1
|
Adugna T, Niu Q, Guan G, Du J, Yang J, Tian Z, Yin H. Advancements in nanoparticle-based vaccine development against Japanese encephalitis virus: a systematic review. Front Immunol 2024; 15:1505612. [PMID: 39759527 PMCID: PMC11695416 DOI: 10.3389/fimmu.2024.1505612] [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: 10/03/2024] [Accepted: 11/25/2024] [Indexed: 01/07/2025] Open
Abstract
Vaccination remains the sole effective strategy for combating Japanese encephalitis (JE). Both inactivated and live attenuated vaccines exhibit robust immunogenicity. However, the production of these conventional vaccine modalities necessitates extensive cultivation of the pathogen, incurring substantial costs and presenting significant biosafety risks. Moreover, the administration of live pathogens poses potential hazards for individuals or animals with compromised immune systems or other health vulnerabilities. Subsequently, ongoing research endeavors are focused on the development of next-generation JE vaccines utilizing nanoparticle (NP) platforms. This systematic review seeks to aggregate the research findings pertaining to NP-based vaccine development against JE. A thorough literature search was conducted across established English-language databases for research articles on JE NP vaccine development published between 2000 and 2023. A total of twenty-eight published studies were selected for detailed analysis in this review. Of these, 16 studies (57.14%) concentrated on virus-like particles (VLPs) employing various structural proteins. Other approaches, including sub-viral particles (SVPs), biopolymers, and both synthetic and inorganic NP platforms, were utilized to a lesser extent. The results of these investigations indicated that, despite variations in the usage of adjuvants, dosages, NP types, antigenic proteins, and animal models employed across different studies, the candidate NP vaccines developed were capable of eliciting enhanced humoral and cellular adaptive immune responses, providing effective protection (70-100%) for immunized mice against lethal challenges posed by virulent Japanese encephalitis virus (JEV). In conclusion, prospective next-generation JE vaccines for humans and animals may emerge from these candidate formulations following further evaluation in subsequent vaccine development phases.
Collapse
Affiliation(s)
- Takele Adugna
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, State Key Laboratory of Veterinary Etiological Biology Project, Yangzhou, China
- College of Veterinary Medicine and Animal Sciences, University of Gondar, Gondar, Ethiopia
| | - Qingli Niu
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, State Key Laboratory of Veterinary Etiological Biology Project, Yangzhou, China
| | - Guiquan Guan
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, State Key Laboratory of Veterinary Etiological Biology Project, Yangzhou, China
| | - Junzheng Du
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, State Key Laboratory of Veterinary Etiological Biology Project, Yangzhou, China
| | - Jifei Yang
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, State Key Laboratory of Veterinary Etiological Biology Project, Yangzhou, China
| | - Zhancheng Tian
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, State Key Laboratory of Veterinary Etiological Biology Project, Yangzhou, China
| | - Hong Yin
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, State Key Laboratory of Veterinary Etiological Biology Project, Yangzhou, China
| |
Collapse
|
2
|
Chege H, Githigia S, Gathumbi J, Chege N, Ojuok R, Odaba J, Mwalimu S, Oboge H, Steinaa L, Nene V, Lacasta A. An Improved Theileria parva Sporozoite Seroneutralization Assay for the Identification of East Coast Fever Immune Correlates. Antibodies (Basel) 2024; 13:100. [PMID: 39727483 DOI: 10.3390/antib13040100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 11/04/2024] [Accepted: 11/20/2024] [Indexed: 12/28/2024] Open
Abstract
BACKGROUND Immune correlates of protection are ideal tools to predict treatment or vaccine efficacy. However, the accuracy of the immune correlate and the capability to robustly predict the outcome of a vaccine candidate are determined by the performance of the in vitro immunoassay used. Several Theileria parva sporozoite seroneutralization assays have previously been used to assess antibody functional activities; however, a common limitation has been the need for fresh material, target cells and sporozoites, and operator-to-operator bias. An improved assay represents a positive step toward overcoming challenges associated with variability and it might provide a more reliable means of establishing an immune correlate with protection after sub-unit vaccine administration. METHODS Herein, we describe key improvements, among them, (1) the use of frozen parasites and target cells to avoid batch-to-batch variations and (2) the development of a new assay read-out based on the detection of infected cells through flow cytometry, instead of the use of Giemsa staining and microscopic evaluation, in order to improve the reproducibility of the results. RESULTS The improved seroneutralization assay is not only able to detect the individual neutralizing capacity of antibodies; it also detects the additive effect of antibody combinations. CONCLUSIONS This effect is described for the first time in Theileria parva and is of great interest for new antigen discovery and/or the epitope discovery of already known antigens like p67, opening a new avenue for the identification of ECF immune correlates of protection and the in vitro down-selection of new Theileria parva vaccine candidates, thereby contributing to reducing the use of animals in challenge experiments.
Collapse
Affiliation(s)
- Hannah Chege
- Animal and Human Health Program, International Livestock Research Institute, Nairobi P.O. Box 30709, Kenya
- Department of Veterinary Pathology, Microbiology and Parasitology, Faculty of Veterinary Medicine, University of Nairobi, Nairobi P.O. Box 29053-00625, Kenya
| | - Samuel Githigia
- Department of Veterinary Pathology, Microbiology and Parasitology, Faculty of Veterinary Medicine, University of Nairobi, Nairobi P.O. Box 29053-00625, Kenya
| | - James Gathumbi
- Department of Veterinary Pathology, Microbiology and Parasitology, Faculty of Veterinary Medicine, University of Nairobi, Nairobi P.O. Box 29053-00625, Kenya
| | - Naomi Chege
- Animal and Human Health Program, International Livestock Research Institute, Nairobi P.O. Box 30709, Kenya
| | - Rose Ojuok
- Animal and Human Health Program, International Livestock Research Institute, Nairobi P.O. Box 30709, Kenya
| | - Josiah Odaba
- Animal and Human Health Program, International Livestock Research Institute, Nairobi P.O. Box 30709, Kenya
| | - Stephen Mwalimu
- Animal and Human Health Program, International Livestock Research Institute, Nairobi P.O. Box 30709, Kenya
| | - Harriet Oboge
- Animal and Human Health Program, International Livestock Research Institute, Nairobi P.O. Box 30709, Kenya
- Department of Veterinary Pathology, Microbiology and Parasitology, Faculty of Veterinary Medicine, University of Nairobi, Nairobi P.O. Box 29053-00625, Kenya
| | - Lucilla Steinaa
- Animal and Human Health Program, International Livestock Research Institute, Nairobi P.O. Box 30709, Kenya
| | - Vishvanath Nene
- Animal and Human Health Program, International Livestock Research Institute, Nairobi P.O. Box 30709, Kenya
| | - Anna Lacasta
- Animal and Human Health Program, International Livestock Research Institute, Nairobi P.O. Box 30709, Kenya
| |
Collapse
|
3
|
Thimmiraju SR, Villar MJ, Kimata JT, Strych U, Bottazzi ME, Hotez PJ, Pollet J. Optimization of Cellular Transduction by the HIV-Based Pseudovirus Platform with Pan-Coronavirus Spike Proteins. Viruses 2024; 16:1492. [PMID: 39339968 PMCID: PMC11437443 DOI: 10.3390/v16091492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 09/05/2024] [Accepted: 09/17/2024] [Indexed: 09/30/2024] Open
Abstract
Over the past three years, new SARS-CoV-2 variants have continuously emerged, evolving to a point where an immune response against the original vaccine no longer provided optimal protection against these new strains. During this time, high-throughput neutralization assays based on pseudoviruses have become a valuable tool for assessing the efficacy of new vaccines, screening updated vaccine candidates against emerging variants, and testing the efficacy of new therapeutics such as monoclonal antibodies. Lentiviral vectors derived from HIV-1 are popular for developing pseudo and chimeric viruses due to their ease of use, stability, and long-term transgene expression. However, the HIV-based platform has lower transduction rates for pseudotyping coronavirus spike proteins than other pseudovirus platforms, necessitating more optimized methods. As the SARS-CoV-2 virus evolved, we produced over 18 variants of the spike protein for pseudotyping with an HIV-based vector, optimizing experimental parameters for their production and transduction. In this article, we present key parameters that were assessed to improve such technology, including (a) the timing and method of collection of pseudovirus supernatant; (b) the timing of host cell transduction; (c) cell culture media replenishment after pseudovirus adsorption; and (d) the centrifugation (spinoculation) parameters of the host cell+ pseudovirus mix, towards improved transduction. Additionally, we found that, for some pseudoviruses, the addition of a cationic polymer (polybrene) to the culture medium improved the transduction process. These findings were applicable across variant spike pseudoviruses that include not only SARS-CoV-2 variants, but also SARS, MERS, Alpha Coronavirus (NL-63), and bat-like coronaviruses. In summary, we present improvements in transduction efficiency, which can broaden the dynamic range of the pseudovirus titration and neutralization assays.
Collapse
Affiliation(s)
- Syamala Rani Thimmiraju
- Texas Children’s Hospital Center for Vaccine Development, Houston, TX 77030, USA; (S.R.T.); (M.J.V.); (U.S.); (M.E.B.); (P.J.H.)
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Maria Jose Villar
- Texas Children’s Hospital Center for Vaccine Development, Houston, TX 77030, USA; (S.R.T.); (M.J.V.); (U.S.); (M.E.B.); (P.J.H.)
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jason T. Kimata
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA;
| | - Ulrich Strych
- Texas Children’s Hospital Center for Vaccine Development, Houston, TX 77030, USA; (S.R.T.); (M.J.V.); (U.S.); (M.E.B.); (P.J.H.)
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Maria Elena Bottazzi
- Texas Children’s Hospital Center for Vaccine Development, Houston, TX 77030, USA; (S.R.T.); (M.J.V.); (U.S.); (M.E.B.); (P.J.H.)
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA;
| | - Peter J. Hotez
- Texas Children’s Hospital Center for Vaccine Development, Houston, TX 77030, USA; (S.R.T.); (M.J.V.); (U.S.); (M.E.B.); (P.J.H.)
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA;
| | - Jeroen Pollet
- Texas Children’s Hospital Center for Vaccine Development, Houston, TX 77030, USA; (S.R.T.); (M.J.V.); (U.S.); (M.E.B.); (P.J.H.)
