS-CoV, MERS-CoV, and SARS-CoV-2-briefly discussing the importance of an appropriately regulated immune response; how the immune response to these highly pathogenic hCoVs might be dysregulated through interferon (IFN) inhibition, antibody-dependent enhancement (ADE), and long non-coding RNA (lncRNA); and how these could link to the ensuing cytokine storm. The treatment approaches to highly pathogenic hCoV infections are discussed and it is suggested that a greater focus be placed on T-cell vaccines that elicit a cell-mediated immune response, using rapamycin as a potential agent to improve vaccine responses in the elderly and obese, and the potential of stapled peptides as antiviral agents.
Collapse
Affiliation(s)
| | - Burtram C. Fielding
- Molecular Biology and Virology Research Laboratory, Department of Medical Biosciences, University of the Western Cape, Cape Town 7535, South Africa;
| |
Collapse
|
39
|
Kumavath R, Barh D, Andrade BS, Imchen M, Aburjaile FF, Ch A, Rodrigues DLN, Tiwari S, Alzahrani KJ, Góes-Neto A, Weener ME, Ghosh P, Azevedo V. The Spike of SARS-CoV-2: Uniqueness and Applications. Front Immunol 2021; 12:663912. [PMID: 34305894 PMCID: PMC8297464 DOI: 10.3389/fimmu.2021.663912] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 06/16/2021] [Indexed: 12/20/2022] Open
Abstract
The Spike (S) protein of the SARS-CoV-2 virus is critical for its ability to attach and fuse into the host cells, leading to infection, and transmission. In this review, we have initially performed a meta-analysis of keywords associated with the S protein to frame the outline of important research findings and directions related to it. Based on this outline, we have reviewed the structure, uniqueness, and origin of the S protein of SARS-CoV-2. Furthermore, the interactions of the Spike protein with host and its implications in COVID-19 pathogenesis, as well as drug and vaccine development, are discussed. We have also summarized the recent advances in detection methods using S protein-based RT-PCR, ELISA, point-of-care lateral flow immunoassay, and graphene-based field-effect transistor (FET) biosensors. Finally, we have also discussed the emerging Spike mutants and the efficacy of the Spike-based vaccines against those strains. Overall, we have covered most of the recent advances on the SARS-CoV-2 Spike protein and its possible implications in countering this virus.
Collapse
Affiliation(s)
- Ranjith Kumavath
- Department of Genomic Science, School of Biological Sciences, Central University of Kerala, Kasaragod, India
| | - Debmalya Barh
- Centre for Genomics and Applied Gene Technology, Institute of Integrative Omics and Applied Biotechnology (IIOAB), Nonakuri, Purba Medinipur, West Bengal, India
- Laboratório de Genética Celular e Molecular, Departamento de Genetica, Ecologia e Evolucao, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Bruno Silva Andrade
- Laboratório de Bioinformática e Química Computacional, Departamento de Ciências Biológicas, Universidade Estadual do Sudoeste da Bahia (UESB), Jequié, Brazil
| | - Madangchanok Imchen
- Department of Genomic Science, School of Biological Sciences, Central University of Kerala, Kasaragod, India
| | - Flavia Figueira Aburjaile
- Laboratório de Genética Celular e Molecular, Departamento de Genetica, Ecologia e Evolucao, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Athira Ch
- Department of Genomic Science, School of Biological Sciences, Central University of Kerala, Kasaragod, India
| | - Diego Lucas Neres Rodrigues
- Laboratório de Genética Celular e Molecular, Departamento de Genetica, Ecologia e Evolucao, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Sandeep Tiwari
- Laboratório de Genética Celular e Molecular, Departamento de Genetica, Ecologia e Evolucao, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Khalid J Alzahrani
- Department of Clinical Laboratories Sciences, College of Applied Medical Sciences, Taif University, Taif, Saudi Arabia
| | - Aristóteles Góes-Neto
- Laboratório de Biologia Molecular e Computacional de Fungos, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil
| | | | - Preetam Ghosh
- Department of Computer Science, Virginia Commonwealth University, Richmond, VA, United States
| | - Vasco Azevedo
- Laboratório de Genética Celular e Molecular, Departamento de Genetica, Ecologia e Evolucao, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
40
|
Raman R, Patel KJ, Ranjan K. COVID-19: Unmasking Emerging SARS-CoV-2 Variants, Vaccines and Therapeutic Strategies. Biomolecules 2021; 11:993. [PMID: 34356617 PMCID: PMC8301790 DOI: 10.3390/biom11070993] [Citation(s) in RCA: 107] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 06/26/2021] [Accepted: 06/29/2021] [Indexed: 02/07/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the etiological agent of the coronavirus disease 2019 (COVID-19) pandemic, which has been a topic of major concern for global human health. The challenge to restrain the COVID-19 pandemic is further compounded by the emergence of several SARS-CoV-2 variants viz. B.1.1.7 (Alpha), B.1.351 (Beta), P1 (Gamma) and B.1.617.2 (Delta), which show increased transmissibility and resistance towards vaccines and therapies. Importantly, there is convincing evidence of increased susceptibility to SARS-CoV-2 infection among individuals with dysregulated immune response and comorbidities. Herein, we provide a comprehensive perspective regarding vulnerability of SARS-CoV-2 infection in patients with underlying medical comorbidities. We discuss ongoing vaccine (mRNA, protein-based, viral vector-based, etc.) and therapeutic (monoclonal antibodies, small molecules, plasma therapy, etc.) modalities designed to curb the COVID-19 pandemic. We also discuss in detail, the challenges posed by different SARS-CoV-2 variants of concern (VOC) identified across the globe and their effects on therapeutic and prophylactic interventions.
