1
|
Wei L, Yu P, Wang H, Liu J. Adeno-associated viral vectors deliver gene vaccines. Eur J Med Chem 2025; 281:117010. [PMID: 39488197 DOI: 10.1016/j.ejmech.2024.117010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 10/24/2024] [Accepted: 10/27/2024] [Indexed: 11/04/2024]
Abstract
Adeno-associated viruses (AAVs) are leading platforms for in vivo delivery of gene therapies, with six licensed AAV-based therapeutics attributed to their non-pathogenic nature, low immunogenicity, and high efficiency. In the realm of gene-based vaccines, one of the most vital therapeutic areas, AAVs are also emerging as promising delivery tools. We scrutinized AAVs, focusing on their virological properties, as well as bioengineering and chemical modifications to demonstrate their significant potential in gene vaccine delivery, and detailing the preparation of AAV particles. Additionally, we summarized the use of AAV vectors in vaccines for both infectious and non-infectious diseases, such as influenza, COVID-19, Alzheimer's disease, and cancer. Furthermore, this review, along with the latest clinical trial updates, provides a comprehensive overview of studies on the potential of using AAV vectors for gene vaccine delivery. It aims to deepen our understanding of the challenges and limitations in nucleic acid delivery and pave the way for future clinical success.
Collapse
Affiliation(s)
- Lai Wei
- College of Life Science and Technology, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Peng Yu
- College of Biotechnology, Tianjin University of Science & Technology, 300457 Tianjin, China
| | - Haomeng Wang
- CanSino (Shanghai) Biological Research Co., Ltd, 201208, Shanghai, China.
| | - Jiang Liu
- Rosalind Franklin Institute, Harwell Campus, OX11 0QS, Oxford, United Kingdom; Department of Pharmacology, University of Oxford, Mansfield Road, OX1 3QT, Oxford, United Kingdom.
| |
Collapse
|
2
|
Wang Z, Zhang Z, Wang Q, Zeng L, Jin J. A nasal spray vaccination device based on Laval nozzle and its experimental test. Sci Rep 2023; 13:6267. [PMID: 37069262 PMCID: PMC10109226 DOI: 10.1038/s41598-023-33452-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 04/13/2023] [Indexed: 04/19/2023] Open
Abstract
In order to realize the application of the nasal spray vaccination in the prevention and protection of respiratory infectious diseases, a nasal spray vaccination device is designed in this paper. The device uses a Laval nozzle structure to generate a high-speed airflow that impinges on the vaccine reagent and forms nebulized particles. Through optimizing of the Laval nozzle structure and testing experiments on spray particle size, spray velocity, spray angle and spray rate, a set of parameters which is applicable to actual nasal spray vaccination is obtained. The experimental results show that when the air source pressure is 2 bar, the spray angle is about 15°, the diameter of the spray particles Dv50 is about 17 μm, the volume fraction of particles with diameter smaller than 10um is about 24%, the spray rate is close to 300 μl/s. The vaccine activity tests demonstrate that under these conditions, not only the biological activity of vaccines is guaranteed, but also the delivery efficiency is well assured.
Collapse
Affiliation(s)
- Zhong Wang
- Guangzhou Laboratory, Guangzhou, 510005, China
| | - Zhengyuan Zhang
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Qian Wang
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510182, China
| | | | - Jian Jin
- Guangzhou Laboratory, Guangzhou, 510005, China.
- School of Mechanical Engineering, Hefei University of Technology, Hefei, 230009, China.
| |
Collapse
|
3
|
Jacob-Dolan C, Yu J, McMahan K, Giffin V, Chandrashekar A, Martinot AJ, Anioke T, Powers OC, Hall K, Hope D, Miller J, Hachmann NP, Chung B, Gardner S, Sellers D, Barrett J, Lewis MG, Andersen H, Kleanthous H, Seo KW, Lee SJ, Park YW, Kim H, Barouch DH. Immunogenicity and protective efficacy of GBP510/AS03 vaccine against SARS-CoV-2 delta challenge in rhesus macaques. NPJ Vaccines 2023; 8:23. [PMID: 36823160 PMCID: PMC9947939 DOI: 10.1038/s41541-023-00622-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 02/09/2023] [Indexed: 02/25/2023] Open
Abstract
Despite the availability of several effective SARS-CoV-2 vaccines, additional vaccines will be required for optimal global vaccination. In this study, we investigate the immunogenicity and protective efficacy of the GBP510 protein subunit vaccine adjuvanted with AS03, which has recently been authorized for marketing in South Korea under the trade name SKYCovioneTM. The antigen in GBP510/AS03 is a two-part recombinant nanoparticle, which displays 60 receptor binding domain (RBD) proteins of SARS-CoV-2 Spike on its surface. In this study we show that GBP510/AS03 induced robust immune responses in rhesus macaques and protected against a high-dose SARS-CoV-2 Delta challenge. We vaccinated macaques with two or three doses of GBP510/AS03 matched to the ancestral Wuhan strain of SARS-CoV-2 or with two doses of GBP510/AS03 matched to the ancestral strain and one dose matched to the Beta strain. Following the challenge with Delta, the vaccinated macaques rapidly controlled the virus in bronchoalveolar lavage and nasal swabs. Binding and neutralizing antibody responses prior to challenge correlated with protection against viral replication postchallenge. These data are consistent with data with this vaccine from the phase 3 clinical trial.
Collapse
Affiliation(s)
- Catherine Jacob-Dolan
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, 02115, USA
| | - Jingyou Yu
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA
| | - Katherine McMahan
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA
| | - Victoria Giffin
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA
| | - Abishek Chandrashekar
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA
| | - Amanda J Martinot
- Tufts Cummings School of Veterinary Medicine, North Grafton, MA, 01536, USA
| | - Tochi Anioke
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA
| | - Olivia C Powers
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA
| | - Kevin Hall
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA
| | - David Hope
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA
| | - Jessica Miller
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA
| | - Nichole P Hachmann
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA
| | - Benjamin Chung
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA
| | - Sarah Gardner
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA
| | - Daniel Sellers
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA
| | - Julia Barrett
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA
| | | | | | | | - Ki-Woen Seo
- Department of Research and Development, SK bioscience 310 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Su Jeen Lee
- Department of Research and Development, SK bioscience 310 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Yong Wook Park
- Department of Research and Development, SK bioscience 310 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Hun Kim
- Department of Research and Development, SK bioscience 310 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Dan H Barouch
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA.
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA.
- Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
4
|
Boley PA, Lee CM, Schrock J, Yadav KK, Patil V, Suresh R, Lu S, Feng MM, Hanson J, Channappanavar R, Kenney SP, Renukaradhya GJ. Enhanced mucosal immune responses and reduced viral load in the respiratory tract of ferrets to intranasal lipid nanoparticle-based SARS-CoV-2 proteins and mRNA vaccines. J Nanobiotechnology 2023; 21:60. [PMID: 36814238 PMCID: PMC9944789 DOI: 10.1186/s12951-023-01816-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 02/14/2023] [Indexed: 02/24/2023] Open
Abstract
BACKGROUND Unlike the injectable vaccines, intranasal lipid nanoparticle (NP)-based adjuvanted vaccine is promising to protect against local infection and viral transmission. Infection of ferrets with SARS-CoV-2 results in typical respiratory disease and pathology akin to in humans, suggesting that the ferret model may be ideal for intranasal vaccine studies. RESULTS We developed SARS-CoV-2 subunit vaccine containing both Spike receptor binding domain (S-RBD) and Nucleocapsid (N) proteins (NP-COVID-Proteins) or their mRNA (NP-COVID-mRNA) and NP-monosodium urate adjuvant. Both the candidate vaccines in intranasal vaccinated aged ferrets substantially reduced the replicating virus in the entire respiratory tract. Specifically, the NP-COVID-Proteins vaccine did relatively better in clearing the virus from the nasal passage early post challenge infection. The immune gene expression in NP-COVID-Proteins vaccinates indicated increased levels of mRNA of IFNα, MCP1 and IL-4 in lungs and nasal turbinates, and IFNγ and IL-2 in lungs; while proinflammatory mediators IL-1β and IL-8 mRNA levels in lungs were downregulated. In NP-COVID-Proteins vaccinated ferrets S-RBD and N protein specific IgG antibodies in the serum were substantially increased at both day post challenge (DPC) 7 and DPC 14, while the virus neutralizing antibody titers were relatively better induced by mRNA versus the proteins-based vaccine. In conclusion, intranasal NP-COVID-Proteins vaccine induced balanced Th1 and Th2 immune responses in the respiratory tract, while NP-COVID-mRNA vaccine primarily elicited antibody responses. CONCLUSIONS Intranasal NP-COVID-Proteins vaccine may be an ideal candidate to elicit increased breadth of immunity against SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Patricia A Boley
- Center for Food Animal Health, Department of Animal Sciences, The Ohio State University, 1680 Madison Avenue, Wooster, OH, 44691, USA
| | - Carolyn M Lee
- Center for Food Animal Health, Department of Animal Sciences, The Ohio State University, 1680 Madison Avenue, Wooster, OH, 44691, USA
| | - Jennifer Schrock
- Center for Food Animal Health, Department of Animal Sciences, The Ohio State University, 1680 Madison Avenue, Wooster, OH, 44691, USA
| | - Kush Kumar Yadav
- Center for Food Animal Health, Department of Animal Sciences, The Ohio State University, 1680 Madison Avenue, Wooster, OH, 44691, USA
| | - Veerupaxagouda Patil
- Center for Food Animal Health, Department of Animal Sciences, The Ohio State University, 1680 Madison Avenue, Wooster, OH, 44691, USA
| | - Raksha Suresh
- Center for Food Animal Health, Department of Animal Sciences, The Ohio State University, 1680 Madison Avenue, Wooster, OH, 44691, USA
| | - Songqing Lu
- Dynamic Entropy Technology LLC, Building B, 1028 W. Nixon St., Pasco, WA, 99301-5216, USA
| | - Maoqi Mark Feng
- Dynamic Entropy Technology LLC, Building B, 1028 W. Nixon St., Pasco, WA, 99301-5216, USA
| | - Juliette Hanson
- Center for Food Animal Health, Department of Animal Sciences, The Ohio State University, 1680 Madison Avenue, Wooster, OH, 44691, USA
| | - Rudra Channappanavar
- Department of Veterinary Pathobiology, Oklahoma Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, OK, 74078, USA
| | - Scott P Kenney
- Center for Food Animal Health, Department of Animal Sciences, The Ohio State University, 1680 Madison Avenue, Wooster, OH, 44691, USA.
| | - Gourapura J Renukaradhya
- Center for Food Animal Health, Department of Animal Sciences, The Ohio State University, 1680 Madison Avenue, Wooster, OH, 44691, USA.
| |
Collapse
|
5
|
Law LH, Huang J, Xiao P, Liu Y, Chen Z, Lai JHC, Han X, Cheng GWY, Tse KH, Chan KWY. Multiple CEST contrast imaging of nose-to-brain drug delivery using iohexol liposomes at 3T MRI. J Control Release 2023; 354:208-220. [PMID: 36623695 DOI: 10.1016/j.jconrel.2023.01.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 12/28/2022] [Accepted: 01/03/2023] [Indexed: 01/11/2023]
Abstract
Image guided nose-to-brain drug delivery provides a non-invasive way to monitor drug delivered to the brain, and the intranasal administration could increase effective dose via bypassing Blood Brain Barrier (BBB). Here, we investigated the imaging of liposome-based drug delivery to the brain via intranasal administration, in which the liposome could penetrate mucus and could be detected by chemical exchange saturation transfer (CEST) magnetic resonance imaging (MRI) at 3T field strength. Liposomes were loaded with a computed tomography (CT) contrast agent, iohexol (Ioh-Lipo), which has specific amide protons exchanging at 4.3 ppm of Z-spectrum (or CEST spectrum). Ioh-Lipo generated CEST contrasts of 35.4% at 4.3 ppm, 1.8% at -3.4 ppm and 20.6% at 1.2 ppm in vitro. After intranasal administration, these specific CEST contrasts were observed in both olfactory bulb (OB) and frontal lobe (FL) in the case of 10% polyethylene glycol (PEG) Ioh-Lipo. We observed obvious increases in CEST contrast in OB half an hour after the injection of 10% PEG Ioh-Lipo, with a percentage increase of 62.0% at 4.3 ppm, 10.9% at -3.4 ppm and 25.7% at 1.2 ppm. Interestingly, the CEST map at 4.3 ppm was distinctive from that at -3.4 pm and 1.2 ppm. The highest contrast of 4.3 ppm was at the external plexiform layer (EPL) and the region between left and right OB (LROB), while the CEST contrast at -3.4 ppm had no significant difference among all investigated regions with slightly higher signal in olfactory limbus (OL, between OB and FL) and FL, as validated with histology. While no substantial increase of CEST contrast at 4.3 ppm, -3.4 ppm or 1.2 ppm was observed in OB and FL when 1% PEG Ioh-Lipo was administered. We demonstrated for the first time the feasibility of non-invasively detecting the nose-to-brain delivery of liposomes using CEST MRI. This multiple-contrast approach is necessary to image the specific distribution of iohexol and liposome simultaneously and independently, especially when designing drug carriers for nose-to-brain drug delivery.
Collapse
Affiliation(s)
- Lok Hin Law
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China
| | - Jianpan Huang
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China
| | - Peng Xiao
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China
| | - Yang Liu
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China
| | - Zilin Chen
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China
| | - Joseph H C Lai
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China
| | - Xiongqi Han
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China
| | - Gerald W Y Cheng
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China
| | - Kai-Hei Tse
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China
| | - Kannie W Y Chan
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China; Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, United States; City University of Hong Kong Shenzhen Research Institute, Shenzhen, China; Tung Biomedical Science Centre, City University of Hong Kong, Hong Kong, China; Hong Kong Centre for Cerebro-cardiovascular Health Engineering, Hong Kong, China.
| |
Collapse
|
6
|
Saint-Pierre Contreras G, Conei Valencia D, Lizama L, Vargas Zuñiga D, Avendaño Carvajal LF, Ampuero Llanos S. An Old Acquaintance: Could Adenoviruses Be Our Next Pandemic Threat? Viruses 2023; 15:330. [PMID: 36851544 PMCID: PMC9966032 DOI: 10.3390/v15020330] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/17/2023] [Accepted: 01/18/2023] [Indexed: 01/26/2023] Open
Abstract
Human adenoviruses (HAdV) are one of the most important pathogens detected in acute respiratory diseases in pediatrics and immunocompromised patients. In 1953, Wallace Rowe described it for the first time in oropharyngeal lymphatic tissue. To date, more than 110 types of HAdV have been described, with different cellular tropisms. They can cause respiratory and gastrointestinal symptoms, even urinary tract inflammation, although most infections are asymptomatic. However, there is a population at risk that can develop serious and even lethal conditions. These viruses have a double-stranded DNA genome, 25-48 kbp, 90 nm in diameter, without a mantle, are stable in the environment, and resistant to fat-soluble detergents. Currently the diagnosis is made with lateral flow immunochromatography or molecular biology through a polymerase chain reaction. This review aimed to highlight the HAdV variability and the pandemic potential that a HAdV3 and 7 recombinant could have considering the aggressive outbreaks produced in health facilities. Herein, we described the characteristics of HAdV, from the infection to treatment, vaccine development, and the evaluation of the social determinants of health associated with HAdV, suggesting the necessary measures for future sanitary control to prevent disasters such as the SARS-CoV-2 pandemic, with an emphasis on the use of recombinant AdV vaccines to control other potential pandemics.