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA;
| |
Collapse
|
4
|
Eliadis P, Mais A, Papazisis A, Loxa EK, Dimitriadis A, Sarrigeorgiou I, Backovic M, Agallou M, Zouridakis M, Karagouni E, Lazaridis K, Mamalaki A, Lymberi P. Novel Competitive ELISA Utilizing Trimeric Spike Protein of SARS-CoV-2, Could Identify More Than RBD-RBM Specific Neutralizing Antibodies in Hybrid Sera. Vaccines (Basel) 2024; 12:914. [PMID: 39204038 PMCID: PMC11359269 DOI: 10.3390/vaccines12080914] [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: 06/17/2024] [Revised: 08/05/2024] [Accepted: 08/06/2024] [Indexed: 09/03/2024] Open
Abstract
Since the initiation of the COVID-19 pandemic, there has been a need for the development of diagnostic methods to determine the factors implicated in mounting an immune response against the virus. The most promising indicator has been suggested to be neutralizing antibodies (nAbs), which mainly block the interaction between the Spike protein (S) of SARS-CoV-2 and the host entry receptor ACE2. In this study, we aimed to develop and optimize conditions of a competitive ELISA to measure serum neutralizing titer, using a recombinant trimeric Spike protein modified to have six additional proline residues (S(6P)-HexaPro) and h-ACE2. The results of our surrogate Virus Neutralizing Assay (sVNA) were compared against the commercial sVNT (cPass, Nanjing GenScript Biotech Co., Nanjing City, China), using serially diluted sera from vaccinees, and a high correlation of ID50-90 titer values was observed between the two assays. Interestingly, when we tested and compared the neutralizing activity of sera from eleven fully vaccinated individuals who subsequently contracted COVID-19 (hybrid sera), we recorded a moderate correlation between the two assays, while higher sera neutralizing titers were measured with sVNA. Our data indicated that the sVNA, as a more biologically relevant model assay that paired the trimeric S(6P) with ACE2, instead of the isolated RBD-ACE2 pairing cPass test, could identify nAbs other than the RBD-RBM specific ones.
Collapse
Affiliation(s)
- Petros Eliadis
- Immunology Laboratory, Immunology Department, Hellenic Pasteur Institute, 11521 Athens, Greece; (A.P.); (E.K.L.); (I.S.); (K.L.)
- Biotechnology Unit, Hellenic Pasteur Institute, 11521 Athens, Greece; (A.D.); (A.M.)
| | - Annie Mais
- Laboratory of Molecular Biology and Immunobiotechnology, Immunology Department, Hellenic Pasteur Institute, 11521 Athens, Greece;
| | - Alexandros Papazisis
- Immunology Laboratory, Immunology Department, Hellenic Pasteur Institute, 11521 Athens, Greece; (A.P.); (E.K.L.); (I.S.); (K.L.)
| | - Eleni K. Loxa
- Immunology Laboratory, Immunology Department, Hellenic Pasteur Institute, 11521 Athens, Greece; (A.P.); (E.K.L.); (I.S.); (K.L.)
| | - Alexios Dimitriadis
- Biotechnology Unit, Hellenic Pasteur Institute, 11521 Athens, Greece; (A.D.); (A.M.)
| | - Ioannis Sarrigeorgiou
- Immunology Laboratory, Immunology Department, Hellenic Pasteur Institute, 11521 Athens, Greece; (A.P.); (E.K.L.); (I.S.); (K.L.)
| | - Marija Backovic
- Institut Pasteur, Unité de Virologie Structurale, Université Paris Cité, CNRS-UMR3569, 75724 Paris, France;
| | - Maria Agallou
- Immunology of Infection Laboratory, Microbiology Department, Hellenic Pasteur Institute, 11521 Athens, Greece; (M.A.); (E.K.)
| | - Marios Zouridakis
- Structural Neurobiology Research Group, Laboratory of Molecular Neurobiology and Immunology, Department of Neurobiology, Hellenic Pasteur Institute, 11521 Athens, Greece;
| | - Evdokia Karagouni
- Immunology of Infection Laboratory, Microbiology Department, Hellenic Pasteur Institute, 11521 Athens, Greece; (M.A.); (E.K.)
| | - Konstantinos Lazaridis
- Immunology Laboratory, Immunology Department, Hellenic Pasteur Institute, 11521 Athens, Greece; (A.P.); (E.K.L.); (I.S.); (K.L.)
- Biotechnology Unit, Hellenic Pasteur Institute, 11521 Athens, Greece; (A.D.); (A.M.)
| | - Avgi Mamalaki
- Biotechnology Unit, Hellenic Pasteur Institute, 11521 Athens, Greece; (A.D.); (A.M.)
- Laboratory of Molecular Biology and Immunobiotechnology, Immunology Department, Hellenic Pasteur Institute, 11521 Athens, Greece;
| | - Peggy Lymberi
- Immunology Laboratory, Immunology Department, Hellenic Pasteur Institute, 11521 Athens, Greece; (A.P.); (E.K.L.); (I.S.); (K.L.)
| |
Collapse
|
5
|
Duan H, Zhang E, Ren G, Cheng Y, Yang B, Liu L, Jolicoeur N, Hu H, Xu Y, Liu B. Exploring immune evasion of SARS-CoV-2 variants using a pseudotyped system. Heliyon 2024; 10:e29939. [PMID: 38699727 PMCID: PMC11063423 DOI: 10.1016/j.heliyon.2024.e29939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 03/18/2024] [Accepted: 04/17/2024] [Indexed: 05/05/2024] Open
Abstract
In the United States, coronavirus disease 2019 (COVID-19) cases have consistently been linked to the prevailing variant XBB.1.5 of SARS-CoV-2 since late 2022. A system has been developed for producing and infecting cells with a pseudovirus (PsV) of SARS-CoV-2 to investigate the infection in a Biosafety Level 2 (BSL-2) laboratory. This system utilizes a lentiviral vector carrying ZsGreen1 and Firefly luciferase (Fluc) dual reporter genes, facilitating the analysis of experimental results. In addition, we have created a panel of PsV variants that depict both previous and presently circulating mutations found in circulating SARS-CoV-2 strains. A series of PsVs includes the prototype SARS-CoV-2, Delta B.1.617.2, BA.5, XBB.1, and XBB.1.5. To facilitate the study of infections caused by different variants of SARS-CoV-2 PsV, we have developed a HEK-293T cell line expressing mCherry and human angiotensin converting enzyme 2 (ACE2). To validate whether different SARS-CoV-2 PsV variants can be used for neutralization assays, we employed serum from rats immunized with the PF-D-Trimer protein vaccine to investigate its inhibitory effect on the infectivity of various SARS-CoV-2 PsV variants. According to our observations, the XBB variant, particularly XBB.1.5, exhibits stronger immune evasion capabilities than the prototype SARS-CoV-2, Delta B.1.617.2, and BA.5 PsV variants. Hence, utilizing the neutralization test, this study has the capability to forecast the effectiveness in preventing future SARS-CoV-2 variants infections.
Collapse
Affiliation(s)
- Haixiao Duan
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, College of Bioengineering, Hubei University of Technology, Wuhan, China
| | - Ershuai Zhang
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, College of Bioengineering, Hubei University of Technology, Wuhan, China
| | - Ge Ren
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, College of Bioengineering, Hubei University of Technology, Wuhan, China
| | - Yining Cheng
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, College of Bioengineering, Hubei University of Technology, Wuhan, China
| | - Binfeng Yang
- Wuhan Binhui Biopharmaceutical Co., Ltd., Wuhan, China
| | - Lirong Liu
- Wuhan Binhui Biopharmaceutical Co., Ltd., Wuhan, China
| | | | - Han Hu
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, College of Bioengineering, Hubei University of Technology, Wuhan, China
| | - Yan Xu
- Wuhan Binhui Biopharmaceutical Co., Ltd., Wuhan, China
| | - Binlei Liu
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, College of Bioengineering, Hubei University of Technology, Wuhan, China
- Wuhan Binhui Biopharmaceutical Co., Ltd., Wuhan, China
| |
Collapse
|
6
|
Lobaina Y, Chen R, Suzarte E, Ai P, Huerta V, Musacchio A, Silva R, Tan C, Martín A, Lazo L, Guillén-Nieto G, Yang K, Perera Y, Hermida L. The Nucleocapsid Protein of SARS-CoV-2, Combined with ODN-39M, Is a Potential Component for an Intranasal Bivalent Vaccine with Broader Functionality. Viruses 2024; 16:418. [PMID: 38543783 PMCID: PMC10976088 DOI: 10.3390/v16030418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 02/29/2024] [Accepted: 03/01/2024] [Indexed: 05/23/2024] Open
Abstract
Despite the rapid development of vaccines against COVID-19, they have important limitations, such as safety issues, the scope of their efficacy, and the induction of mucosal immunity. The present study proposes a potential component for a new generation of vaccines. The recombinant nucleocapsid (N) protein from the SARS-CoV-2 Delta variant was combined with the ODN-39M, a synthetic 39 mer unmethylated cytosine-phosphate-guanine oligodeoxynucleotide (CpG ODN), used as an adjuvant. The evaluation of its immunogenicity in Balb/C mice revealed that only administration by intranasal route induced a systemic cross-reactive, cell-mediated immunity (CMI). In turn, this combination was able to induce anti-N IgA in the lungs, which, along with the specific IgG in sera and CMI in the spleen, was cross-reactive against the nucleocapsid protein of SARS-CoV-1. Furthermore, the nasal administration of the N + ODN-39M preparation, combined with RBD Delta protein, enhanced the local and systemic immune response against RBD, with a neutralizing capacity. Results make the N + ODN-39M preparation a suitable component for a future intranasal vaccine with broader functionality against Sarbecoviruses.
Collapse
Affiliation(s)
- Yadira Lobaina
- Research Department, China-Cuba Biotechnology Joint Innovation Center (CCBJIC), Lengshuitan District, Yongzhou 425000, China; (Y.L.); (R.C.); (P.A.); (V.H.); (A.M.); (R.S.); (C.T.)
- R&D Department, Yongzhou Zhong Gu Biotechnology Co., Ltd., Yangjiaqiao Street, Lengshuitan District, Yongzhou 425000, China
| | - Rong Chen
- Research Department, China-Cuba Biotechnology Joint Innovation Center (CCBJIC), Lengshuitan District, Yongzhou 425000, China; (Y.L.); (R.C.); (P.A.); (V.H.); (A.M.); (R.S.); (C.T.)
- Yongzhou Development and Construction Investment Co., Ltd. (YDCI), Changfeng Industry Park, Yongzhou Economic and Technological Development Zone, No. 1 Liebao Road, Lengshuitan District, Yongzhou 425000, China
| | - Edith Suzarte
- CIGB: Research Department, Center for Genetic Engineering and Biotechnology, Havana 10600, Cuba; (E.S.); (A.M.); (L.L.); (G.G.-N.)
| | - Panchao Ai
- Research Department, China-Cuba Biotechnology Joint Innovation Center (CCBJIC), Lengshuitan District, Yongzhou 425000, China; (Y.L.); (R.C.); (P.A.); (V.H.); (A.M.); (R.S.); (C.T.)
- Yongzhou Development and Construction Investment Co., Ltd. (YDCI), Changfeng Industry Park, Yongzhou Economic and Technological Development Zone, No. 1 Liebao Road, Lengshuitan District, Yongzhou 425000, China
| | - Vivian Huerta
- Research Department, China-Cuba Biotechnology Joint Innovation Center (CCBJIC), Lengshuitan District, Yongzhou 425000, China; (Y.L.); (R.C.); (P.A.); (V.H.); (A.M.); (R.S.); (C.T.)