Collapse
Affiliation(s)
- Renuka Raman
- Department of Surgery, Weill Cornell Medical College, New York, NY 10065, USA;
| | - Krishna J. Patel
- Mount Sinai Innovation Partners, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
| | - Kishu Ranjan
- School of Medicine, Yale University, New Haven, CT 06519, USA
| |
Collapse
|
41
|
Harbour JC, Lyski ZL, Schell JB, Thomas A, Messer WB, Slifka MK, Nolz JC. Cellular and Humoral Immune Responses in Mice Immunized with Vaccinia Virus Expressing the SARS-CoV-2 Spike Protein. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2021; 206:2596-2604. [PMID: 33972374 PMCID: PMC8165000 DOI: 10.4049/jimmunol.2100054] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 03/22/2021] [Indexed: 12/26/2022]
Abstract
The COVID-19 pandemic is a global health emergency, and the development of a successful vaccine will ultimately be required to prevent the continued spread and seasonal recurrence of this disease within the human population. However, very little is known about either the quality of the adaptive immune response or the viral Ag targets that will be necessary to prevent the spread of the infection. In this study, we generated recombinant Vaccinia virus expressing the full-length spike protein from SARS-CoV-2 (VacV-S) to evaluate the cellular and humoral immune response mounted against this viral Ag in mice. Both CD8+ and CD4+ T cells specific to the SARS-CoV-2 spike protein underwent robust expansion, contraction, and persisted for at least 40 d following a single immunization with VacV-S. Vaccination also caused the rapid emergence of spike-specific IgG-neutralizing Abs. Interestingly, both the cellular and humoral immune responses strongly targeted the S1 domain of spike following VacV-S immunization. Notably, immunization with VacV-expressing spike conjugated to the MHC class II invariant chain, a strategy previously reported by us and others to enhance the immunogenicity of antigenic peptides, did not promote stronger spike-specific T cell or Ab responses in vivo. Overall, these findings demonstrate that an immunization approach using VacV or attenuated versions of VacV expressing the native, full-length SARS-CoV-2 spike protein could be used for further vaccine development to prevent the spread of COVID-19.