Collapse
Affiliation(s)
- Gustavo Saint-Pierre Contreras
- Programa de Virología, ICBM, Facultad de Medicina, Universidad de Chile, Independencia 1027, Santiago 8380453, Chile
- Unidad Microbiología, Hospital Barros Luco Trudeau, Servicio de Salud Metropolitano Sur, Santiago 8900000, Chile
| | - Daniel Conei Valencia
- Departamento de Ciencias de la Salud, Universidad de Aysén, Coyhaique 5951537, Chile
| | - Luis Lizama
- Programa de Virología, ICBM, Facultad de Medicina, Universidad de Chile, Independencia 1027, Santiago 8380453, Chile
| | - Daniela Vargas Zuñiga
- Programa de Virología, ICBM, Facultad de Medicina, Universidad de Chile, Independencia 1027, Santiago 8380453, Chile
| | - Luis Fidel Avendaño Carvajal
- Programa de Virología, ICBM, Facultad de Medicina, Universidad de Chile, Independencia 1027, Santiago 8380453, Chile
| | - Sandra Ampuero Llanos
- Programa de Virología, ICBM, Facultad de Medicina, Universidad de Chile, Independencia 1027, Santiago 8380453, Chile
| |
Collapse
|
7
|
Abdolmaleki G, Taheri MA, Paridehpour S, Mohammadi NM, Tabatabaei YA, Mousavi T, Amin M. A comparison between SARS-CoV-1 and SARS-CoV2: an update on current COVID-19 vaccines. Daru 2022; 30:379-406. [PMID: 36050585 PMCID: PMC9436716 DOI: 10.1007/s40199-022-00446-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Accepted: 08/05/2022] [Indexed: 10/31/2022] Open
Abstract
Since the outbreak of the novel coronavirus disease 2019 (COVID-19) in Wuhan, China, many health care systems have been heavily engaged in treating and preventing the disease, and the year 2020 may be called as "historic COVID-19 vaccine breakthrough". Due to the COVID-19 pandemic, many companies have initiated investigations on developing an efficient and safe vaccine against the virus. From Moderna and Pfizer in the United States to PastocoVac in Pasteur Institute of Iran and the University of Oxford in the United Kingdom, different candidates have been introduced to the market. COVID-19 vaccine research has been facilitated based on genome and structural information, bioinformatics predictions, epitope mapping, and data obtained from the previous developments of severe acute respiratory syndrome coronavirus (SARS-CoV or SARS-CoV-1) and middle east respiratory syndrome coronavirus (MERS-CoV) vaccine candidates. SARS-CoV genome sequence is highly homologous to the one in COVID-19 and both viruses use the same receptor, angiotensin-converting enzyme 2 (ACE2). Moreover, the immune system responds to these viruses, partially in the same way. Considering the on-going COVID-19 pandemic and previous attempts to manufacture SARS-CoV vaccines, this paper is going to discuss clinical cases as well as vaccine challenges, including those related to infrastructures, transportation, possible adverse reactions, utilized delivery systems (e.g., nanotechnology and electroporation) and probable vaccine-induced mutations.
Collapse
Affiliation(s)
- Gelareh Abdolmaleki
- Department of Drug and Food Control, Faculty of Pharmacy, Tehran University of Medical Sciences, Room No. 1-221, 16th Azar Street, Tehran, Iran
| | - Mina Azam Taheri
- Department of Drug and Food Control, Faculty of Pharmacy, Tehran University of Medical Sciences, Room No. 1-221, 16th Azar Street, Tehran, Iran
| | - Sarina Paridehpour
- Department of Drug and Food Control, Faculty of Pharmacy, Tehran University of Medical Sciences, Room No. 1-221, 16th Azar Street, Tehran, Iran
| | - Neshaut Mashreghi Mohammadi
- Department of Drug and Food Control, Faculty of Pharmacy, Tehran University of Medical Sciences, Room No. 1-221, 16th Azar Street, Tehran, Iran
- Pharmaceutical Microbiology Group, Pharmaceutical Quality Assurance Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Yasaman Ahmadi Tabatabaei
- Department of Drug and Food Control, Faculty of Pharmacy, Tehran University of Medical Sciences, Room No. 1-221, 16th Azar Street, Tehran, Iran
| | - Taraneh Mousavi
- Toxicology and Diseases Group (TDG), Pharmaceutical Sciences Research Center (PSRC), Tehran University of Medical Sciences, Tehran, Iran
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohsen Amin
- Department of Drug and Food Control, Faculty of Pharmacy, Tehran University of Medical Sciences, Room No. 1-221, 16th Azar Street, Tehran, Iran.
- Pharmaceutical Microbiology Group, Pharmaceutical Quality Assurance Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
8
|
Valenzuela-Fernández A, Cabrera-Rodriguez R, Ciuffreda L, Perez-Yanes S, Estevez-Herrera J, González-Montelongo R, Alcoba-Florez J, Trujillo-González R, García-Martínez de Artola D, Gil-Campesino H, Díez-Gil O, Lorenzo-Salazar JM, Flores C, Garcia-Luis J. Nanomaterials to combat SARS-CoV-2: Strategies to prevent, diagnose and treat COVID-19. Front Bioeng Biotechnol 2022; 10:1052436. [PMID: 36507266 PMCID: PMC9732709 DOI: 10.3389/fbioe.2022.1052436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 11/09/2022] [Indexed: 11/26/2022] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and the associated coronavirus disease 2019 (COVID-19), which severely affect the respiratory system and several organs and tissues, and may lead to death, have shown how science can respond when challenged by a global emergency, offering as a response a myriad of rapid technological developments. Development of vaccines at lightning speed is one of them. SARS-CoV-2 outbreaks have stressed healthcare systems, questioning patients care by using standard non-adapted therapies and diagnostic tools. In this scenario, nanotechnology has offered new tools, techniques and opportunities for prevention, for rapid, accurate and sensitive diagnosis and treatment of COVID-19. In this review, we focus on the nanotechnological applications and nano-based materials (i.e., personal protective equipment) to combat SARS-CoV-2 transmission, infection, organ damage and for the development of new tools for virosurveillance, diagnose and immune protection by mRNA and other nano-based vaccines. All the nano-based developed tools have allowed a historical, unprecedented, real time epidemiological surveillance and diagnosis of SARS-CoV-2 infection, at community and international levels. The nano-based technology has help to predict and detect how this Sarbecovirus is mutating and the severity of the associated COVID-19 disease, thereby assisting the administration and public health services to make decisions and measures for preparedness against the emerging variants of SARS-CoV-2 and severe or lethal COVID-19.
Collapse
Affiliation(s)
- Agustín Valenzuela-Fernández
- Laboratorio de Inmunología Celular y Viral, Unidad de Farmacología, Sección de Medicina, Facultad de Ciencias de la Salud, Universidad de La Laguna, San Cristóbal de La Laguna, Spain
| | - Romina Cabrera-Rodriguez
- Laboratorio de Inmunología Celular y Viral, Unidad de Farmacología, Sección de Medicina, Facultad de Ciencias de la Salud, Universidad de La Laguna, San Cristóbal de La Laguna, Spain
| | - Laura Ciuffreda
- Research Unit, Hospital Universitario N. S. de Candelaria, Santa Cruz de Tenerife, Spain
| | - Silvia Perez-Yanes
- Laboratorio de Inmunología Celular y Viral, Unidad de Farmacología, Sección de Medicina, Facultad de Ciencias de la Salud, Universidad de La Laguna, San Cristóbal de La Laguna, Spain
| | - Judith Estevez-Herrera
- Laboratorio de Inmunología Celular y Viral, Unidad de Farmacología, Sección de Medicina, Facultad de Ciencias de la Salud, Universidad de La Laguna, San Cristóbal de La Laguna, Spain
| | | | - Julia Alcoba-Florez
- Servicio de Microbiología, Hospital Universitario N. S. de Candelaria, Santa Cruz de Tenerife, Spain
| | - Rodrigo Trujillo-González
- Laboratorio de Inmunología Celular y Viral, Unidad de Farmacología, Sección de Medicina, Facultad de Ciencias de la Salud, Universidad de La Laguna, San Cristóbal de La Laguna, Spain
- Departamento de Análisis Matemático, Facultad de Ciencias, Universidad de La Laguna, Santa Cruz de Tenerife, Spain
| | | | - Helena Gil-Campesino
- Servicio de Microbiología, Hospital Universitario N. S. de Candelaria, Santa Cruz de Tenerife, Spain
| | - Oscar Díez-Gil
- Servicio de Microbiología, Hospital Universitario N. S. de Candelaria, Santa Cruz de Tenerife, Spain
| | - José M. Lorenzo-Salazar
- Genomics Division, Instituto Tecnológico y de Energías Renovables, Santa Cruz de Tenerife, Spain
| | - Carlos Flores
- Research Unit, Hospital Universitario N. S. de Candelaria, Santa Cruz de Tenerife, Spain
- Genomics Division, Instituto Tecnológico y de Energías Renovables, Santa Cruz de Tenerife, Spain
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
- Faculty of Health Sciences, University of Fernando Pessoa Canarias, Las Palmas de Gran Canaria, Spain
| | - Jonay Garcia-Luis
- Laboratorio de Inmunología Celular y Viral, Unidad de Farmacología, Sección de Medicina, Facultad de Ciencias de la Salud, Universidad de La Laguna, San Cristóbal de La Laguna, Spain
| |
Collapse
|
9
|
Yaugel-Novoa M, Bourlet T, Paul S. Role of the humoral immune response during COVID-19: guilty or not guilty? Mucosal Immunol 2022; 15:1170-1180. [PMID: 36195658 PMCID: PMC9530436 DOI: 10.1038/s41385-022-00569-w] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 08/07/2022] [Accepted: 09/19/2022] [Indexed: 02/04/2023]
Abstract
Systemic and mucosal humoral immune responses are crucial to fight respiratory viral infections in the current pandemic of COVID-19 caused by the SARS-CoV-2 virus. During SARS-CoV-2 infection, the dynamics of systemic and mucosal antibody infections are affected by patient characteristics, such as age, sex, disease severity, or prior immunity to other human coronaviruses. Patients suffering from severe disease develop higher levels of anti-SARS-CoV-2 antibodies in serum and mucosal tissues than those with mild disease, and these antibodies are detectable for up to a year after symptom onset. In hospitalized patients, the aberrant glycosylation of anti-SARS-CoV-2 antibodies enhances inflammation-associated antibody Fc-dependent effector functions, thereby contributing to COVID-19 pathophysiology. Current vaccines elicit robust humoral immune responses, principally in the blood. However, they are less effective against new viral variants, such as Delta and Omicron. This review provides an overview of current knowledge about the humoral immune response to SARS-CoV-2, with a particular focus on the protective and pathological role of humoral immunity in COVID-19 severity. We also discuss the humoral immune response elicited by COVID-19 vaccination and protection against emerging viral variants.
Collapse
Affiliation(s)
- Melyssa Yaugel-Novoa
- CIRI—Centre International de Recherche en Infectiologie, Team GIMAP (Saint-Etienne), Inserm, U1111, CNRS, UMR5308, ENS Lyon, UJM, Université Claude Bernard Lyon 1, Lyon, France
| | - Thomas Bourlet
- CIRI—Centre International de Recherche en Infectiologie, Team GIMAP (Saint-Etienne), Inserm, U1111, CNRS, UMR5308, ENS Lyon, UJM, Université Claude Bernard Lyon 1, Lyon, France
| | - Stéphane Paul
- CIRI—Centre International de Recherche en Infectiologie, Team GIMAP (Saint-Etienne), Inserm, U1111, CNRS, UMR5308, ENS Lyon, UJM, Université Claude Bernard Lyon 1, Lyon, France,CIC Inserm 1408 Vaccinology, Saint-Etienne, France
| |
Collapse
|
10
|
Adam A, Shi Q, Wang B, Zou J, Mai J, Osman SR, Wu W, Xie X, Aguilar PV, Bao X, Shi PY, Shen H, Wang T. A modified porous silicon microparticle potentiates protective systemic and mucosal immunity for SARS-CoV-2 subunit vaccine. Transl Res 2022; 249:13-27. [PMID: 35688318 PMCID: PMC9173827 DOI: 10.1016/j.trsl.2022.06.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 05/30/2022] [Accepted: 06/02/2022] [Indexed: 11/28/2022]
Abstract
Development of optimal SARS-CoV-2 vaccines to induce potent, long-lasting immunity and provide cross-reactive protection against emerging variants remains a high priority. Here, we report that a modified porous silicon microparticle (mPSM) adjuvant to SARS-CoV-2 receptor-binding domain (RBD) vaccine activated dendritic cells and generated more potent and durable systemic humoral and type 1 helper T (Th) cell- mediated immune responses than alum-formulated RBD following parenteral vaccination, and protected mice from SARS-CoV-2 and Beta variant challenge. Notably, mPSM facilitated the uptake of SARS-CoV-2 RBD antigens by nasal and airway epithelial cells. Parenteral and intranasal prime and boost vaccinations with mPSM-RBD elicited stronger lung resident T and B cells and IgA responses compared to parenteral vaccination alone, which led to markedly diminished viral loads and inflammation in the lung following SARS-CoV-2 Delta variant challenge. Overall, our results suggest that mPSM is effective adjuvant for SARS-CoV-2 subunit vaccine in both systemic and mucosal vaccinations.
Collapse
Key Words
- mpsm, modified porous silicon microparticle
- covid-19, coronavirus disease 2019
- rbd, receptor-binding domain
- sars-cov-2, severe acute respiratory syndrome coronavirus 2
- β-cov, betacoronavirus
- e, envelope
- m, membrane
- n, nucleocapsid
- hace2, human angiotensin-converting enzyme 2
- nabs, neutralizing antibodies
- dc, dendritic cell
- th1, t helper 1
- cpg, cytosine guanosine dinucleotide
- cgamp, cyclic gamp
- bm, bone marrow
- i.p., intraperitoneally
- i.d., intradermally
- i.m., or intramuscularly
- tmb, tetramethylbenzidine
- pbs-t, phosphate-buffered saline containing tween-20
- bal, bronchoalveolar lavage
- hrp, horseradish peroxidase
- elisa, enzyme-linked immunosorbent assay
- elispot, enzyme-linked immune absorbent spot
- sfc, spot-forming cells
- ics, intracellular cytokine staining
- moi, multiplicity of infection
- apc, antigen presenting cells
- mbc, memory b cell
- asc, antibody secreting cells
- prnt, plaque reduction neutralization test
- saec, small airway epithelial cells
- nalt, nasal-associated lymphoid tissue
- ade, antibody-dependent enhancement
- q-pcr, quantitative pcr
Collapse
Affiliation(s)
- Awadalkareem Adam
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, Texas
| | - Qing Shi
- Department of Nanomedicine, Houston Methodist Academic Institute, Houston, Texas
| | - Binbin Wang
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, Texas
| | - Jing Zou
- Department of Biochemistry & Molecular Biology, University of Texas Medical Branch, Galveston, Texas
| | - Junhua Mai
- Department of Nanomedicine, Houston Methodist Academic Institute, Houston, Texas
| | - Samantha R Osman
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, Texas
| | - Wenzhe Wu
- Department of Pediatrics, The University of Texas Medical Branch, Galveston, Texas
| | - Xuping Xie
- Department of Biochemistry & Molecular Biology, University of Texas Medical Branch, Galveston, Texas
| | - Patricia V Aguilar
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas; Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, Texas; Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, Texas
| | - Xiaoyong Bao
- Department of Pediatrics, The University of Texas Medical Branch, Galveston, Texas; Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, Texas
| | - Pei-Yong Shi
- Department of Biochemistry & Molecular Biology, University of Texas Medical Branch, Galveston, Texas; Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, Texas; Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, Texas
| | - Haifa Shen
- Department of Nanomedicine, Houston Methodist Academic Institute, Houston, Texas; Innovative Therapeutic Program, Houston Methodist Cancer Center, Houston, Texas; ImmunoQ Therapeutics, Houston, Texas.
| | - Tian Wang
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, Texas; Department of Pathology, University of Texas Medical Branch, Galveston, Texas; Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, Texas; Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, Texas
| |
Collapse
|
11
|
Davis SK, Selva KJ, Lopez E, Haycroft ER, Lee WS, Wheatley AK, Juno JA, Adair A, Pymm P, Redmond SJ, Gherardin NA, Godfrey DI, Tham W, Kent SJ, Chung AW. Heterologous SARS-CoV-2 IgA neutralising antibody responses in convalescent plasma. Clin Transl Immunology 2022; 11:e1424. [PMID: 36299410 PMCID: PMC9588388 DOI: 10.1002/cti2.1424] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 07/21/2022] [Accepted: 09/28/2022] [Indexed: 12/14/2022] Open
Abstract
Objectives Following infection with SARS-CoV-2, virus-specific antibodies are generated, which can both neutralise virions and clear infection via Fc effector functions. The importance of IgG antibodies for protection and control of SARS-CoV-2 has been extensively reported. By comparison, other antibody isotypes including IgA have been poorly characterised. Methods Here, we characterised plasma IgA from 41 early convalescent COVID-19 subjects for neutralisation and Fc effector functions. Results Convalescent plasma IgA from > 60% of the cohort had the capacity to inhibit the interaction between wild-type RBD and ACE2. Furthermore, a third of the cohort induced stronger IgA-mediated ACE2 inhibition than matched IgG when tested at equivalent concentrations. Plasma IgA and IgG from this cohort broadly recognised similar RBD epitopes and had similar capacities to inhibit ACE2 from binding to 22 of the 23 prevalent RBD mutations assessed. However, plasma IgA was largely incapable of mediating antibody-dependent phagocytosis in comparison with plasma IgG. Conclusion Overall, convalescent plasma IgA contributed to the neutralising antibody response of wild-type SARS-CoV-2 RBD and various RBD mutations. However, this response displayed large heterogeneity and was less potent than IgG.