- CIGB: Research Department, Center for Genetic Engineering and Biotechnology, Havana 10600, Cuba; (E.S.); (A.M.); (L.L.); (G.G.-N.)
| | - Alexis Musacchio
- Research Department, China-Cuba Biotechnology Joint Innovation Center (CCBJIC), Lengshuitan District, Yongzhou 425000, China; (Y.L.); (R.C.); (P.A.); (V.H.); (A.M.); (R.S.); (C.T.)
- CIGB: Research Department, Center for Genetic Engineering and Biotechnology, Havana 10600, Cuba; (E.S.); (A.M.); (L.L.); (G.G.-N.)
| | - Ricardo Silva
- Research Department, China-Cuba Biotechnology Joint Innovation Center (CCBJIC), Lengshuitan District, Yongzhou 425000, China; (Y.L.); (R.C.); (P.A.); (V.H.); (A.M.); (R.S.); (C.T.)
- BCF: R&D Section, Representative Office BCF in China, Jingtai Tower, No. 24 Jianguomen Wai Street, Chaoyang District, Beijing 100022, China
| | - Changyuan Tan
- Research Department, China-Cuba Biotechnology Joint Innovation Center (CCBJIC), Lengshuitan District, Yongzhou 425000, China; (Y.L.); (R.C.); (P.A.); (V.H.); (A.M.); (R.S.); (C.T.)
- Yongzhou Development and Construction Investment Co., Ltd. (YDCI), Changfeng Industry Park, Yongzhou Economic and Technological Development Zone, No. 1 Liebao Road, Lengshuitan District, Yongzhou 425000, China
| | - Alejandro Martín
- CIGB: Research Department, Center for Genetic Engineering and Biotechnology, Havana 10600, Cuba; (E.S.); (A.M.); (L.L.); (G.G.-N.)
| | - Laura Lazo
- CIGB: Research Department, Center for Genetic Engineering and Biotechnology, Havana 10600, Cuba; (E.S.); (A.M.); (L.L.); (G.G.-N.)
| | - Gerardo Guillén-Nieto
- CIGB: Research Department, Center for Genetic Engineering and Biotechnology, Havana 10600, Cuba; (E.S.); (A.M.); (L.L.); (G.G.-N.)
| | - Ke Yang
- Research Department, China-Cuba Biotechnology Joint Innovation Center (CCBJIC), Lengshuitan District, Yongzhou 425000, China; (Y.L.); (R.C.); (P.A.); (V.H.); (A.M.); (R.S.); (C.T.)
- Yongzhou Development and Construction Investment Co., Ltd. (YDCI), Changfeng Industry Park, Yongzhou Economic and Technological Development Zone, No. 1 Liebao Road, Lengshuitan District, Yongzhou 425000, China
| | - Yasser Perera
- Research Department, China-Cuba Biotechnology Joint Innovation Center (CCBJIC), Lengshuitan District, Yongzhou 425000, China; (Y.L.); (R.C.); (P.A.); (V.H.); (A.M.); (R.S.); (C.T.)
- R&D Department, Yongzhou Zhong Gu Biotechnology Co., Ltd., Yangjiaqiao Street, Lengshuitan District, Yongzhou 425000, China
- CIGB: Research Department, Center for Genetic Engineering and Biotechnology, Havana 10600, Cuba; (E.S.); (A.M.); (L.L.); (G.G.-N.)
| | - Lisset Hermida
- Research Department, China-Cuba Biotechnology Joint Innovation Center (CCBJIC), Lengshuitan District, Yongzhou 425000, China; (Y.L.); (R.C.); (P.A.); (V.H.); (A.M.); (R.S.); (C.T.)
- Yongzhou Development and Construction Investment Co., Ltd. (YDCI), Changfeng Industry Park, Yongzhou Economic and Technological Development Zone, No. 1 Liebao Road, Lengshuitan District, Yongzhou 425000, China
- BCF: R&D Section, Representative Office BCF in China, Jingtai Tower, No. 24 Jianguomen Wai Street, Chaoyang District, Beijing 100022, China
| |
Collapse
|
7
|
Zhang D, Kukkar D, Kim KH, Bhatt P. A comprehensive review on immunogen and immune-response proteins of SARS-CoV-2 and their applications in prevention, diagnosis, and treatment of COVID-19. Int J Biol Macromol 2024; 259:129284. [PMID: 38211928 DOI: 10.1016/j.ijbiomac.2024.129284] [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: 09/06/2023] [Revised: 01/03/2024] [Accepted: 01/04/2024] [Indexed: 01/13/2024]
Abstract
Exposure to severe acute respiratory syndrome-corona virus-2 (SARS-CoV-2) prompts humoral immune responses in the human body. As the auxiliary diagnosis of a current infection, the existence of viral proteins can be checked from specific antibodies (Abs) induced by immunogenic viral proteins. For people with a weakened immune system, Ab treatment can help neutralize viral antigens to resist and treat the disease. On the other hand, highly immunogenic viral proteins can serve as effective markers for detecting prior infections. Additionally, the identification of viral particles or the presence of antibodies may help establish an immune defense against the virus. These immunogenic proteins rather than SARS-CoV-2 can be given to uninfected people as a vaccination to improve their coping ability against COVID-19 through the generation of memory plasma cells. In this work, we review immunogenic and immune-response proteins derived from SARS-CoV-2 with regard to their classification, origin, and diverse applications (e.g., prevention (vaccine development), diagnostic testing, and treatment (via neutralizing Abs)). Finally, advanced immunization strategies against COVID-19 are discussed along with the contemporary circumstances and future challenges.
Collapse
Affiliation(s)
- Daohong Zhang
- College of Food Engineering, Ludong University, Yantai 264025, Shandong, China; Bio-Nanotechnology Research Institute, Ludong University, Yantai 264025, Shandong, China
| | - Deepak Kukkar
- Department of Biotechnology, Chandigarh University, Gharuan, Mohali 140413, Punjab, India; University Center for Research and Development, Chandigarh University, Gharuan, Mohali 140413, Punjab, India
| | - Ki-Hyun Kim
- Department of Civil & Environmental Engineering, Hanyang University, 222 Wangsimni-Ro, Seoul 04763, Republic of Korea.
| | - Poornima Bhatt
- Department of Biotechnology, Chandigarh University, Gharuan, Mohali 140413, Punjab, India; University Center for Research and Development, Chandigarh University, Gharuan, Mohali 140413, Punjab, India
| |
Collapse
|
8
|
Ismail S, Unger S, Budylowski P, Poutanen S, Yau Y, Jenkins C, Anwer S, Christie-Holmes N, Kiss A, Mazzulli T, Johnstone J, McGeer A, Whittle W, Parvez B, Gray-Owen SD, Stone D, O'Connor DL. SARS-CoV-2 antibodies and their neutralizing capacity against live virus in human milk after COVID-19 infection and vaccination: prospective cohort studies. Am J Clin Nutr 2024; 119:485-495. [PMID: 38309831 DOI: 10.1016/j.ajcnut.2023.10.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 09/29/2023] [Accepted: 10/02/2023] [Indexed: 02/05/2024] Open
Abstract
BACKGROUND There is limited understanding of the impact of coronavirus disease 2019 (COVID-19) infection and vaccination type and interval on severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) human milk antibodies and their neutralizing capacity. OBJECTIVES These cohort studies aimed to determine the presence of antibodies and live virus neutralizing capacity in milk from females infected with COVID-19, unexposed milk bank donors, and vaccinated females and examine impacts of vaccine interval and type. METHODS Milk was collected from participants infected with COVID-19 during pregnancy or lactation (Cohort-1) and milk bank donors (Cohort-2) from March 2020-July 2021 at 3 sequential 4-wk intervals and COVID-19 vaccinated participants with varying dose intervals (Cohort-3) (January-October 2021). Cohort-1 and Cohort-3 were recruited from Sinai Health (patients) and through social media. Cohort-2 included Ontario Milk Bank donors. Milk was examined for SARS-CoV-2 antibodies and live virus neutralization. RESULTS Of females with COVID-19, 53% (Cohort-1, n = 55) had anti-SARS-CoV-2 IgA antibodies in ≥1 milk sample. IgA+ samples (40%) were more likely neutralizing than IgA- samples (odds ratio [OR]: 2.18; 95% confidence interval [CI]: 1.03, 4.60; P = 0.04); however, 25% of IgA- samples were neutralizing. Both IgA positivity and neutralization decreased ∼6 mo after symptom onset (0-100 compared with 201+ d: IgA OR: 14.30; 95% CI: 1.08, 189.89; P = 0.04; neutralizing OR: 4.30; 95% CI: 1.55, 11.89; P = 0.005). Among milk bank donors (Cohort-2, n = 373), 4.3% had IgA antibodies; 23% of IgA+ samples were neutralizing. Vaccination (Cohort-3, n = 60) with mRNA-1273 and shorter vaccine intervals (3 to <6 wk) resulted in higher IgA and IgG than BNT162b2 (P < 0.04) and longer intervals (6 to <16 wk) (P≤0.02), respectively. Neutralizing capacity increased postvaccination (P = 0.04) but was not associated with antibody positivity. CONCLUSIONS SARS-CoV-2 infection and vaccination (type and interval) impacted milk antibodies; however, antibody presence did not consistently predict live virus neutralization. Although human milk is unequivocally the best way to nourish infants, guidance on protection to infants following maternal infection/vaccination may require more nuanced messaging. This study was registered at clinicaltrials.gov as NCT04453969 and NCT04453982.