Collapse
Affiliation(s)
- Jake C Harbour
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR
| | - Zoe L Lyski
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR
| | - John B Schell
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR
| | - Archana Thomas
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR
| | - William B Messer
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR
- Department of Medicine, Division of Infectious Diseases, Oregon Health & Science University, Portland, OR
- Program in Epidemiology, Oregon Health & Science University-Portland State University School of Public Health, Oregon Health & Science University, Portland, OR
| | - Mark K Slifka
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR
| | - Jeffrey C Nolz
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR;
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR; and
- Department of Radiation Medicine, Oregon Health & Science University, Portland, OR
| |
Collapse
|
42
|
Bisgin A, Sanlioglu AD, Eksi YE, Griffith TS, Sanlioglu S. Current Update on Severe Acute Respiratory Syndrome Coronavirus 2 Vaccine Development with a Special Emphasis on Gene Therapy Viral Vector Design and Construction for Vaccination. Hum Gene Ther 2021; 32:541-562. [PMID: 33858231 DOI: 10.1089/hum.2021.052] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Severe acute respiratory syndrome (SARS) is a newly emerging infectious disease (COVID-19) caused by the novel coronavirus SARS-coronavirus 2 (CoV-2). To combat the devastating spread of SARS-CoV-2, extraordinary efforts from numerous laboratories have focused on the development of effective and safe vaccines. Traditional live-attenuated or inactivated viral vaccines are not recommended for immunocompromised patients as the attenuated virus can still cause disease via phenotypic or genotypic reversion. Subunit vaccines require repeated dosing and adjuvant use to be effective, and DNA vaccines exhibit lower immune responses. mRNA vaccines can be highly unstable under physiological conditions. On the contrary, naturally antigenic viral vectors with well-characterized structure and safety profile serve as among the most effective gene carriers to provoke immune response via heterologous gene transfer. Viral vector-based vaccines induce both an effective cellular immune response and a humoral immune response owing to their natural adjuvant properties via transduction of immune cells. Consequently, viral vectored vaccines carrying the SARS-CoV-2 spike protein have recently been generated and successfully used to activate cytotoxic T cells and develop a neutralizing antibody response. Recent progress in SARS-CoV-2 vaccines, with an emphasis on gene therapy viral vector-based vaccine development, is discussed in this review.
Collapse
Affiliation(s)
- Atil Bisgin
- The Department of Gene and Cell Therapy, Faculty of Medicine, Akdeniz University, Antalya, Turkey
- Department of Medical Genetics, Faculty of Medicine, Cukurova University, Adana, Turkey
| | - Ahter D Sanlioglu
- The Department of Gene and Cell Therapy, Faculty of Medicine, Akdeniz University, Antalya, Turkey
| | - Yunus Emre Eksi
- The Department of Gene and Cell Therapy, Faculty of Medicine, Akdeniz University, Antalya, Turkey
| | - Thomas S Griffith
- The Department of Urology, School of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Salih Sanlioglu
- The Department of Gene and Cell Therapy, Faculty of Medicine, Akdeniz University, Antalya, Turkey
| |
Collapse
|
43
|
Keaney D, Whelan S, Finn K, Lucey B. Misdiagnosis of SARS-CoV-2: A Critical Review of the Influence of Sampling and Clinical Detection Methods. Med Sci (Basel) 2021; 9:36. [PMID: 34070530 PMCID: PMC8162574 DOI: 10.3390/medsci9020036] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/20/2021] [Accepted: 05/21/2021] [Indexed: 01/08/2023] Open
Abstract
SARS-CoV-2 infection has generated the biggest pandemic since the influenza outbreak of 1918-1919. One clear difference between these pandemics has been the ability to test for the presence of the virus or for evidence of infection. This review examined the performance characteristics of sample types via PCR detection of the virus, of antibody testing, of rapid viral antigen detection kits and computerised tomography (CT) scanning. It was found that combined detection approaches, such as the incorporation of CT scans, may reduce the levels of false negatives obtained by PCR detection in both symptomatic and asymptomatic patients, while sputum and oral throat washing sample types should take precedence over swabbing when available. Rt-PCR assays for detection of the virus remain the gold-standard method for SARS-CoV-2 diagnosis and can be used effectively on pooled samples for widespread screening. The novel Oxford antibody assay was found to have the highest sensitivity and specificity of four currently available commercial antibody kits but should only be used during a specific timeframe post-symptom onset. Further research into transmission modes between symptomatic and asymptomatic patients is needed. Analysis of the performance characteristics of different sampling and detection methods for SARS-CoV-2 showed that timing of sampling and testing methods used can greatly influence the rate of false-positive and false-negative test results, thereby influencing viral spread.
Collapse
Affiliation(s)
- Daniel Keaney
- Department of Biological Sciences, Munster Technological University, Bishopstown, T12 P928 Cork, Ireland; (D.K.); (S.W.); (B.L.)
| | - Shane Whelan
- Department of Biological Sciences, Munster Technological University, Bishopstown, T12 P928 Cork, Ireland; (D.K.); (S.W.); (B.L.)
| | - Karen Finn
- Department of Biopharmaceutical and Medical Science, Galway-Mayo Institute of Technology, Old Dublin Road, H91 DCH9 Galway, Ireland
| | - Brigid Lucey
- Department of Biological Sciences, Munster Technological University, Bishopstown, T12 P928 Cork, Ireland; (D.K.); (S.W.); (B.L.)