Collapse
Affiliation(s)
- Samantha K Davis
- Department of Microbiology and ImmunologyThe Peter Doherty Institute for Infection and ImmunityUniversity of MelbourneMelbourneVICAustralia
| | - Kevin John Selva
- Department of Microbiology and ImmunologyThe Peter Doherty Institute for Infection and ImmunityUniversity of MelbourneMelbourneVICAustralia
| | - Ester Lopez
- Department of Microbiology and ImmunologyThe Peter Doherty Institute for Infection and ImmunityUniversity of MelbourneMelbourneVICAustralia
| | - Ebene R Haycroft
- Department of Microbiology and ImmunologyThe Peter Doherty Institute for Infection and ImmunityUniversity of MelbourneMelbourneVICAustralia
| | - Wen Shi Lee
- Department of Microbiology and ImmunologyThe Peter Doherty Institute for Infection and ImmunityUniversity of MelbourneMelbourneVICAustralia
- The Walter and Eliza Hall Institute of Medical ResearchMelbourneVICAustralia
| | - Adam K Wheatley
- Department of Microbiology and ImmunologyThe Peter Doherty Institute for Infection and ImmunityUniversity of MelbourneMelbourneVICAustralia
| | - Jennifer A Juno
- Department of Microbiology and ImmunologyThe Peter Doherty Institute for Infection and ImmunityUniversity of MelbourneMelbourneVICAustralia
| | - Amy Adair
- The Walter and Eliza Hall Institute of Medical ResearchMelbourneVICAustralia
| | - Phillip Pymm
- The Walter and Eliza Hall Institute of Medical ResearchMelbourneVICAustralia
| | - Samuel J Redmond
- Department of Microbiology and ImmunologyThe Peter Doherty Institute for Infection and ImmunityUniversity of MelbourneMelbourneVICAustralia
| | - Nicholas A Gherardin
- Department of Microbiology and ImmunologyThe Peter Doherty Institute for Infection and ImmunityUniversity of MelbourneMelbourneVICAustralia
| | - Dale I Godfrey
- Department of Microbiology and ImmunologyThe Peter Doherty Institute for Infection and ImmunityUniversity of MelbourneMelbourneVICAustralia
| | - Wai‐Hong Tham
- The Walter and Eliza Hall Institute of Medical ResearchMelbourneVICAustralia
| | - Stephen J Kent
- Department of Microbiology and ImmunologyThe Peter Doherty Institute for Infection and ImmunityUniversity of MelbourneMelbourneVICAustralia
- Melbourne Sexual Health Centre and Department of Infectious DiseasesAlfred Hospital and Central Clinical SchoolMonash UniversityMelbourneVICAustralia
| | - Amy W Chung
- Department of Microbiology and ImmunologyThe Peter Doherty Institute for Infection and ImmunityUniversity of MelbourneMelbourneVICAustralia
| |
Collapse
|
12
|
Intranasal application of adeno-associated viruses: a systematic review. Transl Res 2022; 248:87-110. [PMID: 35597541 DOI: 10.1016/j.trsl.2022.05.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 04/12/2022] [Accepted: 05/16/2022] [Indexed: 01/13/2023]
Abstract
Adeno-associated viruses (AAVs) represent some of the most commonly employed vectors for targeted gene delivery and their extensive study has resulted in the approval of multiple gene therapies to treat human diseases. The intranasal route of vector application in gene therapy offers several advantages over traditional ways of administration. In addition to targeting local tissue like the olfactory epithelium, it provides minimally invasive access to various organ systems, including the central nervous system and the respiratory tract. Through a systematic literature review, a total of 53 articles that investigated the intranasal application of AAVs were identified, included, and summarized in this manuscript. Within these studies, AAV-based gene therapy was mainly investigated for its application in various infectious, pulmonary, or neurologic and/or psychiatric diseases. This review gives a comprehensive overview of the current technological state of the art regarding the intranasal application of AAVs for gene transfer and discusses remaining hurdles, which still have to be resolved before this approach can effectively be implemented in the routine clinical setting.
Collapse
|
13
|
Vaccines platforms and COVID-19: what you need to know. Trop Dis Travel Med Vaccines 2022; 8:20. [PMID: 35965345 PMCID: PMC9537331 DOI: 10.1186/s40794-022-00176-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 06/22/2022] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND The novel SARS-CoV-2, responsible for the COVID-19 pandemic, is the third zoonotic coronavirus since the beginning of the 21 first century, and it has taken more than 6 million human lives because of the lack of immunity causing global economic losses. Consequently, developing a vaccine against the virus represents the fastest way to finish the threat and regain some "normality." OBJECTIVE Here, we provide information about the main features of the most important vaccine platforms, some of them already approved, to clear common doubts fostered by widespread misinformation and to reassure the public of the safety of the vaccination process and the different alternatives presented. METHODS Articles published in open access databases until January 2022 were identified using the search terms "SARS-CoV-2," "COVID-19," "Coronavirus," "COVID-19 Vaccines," "Pandemic," COVID-19, and LMICs or their combinations. DISCUSSION Traditional first-generation vaccine platforms, such as whole virus vaccines (live attenuated and inactivated virus vaccines), as well as second-generation vaccines, like protein-based vaccines (subunit and viral vector vaccines), and third-generation vaccines, such as nanoparticle and genetic vaccines (mRNA vaccines), are described. CONCLUSIONS SARS-CoV-2 sequence information obtained in a record time provided the basis for the fast development of a COVID-19 vaccine. The adaptability characteristic of the new generation of vaccines is changing our capability to react to emerging threats to future pandemics. Nevertheless, the slow and unfair distribution of vaccines to low- and middle-income countries and the spread of misinformation are a menace to global health since the unvaccinated will increase the chances for resurgences and the surge of new variants that can escape the current vaccines.
Collapse
|
14
|
Ozger ZB, Cihan P. A novel ensemble fuzzy classification model in SARS-CoV-2 B-cell epitope identification for development of protein-based vaccine. Appl Soft Comput 2022; 116:108280. [PMID: 34931117 PMCID: PMC8673934 DOI: 10.1016/j.asoc.2021.108280] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 11/25/2021] [Accepted: 12/03/2021] [Indexed: 12/23/2022]
Abstract
B-cell epitope prediction research has received growing interest since the development of the first method. B-cell epitope identification with the aid of an accurate prediction method is one of the most important steps in epitope-based vaccine development, immunodiagnostic testing, antibody production, disease diagnosis, and treatment. Nevertheless, using experimental methods in epitope mapping is very time-consuming, costly, and labor-intensive. Therefore, although successful predictions with in silico methods are very important in epitope prediction, there are limited studies in this area. The aim of this study is to propose a new approach for successfully predicting B-cell epitopes for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). In this study, the SARS-CoV B-cell epitope prediction performances of different fuzzy learning classification models genetic cooperative competitive learning (GCCL), fuzzy genetics-based machine learning (GBML), Chi's method (CHI), Ishibuchi's method with weight factor (W), structural learning algorithm on vague environment (SLAVE) and the state-of-the-art ensemble fuzzy classification model were compared. The obtained results showed that the proposed ensemble approach has the lowest error in SARS-CoV B-cell epitope estimation compared to the base fuzzy learners (average error rates; ensemble fuzzy=8.33, GCCL=30.42, GBML=23.82, CHI=29.17, W=46.25, and SLAVE=20.42). SARS-CoV and SARS-CoV-2 have high genome similarities. Therefore, the most successful method determined for SARS-CoV B-cell epitope prediction was used in SARS-CoV-2 cell epitope prediction. Finally, the eventual B-cell epitope prediction results obtained for SARS-CoV-2 with the ensemble fuzzy classification model were compared with the epitope sequences predicted by the BepiPred server and immunoinformatics studies in the literature for the same protein sequences according to VaxiJen 2.0 scores. We hope that the developed epitope prediction method will help design effective vaccines and drugs against future outbreaks of the coronavirus family, especially SARS-CoV-2 and its possible mutations.
Collapse
Affiliation(s)
- Zeynep Banu Ozger
- Department of Computer Engineering, Sutcu Imam University, 46040, Kahramanmaras, Turkey
| | - Pınar Cihan
- Department of Computer Engineering, Tekirdag Namik Kemal University, 59860, Corlu, Tekirdag, Turkey
| |
Collapse
|
15
|
Focosi D, Maggi F, Casadevall A. Mucosal Vaccines, Sterilizing Immunity, and the Future of SARS-CoV-2 Virulence. Viruses 2022; 14:187. [PMID: 35215783 PMCID: PMC8878800 DOI: 10.3390/v14020187] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 01/15/2022] [Accepted: 01/17/2022] [Indexed: 02/01/2023] Open
Abstract
Sterilizing immunity after vaccination is desirable to prevent the spread of infection from vaccinees, which can be especially dangerous in hospital settings while managing frail patients. Sterilizing immunity requires neutralizing antibodies at the site of infection, which for respiratory viruses such as SARS-CoV-2 implies the occurrence of neutralizing IgA in mucosal secretions. Systemic vaccination by intramuscular delivery induces no or low-titer neutralizing IgA against vaccine antigens. Mucosal priming or boosting, is needed to provide sterilizing immunity. On the other side of the coin, sterilizing immunity, by zeroing interhuman transmission, could confine SARS-CoV-2 in animal reservoirs, preventing spontaneous attenuation of virulence in humans as presumably happened with the endemic coronaviruses. We review here the pros and cons of each vaccination strategy, the current mucosal SARS-CoV-2 vaccines under development, and their implications for public health.
Collapse
Affiliation(s)
- Daniele Focosi
- North-Western Tuscany Blood Bank, Pisa University Hospital, 56124 Pisa, Italy
| | - Fabrizio Maggi
- Department of Medicine and Surgery, University of Insubria, 21100 Varese, Italy;
| | - Arturo Casadevall
- Department of Medicine, Johns Hopkins School of Public Health and School of Medicine, Baltimore, MD 21218, USA;
| |
Collapse
|
16
|
Zorgi NE, Meireles LR, Oliveira DBL, Araujo DB, Durigon EL, Andrade Junior HFD. Isolated specific IgA against respiratory viruses, Influenza or SARS-CoV-2, present in the saliva of a fraction of healthy and asymptomatic volunteers. Clinics (Sao Paulo) 2022; 77:100105. [PMID: 36116267 PMCID: PMC9444893 DOI: 10.1016/j.clinsp.2022.100105] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 06/07/2022] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVES Defense against respiratory viruses depends on an immune response present in the mucosa, as saliva IgA secretes antibodies. During the pandemic, such as influenza or SARS-CoV-2, most infected patients are asymptomatic but retain specific antibodies post-infection. The authors evaluated IgG and IgA antibodies against SARS-CoV-2 and influenza in the saliva of asymptomatic volunteers, validated with controls or vaccinated individuals. METHODS The authors detected specific antibodies by validated conventional ELISA using natural SARS-CoV-2 antigens from infected Vero cells or capture-ELISA for influenza using natural antigens of the influenza vaccine. RESULTS Saliva from influenza-vaccinated individuals had more IgA than paired serum, contrary to the findings for specific IgG. In COVID-19-vaccinated samples, specific IgA in saliva increased after vaccination, but IgG levels were high after the first dose. In saliva from the asymptomatic population (226), anti-Influenza IgG was found in 57.5% (130) of samples, higher than IgA, found in 35% (79) of samples. IgA results were similar for SARS-CoV-2, with IgA present in 30% (68) of samples, while IgG was less present, in 44.2% (100) of samples. The proportion of influenza IgG responders was higher than that for SARS-CoV-2 IgG, but both populations presented similar proportions of IgA responders, possibly due to variable memory B cell survival. For both viruses, the authors found an important proportion (> 10%) of IgA+IgG- samples, suggesting the occurrence of humoral immunity directed to the mucosa. CONCLUSION Specific antibodies for respiratory viruses in saliva are found in either infection or vaccination and are a convenient and sensitive diagnostic tool for host immune response.
Collapse
Affiliation(s)
- Nahiara Esteves Zorgi
- Faculdade de Medicina da Universidade São Paulo (FMUSP), São Paulo, SP, Brazil; Laboratório de Protozoologia, Instituto de Medicina Tropical de São Paulo (IMTSP), São Paulo, SP, Brazil
| | - Luciana R Meireles
- Faculdade de Medicina da Universidade São Paulo (FMUSP), São Paulo, SP, Brazil; Laboratório de Protozoologia, Instituto de Medicina Tropical de São Paulo (IMTSP), São Paulo, SP, Brazil
| | | | - Danielle Bastos Araujo
- Instituto de Ciências Biomédicas, Universidade de São Paulo (USP), São Paulo, SP, Brazil
| | - Edson L Durigon
- Instituto de Ciências Biomédicas, Universidade de São Paulo (USP), São Paulo, SP, Brazil
| | - Heitor Franco de Andrade Junior
- Faculdade de Medicina da Universidade São Paulo (FMUSP), São Paulo, SP, Brazil; Laboratório de Protozoologia, Instituto de Medicina Tropical de São Paulo (IMTSP), São Paulo, SP, Brazil.
| |
Collapse
|
17
|
Aravind S, Mathew KA, Madathil BK, Mini S, John A. Current strategies and future perspectives in COVID-19 therapy. STEM CELLS AND COVID-19 2022:169-227. [DOI: 10.1016/b978-0-323-89972-7.00011-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
18
|
Gartlan C, Tipton T, Salguero FJ, Sattentau Q, Gorringe A, Carroll MW. Vaccine-Associated Enhanced Disease and Pathogenic Human Coronaviruses. Front Immunol 2022; 13:882972. [PMID: 35444667 PMCID: PMC9014240 DOI: 10.3389/fimmu.2022.882972] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 03/14/2022] [Indexed: 01/14/2023] Open
Abstract
Vaccine-associated enhanced disease (VAED) is a difficult phenomenon to define and can be confused with vaccine failure. Using studies on respiratory syncytial virus (RSV) vaccination and dengue virus infection, we highlight known and theoretical mechanisms of VAED, including antibody-dependent enhancement (ADE), antibody-enhanced disease (AED) and Th2-mediated pathology. We also critically review the literature surrounding this phenomenon in pathogenic human coronaviruses, including MERS-CoV, SARS-CoV-1 and SARS-CoV-2. Poor quality histopathological data and a lack of consistency in defining severe pathology and VAED in preclinical studies of MERS-CoV and SARS-CoV-1 vaccines in particular make it difficult to interrogate potential cases of VAED. Fortuitously, there have been only few reports of mild VAED in SARS-CoV-2 vaccination in preclinical models and no observations in their clinical use. We describe the problem areas and discuss methods to improve the characterisation of VAED in the future.