Collapse
Affiliation(s)
- Samantha Ismail
- Department of Nutritional Sciences, University of Toronto, Toronto, Canada
| | - Sharon Unger
- Department of Nutritional Sciences, University of Toronto, Toronto, Canada; Rogers Hixon Ontario Human Milk Bank, Sinai Health System, Toronto, Canada; Paediatrics, Sinai Health System, Toronto, Canada
| | - Patrick Budylowski
- Combined Containment Level 3 Unit, University of Toronto, Toronto, Canada; Institute of Medical Sciences, University of Toronto, Toronto, Canada
| | - Susan Poutanen
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada; Department of Microbiology, Sinai Health System/University Health Network, Toronto, Canada
| | - Yvonne Yau
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada; The Hospital for Sick Children Research Institute, Toronto, Canada; Department of Paediatric Laboratory Medicine, The Hospital for Sick Children, Toronto, Canada
| | - Carleigh Jenkins
- Department of Nutritional Sciences, University of Toronto, Toronto, Canada; Rogers Hixon Ontario Human Milk Bank, Sinai Health System, Toronto, Canada
| | - Shaista Anwer
- Department of Microbiology, Sinai Health System/University Health Network, Toronto, Canada
| | | | - Alex Kiss
- Evaluative Clinical Sciences, Sunnybrook Research Institute, Toronto, Canada; Institute of Health Policy Management and Evaluation, University of Toronto, Toronto, Canada
| | - Tony Mazzulli
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada; Department of Microbiology, Sinai Health System/University Health Network, Toronto, Canada
| | - Jennie Johnstone
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada; Department of Microbiology, Sinai Health System/University Health Network, Toronto, Canada
| | - Allison McGeer
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada; Department of Microbiology, Sinai Health System/University Health Network, Toronto, Canada
| | - Wendy Whittle
- Obstetrics and Gynecology, Sinai Health System, Toronto, Canada
| | | | - Scott D Gray-Owen
- Combined Containment Level 3 Unit, University of Toronto, Toronto, Canada; Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - Debbie Stone
- Rogers Hixon Ontario Human Milk Bank, Sinai Health System, Toronto, Canada
| | - Deborah L O'Connor
- Department of Nutritional Sciences, University of Toronto, Toronto, Canada; Rogers Hixon Ontario Human Milk Bank, Sinai Health System, Toronto, Canada; Paediatrics, Sinai Health System, Toronto, Canada; The Hospital for Sick Children Research Institute, Toronto, Canada.
| |
Collapse
|
9
|
Izac JR, Kwee EJ, Gaigalas A, Wang L. Quantitative and Standardized Pseudovirus Neutralization Assay for COVID-19. Methods Mol Biol 2024; 2779:259-271. [PMID: 38526789 DOI: 10.1007/978-1-0716-3738-8_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/27/2024]
Abstract
COVID-19 is a global pandemic caused by the highly infectious SARS-CoV-2 virus. Efforts to combat SARS-CoV-2 infection include mass vaccination and development of monoclonal and convalescent plasma therapeutics that require precise measurements of correlative, functional neutralizing antibodies that prevent virus infection. Developing rapid, safe, easy-to-use, and high-quality neutralization assays are essential for the success of the massive effort. Here, we developed a vesicular stomatitis virus-based neutralization assay that was capable of quantifying varying degrees of neutralization in patient serum samples. This assay has two detection readouts, flow cytometry and live cell imaging. The two readout methods produced consistent values of all 50% neutralization titers, further enhancing measurement confidence on the assay. Moreover, the use of available reference standards such as the World Health Organization International Standard (NIBSC code 20/136) enables quantification and standardization of the pseudovirus neutralization assay with neutralizing antibody titers measured in International Units/mL. Quantitative and standardized neutralization assays are critical for reliable efficacy evaluation and comparison of numerous vaccines and therapeutics.
Collapse
Affiliation(s)
- Jerilyn R Izac
- Biosystem and Biomaterials Division, National Institute of Standards and Technology (NIST), Gaithersburg, MD, USA
| | - Edward J Kwee
- Biosystem and Biomaterials Division, National Institute of Standards and Technology (NIST), Gaithersburg, MD, USA
| | - Adolfas Gaigalas
- Biosystem and Biomaterials Division, National Institute of Standards and Technology (NIST), Gaithersburg, MD, USA
| | - Lili Wang
- Biosystem and Biomaterials Division, National Institute of Standards and Technology (NIST), Gaithersburg, MD, USA.
| |
Collapse
|
10
|
Holdenrieder S, Dos Santos Ferreira CE, Izopet J, Theel ES, Wieser A. Clinical and laboratory considerations: determining an antibody-based composite correlate of risk for reinfection with SARS-CoV-2 or severe COVID-19. Front Public Health 2023; 11:1290402. [PMID: 38222091 PMCID: PMC10788057 DOI: 10.3389/fpubh.2023.1290402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 11/30/2023] [Indexed: 01/16/2024] Open
Abstract
Much of the global population now has some level of adaptive immunity to SARS-CoV-2 induced by exposure to the virus (natural infection), vaccination, or a combination of both (hybrid immunity). Key questions that subsequently arise relate to the duration and the level of protection an individual might expect based on their infection and vaccination history. A multi-component composite correlate of risk (CoR) could inform individuals and stakeholders about protection and aid decision making. This perspective evaluates the various elements that need to be accommodated in the development of an antibody-based composite CoR for reinfection with SARS-CoV-2 or development of severe COVID-19, including variation in exposure dose, transmission route, viral genetic variation, patient factors, and vaccination status. We provide an overview of antibody dynamics to aid exploration of the specifics of SARS-CoV-2 antibody testing. We further discuss anti-SARS-CoV-2 immunoassays, sample matrices, testing formats, frequency of sampling and the optimal time point for such sampling. While the development of a composite CoR is challenging, we provide our recommendations for each of these key areas and highlight areas that require further work to be undertaken.
Collapse
Affiliation(s)
- Stefan Holdenrieder
- Institute of Laboratory Medicine, German Heart Centre Munich, Technical University Munich, Munich, Germany
| | | | - Jacques Izopet
- Laboratory of Virology, Toulouse University Hospital and INFINITY Toulouse Institute for Infections and Inflammatory Diseases, INSERM UMR 1291 CNRS UMR 5051, University Toulouse III, Toulouse, France
| | - Elitza S. Theel
- Division of Clinical Microbiology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States
| | - Andreas Wieser
- Division of Infectious Diseases and Tropical Medicine, University Hospital, LMU Munich, Munich, Germany
- German Centre for Infection Research (DZIF), Munich, Germany
- Faculty of Medicine, Max Von Pettenkofer Institute, LMU Munich, Munich, Germany
- Immunology, Infection and Pandemic Research, Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Munich, Germany
| |
Collapse
|
11
|
Tiara MR, Djauhari H, Rachman FR, Rettob AC, Utami D, Pulungan FCS, Purwanta H, Wisaksana R, Alisjahbana B, Indrati AR. Performance of a Point-of-Care Fluorescence Immunoassay Test to Measure the Anti-Severe Acute Respiratory Syndrome Corona Virus 2 Spike, Receptor Binding Domain Antibody Level. Diagnostics (Basel) 2023; 13:3686. [PMID: 38132270 PMCID: PMC10743294 DOI: 10.3390/diagnostics13243686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 11/01/2023] [Accepted: 12/14/2023] [Indexed: 12/23/2023] Open
Abstract
Quantitative determination of anti-SARS-CoV2-S-RBD is necessary for the evaluation of vaccination effectiveness. The surrogate viral neutralization test (SVNT) is approved for measuring anti-SARS-CoV2-S-RBD, but a point-of-care platform is needed to simplify anti-SARS-CoV-2-S-RBD measurement. We aimed to evaluate the performance of a rapid fluorescent immunoassay-based kit, FastBio-RBDTM, compared to the SVNT. During April-September 2021, we enrolled two groups of subjects, convalescent subjects and subjects without a COVID-19 history. The subjects were tested for the anti-SARS-CoV2-S-RBD antibody using FastBio-RBDTM and the GenScript-cPASSTM SVNT. We measured the correlation coefficient and conducted an ROC analysis to determine the best cut-off value of anti-SARS-CoV2-S-RBD against the SVNT percent inhibition levels of 30% and 60%. We included 109 subjects. Anti-SARS-CoV-2-S-RBD strongly correlated to SVNT % inhibition with an R value of 0.866 (p < 0.0001). The ROC analysis showed that the anti-SARS-CoV-2-S-RBD of 6.71 AU/mL had 95.7% sensitivity and 87.5% specificity to detect a percentage inhibition of 30%. The anti-SARS-CoV-2-S-RBD of 59.76 AU/mL had a sensitivity of 88.1% and specificity of 97.0% to detect a percentage inhibition of 60%. FastBio-RBDTM could determine the presence and level of anti-SARS-CoV-2-S-RBD with good sensitivity and specificity. It has the potential to be deployed in health facilities with limited resources.
Collapse
Affiliation(s)
- Marita Restie Tiara
- Research Center for Care and Control of Infectious Disease, Universitas Padjadjaran, Bandung 40161, West Java, Indonesia
| | - Hofiya Djauhari
- Research Center for Care and Control of Infectious Disease, Universitas Padjadjaran, Bandung 40161, West Java, Indonesia
| | - Febi Ramdhani Rachman
- Research Center for Care and Control of Infectious Disease, Universitas Padjadjaran, Bandung 40161, West Java, Indonesia
| | - Antonius Christianus Rettob
- Research Center for Care and Control of Infectious Disease, Universitas Padjadjaran, Bandung 40161, West Java, Indonesia
| | - Darmastuti Utami
- Research Center for Care and Control of Infectious Disease, Universitas Padjadjaran, Bandung 40161, West Java, Indonesia
| | - Fahda Cintia Suci Pulungan
- Research Center for Care and Control of Infectious Disease, Universitas Padjadjaran, Bandung 40161, West Java, Indonesia
| | - Heru Purwanta
- Research Center for Agroindustry–National Research and Innovation Agency (BRIN), Jakarta 16911, DKI Jakarta, Indonesia
| | - Rudi Wisaksana
- Research Center for Care and Control of Infectious Disease, Universitas Padjadjaran, Bandung 40161, West Java, Indonesia
- Department of Internal Medicine, Hasan Sadikin Hospital, Faculty of Medicine, Universitas Padjadjaran, Bandung 40161, West Java, Indonesia
| | - Bachti Alisjahbana
- Research Center for Care and Control of Infectious Disease, Universitas Padjadjaran, Bandung 40161, West Java, Indonesia
- Department of Internal Medicine, Hasan Sadikin Hospital, Faculty of Medicine, Universitas Padjadjaran, Bandung 40161, West Java, Indonesia
| | - Agnes Rengga Indrati
- Research Center for Care and Control of Infectious Disease, Universitas Padjadjaran, Bandung 40161, West Java, Indonesia
- Department of Clinical Pathology, Hasan Sadikin Hospital, Faculty of Medicine, Universitas Padjadjaran, Bandung 40161, West Java, Indonesia
| |
Collapse
|
12
|
Zak AJ, Hoang T, Yee CM, Rizvi SM, Prabhu P, Wen F. Pseudotyping Improves the Yield of Functional SARS-CoV-2 Virus-like Particles (VLPs) as Tools for Vaccine and Therapeutic Development. Int J Mol Sci 2023; 24:14622. [PMID: 37834067 PMCID: PMC10572262 DOI: 10.3390/ijms241914622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 09/08/2023] [Accepted: 09/16/2023] [Indexed: 10/15/2023] Open
Abstract
Virus-like particles (VLPs) have been proposed as an attractive tool in SARS-CoV-2 vaccine development, both as (1) a vaccine candidate with high immunogenicity and low reactogenicity and (2) a substitute for live virus in functional and neutralization assays. Though multiple SARS-CoV-2 VLP designs have already been explored in Sf9 insect cells, a key parameter ensuring VLPs are a viable platform is the VLP spike yield (i.e., spike protein content in VLP), which has largely been unreported. In this study, we show that the common strategy of producing SARS-CoV-2 VLPs by expressing spike protein in combination with the native coronavirus membrane and/or envelope protein forms VLPs, but at a critically low spike yield (~0.04-0.08 mg/L). In contrast, fusing the spike ectodomain to the influenza HA transmembrane domain and cytoplasmic tail and co-expressing M1 increased VLP spike yield to ~0.4 mg/L. More importantly, this increased yield translated to a greater VLP spike antigen density (~96 spike monomers/VLP) that more closely resembles that of native SARS-CoV-2 virus (~72-144 Spike monomers/virion). Pseudotyping further allowed for production of functional alpha (B.1.1.7), beta (B.1.351), delta (B.1.617.2), and omicron (B.1.1.529) SARS-CoV-2 VLPs that bound to the target ACE2 receptor. Finally, we demonstrated the utility of pseudotyped VLPs to test neutralizing antibody activity using a simple, acellular ELISA-based assay performed at biosafety level 1 (BSL-1). Taken together, this study highlights the advantage of pseudotyping over native SARS-CoV-2 VLP designs in achieving higher VLP spike yield and demonstrates the usefulness of pseudotyped VLPs as a surrogate for live virus in vaccine and therapeutic development against SARS-CoV-2 variants.