| |
Collapse
|
44
|
Arashkia A, Jalilvand S, Mohajel N, Afchangi A, Azadmanesh K, Salehi‐Vaziri M, Fazlalipour M, Pouriayevali MH, Jalali T, Mousavi Nasab SD, Roohvand F, Shoja Z. Severe acute respiratory syndrome-coronavirus-2 spike (S) protein based vaccine candidates: State of the art and future prospects. Rev Med Virol 2021; 31:e2183. [PMID: 33594794 PMCID: PMC7646037 DOI: 10.1002/rmv.2183] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 09/30/2020] [Accepted: 10/01/2020] [Indexed: 01/07/2023]
Abstract
Coronavirus disease 2019 (Covid-19) is caused by severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2) which is responsible for a global pandemic that started in late 2019 in Wuhan, China. To prevent the worldwide spread of this highly pathogenic virus, development of an effective and safe vaccine is urgently needed. The SARS-CoV-2 and SARS-CoV share a high degree of genetic and pathologic identity and share safety and immune-enhancement concerns regarding vaccine development. Prior animal studies with first generation (whole virus-based) preparations of SARS-CoV vaccines (inactivated and attenuated vaccine modalities) indicated the possibility of increased infectivity or eosinophilic infiltration by immunization. Therefore, development of second and third generation safer vaccines (by using modern vaccine platforms) is actively sought for this viral infection. The spike (S) protein of SARS-CoVs is the main determinant of cell entry and tropism and is responsible for facilitating zoonosis into humans and sustained person-to-person transmission. Furthermore, 'S' protein contains multiple neutralizing epitopes that play an essential role in the induction of neutralizing antibodies (nAbs) and protective immunity. Moreover, T-cell responses against the SARS-CoV-2 'S' protein have also been characterized that correlate to the IgG and IgA antibody titres in Covid-19 patients. Thus, S protein is an obvious candidate antigen for inclusion into vaccine platforms against SARS-CoV-2 viral infection. This manuscript reviews different characteristics of S protein, its potency and 'state of the art' of the vaccine development strategies and platforms using this antigen, for construction of a safe and effective SARS-CoV-2 vaccine.
Collapse
MESH Headings
- Antibodies, Viral/biosynthesis
- COVID-19/epidemiology
- COVID-19/immunology
- COVID-19/prevention & control
- COVID-19/virology
- COVID-19 Vaccines/administration & dosage
- COVID-19 Vaccines/biosynthesis
- COVID-19 Vaccines/immunology
- Clinical Trials as Topic
- Genetic Vectors/chemistry
- Genetic Vectors/immunology
- Genome, Viral/immunology
- Humans
- Immunity, Innate/drug effects
- Immunization Schedule
- Immunogenicity, Vaccine
- Pandemics
- Patient Safety
- SARS-CoV-2/drug effects
- SARS-CoV-2/immunology
- SARS-CoV-2/pathogenicity
- Spike Glycoprotein, Coronavirus/chemistry
- Spike Glycoprotein, Coronavirus/genetics
- Spike Glycoprotein, Coronavirus/immunology
- Vaccines, Attenuated
- Vaccines, DNA
- Vaccines, Subunit
Collapse
Affiliation(s)
- Arash Arashkia
- Department of Molecular VirologyPasteur Institute of IranTehranIran
| | - Somayeh Jalilvand
- Department of VirologySchool of Public HealthTehran University of Medical SciencesTehranIran
| | - Nasir Mohajel
- Department of Molecular VirologyPasteur Institute of IranTehranIran
| | - Atefeh Afchangi
- Department of VirologySchool of Public HealthTehran University of Medical SciencesTehranIran
| | | | - Mostafa Salehi‐Vaziri
- Department of Arboviruses and Viral Hemorrhagic Fevers (National Ref Lab)Pasteur Institute of IranTehranIran
| | - Mehdi Fazlalipour
- Department of Arboviruses and Viral Hemorrhagic Fevers (National Ref Lab)Pasteur Institute of IranTehranIran
| | | | - Tahmineh Jalali
- Department of Arboviruses and Viral Hemorrhagic Fevers (National Ref Lab)Pasteur Institute of IranTehranIran
| | - Seyed Dawood Mousavi Nasab
- Department of Research and DevelopmentProduction and Research ComplexPasteur Institute of IranTehranIran
| | - Farzin Roohvand
- Department of Molecular VirologyPasteur Institute of IranTehranIran
| | - Zabihollah Shoja
- Department of Molecular VirologyPasteur Institute of IranTehranIran
| | | |
Collapse
|
45
|
Zhang Y, Gargan S, Lu Y, Stevenson NJ. An Overview of Current Knowledge of Deadly CoVs and Their Interface with Innate Immunity. Viruses 2021; 13:560. [PMID: 33810391 PMCID: PMC8066579 DOI: 10.3390/v13040560] [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: 02/08/2021] [Revised: 03/12/2021] [Accepted: 03/16/2021] [Indexed: 02/06/2023] Open