Collapse
Affiliation(s)
- Cillian Gartlan
- Wellcome Centre for Human Genetics and Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Tom Tipton
- Wellcome Centre for Human Genetics and Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Francisco J Salguero
- Research and Evaluation, UK Health Security Agency, Porton Down, Salisbury, United Kingdom
| | - Quentin Sattentau
- The Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Andrew Gorringe
- Research and Evaluation, UK Health Security Agency, Porton Down, Salisbury, United Kingdom
| | - Miles W Carroll
- Wellcome Centre for Human Genetics and Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
19
|
Nistri R, Barbuti E, Rinaldi V, Tufano L, Pozzilli V, Ianniello A, Marinelli F, De Luca G, Prosperini L, Tomassini V, Pozzilli C. Case Report: Multiple Sclerosis Relapses After Vaccination Against SARS-CoV2: A Series of Clinical Cases. Front Neurol 2021; 12:765954. [PMID: 34744992 PMCID: PMC8569136 DOI: 10.3389/fneur.2021.765954] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 09/21/2021] [Indexed: 11/26/2022] Open
Abstract
Objective: To describe a temporal association between COVID-19 vaccine administration and multiple sclerosis (MS) relapses. Methods: This case series study was collected in four MS Centres in Central Italy, using data from 16 MS patients who received COVID-19 vaccination and presented both clinically and radiologically confirmed relapses between March and June 2021. We collected patients' relevant medical history, including demographics, MS clinical course, disease-modifying treatment (DMT) received (if applicable), and data from MRI scans obtained after the COVID-19 vaccination. Results: Three out of 16 patients received a diagnosis of MS with a first episode occurring after COVID-19 vaccination; 13 had already a diagnosis of MS and, among them, 9 were on treatment with DMTs. Ten patients received BNT162b2/Pfizer-BioNTech, 2 patients mRNA-1273/Moderna, and 4 patients ChAdOx1 nCoV-19/AstraZeneca. All MS relapses occurred from 3 days to 3 weeks after receiving the first dose of the COVID-19 vaccination or the booster. All patients had evidence of radiological activity on MRI. Discussion: Clinical and radiological findings in these cohort of MS patients confirmed disease re/activation and suggested a temporal association between disease activity and COVID-19 vaccination. The nature of this temporal association, whether causative or incidental, remains to be established.
Collapse
Affiliation(s)
- Riccardo Nistri
- Neurology Unit, Sant'Andrea Hospital, Sapienza University, Rome, Italy
| | - Elena Barbuti
- Neurology Unit, Sant'Andrea Hospital, Sapienza University, Rome, Italy
| | - Virginia Rinaldi
- Neurology Unit, Sant'Andrea Hospital, Sapienza University, Rome, Italy
| | - Laura Tufano
- Neurology Unit, Sant'Andrea Hospital, Sapienza University, Rome, Italy
| | - Valeria Pozzilli
- Institute of Advanced Biomedical Technologies (ITAB), Department of Neurosciences, Imaging and Clinical Sciences, University G. d'Annunzio of Chieti-Pescara, Chieti, Italy.,MS Centre, Department of Clinical Neurology, SS. Annunziata University Hospital, Chieti, Italy
| | | | - Fabiana Marinelli
- MS Centre, Department of Neurology, Fabrizio Spaziani Hospital, Frosinone, Italy
| | - Giovanna De Luca
- MS Centre, Department of Clinical Neurology, SS. Annunziata University Hospital, Chieti, Italy
| | - Luca Prosperini
- MS Centre, Department of Neurosciences, S. Camillo-Forlanini Hospital, Rome, Italy
| | - Valentina Tomassini
- Institute of Advanced Biomedical Technologies (ITAB), Department of Neurosciences, Imaging and Clinical Sciences, University G. d'Annunzio of Chieti-Pescara, Chieti, Italy.,MS Centre, Department of Clinical Neurology, SS. Annunziata University Hospital, Chieti, Italy
| | - Carlo Pozzilli
- Neurology Unit, Sant'Andrea Hospital, Sapienza University, Rome, Italy.,MS Centre, Sant'Andrea Hospital, Sapienza University, Rome, Italy
| |
Collapse
|
20
|
Centonze D, Rocca MA, Gasperini C, Kappos L, Hartung HP, Magyari M, Oreja-Guevara C, Trojano M, Wiendl H, Filippi M. Disease-modifying therapies and SARS-CoV-2 vaccination in multiple sclerosis: an expert consensus. J Neurol 2021; 268:3961-3968. [PMID: 33844056 PMCID: PMC8038920 DOI: 10.1007/s00415-021-10545-2] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 04/01/2021] [Accepted: 04/03/2021] [Indexed: 12/12/2022]
Abstract
Coronavirus disease (COVID-19) appeared in December 2019 in the Chinese city of Wuhan and has quickly become a global pandemic. The disease is caused by the severe acute respiratory syndrome coronavirus type-2 (SARS-CoV-2), an RNA beta coronavirus phylogenetically similar to SARS coronavirus. To date, more than 132 million cases of COVID19 have been recorded in the world, of which over 2.8 million were fatal ( https://coronavirus.jhu.edu/map.html ). A huge vaccination campaign has started around the world since the end of 2020. The availability of vaccines has raised some concerns among neurologists regarding the safety and efficacy of vaccination in patients with multiple sclerosis (MS) taking immunomodulatory or immunosuppressive therapies.
Collapse
Affiliation(s)
- Diego Centonze
- Department of Systems Medicine, Tor Vergata University, Rome, Italy
- Unit of Neurology, IRCCS Neuromed, Pozzilli (IS), Italy
| | - Maria A Rocca
- MS Center and Neurology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina, 60, 20132, Milan, Italy
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Claudio Gasperini
- Department of Neurosciences, San Camillo Forlanini Hospital, Rome, Italy
| | - Ludwig Kappos
- MS Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), Departments of Medicine, Clinical Research and Biomedicine and Biomedical Engineering, University Hospital and University of Basel, Basel, Switzerland
| | - Hans-Peter Hartung
- Department of Neurology, Medical Faculty, Heinrich-Heine University, University Hospital Duesseldorf, Düsseldorf, Germany
- Brain and Mind Centre, University of Sydney, Sydney, Australia
- Department of Neurology, Medical University of Vienna, Wien, Austria
| | - Melinda Magyari
- Danish Multiple Sclerosis Center, Department of Neurology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Celia Oreja-Guevara
- Department of Neurology, Hospital Clínico San Carlos, IdISSC, Madrid, Spain
- Departamento de Medicina, Facultad de Medicina, Universidad Complutense de Madrid (UCM), Madrid, Spain
| | - Maria Trojano
- Neurology and Neurophysiopathology Unit, Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari "Aldo Moro", Bari, Italy
| | - Heinz Wiendl
- Department of Neurology, University Hospital Münster, Münster, Germany
| | - Massimo Filippi
- MS Center and Neurology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina, 60, 20132, Milan, Italy.
- Neurorehabilitation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy.
- Neurophysiology Service, IRCCS San Raffaele Scientific Institute, Milan, Italy.
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy.
- Vita-Salute San Raffaele University, Milan, Italy.
| |
Collapse
|
21
|
Shah SM, Alsaab HO, Rawas-Qalaji MM, Uddin MN. A Review on Current COVID-19 Vaccines and Evaluation of Particulate Vaccine Delivery Systems. Vaccines (Basel) 2021; 9:vaccines9101086. [PMID: 34696194 PMCID: PMC8540464 DOI: 10.3390/vaccines9101086] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 09/20/2021] [Accepted: 09/24/2021] [Indexed: 12/21/2022] Open
Abstract
First detected in Wuhan, China, a highly contagious coronavirus, severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), also known as COVID-19, spread globally in December of 2019. As of 19 September 2021, approximately 4.5 million people have died globally, and 215 million active cases have been reported. To date, six vaccines have been developed and approved for human use. However, current production and supply capabilities are unable to meet global demands to immunize the entire world population. Only a few countries have been able to successfully vaccinate many of their residents. Therefore, an alternative vaccine that can be prepared in an easy and cost-effective manner is urgently needed. A vaccine that could be prepared in this manner, as well as can be preserved and transported at room temperature, would be of great benefit to public health. It is possible to develop such an alternative vaccine by using nano- or microparticle platforms. These platforms address most of the existing vaccine limitations as they are stable at room temperature, are inexpensive to produce and distribute, can be administered orally, and do not require cold chain storage for transportation or preservation. Particulate vaccines can be administered as either oral solutions or in sublingual or buccal film dosage forms. Besides improved patient compliance, the major advantage of oral, sublingual, and buccal routes of administration is that they can elicit mucosal immunity. Mucosal immunity, along with systemic immunity, can be a strong defense against SARS-CoV-2 as the virus enters the system through inhalation or saliva. This review discusses the possibility to produce a particulate COVID vaccine by using nano- or microparticles as platforms for oral administration or in sublingual or buccal film dosage forms in order to accelerate global vaccination.
Collapse
Affiliation(s)
- Sarthak M. Shah
- College of Pharmacy, Mercer University, Atlanta, GA 31207, USA;
| | - Hashem O. Alsaab
- Department of Pharmaceutics and Pharmaceutical Technology, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia;
| | - Mutasem M. Rawas-Qalaji
- College of Pharmacy, University of Sharjah, Sharjah 26666, United Arab Emirates;
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah 26666, United Arab Emirates
- Dr. Kiran C. Patel College of Allopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL 27272, USA
| | - Mohammad N. Uddin
- College of Pharmacy, Mercer University, Atlanta, GA 31207, USA;
- Correspondence: ; Tel.: +1-678-547-6224
| |
Collapse
|
22
|
Fröberg J, Gillard J, Philipsen R, Lanke K, Rust J, van Tuijl D, Teelen K, Bousema T, Simonetti E, van der Gaast-de Jongh CE, Bos M, van Kuppeveld FJ, Bosch BJ, Nabuurs-Franssen M, van der Geest-Blankert N, van Daal C, Huynen MA, de Jonge MI, Diavatopoulos DA. SARS-CoV-2 mucosal antibody development and persistence and their relation to viral load and COVID-19 symptoms. Nat Commun 2021; 12:5621. [PMID: 34556667 PMCID: PMC8460778 DOI: 10.1038/s41467-021-25949-x] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 09/08/2021] [Indexed: 12/27/2022] Open
Abstract
Although serological studies have shown that antibodies against SARS-CoV-2 play an important role in protection against (re)infection, the dynamics of mucosal antibodies during primary infection and their potential impact on viral load and the resolution of disease symptoms remain unclear. During the first pandemic wave, we assessed the longitudinal nasal antibody response in index cases with mild COVID-19 and their household contacts. Nasal and serum antibody responses were analysed for up to nine months. Higher nasal receptor binding domain and spike protein-specific antibody levels at study inclusion were associated with lower viral load. Older age was correlated with more frequent COVID-19 related symptoms. Receptor binding domain and spike protein-specific mucosal antibodies were associated with the resolution of systemic, but not respiratory symptoms. Finally, receptor binding domain and spike protein-specific mucosal antibodies remained elevated up to nine months after symptom onset.
Collapse
Affiliation(s)
- Janeri Fröberg
- Radboud University Medical Centre, Radboud Institute for Molecular Life Sciences, Laboratory of Medical Immunology, Section Paediatric Infectious Diseases, 6525 GA, Nijmegen, The Netherlands
- Radboud Center for Infectious Diseases, Radboudumc, Nijmegen, The Netherlands
| | - Joshua Gillard
- Radboud University Medical Centre, Radboud Institute for Molecular Life Sciences, Laboratory of Medical Immunology, Section Paediatric Infectious Diseases, 6525 GA, Nijmegen, The Netherlands
- Radboud Center for Infectious Diseases, Radboudumc, Nijmegen, The Netherlands
- Centre for molecular and Biomolecular Informatics, Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre Nijmegen, 6525 GA, Nijmegen, The Netherlands
| | - Ria Philipsen
- Radboud University Medical Centre, Radboud Institute for Molecular Life Sciences, Laboratory of Medical Immunology, Section Paediatric Infectious Diseases, 6525 GA, Nijmegen, The Netherlands
- Radboud Center for Infectious Diseases, Radboudumc, Nijmegen, The Netherlands
- RTC CS Radboud Technology Center Clinical Studies, Radboudumc, Nijmegen, The Netherlands
| | - Kjerstin Lanke
- Radboud Center for Infectious Diseases, Radboudumc, Nijmegen, The Netherlands
| | - Joyce Rust
- Radboud University Medical Centre, Radboud Institute for Molecular Life Sciences, Laboratory of Medical Immunology, Section Paediatric Infectious Diseases, 6525 GA, Nijmegen, The Netherlands
- Radboud Center for Infectious Diseases, Radboudumc, Nijmegen, The Netherlands
- RTC CS Radboud Technology Center Clinical Studies, Radboudumc, Nijmegen, The Netherlands
| | - Diana van Tuijl
- Radboud University Medical Centre, Radboud Institute for Molecular Life Sciences, Laboratory of Medical Immunology, Section Paediatric Infectious Diseases, 6525 GA, Nijmegen, The Netherlands
- Radboud Center for Infectious Diseases, Radboudumc, Nijmegen, The Netherlands
- RTC CS Radboud Technology Center Clinical Studies, Radboudumc, Nijmegen, The Netherlands
| | - Karina Teelen
- Radboud Center for Infectious Diseases, Radboudumc, Nijmegen, The Netherlands
| | - Teun Bousema
- Radboud Center for Infectious Diseases, Radboudumc, Nijmegen, The Netherlands
| | - Elles Simonetti
- Radboud University Medical Centre, Radboud Institute for Molecular Life Sciences, Laboratory of Medical Immunology, Section Paediatric Infectious Diseases, 6525 GA, Nijmegen, The Netherlands
- Radboud Center for Infectious Diseases, Radboudumc, Nijmegen, The Netherlands
| | - Christa E van der Gaast-de Jongh
- Radboud University Medical Centre, Radboud Institute for Molecular Life Sciences, Laboratory of Medical Immunology, Section Paediatric Infectious Diseases, 6525 GA, Nijmegen, The Netherlands
- Radboud Center for Infectious Diseases, Radboudumc, Nijmegen, The Netherlands
| | - Mariska Bos
- Radboud University Medical Centre, Radboud Institute for Molecular Life Sciences, Laboratory of Medical Immunology, Section Paediatric Infectious Diseases, 6525 GA, Nijmegen, The Netherlands
- Radboud Center for Infectious Diseases, Radboudumc, Nijmegen, The Netherlands
| | - Frank J van Kuppeveld
- Utrecht University, Faculty of Veterinary Medicine, Department of Biomolecular Health Sciences, Division Infectious Diseases and Immunology, Utrecht, The Netherlands
| | - Berend-Jan Bosch
- Utrecht University, Faculty of Veterinary Medicine, Department of Biomolecular Health Sciences, Division Infectious Diseases and Immunology, Utrecht, The Netherlands
| | - Marrigje Nabuurs-Franssen
- Department of Medical Microbiology and Infectious Diseases, Canisius Wilhelmina Hospital, Nijmegen, The Netherlands
| | | | - Charlotte van Daal
- Department of Occupational Health, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Martijn A Huynen
- Centre for molecular and Biomolecular Informatics, Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre Nijmegen, 6525 GA, Nijmegen, The Netherlands
| | - Marien I de Jonge
- Radboud University Medical Centre, Radboud Institute for Molecular Life Sciences, Laboratory of Medical Immunology, Section Paediatric Infectious Diseases, 6525 GA, Nijmegen, The Netherlands
- Radboud Center for Infectious Diseases, Radboudumc, Nijmegen, The Netherlands
| | - Dimitri A Diavatopoulos
- Radboud University Medical Centre, Radboud Institute for Molecular Life Sciences, Laboratory of Medical Immunology, Section Paediatric Infectious Diseases, 6525 GA, Nijmegen, The Netherlands.