Collapse
Affiliation(s)
| | | | | | | | | | - Fei Wen
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA (P.P.)
| |
Collapse
|
13
|
Roper KJ, Thomas J, Albalawi W, Maddocks E, Dobson S, Alshehri A, Barone FG, Baltazar M, Semple MG, Ho A, Turtle L, Paxton WA, Pollakis G. Quantifying neutralising antibody responses against SARS-CoV-2 in dried blood spots (DBS) and paired sera. Sci Rep 2023; 13:15014. [PMID: 37697014 PMCID: PMC10495436 DOI: 10.1038/s41598-023-41928-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 09/04/2023] [Indexed: 09/13/2023] Open
Abstract
The ongoing SARS-CoV-2 pandemic was initially managed by non-pharmaceutical interventions such as diagnostic testing, isolation of positive cases, physical distancing and lockdowns. The advent of vaccines has provided crucial protection against SARS-CoV-2. Neutralising antibody (nAb) responses are a key correlate of protection, and therefore measuring nAb responses is essential for monitoring vaccine efficacy. Fingerstick dried blood spots (DBS) are ideal for use in large-scale sero-surveillance because they are inexpensive, offer the option of self-collection and can be transported and stored at ambient temperatures. Such advantages also make DBS appealing to use in resource-limited settings and in potential future pandemics. In this study, nAb responses in sera, venous blood and fingerstick blood stored on filter paper were measured. Samples were collected from SARS-CoV-2 acutely infected individuals, SARS-CoV-2 convalescent individuals and SARS-CoV-2 vaccinated individuals. Good agreement was observed between the nAb responses measured in eluted DBS and paired sera. Stability of nAb responses was also observed in sera stored on filter paper at room temperature for 28 days. Overall, this study provides support for the use of filter paper as a viable sample collection method to study nAb responses.
Collapse
Affiliation(s)
- Kelly J Roper
- Department of Clinical Infection, Microbiology and Immunology (CIMI), Institute of Infection, Veterinary and Ecological Sciences (IVES), University of Liverpool, Liverpool, L69 7BE, UK
- Faculty of Health and Life Sciences, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Jordan Thomas
- Department of Clinical Infection, Microbiology and Immunology (CIMI), Institute of Infection, Veterinary and Ecological Sciences (IVES), University of Liverpool, Liverpool, L69 7BE, UK
- Faculty of Health and Life Sciences, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Wejdan Albalawi
- Department of Clinical Infection, Microbiology and Immunology (CIMI), Institute of Infection, Veterinary and Ecological Sciences (IVES), University of Liverpool, Liverpool, L69 7BE, UK
- Faculty of Health and Life Sciences, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Emily Maddocks
- Department of Clinical Infection, Microbiology and Immunology (CIMI), Institute of Infection, Veterinary and Ecological Sciences (IVES), University of Liverpool, Liverpool, L69 7BE, UK
- Faculty of Health and Life Sciences, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Susan Dobson
- Department of Clinical Infection, Microbiology and Immunology (CIMI), Institute of Infection, Veterinary and Ecological Sciences (IVES), University of Liverpool, Liverpool, L69 7BE, UK
- Faculty of Health and Life Sciences, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Abdullateef Alshehri
- Department of Clinical Infection, Microbiology and Immunology (CIMI), Institute of Infection, Veterinary and Ecological Sciences (IVES), University of Liverpool, Liverpool, L69 7BE, UK
- Faculty of Health and Life Sciences, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Francesco G Barone
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology (ISMIB), University of Liverpool, Liverpool, L69 3BX, UK
| | - Murielle Baltazar
- Department of Clinical Infection, Microbiology and Immunology (CIMI), Institute of Infection, Veterinary and Ecological Sciences (IVES), University of Liverpool, Liverpool, L69 7BE, UK
- Faculty of Health and Life Sciences, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Malcolm G Semple
- Department of Clinical Infection, Microbiology and Immunology (CIMI), Institute of Infection, Veterinary and Ecological Sciences (IVES), University of Liverpool, Liverpool, L69 7BE, UK
- Faculty of Health and Life Sciences, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
- Respiratory Medicine, Alder Hey Children's Hospital, Institute in The Park, University of Liverpool, Liverpool, UK
| | - Antonia Ho
- MRC-University of Glasgow Centre for Virus Research, 464 Bearsden Road, Glasgow, G61 1QH, UK
| | - Lance Turtle
- Department of Clinical Infection, Microbiology and Immunology (CIMI), Institute of Infection, Veterinary and Ecological Sciences (IVES), University of Liverpool, Liverpool, L69 7BE, UK
- Faculty of Health and Life Sciences, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - William A Paxton
- Department of Clinical Infection, Microbiology and Immunology (CIMI), Institute of Infection, Veterinary and Ecological Sciences (IVES), University of Liverpool, Liverpool, L69 7BE, UK
- Faculty of Health and Life Sciences, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
- NIHR Health Protection Research Unit in Emerging and Zoonotic Infections, Liverpool, UK
| | - Georgios Pollakis
- Department of Clinical Infection, Microbiology and Immunology (CIMI), Institute of Infection, Veterinary and Ecological Sciences (IVES), University of Liverpool, Liverpool, L69 7BE, UK.
- Faculty of Health and Life Sciences, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK.
- NIHR Health Protection Research Unit in Emerging and Zoonotic Infections, Liverpool, UK.
| |
Collapse
|
14
|
Izac JR, Kwee EJ, Tian L, Elsheikh E, Gaigalas AK, Elliott JT, Wang L. Development of a Cell-Based SARS-CoV-2 Pseudovirus Neutralization Assay Using Imaging and Flow Cytometry Analysis. Int J Mol Sci 2023; 24:12332. [PMID: 37569707 PMCID: PMC10418775 DOI: 10.3390/ijms241512332] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 07/28/2023] [Accepted: 07/31/2023] [Indexed: 08/13/2023] Open
Abstract
COVID-19 is an ongoing, global pandemic caused by the novel, highly infectious SARS-CoV-2 virus. Efforts to mitigate the effects of SARS-CoV-2, such as mass vaccination and development of monoclonal therapeutics, require precise measurements of correlative, functional neutralizing antibodies that block virus infection. The development of rapid, safe, and easy-to-use neutralization assays is essential for faster diagnosis and treatment. Here, we developed a vesicular stomatitis virus (VSV)-based neutralization assay with two readout methods, imaging and flow cytometry, that were capable of quantifying varying degrees of neutralization in patient serum samples. We tested two different spike-pseudoviruses and conducted a time-course assay at multiple multiplicities of infection (MOIs) to optimize the assay workflow. The results of this assay correlate with the results of previously developed serology and surrogate neutralization assays. The two pseudovirus readout methods produced similar values of 50% neutralization titer values. Harvest-free in situ readouts for live-cell imaging and high-throughput analysis results for flow cytometry can provide unique capabilities for fast evaluation of neutralization, which is critical for the mitigation of future pandemics.
Collapse
Affiliation(s)
- Jerilyn R. Izac
- Biosystems and Biomaterials Division, National Institute of Standards and Technology (NIST), Gaithersburg, MD 20899, USA; (L.T.); (E.E.); (A.K.G.); (J.T.E.); (L.W.)
| | - Edward J. Kwee
- Biosystems and Biomaterials Division, National Institute of Standards and Technology (NIST), Gaithersburg, MD 20899, USA; (L.T.); (E.E.); (A.K.G.); (J.T.E.); (L.W.)
| | | | | | | | | | | |
Collapse
|
15
|
Montero S, Urrunaga-Pastor D, Soto-Becerra P, Cvetkovic-Vega A, Guillermo-Roman M, Figueroa-Montes L, Sagástegui AA, Alvizuri-Pastor S, Contreras-Macazana RM, Apolaya-Segura M, Díaz-Vélez C, Maguiña JL. Humoral response after a BNT162b2 heterologous third dose of COVID-19 vaccine following two doses of BBIBP-CorV among healthcare personnel in Peru. Vaccine X 2023; 14:100311. [PMID: 37207103 PMCID: PMC10162476 DOI: 10.1016/j.jvacx.2023.100311] [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: 10/05/2022] [Revised: 04/14/2023] [Accepted: 04/30/2023] [Indexed: 05/21/2023] Open
Abstract
Background The inactivated virus vaccine, BBIBP-CorV, was principally distributed across low- and middle-income countries as primary vaccination strategy to prevent poor COVID-19 outcomes. Limited information is available regarding its effect on heterologous boosting. We aim to evaluate the immunogenicity and reactogenicity of a third booster dose of BNT162b2 following a double BBIBP-CorV regime. Methods We conducted a cross-sectional study among healthcare providers from several healthcare facilities of the Seguro Social de Salud del Perú - ESSALUD. We included participants two-dose BBIBP-CorV vaccinated who presented a three-dose vaccination card at least 21 days passed since the vaccinees received their third dose and were willing to provide written informed consent. Antibodies were determined using LIAISON® SARS-CoV-2 TrimericS IgG (DiaSorin Inc., Stillwater, USA). Factors potentially associated with immunogenicity, and adverse events, were considered. We used a multivariable fractional polynomial modeling approach to estimate the association between anti-SARS-CoV-2 IgG antibodies' geometric mean (GM) ratios and related predictors. Results We included 595 subjects receiving a third dose with a median (IQR) age of 46 [37], [54], from which 40% reported previous SARS-CoV-2 infection. The overall geometric mean (IQR) of anti-SARS-CoV-2 IgG antibodies was 8,410 (5,115 - 13,000) BAU/mL. Prior SARS-CoV-2 history and full/part-time in-person working modality were significantly associated with greater GM. Conversely, time from boosting to IgG measure was associated with lower GM levels. We found 81% of reactogenicity in the study population; younger age and being a nurse were associated with a lower incidence of adverse events. Conclusions Among healthcare providers, a booster dose of BNT162b2 following a full BBIBP-CorV regime provided high humoral immune protection. Thus, SARS-CoV-2 previous exposure and working in person displayed as determinants that increase anti-SARS-CoV-2 IgG antibodies.