Abstract
Coronaviruses are a large family of zoonotic RNA viruses, whose infection can lead to mild or lethal respiratory tract disease. Severe Acute Respiratory Syndrome-Coronavirus-1 (SARS-CoV-1) first emerged in Guangdong, China in 2002 and spread to 29 countries, infecting 8089 individuals and causing 774 deaths. In 2012, Middle East Respiratory Syndrome-Coronavirus (MERS-CoV) emerged in Saudi Arabia and has spread to 27 countries, with a mortality rate of ~34%. In 2019, SARS-CoV-2 emerged and has spread to 220 countries, infecting over 100,000,000 people and causing more than 2,000,000 deaths to date. These three human coronaviruses cause diseases of varying severity. Most people develop mild, common cold-like symptoms, while some develop acute respiratory distress syndrome (ARDS). The success of all viruses, including coronaviruses, relies on their evolved abilities to evade and modulate the host anti-viral and pro-inflammatory immune responses. However, we still do not fully understand the transmission, phylogeny, epidemiology, and pathogenesis of MERS-CoV and SARS-CoV-1 and -2. Despite the rapid application of a range of therapies for SARS-CoV-2, such as convalescent plasma, remdesivir, hydroxychloroquine and type I interferon, no fully effective treatment has been determined. Remarkably, COVID-19 vaccine research and development have produced several offerings that are now been administered worldwide. Here, we summarise an up-to-date understanding of epidemiology, immunomodulation and ongoing anti-viral and immunosuppressive treatment strategies. Indeed, understanding the interplay between coronaviruses and the anti-viral immune response is crucial to identifying novel targets for therapeutic intervention, which may even prove invaluable for the control of future emerging coronavirus.
Collapse
Affiliation(s)
- Yamei Zhang
- Viral Immunology Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, D02 R590 Dublin, Ireland; (Y.Z.); (S.G.)
| | - Siobhan Gargan
- Viral Immunology Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, D02 R590 Dublin, Ireland; (Y.Z.); (S.G.)
| | - Yongxu Lu
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK;
| | - Nigel J. Stevenson
- Viral Immunology Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, D02 R590 Dublin, Ireland; (Y.Z.); (S.G.)
- Viral Immunology Group, Royal College of Surgeons in Ireland—Medical University of Bahrain, Adliya 15503, Bahrain
| |
Collapse
|
46
|
Liu R, Americo JL, Cotter CA, Earl PL, Erez N, Peng C, Moss B. One or two injections of MVA-vectored vaccine shields hACE2 transgenic mice from SARS-CoV-2 upper and lower respiratory tract infection. Proc Natl Acad Sci U S A 2021; 118:e2026785118. [PMID: 33688035 PMCID: PMC8000198 DOI: 10.1073/pnas.2026785118] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Modified vaccinia virus Ankara (MVA) is a replication-restricted smallpox vaccine, and numerous clinical studies of recombinant MVAs (rMVAs) as vectors for prevention of other infectious diseases, including COVID-19, are in progress. Here, we characterize rMVAs expressing the S protein of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Modifications of full-length S individually or in combination included two proline substitutions, mutations of the furin recognition site, and deletion of the endoplasmic retrieval signal. Another rMVA in which the receptor binding domain (RBD) is flanked by the signal peptide and transmembrane domains of S was also constructed. Each modified S protein was displayed on the surface of rMVA-infected cells and was recognized by anti-RBD antibody and soluble hACE2 receptor. Intramuscular injection of mice with the rMVAs induced antibodies, which neutralized a pseudovirus in vitro and, upon passive transfer, protected hACE2 transgenic mice from lethal infection with SARS-CoV-2, as well as S-specific CD3+CD8+IFNγ+ T cells. Antibody boosting occurred following a second rMVA or adjuvanted purified RBD protein. Immunity conferred by a single vaccination of hACE2 mice prevented morbidity and weight loss upon intranasal infection with SARS-CoV-2 3 wk or 7 wk later. One or two rMVA vaccinations also prevented detection of infectious SARS-CoV-2 and subgenomic viral mRNAs in the lungs and greatly reduced induction of cytokine and chemokine mRNAs. A low amount of virus was found in the nasal turbinates of only one of eight rMVA-vaccinated mice on day 2 and none later. Detection of low levels of subgenomic mRNAs in turbinates indicated that replication was aborted in immunized animals.