- Radboud Center for Infectious Diseases, Radboudumc, Nijmegen, The Netherlands.
| |
Collapse
|
23
|
Shahnaij M, Iyori M, Mizukami H, Kajino M, Yamagoshi I, Syafira I, Yusuf Y, Fujiwara K, Yamamoto DS, Kato H, Ohno N, Yoshida S. Liver-Directed AAV8 Booster Vaccine Expressing Plasmodium falciparum Antigen Following Adenovirus Vaccine Priming Elicits Sterile Protection in a Murine Model. Front Immunol 2021; 12:612910. [PMID: 34248928 PMCID: PMC8261234 DOI: 10.3389/fimmu.2021.612910] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 06/04/2021] [Indexed: 12/12/2022] Open
Abstract
Hepatocyte infection by malaria sporozoites is a bottleneck in the life-cycle of Plasmodium spp. including P. falciparum, which causes the most lethal form of malaria. Therefore, developing an effective vaccine capable of inducing the strong humoral and cellular immune responses necessary to block the pre-erythrocytic stage has potential to overcome the spatiotemporal hindrances pertaining to parasite biology and hepatic microanatomy. We recently showed that when combined with a human adenovirus type 5 (AdHu5)-priming vaccine, adeno-associated virus serotype 1 (AAV1) is a potent booster malaria vaccine vector capable of inducing strong and long-lasting protective immune responses in a rodent malaria model. Here, we evaluated the protective efficacy of a hepatotropic virus, adeno-associated virus serotype 8 (AAV8), as a booster vector because it can deliver a transgene potently and rapidly to the liver, the organ malaria sporozoites initially infect and multiply in following sporozoite injection by the bite of an infected mosquito. We first generated an AAV8-vectored vaccine expressing P. falciparum circumsporozoite protein (PfCSP). Intravenous (i.v.) administration of AAV8-PfCSP to mice initially primed with AdHu5-PfCSP resulted in a hepatocyte transduction rate ~2.5 times above that seen with intramuscular (i.m.) administration. This immunization regimen provided a better protection rate (100% sterile protection) than that of the i.m. AdHu5-prime/i.m. AAV8-boost regimen (60%, p < 0.05), i.m. AdHu5-prime/i.v. AAV1-boost (78%), or i.m. AdHu5-prime/i.m. AAV1-boost (80%) against challenge with transgenic PfCSP-expressing P. berghei sporozoites. Compared with the i.m. AdHu5-prime/i.v. AAV1-boost regimen, three other regimens induced higher levels of PfCSP-specific humoral immune responses. Importantly, a single i.v. dose of AAV8-PfCSP recruited CD8+ T cells, especially resident memory CD8+ T cells, in the liver. These data suggest that boost with i.v. AAV8-PfCSP can improve humoral and cellular immune responses in BALB/c mice. Therefore, this regimen holds great promise as a next-generation platform for the development of an effective malaria vaccine.
Collapse
Affiliation(s)
- Mohammad Shahnaij
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kanazawa University, Kanazawa, Japan
| | - Mitsuhiro Iyori
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kanazawa University, Kanazawa, Japan
| | - Hiroaki Mizukami
- Division of Gene Therapy, Jichi Medical University, Shimotsuke, Japan
| | - Mayu Kajino
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kanazawa University, Kanazawa, Japan
| | - Iroha Yamagoshi
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kanazawa University, Kanazawa, Japan
| | - Intan Syafira
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kanazawa University, Kanazawa, Japan
| | - Yenni Yusuf
- Department of Parasitology, Faculty of Medicine, University of Hasanuddin, Makassar, Indonesia
| | - Ken Fujiwara
- Division of Histology and Cell Biology, School of Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Daisuke S Yamamoto
- Division of Medical Zoology, Department of Infection and Immunity, Jichi Medical University, Shimotsuke, Japan
| | - Hirotomo Kato
- Division of Medical Zoology, Department of Infection and Immunity, Jichi Medical University, Shimotsuke, Japan
| | - Nobuhiko Ohno
- Division of Histology and Cell Biology, School of Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Shigeto Yoshida
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kanazawa University, Kanazawa, Japan
| |
Collapse
|
24
|
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: 1.5] [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
|
25
|
Leventhal SS, Clancy C, Erasmus J, Feldmann H, Hawman DW. An Intramuscular DNA Vaccine for SARS-CoV-2 Decreases Viral Lung Load but Not Lung Pathology in Syrian Hamsters. Microorganisms 2021; 9:1040. [PMID: 34065996 PMCID: PMC8151856 DOI: 10.3390/microorganisms9051040] [Citation(s) in RCA: 4] [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: 04/13/2021] [Revised: 05/03/2021] [Accepted: 05/07/2021] [Indexed: 12/23/2022] Open
Abstract
The 2019 novel coronavirus, SARS-CoV-2, first reported in December 2019, has infected over 102 million people around the world as of February 2021 and thus calls for rapid development of safe and effective interventions, namely vaccines. In our study, we evaluated a DNA vaccine against SARS-CoV-2 in the Syrian hamster model. Hamsters were vaccinated with a DNA-plasmid encoding the SARS-CoV-2 full length spike open reading frame (ORF) to induce host cells to produce spike protein and protective immune responses before exposure to infectious virus. We tested this vaccine candidate by both intranasal (IN) and intramuscular (IM) routes of administration and complexing with and without an in vivo delivery reagent. Hamsters receiving prime-boost-boost IM-only vaccinations recovered body weight quicker, had decreased lung viral loads, and increased SARS-CoV-2-specific antibody titers compared to control vaccinated animals but, surprisingly, lung pathology was as severe as sham vaccinated controls. The IM/IN combination group showed no efficacy in reducing lung virus titers or pathology. With increasing public health need for rapid and effective interventions, our data demonstrate that in some vaccine contexts, significant antibody responses and decreased viral loads may not be sufficient to prevent lung pathology.
Collapse
Affiliation(s)
- Shanna S. Leventhal
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT 59840, USA; (S.S.L.); (C.C.); (H.F.)
| | - Chad Clancy
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT 59840, USA; (S.S.L.); (C.C.); (H.F.)
- Rocky Mountain Veterinary Branch, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT 59840, USA
| | - Jesse Erasmus
- Department of Microbiology, University of Washington School of Medicine, Seattle, WA 98195, USA;
- HDT Bio, Seattle, WA 98102, USA
| | - Heinz Feldmann
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT 59840, USA; (S.S.L.); (C.C.); (H.F.)
| | - David W. Hawman
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT 59840, USA; (S.S.L.); (C.C.); (H.F.)
| |
Collapse
|
26
|
Zhan W, Muhuri M, Tai PWL, Gao G. Vectored Immunotherapeutics for Infectious Diseases: Can rAAVs Be The Game Changers for Fighting Transmissible Pathogens? Front Immunol 2021; 12:673699. [PMID: 34046041 PMCID: PMC8144494 DOI: 10.3389/fimmu.2021.673699] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 04/23/2021] [Indexed: 01/08/2023] Open
Abstract
Conventional vaccinations and immunotherapies have encountered major roadblocks in preventing infectious diseases like HIV, influenza, and malaria. These challenges are due to the high genomic variation and immunomodulatory mechanisms inherent to these diseases. Passive transfer of broadly neutralizing antibodies may offer partial protection, but these treatments require repeated dosing. Some recombinant viral vectors, such as those based on lentiviruses and adeno-associated viruses (AAVs), can confer long-term transgene expression in the host after a single dose. Particularly, recombinant (r)AAVs have emerged as favorable vectors, given their high in vivo transduction efficiency, proven clinical efficacy, and low immunogenicity profiles. Hence, rAAVs are being explored to deliver recombinant antibodies to confer immunity against infections or to diminish the severity of disease. When used as a vaccination vector for the delivery of antigens, rAAVs enable de novo synthesis of foreign proteins with the conformation and topology that resemble those of natural pathogens. However, technical hurdles like pre-existing immunity to the rAAV capsid and production of anti-drug antibodies can reduce the efficacy of rAAV-vectored immunotherapies. This review summarizes rAAV-based prophylactic and therapeutic strategies developed against infectious diseases that are currently being tested in pre-clinical and clinical studies. Technical challenges and potential solutions will also be discussed.
Collapse
Affiliation(s)
- Wei Zhan
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, United States
- VIDE Program, University of Massachusetts Medical School, Worcester, MA, United States
| | - Manish Muhuri
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, United States
- VIDE Program, University of Massachusetts Medical School, Worcester, MA, United States
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, United States
| | - Phillip W. L. Tai
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, United States
- VIDE Program, University of Massachusetts Medical School, Worcester, MA, United States
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, United States
| | - Guangping Gao
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, United States
- VIDE Program, University of Massachusetts Medical School, Worcester, MA, United States
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, United States
- Li Weibo Institute for Rare Diseases Research, University of Massachusetts Medical School, Worcester, MA, United States
| |
Collapse
|
27
|
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: 7.8] [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
|
28
|
Kovács OT, Soltész-Katona E, Marton N, Baricza E, Hunyady L, Turu G, Nagy G. Impact of Medium-Sized Extracellular Vesicles on the Transduction Efficiency of Adeno-Associated Viruses in Neuronal and Primary Astrocyte Cell Cultures. Int J Mol Sci 2021; 22:ijms22084221. [PMID: 33921740 PMCID: PMC8073863 DOI: 10.3390/ijms22084221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 04/13/2021] [Accepted: 04/15/2021] [Indexed: 11/16/2022] Open
Abstract
(1) Adeno-associated viruses (AAV) are safe and efficient gene therapy vectors with promising results in the treatment of several diseases. Extracellular vesicles (EV) are phospholipid bilayer-surrounded structures carrying several types of lipids, proteins, and nucleic acids with the ability to cross biological barriers. EV-associated AAVs might serve as new and efficient gene therapy vectors considering that they carry the benefits of both AAVs and EVs. (2) We tested vesicle-associated AAVs and vesicles mixed with AAVs on two major cell types of the central nervous system: a neural cell line (N2A) and primary astrocyte cells. (3) In contrast to previously published in vivo observations, the extracellular vesicle packaging did not improve but, in the case of primary astrocyte cells, even inhibited the infection capacity of the AAV particles. The observed effect was not due to the inhibitory effects of the vesicles themselves, since mixing the AAVs with extracellular vesicles did not change the effectiveness. (4) Our results suggest that improvement of the in vivo efficacy of the EV-associated AAV particles is not due to the enhanced interaction between the AAV and the target cells, but most likely to the improved delivery of the AAVs through tissue barriers and to the shielding of AAVs from neutralizing antibodies.
Collapse
Affiliation(s)
- Orsolya Tünde Kovács
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Nagyvárad tér 4, 1089 Budapest, Hungary; (O.T.K.); (E.B.)
- Department of Physiology, Semmelweis University, Tűzoltó street 37-47, 1094 Budapest, Hungary; (E.S.-K.); (L.H.)
| | - Eszter Soltész-Katona
- Department of Physiology, Semmelweis University, Tűzoltó street 37-47, 1094 Budapest, Hungary; (E.S.-K.); (L.H.)
| | - Nikolett Marton
- Jahn Ferenc Dél-pesti Hospital, Department of Radiology, Köves street 1, 1204 Budapest, Hungary;
| | - Eszter Baricza
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Nagyvárad tér 4, 1089 Budapest, Hungary; (O.T.K.); (E.B.)
| | - László Hunyady
- Department of Physiology, Semmelweis University, Tűzoltó street 37-47, 1094 Budapest, Hungary; (E.S.-K.); (L.H.)
- MTA-SE Laboratory of Molecular Physiology, Eötvös Loránd Research Network, 1085 Budapest, Hungary
| | - Gábor Turu
- Department of Physiology, Semmelweis University, Tűzoltó street 37-47, 1094 Budapest, Hungary; (E.S.-K.); (L.H.)
- MTA-SE Laboratory of Molecular Physiology, Eötvös Loránd Research Network, 1085 Budapest, Hungary
- Correspondence: (G.T.); (G.N.)
| | - György Nagy
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Nagyvárad tér 4, 1089 Budapest, Hungary; (O.T.K.); (E.B.)
- Department of Rheumatology & Clinical Immunology, Semmelweis University, Árpád fejedelem street 7, 1023 Budapest, Hungary
- Correspondence: (G.T.); (G.N.)
| |
Collapse
|
29
|
Grun MK, Suberi A, Shin K, Lee T, Gomerdinger V, Moscato ZM, Piotrowski-Daspit AS, Saltzman WM. PEGylation of poly(amine-co-ester) polyplexes for tunable gene delivery. Biomaterials 2021; 272:120780. [PMID: 33813260 DOI: 10.1016/j.biomaterials.2021.120780] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 03/19/2021] [Accepted: 03/20/2021] [Indexed: 01/17/2023]
Abstract
There is growing interest in PEGylation of cationic polymeric vehicles for gene delivery in order to improve vehicle stability and reduce toxicity, but little is known about the effects of PEG coatings on transfection. We used a polymer from the poly(amine-co-ester) (PACE) family blended with PEG-conjugated PACE at different ratios in order to explore the effects of polyplex PEGylation on the transfection efficiency of plasmid DNA, mRNA, and siRNA in vitro and mRNA in vivo. We discovered that concentrations of PACE-PEG as low as 0.25% by weight improved polyplex stability but also inhibited transfection in vitro. In vivo, the effect of PACE-PEG incorporation on mRNA transfection varied by delivery route; the addition of PACE-PEG improved local delivery to the lung, but PEGylation had little effect on intravenous systemic delivery. By both delivery routes, transfection was inhibited at concentrations higher than 5 wt% PACE-PEG. These results demonstrate that excess PEGylation can be detrimental to vehicle function, and suggest that PEGylation of cationic vehicles must be optimized by PEG content, cargo type, and delivery route.
Collapse
Affiliation(s)
- Molly K Grun
- Department of Chemical & Environmental Engineering, Yale University, New Haven, CT, 06511, USA
| | - Alexandra Suberi
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06511, USA
| | - Kwangsoo Shin
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06511, USA
| | - Teresa Lee
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06511, USA
| | - Victoria Gomerdinger
- Department of Chemical & Environmental Engineering, Yale University, New Haven, CT, 06511, USA
| | - Zoe M Moscato
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06511, USA
| | | | - W Mark Saltzman
- Department of Chemical & Environmental Engineering, Yale University, New Haven, CT, 06511, USA; Department of Biomedical Engineering, Yale University, New Haven, CT, 06511, USA; Department of Cellular & Molecular Physiology, Yale University, New Haven, CT, 06511, USA; Department of Dermatology, Yale School of Medicine, New Haven, CT, 06510, USA.
| |
Collapse
|
30
|
Du Y, Xu Y, Feng J, Hu L, Zhang Y, Zhang B, Guo W, Mai R, Chen L, Fang J, Zhang H, Peng T. Intranasal administration of a recombinant RBD vaccine induced protective immunity against SARS-CoV-2 in mouse. Vaccine 2021; 39:2280-2287. [PMID: 33731271 PMCID: PMC7934688 DOI: 10.1016/j.vaccine.2021.03.006] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 01/06/2021] [Accepted: 03/02/2021] [Indexed: 12/13/2022]
Abstract
The emergence of the global Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) pandemic underscores the importance of the rapid development of a non-invasive vaccine that can be easily administered. A vaccine administered by nasal delivery is endowed with such characteristics against respiratory viruses. In this study, we generated a recombinant SARS-CoV-2 receptor-binding domain (RBD)-based subunit vaccine. Mice were immunized via intranasal inoculation, microneedle-intradermal injection, or intramuscular injection, after which the RBD-specific immune responses were compared. Results showed that when administrated intranasally, the vaccine elicited a robust systemic humoral immunity with high titers of IgG antibodies and neutralizing antibodies as well as a significant mucosal immunity. Besides, antigen-specific T cell responses were also analyzed. These results indicated that the non-invasive intranasal administration should be explored for the future SARS-CoV-2 vaccine design.