Collapse
Affiliation(s)
- Stephanie Montero
- Instituto de Evaluación de Tecnologías en Salud e Investigación - IETSI, ESSALUD, Lima, Peru
| | - Diego Urrunaga-Pastor
- Instituto de Evaluación de Tecnologías en Salud e Investigación - IETSI, ESSALUD, Lima, Peru
- Unidad para la Generación y Síntesis de Evidencias en Salud, Universidad San Ignacio de Loyola (USIL), Lima, Peru
| | - Percy Soto-Becerra
- Instituto de Evaluación de Tecnologías en Salud e Investigación - IETSI, ESSALUD, Lima, Peru
- Universidad Continental, Huancayo, Perú
| | - Aleksandar Cvetkovic-Vega
- Instituto de Evaluación de Tecnologías en Salud e Investigación - IETSI, ESSALUD, Lima, Peru
- Facultad de Medicina Humana, Universidad Privada Antenor Orrego, Trujillo, Peru
| | - Martina Guillermo-Roman
- Instituto de Evaluación de Tecnologías en Salud e Investigación - IETSI, ESSALUD, Lima, Peru
| | | | | | | | | | - Moisés Apolaya-Segura
- Instituto de Evaluación de Tecnologías en Salud e Investigación - IETSI, ESSALUD, Lima, Peru
- Facultad de Medicina Humana, Universidad Privada Antenor Orrego, Trujillo, Peru
| | - Cristian Díaz-Vélez
- Instituto de Evaluación de Tecnologías en Salud e Investigación - IETSI, ESSALUD, Lima, Peru
- Facultad de Medicina Humana, Universidad Privada Antenor Orrego, Trujillo, Peru
| | - Jorge L. Maguiña
- Instituto de Evaluación de Tecnologías en Salud e Investigación - IETSI, ESSALUD, Lima, Peru
- Facultad de Ciencias de la Salud, Universidad Científica del Sur, Lima, Peru
| |
Collapse
|
16
|
Moulana A, Dupic T, Phillips AM, Desai MM. Genotype-phenotype landscapes for immune-pathogen coevolution. Trends Immunol 2023; 44:384-396. [PMID: 37024340 PMCID: PMC10147585 DOI: 10.1016/j.it.2023.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 03/08/2023] [Accepted: 03/09/2023] [Indexed: 04/07/2023]
Abstract
Our immune systems constantly coevolve with the pathogens that challenge them, as pathogens adapt to evade our defense responses, with our immune repertoires shifting in turn. These coevolutionary dynamics take place across a vast and high-dimensional landscape of potential pathogen and immune receptor sequence variants. Mapping the relationship between these genotypes and the phenotypes that determine immune-pathogen interactions is crucial for understanding, predicting, and controlling disease. Here, we review recent developments applying high-throughput methods to create large libraries of immune receptor and pathogen protein sequence variants and measure relevant phenotypes. We describe several approaches that probe different regions of the high-dimensional sequence space and comment on how combinations of these methods may offer novel insight into immune-pathogen coevolution.
Collapse
Affiliation(s)
- Alief Moulana
- Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA 02138, USA
| | - Thomas Dupic
- Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA 02138, USA
| | - Angela M Phillips
- Department of Microbiology and Immunology, University of California at San Francisco, San Francisco, CA 94143, USA
| | - Michael M Desai
- Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA 02138, USA; Department of Physics, Harvard University, Cambridge, MA 02138, USA; NSF-Simons Center for Mathematical and Statistical Analysis of Biology, Harvard University, Cambridge, MA 02138, USA; Quantitative Biology Initiative, Harvard University, Cambridge, MA 02138, USA.
| |
Collapse
|
17
|
Vilca-Alosilla JJ, Candia-Puma MA, Coronel-Monje K, Goyzueta-Mamani LD, Galdino AS, Machado-de-Ávila RA, Giunchetti RC, Ferraz Coelho EA, Chávez-Fumagalli MA. A Systematic Review and Meta-Analysis Comparing the Diagnostic Accuracy Tests of COVID-19. Diagnostics (Basel) 2023; 13:diagnostics13091549. [PMID: 37174941 PMCID: PMC10177430 DOI: 10.3390/diagnostics13091549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 04/06/2023] [Accepted: 04/11/2023] [Indexed: 05/15/2023] Open
Abstract
In this paper, we present a systematic review and meta-analysis that aims to evaluate the reliability of coronavirus disease diagnostic tests in 2019 (COVID-19). This article seeks to describe the scientific discoveries made because of diagnostic tests conducted in recent years during the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic. Between 2020 and 2021, searches for published papers on the COVID-19 diagnostic were made in the PubMed database. Ninety-nine scientific articles that satisfied the requirements were analyzed and included in the meta-analysis, and the specificity and sensitivity of the diagnostic accuracy were assessed. When compared to serological tests such as the enzyme-linked immunosorbent assay (ELISA), chemiluminescence immunoassay (CLIA), lateral flow immunoassay (LFIA), and chemiluminescent microparticle immunoassay (CMIA), molecular tests such as reverse transcription polymerase chain reaction (RT-PCR), reverse transcription loop-mediated isothermal amplification (RT-LAMP), and clustered regularly interspaced short palindromic repeats (CRISPR) performed better in terms of sensitivity and specificity. Additionally, the area under the curve restricted to the false-positive rates (AUCFPR) of 0.984 obtained by the antiviral neutralization bioassay (ANB) diagnostic test revealed significant potential for the identification of COVID-19. It has been established that the various diagnostic tests have been effectively adapted for the detection of SARS-CoV-2; nevertheless, their performance still must be enhanced to contain potential COVID-19 outbreaks, which will also help contain potential infectious agent outbreaks in the future.
Collapse
Affiliation(s)
- Juan Jeferson Vilca-Alosilla
- Computational Biology and Chemistry Research Group, Vicerrectorado de Investigación, Universidad Católica de Santa María, Arequipa 04000, Peru
- Facultad de Ciencias Farmacéuticas, Bioquímicas y Biotecnológicas, Universidad Católica de Santa María, Arequipa 04000, Peru
| | - Mayron Antonio Candia-Puma
- Computational Biology and Chemistry Research Group, Vicerrectorado de Investigación, Universidad Católica de Santa María, Arequipa 04000, Peru
- Facultad de Ciencias Farmacéuticas, Bioquímicas y Biotecnológicas, Universidad Católica de Santa María, Arequipa 04000, Peru
| | - Katiusca Coronel-Monje
- Computational Biology and Chemistry Research Group, Vicerrectorado de Investigación, Universidad Católica de Santa María, Arequipa 04000, Peru
- Facultad de Ciencias Farmacéuticas, Bioquímicas y Biotecnológicas, Universidad Católica de Santa María, Arequipa 04000, Peru
| | - Luis Daniel Goyzueta-Mamani
- Computational Biology and Chemistry Research Group, Vicerrectorado de Investigación, Universidad Católica de Santa María, Arequipa 04000, Peru
- Sustainable Innovative Biomaterials Department, Le Qara Research Center, Arequipa 04000, Peru
| | - Alexsandro Sobreira Galdino
- Laboratório de Biotecnologia de Microrganismos, Universidade Federal São João Del-Rei, Divinópolis 35501-296, MG, Brazil
| | | | - Rodolfo Cordeiro Giunchetti
- Laboratório de Biologia das Interações Celulares, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil
- Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais, INCT-DT, Salvador 40015-970, BA, Brazil
| | - Eduardo Antonio Ferraz Coelho
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil
- Departamento de Patologia Clínica, COLTEC, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil
| | - Miguel Angel Chávez-Fumagalli
- Computational Biology and Chemistry Research Group, Vicerrectorado de Investigación, Universidad Católica de Santa María, Arequipa 04000, Peru
| |
Collapse
|
18
|
Schwarze M, Luo J, Brakel A, Krizsan A, Lakowa N, Grünewald T, Lehmann C, Wolf J, Borte S, Milkovska-Stamenova S, Gabert J, Scholz M, Hoffmann R. Evaluation of S- and M-Proteins Expressed in Escherichia coli and HEK Cells for Serological Detection of Antibodies in Response to SARS-CoV-2 Infections and mRNA-Based Vaccinations. Pathogens 2022; 11:pathogens11121515. [PMID: 36558849 PMCID: PMC9782079 DOI: 10.3390/pathogens11121515] [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: 11/04/2022] [Revised: 11/28/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022] Open
Abstract
This study investigated the IgG and IgA antibody response against recombinant S1 and receptor binding domains (RBD) of the spike (S-) protein and the membrane (M-) protein using a set of 115 serum samples collected from patients infected with SARS-CoV-2 in Germany before April 2021 using protein and peptide ELISA. As S1- and RBD-proteins expressed in Escherichia coli provided poor sensitivities in ELISA, they were replaced by proteins expressed in HEK cells. The RBD-ELISA provided a sensitivity of 90.6% (N = 85) for samples collected from patients with confirmed SARS-CoV-2 infections more than 14 days after symptom onset or a positive PCR test. In population-based controls, the specificity was 97.9% (N = 94). In contrast, the sensitivities were only 41.2% and 72.6% for M- and N-proteins, respectively, while the specificities were 88.5% and 100%, respectively. Considering also 20 samples collected during the first two weeks of symptom onset or PCR confirmation, the sensitivity of RBD- and N-protein ELISA decreased to 82.6% and 72.6%, respectively. The combination of two data sets, i.e., N- and RBD-, N- and M-, or RBD- and M-proteins increased the sensitivity to 85.8%, 77.9%, and 87.8%, respectively. Peptide mapping mostly confirmed epitopes previously reported for S1- and M-proteins, but they were only recognized by a few samples already tested positive in the corresponding protein ELISA indicating that peptide-based assays will not improve the diagnostic sensitivity.