Collapse
Affiliation(s)
- Ruikang Liu
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892
| | - Jeffrey L Americo
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892
| | - Catherine A Cotter
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892
| | - Patricia L Earl
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892
| | - Noam Erez
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892
| | - Chen Peng
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892
| | - Bernard Moss
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892
| |
Collapse
|
47
|
Srivastava V, Niu L, Phadke KS, Bellaire BH, Cho MW. Induction of Potent and Durable Neutralizing Antibodies Against SARS-CoV-2 Using a Receptor Binding Domain-Based Immunogen. Front Immunol 2021; 12:637982. [PMID: 33777030 PMCID: PMC7991075 DOI: 10.3389/fimmu.2021.637982] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 02/15/2021] [Indexed: 01/04/2023] Open
Abstract
A novel betacoronavirus (SARS-CoV-2) that causes severe pneumonia emerged through zoonosis in late 2019. The disease, referred to as COVID-19, has an alarming mortality rate and it is having a devastating effect on the global economy and public health systems. A safe, effective vaccine is urgently needed to halt this pandemic. In this study, immunogenicity of the receptor binding domain (RBD) of spike (S) glycoprotein was examined in mice. Animals were immunized with recombinant RBD antigen intraperitoneally using three different adjuvants (Zn-chitosan, Alhydrogel, and Adju-Phos), and antibody responses were followed for over 5 months. Results showed that potent neutralizing antibodies (nAbs) can be induced with 70% neutralization titer (NT70) of ~14,580 against live, infectious viruses. Although antigen-binding antibody titers decreased gradually over time, sufficiently protective levels of nAbs persisted (NT80 >2,430) over the 5-month observation period. Results also showed that adjuvants have profound effects on kinetics of nAb induction, total antibody titers, antibody avidity, antibody longevity, and B-cell epitopes targeted by the immune system. In conclusion, a recombinant subunit protein immunogen based on the RBD is a highly promising vaccine candidate. Continued evaluation of RBD immunogenicity using different adjuvants and vaccine regimens could further improve vaccine efficacy.
Collapse
Affiliation(s)
- Vikram Srivastava
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
| | - Ling Niu
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
| | - Kruttika S. Phadke
- Department of Veterinary Microbiology and Preventive Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
- Interdepartmental Microbiology Program, Iowa State University, Ames, IA, United States
| | - Bryan H. Bellaire
- Department of Veterinary Microbiology and Preventive Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
- Interdepartmental Microbiology Program, Iowa State University, Ames, IA, United States
| | - Michael W. Cho
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
- Interdepartmental Microbiology Program, Iowa State University, Ames, IA, United States
- NeoVaxSyn, Inc., Ames, IA, United States
| |
Collapse
|
48
|
Routhu NK, Cheedarla N, Gangadhara S, Bollimpelli VS, Boddapati AK, Shiferaw A, Rahman SA, Sahoo A, Edara VV, Lai L, Floyd K, Wang S, Fischinger S, Atyeo C, Shin SA, Gumber S, Kirejczyk S, Cohen J, Jean SM, Wood JS, Connor-Stroud F, Stammen RL, Upadhyay AA, Pellegrini K, Montefiori D, Shi PY, Menachery VD, Alter G, Vanderford TH, Bosinger SE, Suthar MS, Amara RR. A modified vaccinia Ankara vector-based vaccine protects macaques from SARS-CoV-2 infection, immune pathology, and dysfunction in the lungs. Immunity 2021; 54:542-556.e9. [PMID: 33631118 PMCID: PMC7859620 DOI: 10.1016/j.immuni.2021.02.001] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 12/04/2020] [Accepted: 01/29/2021] [Indexed: 02/07/2023]
Abstract
A combination of vaccination approaches will likely be necessary to fully control the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic. Here, we show that modified vaccinia Ankara (MVA) vectors expressing membrane-anchored pre-fusion stabilized spike (MVA/S) but not secreted S1 induced strong neutralizing antibody responses against SARS-CoV-2 in mice. In macaques, the MVA/S vaccination induced strong neutralizing antibodies and CD8+ T cell responses, and conferred protection from SARS-CoV-2 infection and virus replication in the lungs as early as day 2 following intranasal and intratracheal challenge. Single-cell RNA sequencing analysis of lung cells on day 4 after infection revealed that MVA/S vaccination also protected macaques from infection-induced inflammation and B cell abnormalities and lowered induction of interferon-stimulated genes. These results demonstrate that MVA/S vaccination induces neutralizing antibodies and CD8+ T cells in the blood and lungs and is a potential vaccine candidate for SARS-CoV-2.