Collapse
Affiliation(s)
- Yingying Du
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Yuhua Xu
- Guangdong South China Vaccine, Guangzhou, China
| | - Jin Feng
- Guangdong South China Vaccine, Guangzhou, China
| | - Longbo Hu
- Sino-French Hoffmann Institute of Immunology, State Key Laboratory of Respiratory Disease, College of Basic Medical Science, Guangzhou Medical University, Guangzhou, China
| | - Yanan Zhang
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Bo Zhang
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Weili Guo
- Guangdong South China Vaccine, Guangzhou, China
| | - Runming Mai
- Guangdong South China Vaccine, Guangzhou, China
| | - Liyun Chen
- Guangdong South China Vaccine, Guangzhou, China
| | - Jianmin Fang
- Sino-French Hoffmann Institute of Immunology, State Key Laboratory of Respiratory Disease, College of Basic Medical Science, Guangzhou Medical University, Guangzhou, China
| | - Hui Zhang
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, China.
| | - Tao Peng
- Guangdong South China Vaccine, Guangzhou, China; Sino-French Hoffmann Institute of Immunology, State Key Laboratory of Respiratory Disease, College of Basic Medical Science, Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
31
|
Abdullahi IN, Emeribe AU, Adekola HA, Abubakar SD, Dangana A, Shuwa HA, Nwoba ST, Mustapha JO, Haruna MT, Olowookere KA, Animasaun OS, Ugwu CE, Onoja SO, Gadama AS, Mohammed M, Daneji IM, Amadu DO, Ghamba PE, Onukegbe NB, Shehu MS, Isomah C, Babayo A, Ahmad AEF. Leveraging on the genomics and immunopathology of SARS-CoV-2 for vaccines development: prospects and challenges. Hum Vaccin Immunother 2021; 17:620-637. [PMID: 32936732 PMCID: PMC7993231 DOI: 10.1080/21645515.2020.1812313] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 08/10/2020] [Accepted: 08/11/2020] [Indexed: 12/13/2022] Open
Abstract
The incidence and case-fatality rates (CFRs) of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) infection, the etiological agent for Coronavirus Disease 2019 (COVID-19), have been rising unabated. Even though the entire world has been implementing infection prevention and control measures, the pandemic continues to spread. It has been widely accepted that preventive vaccination strategies are the public health measures for countering this pandemic. This study critically reviews the latest scientific advancement in genomics, replication pattern, pathogenesis, and immunopathology of SARS-CoV-2 infection and how these concepts could be used in the development of vaccines. We also offer a detailed discussion on the anticipated potency, efficacy, safety, and pharmaco-economic issues that are and will be associated with candidate COVID-19 vaccines.
Collapse
Affiliation(s)
- Idris Nasir Abdullahi
- Department of Medical Laboratory Science, Faculty of Allied Health Sciences, College of Medical Sciences, Ahmadu Bello University, Zaria, Nigeria
| | - Anthony Uchenna Emeribe
- Department of Medical Laboratory Science, Faculty of Allied Medical Sciences, University of Calabar, Calabar, Nigeria
| | | | - Sharafudeen Dahiru Abubakar
- Department of Medical Laboratory Science, Faculty of Allied Health Sciences, College of Medical Sciences, Ahmadu Bello University, Zaria, Nigeria
| | - Amos Dangana
- Department of Medical Laboratory Services, University of Abuja Teaching Hospital, Gwagwalada, Abuja, Nigeria
| | - Halima Ali Shuwa
- Lydia Becker Institute of Immunology, Manchester Collaborative Center for Inflammation Research, Faculty of Biology, Medicine and Health, The University of Manchester, UK
| | | | - Jelili Olaide Mustapha
- Biological Sciences Department, Faculty of Science, University of Alberta, Edmonton, Canada
| | | | - Kafayat Adepeju Olowookere
- Department of Medical Laboratory Services, Ladoke Akintola University of Technology Teaching Hospital, Ogbomoso, Nigeria
| | - Olawale Sunday Animasaun
- Nigeria Field Epidemiology and Laboratory Training Programme, African Field Epidemiology Network, Abuja, Nigeria
| | - Charles Egede Ugwu
- Department of Medical Laboratory Science, Ebonyi State University, Abakaliki, Nigeria
| | | | - Abdullahi Sani Gadama
- Department of Medical Microbiology and Parasitology, Faculty of Clinical Sciences, Bayero University, Kano, Nigeria
| | - Musa Mohammed
- Department of Medicine, Immunology Unit, Ahmadu Bello University, Zaria, Nigeria
| | - Isa Muhammad Daneji
- Department of Medical Microbiology and Parasitology, Faculty of Clinical Sciences, Bayero University, Kano, Nigeria
| | - Dele Ohinoyi Amadu
- Department of Medical Microbiology and Parasitology, University of Ilorin Teaching Hospital, Ilorin, Nigeria
| | - Peter Elisha Ghamba
- WHO National Polio Reference Laboratory, University of Maiduguri Teaching Hospital, Maiduguri, Nigeria
| | | | - Muhammad Sagir Shehu
- Medical Laboratory Department, College of Health Technology, Ningi, Bauchi State, Nigeria
| | - Chiladi Isomah
- Medical Laboratory Science Department, Rivers State University, Port Harcourt, Nigeria
| | - Adamu Babayo
- Department of Medical Microbiology and Parasitology, Faculty of Clinical Sciences, Bayero University, Kano, Nigeria
| | - Abdurrahman El-Fulaty Ahmad
- Department of Medical Laboratory Science, Faculty of Allied Health Sciences, College of Medical Sciences, Ahmadu Bello University, Zaria, Nigeria
| |
Collapse
|
32
|
Tan Y, Tang F. SARS-CoV-2-mediated immune system activation and potential application in immunotherapy. Med Res Rev 2021; 41:1167-1194. [PMID: 33185926 DOI: 10.1002/med.21756] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 10/23/2020] [Accepted: 11/02/2020] [Indexed: 12/13/2022]
Abstract
Although novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-mediated pulmonary inflammation has recently attracted great attention, its pathology and pathogenesis are not clear. Notably, due to both its high infective and pathogenicity, SARS-CoV-2 infection may cause a severe sometimes fatal respiratory disease. A specific vaccine, which relies on the analysis of SARS-CoV-2 structural protein-derived antigenic peptides, is indispensable for restraining the spread and reducing the mortality of SARS-CoV-2. SARS-CoV-2 infections activate cytototxic, myeloid-derived suppressor cells, dendritic cells, macrophages, as well as natural killer, B, helper T, and regulatory T cells, thus further stimulating innate and antigen-specific immune responses. Nevertheless, many immune effector cells cause hyperinflammation and pulmonary immunopathology by releasing proinflammatory cytokines and chemokines, including interferon (IFN)-α, IFN-β, IFN-γ, monocyte chemoattractant protein-1, macrophage inflammatory protein (MIP)-1A, MIP1B, interleukin (IL)-1, IL-2, IL-4, IL-6, IL-7, IL-8, IL-9, IL-12, IL-17, and IL-18, platelet-derived growth factor, fibroblast growth factor, tumor necrosis factor-α, and induced protein 10. Interestingly, related products derived from SARS-CoV-2 are likely to trigger immune evasion. Therefore, investigating SARS-CoV-2-specific vaccines, blocking immunopathology, and prohibiting immune evasion are urgently required for treating SARS-CoV-2 infection. In this review, we emphatically illuminated the development of a SARS-CoV-2-specific vaccine based on the analysis of epitopes, also expounding the molecular mechanisms of SARS-CoV-2-mediated cytokine release syndrome. Furthermore, we comprehensively discussed SARS-CoV-2-associated immune evasion and lung immunopathology. Lastly, potential therapeutic strategies against SARS-CoV-2 were explored.
Collapse
Affiliation(s)
- Yuan Tan
- Department of Clinical Laboratory, Hunan Key Laboratory of Oncotarget Gene, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Faqin Tang
- Department of Clinical Laboratory, Hunan Key Laboratory of Oncotarget Gene, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| |
Collapse
|
33
|
Su S, Du L, Jiang S. Learning from the past: development of safe and effective COVID-19 vaccines. Nat Rev Microbiol 2021; 19:211-219. [PMID: 33067570 PMCID: PMC7566580 DOI: 10.1038/s41579-020-00462-y] [Citation(s) in RCA: 114] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/23/2020] [Indexed: 01/29/2023]
Abstract
The rapid spread of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has elicited an equally rapid response aiming to develop a COVID-19 vaccine. These efforts are encouraging; however, comprehensive efficacy and safety evaluations are essential in the development of a vaccine, and we can learn from previous vaccine development campaigns. In this Perspective, we summarize examples of vaccine-associated disease enhancement in the history of developing vaccines against respiratory syncytial virus, dengue virus, SARS-CoV and Middle East respiratory syndrome coronavirus, which highlight the importance of a robust safety and efficacy profile, and present recommendations for preclinical and clinical evaluation of COVID-19 vaccine candidates as well as for vaccine design and optimization.
Collapse
Affiliation(s)
- Shan Su
- Key Laboratory of Medical Molecular Virology (MOE/MOH/CAM), School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Lanying Du
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY, USA
| | - Shibo Jiang
- Key Laboratory of Medical Molecular Virology (MOE/MOH/CAM), School of Basic Medical Sciences, Fudan University, Shanghai, China.
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY, USA.
| |
Collapse
|
34
|
Lutz H, Popowski KD, Dinh PUC, Cheng K. Advanced Nanobiomedical Approaches to Combat Coronavirus Disease of 2019. ADVANCED NANOBIOMED RESEARCH 2021; 1:2000063. [PMID: 33681865 PMCID: PMC7917381 DOI: 10.1002/anbr.202000063] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 12/17/2020] [Indexed: 12/14/2022] Open
Abstract
New infectious diseases are making themselves known as the human population grows, expands into new regions, and becomes more dense, increasing contact with each other and animal populations. Ease of travel has also increased infectious disease transmission and has now culminated into a global pandemic. The emergence of the novel coronavirus severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in December 2019 has already infected over 83.7 million people and caused over 1.8 million deaths. While there have been vaccine candidates produced and supportive care implemented, the world is impatiently waiting for a commercially approved vaccine and treatment for the coronavirus disease of 2019 (COVID-19). The different vaccine types investigated for the prevention of COVID-19 all have great promise but face safety obstacles that must be first addressed. Some vaccine candidates of key interest are whole inactivated viruses, adeno-associated viruses, virus-like particles, and lipid nanoparticles. This review examines nanobiomedical techniques for combatting COVID-19 in terms of vaccines and therapeutics.
Collapse
Affiliation(s)
- Halle Lutz
- Department of Molecular Biomedical SciencesNorth Carolina State UniversityRaleighNC27607USA
- Comparative Medicine InstituteNorth Carolina State UniversityRaleighNC27607USA
| | - Kristen D. Popowski
- Department of Molecular Biomedical SciencesNorth Carolina State UniversityRaleighNC27607USA
- Comparative Medicine InstituteNorth Carolina State UniversityRaleighNC27607USA
| | - Phuong-Uyen C. Dinh
- Department of Molecular Biomedical SciencesNorth Carolina State UniversityRaleighNC27607USA
- Comparative Medicine InstituteNorth Carolina State UniversityRaleighNC27607USA
| | - Ke Cheng
- Department of Molecular Biomedical SciencesNorth Carolina State UniversityRaleighNC27607USA
- Comparative Medicine InstituteNorth Carolina State UniversityRaleighNC27607USA
- Joint Department of Biomedical EngineeringUniversity of North Carolina at Chapel Hill/North Carolina State UniversityRaleigh/Chapel HillNC27607/27599USA
- Division of Pharmacoengineering and Molecular PharmaceuticsUniversity of North Carolina at Chapel HillChapel HillNC27599USA
| |
Collapse
|
35
|
Singh R, Kang A, Luo X, Jeyanathan M, Gillgrass A, Afkhami S, Xing Z. COVID-19: Current knowledge in clinical features, immunological responses, and vaccine development. FASEB J 2021; 35:e21409. [PMID: 33577115 PMCID: PMC7898934 DOI: 10.1096/fj.202002662r] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 01/12/2021] [Accepted: 01/19/2021] [Indexed: 01/08/2023]
Abstract
The COVID-19 pandemic has unfolded to be the most challenging global health crisis in a century. In 11 months since its first emergence, according to WHO, the causative infectious agent SARS-CoV-2 has infected more than 100 million people and claimed more than 2.15 million lives worldwide. Moreover, the world has raced to understand the virus and natural immunity and to develop vaccines. Thus, within a short 11 months a number of highly promising COVID-19 vaccines were developed at an unprecedented speed and are now being deployed via emergency use authorization for immunization. Although a considerable number of review contributions are being published, all of them attempt to capture only a specific aspect of COVID-19 or its therapeutic approaches based on ever-expanding information. Here, we provide a comprehensive overview to conceptually thread together the latest information on global epidemiology and mitigation strategies, clinical features, viral pathogenesis and immune responses, and the current state of vaccine development.
Collapse
Affiliation(s)
- Ramandeep Singh
- McMaster Immunology Research CentreM. G. DeGroote Institute for Infectious Disease Research & Department of MedicineMcMaster UniversityHamiltonONCanada
| | - Alisha Kang
- McMaster Immunology Research CentreM. G. DeGroote Institute for Infectious Disease Research & Department of MedicineMcMaster UniversityHamiltonONCanada
| | - Xiangqian Luo
- McMaster Immunology Research CentreM. G. DeGroote Institute for Infectious Disease Research & Department of MedicineMcMaster UniversityHamiltonONCanada
- Department of Pediatric OtolaryngologyShenzhen HospitalSouthern Medical UniversityShenzhenChina
| | - Mangalakumari Jeyanathan
- McMaster Immunology Research CentreM. G. DeGroote Institute for Infectious Disease Research & Department of MedicineMcMaster UniversityHamiltonONCanada
| | - Amy Gillgrass
- McMaster Immunology Research CentreM. G. DeGroote Institute for Infectious Disease Research & Department of MedicineMcMaster UniversityHamiltonONCanada
| | - Sam Afkhami
- McMaster Immunology Research CentreM. G. DeGroote Institute for Infectious Disease Research & Department of MedicineMcMaster UniversityHamiltonONCanada
| | - Zhou Xing
- McMaster Immunology Research CentreM. G. DeGroote Institute for Infectious Disease Research & Department of MedicineMcMaster UniversityHamiltonONCanada
| |
Collapse
|
36
|
Bailey KL, Samuelson DR, Wyatt TA. Alcohol use disorder: A pre-existing condition for COVID-19? Alcohol 2021; 90:11-17. [PMID: 33080339 PMCID: PMC7568767 DOI: 10.1016/j.alcohol.2020.10.003] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 10/02/2020] [Accepted: 10/05/2020] [Indexed: 02/07/2023]
Abstract
Alcohol misuse is long established as a contributor to the pathophysiology of the lung. The intersection of multi-organ responses to alcohol-mediated tissue injury likely contributes to the modulation of lung in response to injury. Indeed, the negative impact of alcohol on susceptibility to infection and on lung barrier function is now well documented. Thus, the alcohol lung represents a very likely comorbidity for the negative consequences of both COVID-19 susceptibility and severity. In this review, we present the known alcohol misuse ramifications on the lung in the context of the current coronavirus pandemic.
Collapse
Affiliation(s)
- Kristina L Bailey
- Research Service, Department of Veterans Affairs Omaha-Western Iowa Health Care System, Omaha, NE, 68105, United States; Pulmonary, Critical Care and Sleep, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, 68198-5910, United States.
| | - Derrick R Samuelson
- Pulmonary, Critical Care and Sleep, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, 68198-5910, United States.
| | - Todd A Wyatt
- Research Service, Department of Veterans Affairs Omaha-Western Iowa Health Care System, Omaha, NE, 68105, United States; Pulmonary, Critical Care and Sleep, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, 68198-5910, United States; Department of Environmental, Agricultural and Occupational Health, University of Nebraska Medical Center, Omaha, NE, 68198-5910, United States.
| |
Collapse
|
37
|
Butler SE, Crowley AR, Natarajan H, Xu S, Weiner JA, Bobak CA, Mattox DE, Lee J, Wieland-Alter W, Connor RI, Wright PF, Ackerman ME. Distinct Features and Functions of Systemic and Mucosal Humoral Immunity Among SARS-CoV-2 Convalescent Individuals. Front Immunol 2021; 11:618685. [PMID: 33584712 PMCID: PMC7876222 DOI: 10.3389/fimmu.2020.618685] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Accepted: 12/21/2020] [Indexed: 12/18/2022] Open
Abstract
Understanding humoral immune responses to SARS-CoV-2 infection will play a critical role in the development of vaccines and antibody-based interventions. We report systemic and mucosal antibody responses in convalescent individuals who experienced varying severity of disease. Whereas assessment of neutralization and antibody-mediated effector functions revealed polyfunctional antibody responses in serum, only robust neutralization and phagocytosis were apparent in nasal wash samples. Serum neutralization and effector functions correlated with systemic SARS-CoV-2-specific IgG response magnitude, while mucosal neutralization was associated with nasal SARS-CoV-2-specific IgA. Antibody depletion experiments support the mechanistic relevance of these correlations. Associations between nasal IgA responses, virus neutralization at the mucosa, and less severe disease suggest the importance of assessing mucosal immunity in larger natural infection cohorts. Further characterization of antibody responses at the portal of entry may define their ability to contribute to protection from infection or reduced risk of hospitalization, informing public health assessment strategies and vaccine development efforts.