Collapse
Affiliation(s)
- Mandy Schwarze
- Institute of Bioanalytical Chemistry, Faculty of Chemistry and Mineralogy, Universität Leipzig, 04103 Leipzig, Germany
- Center for Biotechnology and Biomedicine, Universität Leipzig, 04103 Leipzig, Germany
| | - Ji Luo
- Institute of Bioanalytical Chemistry, Faculty of Chemistry and Mineralogy, Universität Leipzig, 04103 Leipzig, Germany
- Center for Biotechnology and Biomedicine, Universität Leipzig, 04103 Leipzig, Germany
- Adversis Pharma GmbH, 04103 Leipzig, Germany
| | - Alexandra Brakel
- Institute of Bioanalytical Chemistry, Faculty of Chemistry and Mineralogy, Universität Leipzig, 04103 Leipzig, Germany
- Center for Biotechnology and Biomedicine, Universität Leipzig, 04103 Leipzig, Germany
| | - Andor Krizsan
- Institute of Bioanalytical Chemistry, Faculty of Chemistry and Mineralogy, Universität Leipzig, 04103 Leipzig, Germany
- Center for Biotechnology and Biomedicine, Universität Leipzig, 04103 Leipzig, Germany
| | - Nicole Lakowa
- Klinik für Infektions- und Tropenmedizin, Klinikum Chemnitz gGmbH, 09113 Chemnitz, Germany
| | - Thomas Grünewald
- Klinik für Infektions- und Tropenmedizin, Klinikum Chemnitz gGmbH, 09113 Chemnitz, Germany
| | - Claudia Lehmann
- Laboratory for Transplantation Immunology, Institute for Transfusion Medicine, University Hospital Leipzig, 04103 Leipzig, Germany
| | - Johannes Wolf
- Department of Laboratory Medicine, Hospital St. Georg gGmbH, 04129 Leipzig, Germany
- Immuno Deficiency Center Leipzig, Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiency Diseases, Hospital St. Georg gGmbH, 04129 Leipzig, Germany
| | - Stephan Borte
- Department of Laboratory Medicine, Hospital St. Georg gGmbH, 04129 Leipzig, Germany
- Immuno Deficiency Center Leipzig, Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiency Diseases, Hospital St. Georg gGmbH, 04129 Leipzig, Germany
| | - Sanja Milkovska-Stamenova
- Institute of Bioanalytical Chemistry, Faculty of Chemistry and Mineralogy, Universität Leipzig, 04103 Leipzig, Germany
- Center for Biotechnology and Biomedicine, Universität Leipzig, 04103 Leipzig, Germany
- Adversis Pharma GmbH, 04103 Leipzig, Germany
| | - Jörg Gabert
- Adversis Pharma GmbH, 04103 Leipzig, Germany
| | - Markus Scholz
- Institute for Medical Informatics, Statistics and Epidemiology, Universität Leipzig, 04107 Leipzig, Germany
- LIFE Research Center of Civilization Diseases, Universität Leipzig, 04103 Leipzig, Germany
| | - Ralf Hoffmann
- Institute of Bioanalytical Chemistry, Faculty of Chemistry and Mineralogy, Universität Leipzig, 04103 Leipzig, Germany
- Center for Biotechnology and Biomedicine, Universität Leipzig, 04103 Leipzig, Germany
- Correspondence:
| |
Collapse
|
19
|
Atti A, Insalata F, Carr EJ, Otter AD, Castillo-Olivares J, Wu M, Harvey R, Howell M, Chan A, Lyall J, Temperton N, Cantoni D, da Costa K, Nadesalingam A, Taylor-Kerr A, Hettiarachchi N, Tranquillini C, Hewson J, Cole MJ, Foulkes S, Munro K, Monk EJM, Milligan ID, Linley E, Chand MA, Brown CS, Islam J, Semper A, Charlett A, Heeney JL, Beale R, Zambon M, Hopkins S, Brooks T, Hall V. Antibody correlates of protection from SARS-CoV-2 reinfection prior to vaccination: A nested case-control within the SIREN study. J Infect 2022; 85:545-556. [PMID: 36089104 PMCID: PMC9458758 DOI: 10.1016/j.jinf.2022.09.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 09/05/2022] [Indexed: 12/15/2022]
Abstract
OBJECTIVES To investigate serological differences between SARS-CoV-2 reinfection cases and contemporary controls, to identify antibody correlates of protection against reinfection. METHODS We performed a case-control study, comparing reinfection cases with singly infected individuals pre-vaccination, matched by gender, age, region and timing of first infection. Serum samples were tested for anti-SARS-CoV-2 spike (anti-S), anti-SARS-CoV-2 nucleocapsid (anti-N), live virus microneutralisation (LV-N) and pseudovirus microneutralisation (PV-N). Results were analysed using fixed effect linear regression and fitted into conditional logistic regression models. RESULTS We identified 23 cases and 92 controls. First infections occurred before November 2020; reinfections occurred before February 2021, pre-vaccination. Anti-S levels, LV-N and PV-N titres were significantly lower among cases; no difference was found for anti-N levels. Increasing anti-S levels were associated with reduced risk of reinfection (OR 0·63, CI 0·47-0·85), but no association for anti-N levels (OR 0·88, CI 0·73-1·05). Titres >40 were correlated with protection against reinfection for LV-N Wuhan (OR 0·02, CI 0·001-0·31) and LV-N Alpha (OR 0·07, CI 0·009-0·62). For PV-N, titres >100 were associated with protection against Wuhan (OR 0·14, CI 0·03-0·64) and Alpha (0·06, CI 0·008-0·40). CONCLUSIONS Before vaccination, protection against SARS-CoV-2 reinfection was directly correlated with anti-S levels, PV-N and LV-N titres, but not with anti-N levels. Detectable LV-N titres were sufficient for protection, whilst PV-N titres >100 were required for a protective effect. TRIAL REGISTRATION NUMBER ISRCTN11041050.
Collapse
Affiliation(s)
- Ana Atti
- UK Health Security Agency, Smith Square, London SW1P, UK.
| | | | - Edward J Carr
- The Francis Crick Institute, 1 Midland Rd, London NW1 1AT, UK
| | - Ashley D Otter
- UK Health Security Agency, Porton Down, Salisbury SP4 0JG, UK
| | - Javier Castillo-Olivares
- Laboratory of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Cambridge University, Madingley Road, Cambridge CB3 0ES, UK
| | - Mary Wu
- The Francis Crick Institute, 1 Midland Rd, London NW1 1AT, UK
| | - Ruth Harvey
- The Francis Crick Institute, 1 Midland Rd, London NW1 1AT, UK
| | - Michael Howell
- The Francis Crick Institute, 1 Midland Rd, London NW1 1AT, UK
| | - Andrew Chan
- Laboratory of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Cambridge University, Madingley Road, Cambridge CB3 0ES, UK
| | - Jonathan Lyall
- Laboratory of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Cambridge University, Madingley Road, Cambridge CB3 0ES, UK
| | - Nigel Temperton
- Viral Pseudotype Unit, Medway School of Pharmacy, Universities of Greenwich and Kent at Medway, Central Ave, Gillingham, Chatham ME4 4BF, UK
| | - Diego Cantoni
- Viral Pseudotype Unit, Medway School of Pharmacy, Universities of Greenwich and Kent at Medway, Central Ave, Gillingham, Chatham ME4 4BF, UK
| | - Kelly da Costa
- Viral Pseudotype Unit, Medway School of Pharmacy, Universities of Greenwich and Kent at Medway, Central Ave, Gillingham, Chatham ME4 4BF, UK
| | - Angalee Nadesalingam
- Laboratory of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Cambridge University, Madingley Road, Cambridge CB3 0ES, UK
| | | | | | | | | | | | - Sarah Foulkes
- UK Health Security Agency, Smith Square, London SW1P, UK
| | - Katie Munro
- UK Health Security Agency, Smith Square, London SW1P, UK
| | | | | | - Ezra Linley
- Manchester Royal Infirmary, UK Health Security Agency, Oxford Road, Manchester M139WL, UK
| | - Meera A Chand
- UK Health Security Agency, Smith Square, London SW1P, UK
| | - Colin S Brown
- UK Health Security Agency, Smith Square, London SW1P, UK; The National Institute for Health Research Health Protection Research (NIHR) Unit in Healthcare Associated Infections and Antimicrobial Resistance at the University of Oxford, Old Road Campus, Headington, Oxford OX3 7BN, UK
| | - Jasmin Islam
- UK Health Security Agency, Smith Square, London SW1P, UK
| | - Amanda Semper
- UK Health Security Agency, Smith Square, London SW1P, UK
| | - Andre Charlett
- UK Health Security Agency, Smith Square, London SW1P, UK; NIHR Health Protection Research Unit in Behavioural Science and Evaluation at University of Bristol in partnership with Public Health England, Queens Road, Bristol BS8 1QU, UK; NIHR Health Protection Research Unit in Immunisation at the London School of Hygiene and Tropical Medicine in partnership with Public Health England, Keppel St, London WC1E 7HT, UK
| | | | - Rupert Beale
- The Francis Crick Institute, 1 Midland Rd, London NW1 1AT, UK
| | - Maria Zambon
- UK Health Security Agency, Smith Square, London SW1P, UK
| | - Susan Hopkins
- UK Health Security Agency, Smith Square, London SW1P, UK; The National Institute for Health Research Health Protection Research (NIHR) Unit in Healthcare Associated Infections and Antimicrobial Resistance at the University of Oxford, Old Road Campus, Headington, Oxford OX3 7BN, UK
| | - Tim Brooks
- UK Health Security Agency, Smith Square, London SW1P, UK
| | - Victoria Hall
- UK Health Security Agency, Smith Square, London SW1P, UK; The National Institute for Health Research Health Protection Research (NIHR) Unit in Healthcare Associated Infections and Antimicrobial Resistance at the University of Oxford, Old Road Campus, Headington, Oxford OX3 7BN, UK
| |
Collapse
|
20
|
The Bovine Seminal Plasma Protein PDC-109 Possesses Pan-Antiviral Activity. Viruses 2022; 14:v14092031. [PMID: 36146836 PMCID: PMC9504757 DOI: 10.3390/v14092031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 09/08/2022] [Accepted: 09/08/2022] [Indexed: 11/16/2022] Open
Abstract
Mammalian seminal plasma contains a multitude of bioactive components, including lipids, glucose, mineral elements, metabolites, proteins, cytokines, and growth factors, with various functions during insemination and fertilization. The seminal plasma protein PDC-109 is one of the major soluble components of the bovine ejaculate and is crucially important for sperm motility, capacitation, and acrosome reaction. A hitherto underappreciated function of seminal plasma is its anti-microbial and antiviral activity, which may limit the sexual transmission of infectious diseases during intercourse. We have recently discovered that PDC-109 inhibits the membrane fusion activity of influenza virus particles and significantly impairs viral infections at micromolar concentrations. Here we investigated whether the antiviral activity of PDC-109 is restricted to Influenza or if other mammalian viruses are similarly affected. We focused on Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2), the etiological agent of the Coronavirus Disease 19 (COVID-19), thoroughly assessing PDC-109 inhibition with SARS-CoV-2 Spike (S)-pseudotyped reporter virus particles, but also live-virus infections. Consistent with our previous publications, we found significant virus inhibition, albeit accompanied by substantial cytotoxicity. However, using time-of-addition experiments we discovered a treatment regimen that enables virus suppression without affecting cell viability. We furthermore demonstrated that PDC-109 is also able to impair infections mediated by the VSV glycoprotein (VSVg), thus indicating a broad pan-antiviral activity against multiple virus species and families.