Collapse
MESH Headings
- Animals
- Antibodies, Neutralizing/immunology
- Antibodies, Viral/immunology
- Antigens, Viral/genetics
- Antigens, Viral/immunology
- COVID-19/immunology
- COVID-19/pathology
- COVID-19/prevention & control
- COVID-19/virology
- COVID-19 Vaccines/genetics
- COVID-19 Vaccines/immunology
- Disease Models, Animal
- Gene Expression
- Gene Order
- Genetic Vectors/genetics
- Immunophenotyping
- Lung/immunology
- Lung/pathology
- Lung/virology
- Macaca
- Macrophages, Alveolar/immunology
- Macrophages, Alveolar/metabolism
- Macrophages, Alveolar/pathology
- Mice
- SARS-CoV-2/immunology
- Spike Glycoprotein, Coronavirus/genetics
- Spike Glycoprotein, Coronavirus/immunology
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
- Vaccination/methods
- Vaccines, DNA/genetics
- Vaccines, DNA/immunology
- Vaccinia virus/genetics
Collapse
Affiliation(s)
- Nanda Kishore Routhu
- Emory Vaccine Center, Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA; Department of Microbiology and Immunology, Emory School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Narayanaiah Cheedarla
- Emory Vaccine Center, Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA; Department of Microbiology and Immunology, Emory School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Sailaja Gangadhara
- Emory Vaccine Center, Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA; Department of Microbiology and Immunology, Emory School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Venkata Satish Bollimpelli
- Emory Vaccine Center, Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA; Department of Microbiology and Immunology, Emory School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Arun K Boddapati
- Emory Vaccine Center, Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA; Department of Pathology, Emory School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Ayalnesh Shiferaw
- Emory Vaccine Center, Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA; Department of Microbiology and Immunology, Emory School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Sheikh Abdul Rahman
- Emory Vaccine Center, Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA; Department of Microbiology and Immunology, Emory School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Anusmita Sahoo
- Emory Vaccine Center, Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA; Department of Microbiology and Immunology, Emory School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Venkata Viswanadh Edara
- Emory Vaccine Center, Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA; Department of Pediatrics, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Lilin Lai
- Emory Vaccine Center, Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA; Department of Pediatrics, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Katharine Floyd
- Emory Vaccine Center, Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA; Department of Pediatrics, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Shelly Wang
- Emory Vaccine Center, Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | | | - Caroline Atyeo
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Sally A Shin
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Sanjeev Gumber
- Division of Pathology, Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Shannon Kirejczyk
- Division of Pathology, Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Joyce Cohen
- Division of Animal Resources, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Sherrie M Jean
- Division of Animal Resources, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Jennifer S Wood
- Division of Animal Resources, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Fawn Connor-Stroud
- Division of Animal Resources, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Rachelle L Stammen
- Division of Animal Resources, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Amit A Upadhyay
- Emory Vaccine Center, Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Kathryn Pellegrini
- Emory Vaccine Center, Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - David Montefiori
- Department of Surgery, Duke University School of Medicine, Durham, NC 27710, USA
| | - Pei-Yong Shi
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, TX, USA
| | - Vineet D Menachery
- Department of Microbiology and Immunology, The University of Texas Medical Branch, Galveston, TX, USA
| | - Galit Alter
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Thomas H Vanderford
- Emory Vaccine Center, Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Steven E Bosinger
- Emory Vaccine Center, Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA; Department of Pathology, Emory School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Mehul S Suthar
- Emory Vaccine Center, Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA; Department of Pediatrics, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Rama Rao Amara
- Emory Vaccine Center, Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA; Department of Microbiology and Immunology, Emory School of Medicine, Emory University, Atlanta, GA 30322, USA.