Collapse
Affiliation(s)
- Savannah E. Butler
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Dartmouth College, Hanover, NH, United States
| | - Andrew R. Crowley
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Dartmouth College, Hanover, NH, United States
| | - Harini Natarajan
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Dartmouth College, Hanover, NH, United States
| | - Shiwei Xu
- Program in Quantitative and Biology Sciences, Dartmouth College, Hanover, NH, United States
| | - Joshua A. Weiner
- Thayer School of Engineering, Dartmouth College, Hanover, NH, United States
| | - Carly A. Bobak
- Program in Quantitative and Biology Sciences, Dartmouth College, Hanover, NH, United States
| | - Daniel E. Mattox
- Department of Computer Science, Dartmouth College, Hanover, NH, United States
| | - Jiwon Lee
- Thayer School of Engineering, Dartmouth College, Hanover, NH, United States
| | - Wendy Wieland-Alter
- Department of Pediatrics, Geisel School of Medicine at Dartmouth, Dartmouth-Hitchcock Medical Center, Lebanon, NH, United States
| | - Ruth I. Connor
- Department of Pediatrics, Geisel School of Medicine at Dartmouth, Dartmouth-Hitchcock Medical Center, Lebanon, NH, United States
| | - Peter F. Wright
- Department of Pediatrics, Geisel School of Medicine at Dartmouth, Dartmouth-Hitchcock Medical Center, Lebanon, NH, United States
| | - Margaret E. Ackerman
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Dartmouth College, Hanover, NH, United States
- Program in Quantitative and Biology Sciences, Dartmouth College, Hanover, NH, United States
- Thayer School of Engineering, Dartmouth College, Hanover, NH, United States
| |
Collapse
|
38
|
Zhou D, Tian X, Qi R, Peng C, Zhang W. Identification of 22 N-glycosites on spike glycoprotein of SARS-CoV-2 and accessible surface glycopeptide motifs: Implications for vaccination and antibody therapeutics. Glycobiology 2021; 31:69-80. [PMID: 32518941 PMCID: PMC7313968 DOI: 10.1093/glycob/cwaa052] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 05/28/2020] [Accepted: 06/01/2020] [Indexed: 12/17/2022] Open
Abstract
Coronaviruses hijack human enzymes to assemble the sugar coat on their spike glycoproteins. The mechanisms by which human antibodies may recognize the antigenic viral peptide epitopes hidden by the sugar coat are unknown. Glycosylation by insect cells differs from the native form produced in human cells, but insect cell-derived influenza vaccines have been approved by the US Food and Drug Administration. In this study, we analyzed recombinant severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike protein secreted from BTI-Tn-5B1-4 insect cells, by trypsin and chymotrypsin digestion followed by mass spectrometry analysis. We acquired tandem mass spectrometry (MS/MS) spectrums for glycopeptides of all 22 predicted N-glycosylated sites. We further analyzed the surface accessibility of spike proteins according to cryogenic electron microscopy and homolog-modeled structures and available antibodies that bind to SARS-CoV-1. All 22 N-glycosylated sites of SARS-CoV-2 are modified by high-mannose N-glycans. MS/MS fragmentation clearly established the glycopeptide identities. Electron densities of glycans cover most of the spike receptor-binding domain of SARS-CoV-2, except YQAGSTPCNGVEGFNCYFPLQSYGFQPTNGVGYQ, similar to a region FSPDGKPCTPPALNCYWPLNDYGFYTTTGIGYQ in SARS-CoV-1. Other surface-exposed domains include those located on central helix, connecting region, heptad repeats and N-terminal domain. Because the majority of antibody paratopes bind to the peptide portion with or without sugar modification, we propose a snake-catching model for predicted paratopes: a minimal length of peptide is first clamped by a paratope and sugar modifications close to the peptide either strengthen or do not hinder the binding.
Collapse
Affiliation(s)
- Dapeng Zhou
- Tongji University School of Medicine, 1239 Siping Road, Shanghai 200092, China.,Shanghai Pudong New Area Mental Health Center affiliated with Tongji University School of Medicine, 165 Sanlin Road, Shanghai 200124, China
| | - Xiaoxu Tian
- National Facility for Protein Science in Shanghai, Zhangjiang Lab, Shanghai Advanced Research Institute, Chinese Academy of Science, 333 Haike Road, Shanghai 201210, China
| | - Ruibing Qi
- Innovation Team of Small Animal Infectious Disease, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, 518 Ziyue Road, Shanghai 200241, China
| | - Chao Peng
- National Facility for Protein Science in Shanghai, Zhangjiang Lab, Shanghai Advanced Research Institute, Chinese Academy of Science, 333 Haike Road, Shanghai 201210, China
| | - Wen Zhang
- Fudan University Pudong Medical Center, Institutes of Biomedical Sciences, 200433 Gongwei Road, Shanghai, China.,Department of Systems Biology for Medicine, Shanghai Medical College, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| |
Collapse
|
39
|
Domains and Functions of Spike Protein in Sars-Cov-2 in the Context of Vaccine Design. Viruses 2021; 13:v13010109. [PMID: 33466921 PMCID: PMC7829931 DOI: 10.3390/v13010109] [Citation(s) in RCA: 201] [Impact Index Per Article: 50.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 01/10/2021] [Accepted: 01/12/2021] [Indexed: 12/11/2022] Open
Abstract
The spike protein in SARS-CoV-2 (SARS-2-S) interacts with the human ACE2 receptor to gain entry into a cell to initiate infection. Both Pfizer/BioNTech's BNT162b2 and Moderna's mRNA-1273 vaccine candidates are based on stabilized mRNA encoding prefusion SARS-2-S that can be produced after the mRNA is delivered into the human cell and translated. SARS-2-S is cleaved into S1 and S2 subunits, with S1 serving the function of receptor-binding and S2 serving the function of membrane fusion. Here, I dissect in detail the various domains of SARS-2-S and their functions discovered through a variety of different experimental and theoretical approaches to build a foundation for a comprehensive mechanistic understanding of how SARS-2-S works to achieve its function of mediating cell entry and subsequent cell-to-cell transmission. The integration of structure and function of SARS-2-S in this review should enhance our understanding of the dynamic processes involving receptor binding, multiple cleavage events, membrane fusion, viral entry, as well as the emergence of new viral variants. I highlighted the relevance of structural domains and dynamics to vaccine development, and discussed reasons for the spike protein to be frequently featured in the conspiracy theory claiming that SARS-CoV-2 is artificially created.
Collapse
|
40
|
Abstract
The newly emerged severe acute respiratory syndrome-related coronavirus-2 (SARS-CoV-2) has recently caused pandemic Coronavirus Disease-2019 (COVID-19). Considering the serious medical, economic and social consequences of this pandemic and the lack of definite medication and vaccine it is necessary to describe natural immune responses to the SARS-CoV-2 in order to exploit them for treating the patients and monitoring the general population. Moreover, detecting the most immunogenic antigens of the virus is fundamental for designing effective vaccines. Antibodies being valuable for diagnostic therapeutic and protective purposes are suitable to be addressed in this context. Herein, we have summarized the findings of serological investigations and the outcomes of neutralizing antibodies administration in COVID-19 patients.
Collapse
Affiliation(s)
- Sara Assadiasl
- Molecular Immunology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Yousef Fatahi
- Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.,Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahdi Zavvar
- Department of Medical Laboratory Sciences, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Hossein Nicknam
- Molecular Immunology Research Center, Tehran University of Medical Sciences, Tehran, Iran.,Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
41
|
Mudgal R, Nehul S, Tomar S. Prospects for mucosal vaccine: shutting the door on SARS-CoV-2. Hum Vaccin Immunother 2020; 16:2921-2931. [PMID: 32931361 PMCID: PMC7544966 DOI: 10.1080/21645515.2020.1805992] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 07/19/2020] [Accepted: 08/03/2020] [Indexed: 12/25/2022] Open
Abstract
The sudden emergence of a highly transmissible and pathogenic coronavirus SARS-CoV-2 in December 2019 from China and its rapid global spread has posed an international health emergency. The rapid development of an effective vaccine is imperative to control the spread of SARS-CoV-2. A number of concurrent efforts to find an effective therapeutic agent or vaccine for COVID-19 (coronavirus disease 2019) are being undertaken globally. Oral and nasal mucosal surfaces serve as the primary portal of entry for pathogens like coronaviruses in the human body. As evidenced by studies on similar coronaviruses (SARS-CoV and MERS-CoV), mucosal vaccination can provide a safe and effective means for the induction of long-lasting systemic and mucosal immunity to confer protection against SARS-CoV-2. This article summarizes the approaches to an effective mucosal vaccine formulation which can be a rewarding approach to combat the unprecedented threat posed by this emerging global pandemic.
Collapse
Affiliation(s)
- Rajat Mudgal
- Department of Biotechnology, Indian Institute of Technology Roorkee, Roorkee, India
| | - Sanketkumar Nehul
- Department of Biotechnology, Indian Institute of Technology Roorkee, Roorkee, India
| | - Shailly Tomar
- Department of Biotechnology, Indian Institute of Technology Roorkee, Roorkee, India
| |
Collapse
|
42
|
Korang SK, Juul S, Nielsen EE, Feinberg J, Siddiqui F, Ong G, Klingenberg S, Veroniki AA, Bu F, Thabane L, Thomsen AR, Jakobsen JC, Gluud C. Vaccines to prevent COVID-19: a protocol for a living systematic review with network meta-analysis including individual patient data (The LIVING VACCINE Project). Syst Rev 2020; 9:262. [PMID: 33218366 PMCID: PMC7678579 DOI: 10.1186/s13643-020-01516-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 10/27/2020] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes coronavirus disease 2019 (COVID-19) which has rapidly spread worldwide. Several human randomized clinical trials assessing potential vaccines are currently underway. There is an urgent need for a living systematic review that continuously assesses the beneficial and harmful effects of all available vaccines for COVID-19. METHODS/DESIGN We will conduct a living systematic review based on searches of major medical databases (e.g., MEDLINE, EMBASE, CENTRAL) and clinical trial registries from their inception onwards to identify relevant randomized clinical trials. We will update the literature search once a week to continuously assess if new evidence is available. Two review authors will independently extract data and conduct risk of bias assessments. We will include randomized clinical trials comparing any vaccine aiming to prevent COVID-19 (including but not limited to messenger RNA; DNA; non-replicating viral vector; replicating viral vector; inactivated virus; protein subunit; dendritic cell; other vaccines) with any comparator (placebo; "active placebo;" no intervention; standard care; an "active" intervention; another vaccine for COVID-19) for participants in all age groups. Primary outcomes will be all-cause mortality; a diagnosis of COVID-19; and serious adverse events. Secondary outcomes will be quality of life and non-serious adverse events. The living systematic review will include aggregate data meta-analyses, trial sequential analyses, network meta-analyses, and individual patient data meta-analyses. Within-study bias will be assessed using Cochrane risk of bias tool. The Grading of Recommendations, Assessment, Development and Evaluations (GRADE) and Confidence in Network Meta-Analysis (CINeMA) approaches will be used to assess certainty of evidence. Observational studies describing harms identified during the search for trials will also be included and described and analyzed separately. DISCUSSION COVID-19 has become a pandemic with substantial mortality. A living systematic review assessing the beneficial and harmful effects of different vaccines is urgently needed. This living systematic review will regularly inform best practice in vaccine prevention and clinical research of this highly prevalent disease. SYSTEMATIC REVIEW REGISTRATION PROSPERO CRD42020196492.
Collapse
Affiliation(s)
- Steven Kwasi Korang
- Copenhagen Trial Unit, Centre for Clinical Intervention Research, Department 7812, Rigshospitalet, Copenhagen University Hospital, Blegdamsvej 9, 2100 Copenhagen, Denmark
| | - Sophie Juul
- Copenhagen Trial Unit, Centre for Clinical Intervention Research, Department 7812, Rigshospitalet, Copenhagen University Hospital, Blegdamsvej 9, 2100 Copenhagen, Denmark
| | - Emil Eik Nielsen
- Copenhagen Trial Unit, Centre for Clinical Intervention Research, Department 7812, Rigshospitalet, Copenhagen University Hospital, Blegdamsvej 9, 2100 Copenhagen, Denmark
| | - Joshua Feinberg
- Copenhagen Trial Unit, Centre for Clinical Intervention Research, Department 7812, Rigshospitalet, Copenhagen University Hospital, Blegdamsvej 9, 2100 Copenhagen, Denmark
| | - Faiza Siddiqui
- Copenhagen Trial Unit, Centre for Clinical Intervention Research, Department 7812, Rigshospitalet, Copenhagen University Hospital, Blegdamsvej 9, 2100 Copenhagen, Denmark
| | - Giok Ong
- Systematic Review Initiative, NHS Blood and Transplant, John Radcliffe Hospital, Headley Way, Oxford, OX3 9BQ UK
| | - Sarah Klingenberg
- Copenhagen Trial Unit, Centre for Clinical Intervention Research, Department 7812, Rigshospitalet, Copenhagen University Hospital, Blegdamsvej 9, 2100 Copenhagen, Denmark
- The Cochrane Hepato-Biliary Group, Copenhagen Trial Unit, Centre for Clinical Intervention Research, Department 7812, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Areti Angeliki Veroniki
- Department of Primary Education, School of Education, University of Ioannina, Ioannina, Greece
- Knowledge Translation Program, Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Toronto, Ontario Canada
| | - Fanlong Bu
- Centre for Evidence-based Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Lehana Thabane
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Ontario Canada
| | - Allan Randrup Thomsen
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Janus C. Jakobsen
- Copenhagen Trial Unit, Centre for Clinical Intervention Research, Department 7812, Rigshospitalet, Copenhagen University Hospital, Blegdamsvej 9, 2100 Copenhagen, Denmark
- The Cochrane Hepato-Biliary Group, Copenhagen Trial Unit, Centre for Clinical Intervention Research, Department 7812, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
- Department of Regional Health Research, The Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| | - Christian Gluud
- Copenhagen Trial Unit, Centre for Clinical Intervention Research, Department 7812, Rigshospitalet, Copenhagen University Hospital, Blegdamsvej 9, 2100 Copenhagen, Denmark
- The Cochrane Hepato-Biliary Group, Copenhagen Trial Unit, Centre for Clinical Intervention Research, Department 7812, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| |
Collapse
|
43
|
COVID-19: Current Developments and Further Opportunities in Drug Delivery and Therapeutics. Pharmaceutics 2020; 12:pharmaceutics12100945. [PMID: 33023033 PMCID: PMC7601382 DOI: 10.3390/pharmaceutics12100945] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 09/28/2020] [Accepted: 09/29/2020] [Indexed: 12/15/2022] Open
Abstract
SARS-CoV-2 has affected people from all age groups, races and ethnicities. Given that many infected individuals are asymptomatic, they transmit the disease to others unknowingly, which has resulted in the spread of infection at an alarming rate. This review aims to provide an overview of the pathophysiology, preventive measures to reduce the disease spread, therapies currently in use, an update on vaccine development and opportunities for vaccine delivery. The World Health Organization has advised several precautions including social distancing, hand washing and the use of PPE including gloves and face masks for minimizing the spread of SARS-CoV-2 infection. At present, several antiviral therapies previously approved for other infections are being repositioned to study their efficacy against SARS-CoV-2. In addition, some medicines (i.e., remdesivir, chloroquine, hydroxychloroquine) have received emergency use authorisation from the FDA. Plasma therapy has also been authorised for emergency use for the treatment of COVID-19 on a smaller scale. However, no vaccine has been approved so far against this virus. Nevertheless, several potential vaccine targets have been reported, and development of different types of vaccines including DNA, mRNA, viral vector, inactivated, subunit and vaccine-like particles is in process. It is concluded that a suitable candidate delivered through an advanced drug delivery approach would effectively boost the immune system against this coronavirus.
Collapse
|
44
|
Travis CR. As Plain as the Nose on Your Face: The Case for A Nasal (Mucosal) Route of Vaccine Administration for Covid-19 Disease Prevention. Front Immunol 2020; 11:591897. [PMID: 33117404 PMCID: PMC7561361 DOI: 10.3389/fimmu.2020.591897] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 09/17/2020] [Indexed: 12/30/2022] Open
|
45
|
Dagotto G, Yu J, Barouch DH. Approaches and Challenges in SARS-CoV-2 Vaccine Development. Cell Host Microbe 2020; 28:364-370. [PMID: 32798444 PMCID: PMC7416703 DOI: 10.1016/j.chom.2020.08.002] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 08/04/2020] [Accepted: 08/05/2020] [Indexed: 02/06/2023]
Abstract
The explosive spread of SARS-CoV-2 suggests that a vaccine will be required to end this global pandemic. Progress in SARS-CoV-2 vaccine development to date has been faster than for any other pathogen in history. Multiple SARS-CoV-2 vaccine candidates have been evaluated in preclinical models and are currently in clinical trials. In this Perspective, we discuss three topics that are critical for SARS-CoV-2 vaccine development: antigen selection and engineering, preclinical challenge studies in non-human primate models, and immune correlates of protection.
Collapse
MESH Headings
- Animals
- Antigens, Viral/chemistry
- Antigens, Viral/genetics
- Betacoronavirus/genetics
- Betacoronavirus/immunology
- COVID-19
- COVID-19 Vaccines
- Coronavirus Infections/epidemiology
- Coronavirus Infections/immunology
- Coronavirus Infections/prevention & control
- Host Microbial Interactions/immunology
- Humans
- Immunity, Cellular
- Immunity, Humoral
- Immunity, Innate
- Models, Animal
- Pandemics/prevention & control
- Pneumonia, Viral/epidemiology
- Pneumonia, Viral/immunology
- Pneumonia, Viral/prevention & control
- Primates
- SARS-CoV-2
- Spike Glycoprotein, Coronavirus/chemistry
- Spike Glycoprotein, Coronavirus/genetics
- Spike Glycoprotein, Coronavirus/immunology
- Viral Vaccines/administration & dosage
- Viral Vaccines/immunology
- Viral Vaccines/isolation & purification
Collapse
Affiliation(s)
- Gabriel Dagotto
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Jingyou Yu
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Dan H Barouch
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA; Harvard Medical School, Boston, MA 02115, USA; Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA; Massachusetts Consortium on Pathogen Readiness, Boston, MA 02215, USA.
| |
Collapse
|
46
|
Chen Y, Tong X, Li Y, Gu B, Yan J, Liu Y, Shen H, Huang R, Wu C. A comprehensive, longitudinal analysis of humoral responses specific to four recombinant antigens of SARS-CoV-2 in severe and non-severe COVID-19 patients. PLoS Pathog 2020; 16:e1008796. [PMID: 32913364 PMCID: PMC7482996 DOI: 10.1371/journal.ppat.1008796] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 07/09/2020] [Indexed: 12/20/2022] Open
Abstract
There is an urgent need for effective treatment and preventive vaccine to contain this devastating global pandemic, which requires a comprehensive understanding of humoral responses specific to SARS-CoV-2 during the disease progression and convalescent phase of COVID-19 patients. We continuously monitored the serum IgM and IgG responses specific to four SARS-CoV-2 related antigens, including the nucleoprotein (NP), receptor binding domain (RBD), S1 protein, and ectodomain (ECD) of the spike protein among non-severe and severe COVID-19 patients for seven weeks since disease onset. Most patients generated humoral responses against NP and spike protein-related antigens but with their distinct kinetics profiles. Combined detection of NP and ECD antigens as detecting antigen synergistically improved the sensitivity of the serological assay, compared to that of using NP or RBD as detection antigen. 80.7% of convalescent sera from COVID-19 patients revealed that the varying extents of neutralization activities against SARS-CoV-2. S1-specific and ECD-specific IgA responses were strongly correlated with the neutralization activities in non-severe patients, but not in severe patients. Moreover, the neutralizing activities of the convalescent sera were shown to significantly decline during the period between 21 days to 28 days after hospital discharge, accompanied by a substantial drop in RBD-specific IgA response. Our data provide evidence that are crucial for serological testing, antibody-based intervention, and vaccine design of COVID-19.
Collapse
Affiliation(s)
- Yuxin Chen
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Xin Tong
- Department of Infectious Diseases, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Yang Li
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Bin Gu
- Medical Technology School of Xuzhou Medical University, Xuzhou Key Laboratory of Laboratory Diagnostics, Xuzhou, Jiangsu, China
- Department of Laboratory Medicine, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Jiawei Yan
- Department of Laboratory Medicine, Xuzhou Infectious Disease Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yong Liu
- Department of Experimental Medicine, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Han Shen
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Rui Huang
- Department of Infectious Diseases, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Chao Wu
- Department of Infectious Diseases, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing, Jiangsu, China
| |
Collapse
|
47
|
Thames A, Wolniak KL, Stupp SI, Jewett MC. Principles Learned from the International Race to Develop a Safe and Effective COVID-19 Vaccine. ACS CENTRAL SCIENCE 2020; 6:1341-1347. [PMID: 32868999 PMCID: PMC7374935 DOI: 10.1021/acscentsci.0c00644] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Indexed: 05/05/2023]
Abstract
Vaccines against COVID-19 have the potential to protect people before they are exposed to the infective form of the virus. However, because of the involvement of pathogenic immune processes in many severe presentations of COVID-19, eliciting an immune response with a vaccine must strike a delicate balance to achieve viral clearance without also inducing immune-mediated harm. This Outlook synthesizes current laboratory findings to define which parts of the immune system help with recovery from and protection against the virus and which can lead to adverse outcomes. To inform our understanding, we analyze research about the immune mechanisms implicated in SARS-CoV, from the 2003 outbreak, and SARS-CoV-2, the virus causing COVID-19. The impact of how innate immunity, humoral immunity, and cell-mediated immunity play a role in a harmful versus helpful response is discussed, establishing principles to guide the development and evaluation of a safe but effective COVID-19 vaccine. The principles derived include (i) targeting the appropriate specificity and effector function of the humoral response, (ii) eliciting a T cell response, especially a cytotoxic T cell response, to achieve safe, yet effective, immune protection from COVID-19, and (iii) monitoring for the possibility of acute lung injury during SARS-CoV-2 infection post-vaccination in preclinical and clinical studies. These principles can not only guide efforts toward a safe and effective COVID-19 vaccine, but also the development of effective vaccines for viral pandemics to come.
Collapse
Affiliation(s)
- Ariel
H. Thames
- Medical
Scientist Training Program, Northwestern
University Feinberg School of Medicine, Chicago, Illinois 60611, United States
- Center
for Synthetic Biology, Northwestern University, 2145 Sheridan Road, Tech B486, Evanston, Illinois 60208, United States
- Interdisciplinary
Biological Sciences Program, Northwestern
University, Evanston, Illinois 60208, United States
| | - Kristy L. Wolniak
- Division
of Hematopathology, Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, United States
| | - Samuel I. Stupp
- Simpson
Querrey Institute, Northwestern University, Chicago, Illinois 60611, United States
- Department
of Biomedical Engineering, Northwestern
University, Evanston, Illinois 60208, United States
- Department
of Chemistry, Northwestern University, Evanston, Illinois 60208, United States
- Department
of Materials Science and Engineering, Northwestern
University, Evanston, Illinois 60208, United States
- Department
of Medicine, Northwestern University Feinberg
School of Medicine, Chicago, Illinois 60611, United States
| | - Michael C. Jewett
- Medical
Scientist Training Program, Northwestern
University Feinberg School of Medicine, Chicago, Illinois 60611, United States
- Center
for Synthetic Biology, Northwestern University, 2145 Sheridan Road, Tech B486, Evanston, Illinois 60208, United States
- Interdisciplinary
Biological Sciences Program, Northwestern
University, Evanston, Illinois 60208, United States
- Simpson
Querrey Institute, Northwestern University, Chicago, Illinois 60611, United States
- Department
of Chemical and Biological Engineering, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United
States
| |
Collapse
|
48
|
Ejemel M, Li Q, Hou S, Schiller ZA, Tree JA, Wallace A, Amcheslavsky A, Kurt Yilmaz N, Buttigieg KR, Elmore MJ, Godwin K, Coombes N, Toomey JR, Schneider R, Ramchetty AS, Close BJ, Chen DY, Conway HL, Saeed M, Ganesa C, Carroll MW, Cavacini LA, Klempner MS, Schiffer CA, Wang Y. A cross-reactive human IgA monoclonal antibody blocks SARS-CoV-2 spike-ACE2 interaction. Nat Commun 2020; 11:4198. [PMID: 32826914 PMCID: PMC7442812 DOI: 10.1038/s41467-020-18058-8] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 08/03/2020] [Indexed: 01/19/2023] Open
Abstract
COVID-19 caused by SARS-CoV-2 has become a global pandemic requiring the development of interventions for the prevention or treatment to curtail mortality and morbidity. No vaccine to boost mucosal immunity, or as a therapeutic, has yet been developed to SARS-CoV-2. In this study, we discover and characterize a cross-reactive human IgA monoclonal antibody, MAb362. MAb362 binds to both SARS-CoV and SARS-CoV-2 spike proteins and competitively blocks ACE2 receptor binding, by overlapping the ACE2 structural binding epitope. Furthermore, MAb362 IgA neutralizes both pseudotyped SARS-CoV and SARS-CoV-2 in 293 cells expressing ACE2. When converted to secretory IgA, MAb326 also neutralizes authentic SARS-CoV-2 virus while the IgG isotype shows no neutralization. Our results suggest that SARS-CoV-2 specific IgA antibodies, such as MAb362, may provide effective immunity against SARS-CoV-2 by inducing mucosal immunity within the respiratory system, a potentially critical feature of an effective vaccine. Here, Ejemel et al. report the identification and characterization of a cross-neutralizing human IgA monoclonal antibody, named MAb362, that binds the receptor-binding domain of SARS-CoV-2 Spike, blocking its interaction with the ACE2 host receptor.
Collapse
Affiliation(s)
- Monir Ejemel
- MassBiologics of the University of Massachusetts Medical School, Boston, MA, USA
| | - Qi Li
- MassBiologics of the University of Massachusetts Medical School, Boston, MA, USA
| | - Shurong Hou
- Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Boston, MA, USA
| | - Zachary A Schiller
- MassBiologics of the University of Massachusetts Medical School, Boston, MA, USA
| | - Julia A Tree
- National Infection Service, Public Health England, Porton Down, Salisbury, Wiltshire, UK
| | - Aaron Wallace
- MassBiologics of the University of Massachusetts Medical School, Boston, MA, USA
| | - Alla Amcheslavsky
- MassBiologics of the University of Massachusetts Medical School, Boston, MA, USA
| | - Nese Kurt Yilmaz
- Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Boston, MA, USA
| | - Karen R Buttigieg
- National Infection Service, Public Health England, Porton Down, Salisbury, Wiltshire, UK
| | - Michael J Elmore
- National Infection Service, Public Health England, Porton Down, Salisbury, Wiltshire, UK
| | - Kerry Godwin
- National Infection Service, Public Health England, Porton Down, Salisbury, Wiltshire, UK
| | - Naomi Coombes
- National Infection Service, Public Health England, Porton Down, Salisbury, Wiltshire, UK
| | - Jacqueline R Toomey
- MassBiologics of the University of Massachusetts Medical School, Boston, MA, USA
| | - Ryan Schneider
- MassBiologics of the University of Massachusetts Medical School, Boston, MA, USA
| | - Anudeep S Ramchetty
- MassBiologics of the University of Massachusetts Medical School, Boston, MA, USA
| | - Brianna J Close
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA
| | - Da-Yuan Chen
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA
| | - Hasahn L Conway
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA
| | - Mohsan Saeed
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA
| | - Chandrashekar Ganesa
- MassBiologics of the University of Massachusetts Medical School, Boston, MA, USA
| | - Miles W Carroll
- National Infection Service, Public Health England, Porton Down, Salisbury, Wiltshire, UK
| | - Lisa A Cavacini
- MassBiologics of the University of Massachusetts Medical School, Boston, MA, USA.
| | - Mark S Klempner
- MassBiologics of the University of Massachusetts Medical School, Boston, MA, USA.
| | - Celia A Schiffer
- Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Boston, MA, USA.
| | - Yang Wang
- MassBiologics of the University of Massachusetts Medical School, Boston, MA, USA.
| |
Collapse
|
49
|
Al-Kassmy J, Pedersen J, Kobinger G. Vaccine Candidates against Coronavirus Infections. Where Does COVID-19 Stand? Viruses 2020; 12:E861. [PMID: 32784685 PMCID: PMC7472384 DOI: 10.3390/v12080861] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 08/03/2020] [Accepted: 08/05/2020] [Indexed: 12/16/2022] Open
Abstract
Seven years after the Middle East respiratory syndrome (MERS) outbreak, a new severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) made its first appearance in a food market in Wuhan, China, drawing an entirely new course to our lives. As the virus belongs to the same genus of MERS and SARS, researchers have been trying to draw lessons from previous outbreaks to find a potential cure. Although there were five Phase I human vaccine trials against SARS and MERS, the lack of data in humans provided us with limited benchmarks that could help us design a new vaccine for Coronavirus disease 2019 (COVID-19). In this review, we showcase the similarities in structures of virus components between SARS-CoV, MERS-CoV, and SARS-CoV-2 in areas relevant to vaccine design. Using the ClinicalTrials.gov and World Health Organization (WHO) databases, we shed light on the 16 current approved clinical trials worldwide in search for a COVID-19 vaccine. The different vaccine platforms being tested are Bacillus Calmette-Guérin (BCG) vaccines, DNA and RNA-based vaccines, inactivated vaccines, protein subunits, and viral vectors. By thoroughly analyzing different trials and platforms, we also discuss the advantages and disadvantages of using each type of vaccine and how they can contribute to the design of an adequate vaccine for COVID-19. Studying past efforts invested in conducting vaccine trials for MERS and SARS will provide vital insights regarding the best approach to designing an effective vaccine against COVID-19.
Collapse
Affiliation(s)
- Jawad Al-Kassmy
- Department of Experimental Surgery, McGill University, Montreal General Hospital 1650 Cedar Avenue, Montreal, QC H3G 1A4, Canada
| | - Jannie Pedersen
- Axe des Maladies Infectieuses et Immunitaires, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec, QC G1V 4G2, Canada; (J.P.); (G.K.)
| | - Gary Kobinger
- Axe des Maladies Infectieuses et Immunitaires, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec, QC G1V 4G2, Canada; (J.P.); (G.K.)
- Département de Microbiologie-Infectiologie et Immunologie, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada
- Department of Medical Microbiology, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA 19104-4238, USA
| |
Collapse
|
50
|
Butler SE, Crowley AR, Natarajan H, Xu S, Weiner JA, Lee J, Wieland-Alter WF, Connor RI, Wright PF, Ackerman ME. Features and Functions of Systemic and Mucosal Humoral Immunity Among SARS-CoV-2 Convalescent Individuals. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2020:2020.08.05.20168971. [PMID: 32793926 PMCID: PMC7418747 DOI: 10.1101/2020.08.05.20168971] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Understanding humoral immune responses to SARS-CoV-2 infection will play a critical role in the development of vaccines and antibody-based interventions. We report systemic and mucosal antibody responses in convalescent individuals who experienced varying disease severity. Robust antibody responses to diverse SARS-CoV-2 antigens and evidence of elevated responses to endemic CoV were observed among convalescent donors. SARS-CoV-2-specific IgA and IgG responses were often negatively correlated, particularly in mucosal samples, suggesting subject-intrinsic biases in isotype switching. Assessment of antibody-mediated effector functions revealed an inverse correlation between systemic and mucosal neutralization activity and site-dependent differences in the isotype of neutralizing antibodies. Serum neutralization correlated with systemic anti-SARS-CoV-2 IgG and IgM response magnitude, while mucosal neutralization was associated with nasal SARS-CoV-2-specific IgA. These findings begin to map how diverse Ab characteristics relate to Ab functions and outcomes of infection, informing public health assessment strategies and vaccine development efforts.
Collapse
|