| |
Collapse
|
49
|
Molaei S, Dadkhah M, Asghariazar V, Karami C, Safarzadeh E. The immune response and immune evasion characteristics in SARS-CoV, MERS-CoV, and SARS-CoV-2: Vaccine design strategies. Int Immunopharmacol 2021; 92:107051. [PMID: 33429331 PMCID: PMC7522676 DOI: 10.1016/j.intimp.2020.107051] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 09/24/2020] [Accepted: 09/24/2020] [Indexed: 01/25/2023]
Abstract
The worldwide outbreak of SARS-CoV-2, severe acute respiratory syndrome coronavirus 2 as a novel human coronavirus, was the worrying news at the beginning of 2020. Since its emergence complicated more than 870,000 individuals and led to more than 43,000 deaths worldwide. Considering to the potential threat of a pandemic and transmission severity of it, there is an urgent need to evaluate and realize this new virus's structure and behavior and the immunopathology of this disease to find potential therapeutic protocols and to design and develop effective vaccines. This disease is able to agitate the response of the immune system in the infected patients, so ARDS, as a common consequence of immunopathological events for infections with Middle East respiratory syndrome coronavirus (MERS-CoV), SARS-CoV, and SARS-CoV-2, could be the main reason for death. Here, we summarized the immune response and immune evasion characteristics in SARS-CoV, MERS-CoV, and SARS-CoV-2 and therapeutic and prophylactic strategies with a focus on vaccine development and its challenges.
Collapse
Affiliation(s)
- Soheila Molaei
- Deputy of Research & Technology, Ardabil University of Medical Sciences, Ardabil, Iran; Pharmaceutical Sciences Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Masoomeh Dadkhah
- Pharmaceutical Sciences Research Center, Ardabil University of Medical Sciences, Ardabil, Iran; Department of Pharmacology, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Vahid Asghariazar
- Deputy of Research & Technology, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Chiman Karami
- Department of Microbiology, Parasitology, and Immunology, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Elham Safarzadeh
- Department of Microbiology, Parasitology, and Immunology, Ardabil University of Medical Sciences, Ardabil, Iran.
| |
Collapse
|
50
|
ElBagoury M, Tolba MM, Nasser HA, Jabbar A, Elagouz AM, Aktham Y, Hutchinson A. The find of COVID-19 vaccine: Challenges and opportunities. J Infect Public Health 2021; 14:389-416. [PMID: 33647555 PMCID: PMC7773313 DOI: 10.1016/j.jiph.2020.12.025] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 11/30/2020] [Accepted: 12/20/2020] [Indexed: 12/19/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus (SARS-CoV-2), a novel corona virus, causing COVID-19 with Flu-like symptoms is the first alarming pandemic of the third millennium. SARS-CoV-2 belongs to beta coronavirus as Middle East respiratory syndrome coronavirus (MERS-CoV). Pandemic COVID-19 owes devastating mortality and destructively exceptional consequences on Socio-Economics life around the world. Therefore, the current review is redirected to the scientific community to owe comprehensive visualization about SARS-CoV-2 to tackle the current pandemic. As systematically shown through the current review, it indexes unmet medical problem of COVID-19 in view of public health and vaccination discovery for the infectious SARS-CoV-2; it is currently under-investigational therapeutic protocols, and next possible vaccines. Furthermore, the review extensively reports the precautionary measures to achieve" COVID-19/Flatten the curve". It is concluded that vaccines formulation within exceptional no time in this pandemic is highly recommended, via following the same protocols of previous pandemics; MERS-CoV and SARS-CoV, and excluding some initial steps of vaccination development process.
Collapse
Affiliation(s)
- Marwan ElBagoury
- University of South Wales, Pontypridd, Wales, United Kingdom; The Student Science and Technology Online Research Coop, Ontario, Canada.
| | - Mahmoud M Tolba
- Pharmaceutical division, ministry of health and population, Cairo, Egypt
| | - Hebatallah A Nasser
- Microbiology and Public Health Department, Faculty of Pharmacy, Heliopolis University, Cairo, Egypt
| | - Abdul Jabbar
- Department of Clinical Medicine, University of Veterinary and Animal Sciences, Lahore Punjab Pakistan
| | - Ahmed M Elagouz
- University of South Wales, Pontypridd, Wales, United Kingdom
| | - Yahia Aktham
- University of South Wales, Pontypridd, Wales, United Kingdom
| | - Amy Hutchinson
- The Student Science and Technology Online Research Coop, Ontario, Canada; McMaster University, Hamilton, Canada
| |
Collapse
|