Collapse
|
21
|
Bhiman JN, Moore PL. Leveraging South African HIV research to define SARS-CoV-2 immunity triggered by sequential variants of concern. Immunol Rev 2022; 310:61-75. [PMID: 35599324 PMCID: PMC9349367 DOI: 10.1111/imr.13086] [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] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the virus that causes coronavirus disease 2019 (COVID-19), has shifted our paradigms about B cell immunity and the goals of vaccination for respiratory viruses. The development of population immunity, through responses directed to highly immunogenic regions of this virus, has been a strong driving force in the emergence of progressively mutated variants. This review highlights how the strength of the existing global virology and immunology networks built for HIV vaccine research enabled rapid adaptation of techniques, assays, and skill sets, to expeditiously respond to the SARS-CoV-2 pandemic. Allying real-time genomic surveillance to immunological platforms enabled the characterization of immune responses elicited by infection with distinct variants, in sequential epidemic waves, as well as studies of vaccination and hybrid immunity (combination of infection- and vaccination-induced immunity). These studies have shown that consecutive variants of concern have steadily diminished the ability of vaccines to prevent infection, but that increasing levels of hybrid immunity result in higher frequencies of cross-reactive responses. Ultimately, this rapid pivot from HIV to SARS-CoV-2 enabled a depth of understanding of the SARS-CoV-2 antigenic vulnerabilities as population immunity expanded and diversified, providing key insights for future responses to the SARS-CoV-2 pandemic.
Collapse
Affiliation(s)
- Jinal N Bhiman
- National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg, South Africa
- SAMRC Antibody Immunity Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg, South Africa
| | - Penny L Moore
- National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg, South Africa
- SAMRC Antibody Immunity Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg, South Africa
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
- Centre for the AIDS Programme of Research in South Africa, Durban, South Africa
| |
Collapse
|
22
|
Jug U, Naumoska K, Malovrh T. Japanese Knotweed Rhizome Bark Extract Inhibits Live SARS-CoV-2 In Vitro. Bioengineering (Basel) 2022; 9:bioengineering9090429. [PMID: 36134975 PMCID: PMC9495978 DOI: 10.3390/bioengineering9090429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/12/2022] [Accepted: 08/23/2022] [Indexed: 11/25/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19), a viral infectious respiratory disease, is caused by highly contagious severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and is responsible for the ongoing COVID-19 pandemic. Since very few drugs are known to be effective against SARS-CoV-2, there is a general need for new therapeutics, including plant-based drugs, for the prophylaxis and treatment of infections. In the current study, the activity of a 70% ethanolic(aq) extract of the rhizome bark of Japanese knotweed, an invasive alien plant species, was tested for the first time against the wild-type SARS-CoV-2 virus using a specific and robust virus neutralization test (VNT) on Vero-E6 cells, which best mimics the mechanism of real virus−host interaction. A statistically significant antiviral effect against SARS-CoV-2 (p-value < 0.05) was observed for the 50.8 µg mL−1 extract solution in cell medium. A suitable extract preparation was described to avoid loss of polyphenols throughout filtration of the extract, which was dissolved in cell medium containing fetal bovine serum (FBS). The significance of the differences between the sums of the test and control groups in the incidence of cytopathic effects (CPE) was determined using the one-way ANOVA test. A dose−response relationship was observed, with the cytotoxic effect occurring at higher concentrations of the extract (≥101.6 µg mL−1). The obtained results suggest possible use of this plant material for the production of various products (e.g., packaging, hygiene products, biodisinfectants, etc.) that would be useful against the spread of and for self-protection against COVID-19.
Collapse
Affiliation(s)
- Urška Jug
- Laboratory for Food Chemistry, Department of Analytical Chemistry, National Institute of Chemistry, Hajdrihova 19, 1000 Ljubljana, Slovenia
- Correspondence: (U.J.); (K.N.); (T.M.); Tel.: +386-1-4760-521 (U.J. & K.N.); +386-1-4779-824 (T.M.)
| | - Katerina Naumoska
- Laboratory for Food Chemistry, Department of Analytical Chemistry, National Institute of Chemistry, Hajdrihova 19, 1000 Ljubljana, Slovenia
- Correspondence: (U.J.); (K.N.); (T.M.); Tel.: +386-1-4760-521 (U.J. & K.N.); +386-1-4779-824 (T.M.)
| | - Tadej Malovrh
- Veterinary Faculty, University of Ljubljana, Gerbičeva ulica 60, 1000 Ljubljana, Slovenia
- Correspondence: (U.J.); (K.N.); (T.M.); Tel.: +386-1-4760-521 (U.J. & K.N.); +386-1-4779-824 (T.M.)
| |
Collapse
|
23
|
Zhang W, Li D, Xu B, Xu L, Lyu Q, Liu X, Li Z, Zhang J, Sun W, Ma Q, Qiao L, Liao P. Serum peptidome profiles immune response of COVID-19 Vaccine administration. Front Immunol 2022; 13:956369. [PMID: 36091008 PMCID: PMC9450691 DOI: 10.3389/fimmu.2022.956369] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 08/01/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundCoronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused significant loss of life and property. In response to the serious pandemic, recently developed vaccines against SARS-CoV-2 have been administrated to the public. Nevertheless, the research on human immunization response against COVID-19 vaccines is insufficient. Although much information associated with vaccine efficacy, safety and immunogenicity has been reported by pharmaceutical companies based on laboratory studies and clinical trials, vaccine evaluation needs to be extended further to better understand the effect of COVID-19 vaccines on human beings.MethodsWe performed a comparative peptidome analysis on serum samples from 95 participants collected at four time points before and after receiving CoronaVac. The collected serum samples were analyzed by matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF MS) to profile the serum peptides, and also subjected to humoral and cellular immune response analyses to obtain typical immunogenicity information.ResultsSignificant difference in serum peptidome profiles by MALDI-TOF MS was observed after vaccination. By supervised statistical analysis, a total of 13 serum MALDI-TOF MS feature peaks were obtained on day 28 and day 42 of vaccination. The feature peaks were identified as component C1q receptor, CD59 glycoprotein, mannose-binding protein C, platelet basic protein, CD99 antigen, Leucine-rich alpha-2-glycoprotein, integral membrane protein 2B, platelet factor 4 and hemoglobin subunits. Combining with immunogenicity analysis, the study provided evidence for the humoral and cellular immune responses activated by CoronaVac. Furthermore, we found that it is possible to distinguish neutralizing antibody (NAbs)-positive from NAbs-negative individuals after complete vaccination using the serum peptidome profiles by MALDI-TOF MS together with machine learning methods, including random forest (RF), partial least squares-discriminant analysis (PLS-DA), linear support vector machine (SVM) and logistic regression (LR).ConclusionsThe study shows the promise of MALDI-TOF MS-based serum peptidome analysis for the assessment of immune responses activated by COVID-19 vaccination, and discovered a panel of serum peptides biomarkers for COVID-19 vaccination and for NAbs generation. The method developed in this study can help not only in the development of new vaccines, but also in the post-marketing evaluation of developed vaccines.
Collapse
Affiliation(s)
- Wenjia Zhang
- Department of Clinical Laboratory, Chongqing General Hospital, Chongqing, China
| | - Dandan Li
- Department of Chemistry, Fudan University, Shanghai, China
| | - Bin Xu
- Bioyong Technologics, Inc., Beijing, China
| | - Lanlan Xu
- Department of Clinical Laboratory, Chongqing General Hospital, Chongqing, China
| | - Qian Lyu
- Bioyong Technologics, Inc., Beijing, China
| | - Xiangyi Liu
- Department of Laboratory Medicine, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Zhijie Li
- Department of Clinical Laboratory, Chongqing General Hospital, Chongqing, China
| | - Jian Zhang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Wei Sun
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Qingwei Ma
- Bioyong Technologics, Inc., Beijing, China
| | - Liang Qiao
- Department of Chemistry, Fudan University, Shanghai, China
- *Correspondence: Pu Liao, ; Liang Qiao,
| | - Pu Liao
- Department of Clinical Laboratory, Chongqing General Hospital, Chongqing, China
- *Correspondence: Pu Liao, ; Liang Qiao,
| |
Collapse
|
24
|
Psaridi L, Maltezou HC, Simonidou S, Lialliou I, Athanasopoulou D, Haila Z, Kyrimi A, Giannopoulou I, Giannousa S, Pseimada M, Christofilea O, Dounias G, Lanitis S, Kremasmenou E. Neutralizing antibody responses in healthcare personnel after three doses of mRNA BNT162b2 vaccine and association with baseline characteristics and past SARS-CoV-2 infection. Vaccine 2022; 40:5752-5756. [PMID: 36008235 PMCID: PMC9388445 DOI: 10.1016/j.vaccine.2022.08.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 07/12/2022] [Accepted: 08/15/2022] [Indexed: 11/24/2022]
Abstract
Aim To estimate neutralizing antibody (NAb) immunity against SARS-CoV-2 in 739 healthcare personnel (HCP) vaccinated with three doses of BNT162b2 mRNA vaccine. Methods Serum samples were collected at 3, 6, and 9 months after the second vaccine dose and at 7–55 days after the third dose. Samples were tested for NAbs against SARS-CoV-2 receptor binding domain. Results The mean inhibition rates at 3, 6, and 9 months after the second dose were 86.33%, 73.38%, and 61.18%, and increased to 95.57% after the booster dose. Younger HCP and HCP with past SARS-CoV-2 infection had higher inhibition rates while there was an inverse correlation between NAb levels and comorbidities or tobacco use (p-values < 0.001). Increased NAb titers were also noticed in women (p-value = 0.033), especially at the end of the 9-month study period. Conclusion NAb levels increased considerably after a booster mRNA vaccine dose. Host factors and past SARS-CoV-2 infection influence NAb titers.
Collapse
Affiliation(s)
- Loukia Psaridi
- Department of Immunology, Red Cross General Hospital of Athens, Greece
| | - Helena C Maltezou
- Directorate of Research, Studies, and Documentation, National Public Health Organization, Athens, Greece.
| | - Sofia Simonidou
- Department of Immunology, Red Cross General Hospital of Athens, Greece
| | - Ioanna Lialliou
- Department of Immunology, Red Cross General Hospital of Athens, Greece
| | | | - Zoi Haila
- Department of Immunology, Red Cross General Hospital of Athens, Greece
| | - Areti Kyrimi
- Department of Immunology, Red Cross General Hospital of Athens, Greece
| | | | | | - Maria Pseimada
- Department of Immunology, Red Cross General Hospital of Athens, Greece
| | | | - George Dounias
- Department of Public Health Policies, School of Public Health, University of West Attica, Athens, Greece
| | - Sophocles Lanitis
- 2(nd) Surgical Department and Unit of Surgical Oncology, Red Cross General Hospital of Athens, Athens, Greece
| | | |
Collapse
|
25
|
Jeremiah SS, Miyakawa K, Ryo A. Detecting SARS-CoV-2 neutralizing immunity: highlighting the potential of split nanoluciferase technology. J Mol Cell Biol 2022; 14:mjac023. [PMID: 35416249 PMCID: PMC9387144 DOI: 10.1093/jmcb/mjac023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 02/22/2022] [Accepted: 03/01/2022] [Indexed: 11/24/2022] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic has progressed over 2 years since its onset causing significant health concerns all over the world and is currently curtailed by mass vaccination. Immunity acquired against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) can be following either infection or vaccination. However, one can never be sure whether the acquired immunity is adequate to protect the individual from subsequent infection because of three important factors: individual variations in humoral response dynamics, waning of protective antibodies over time, and the emergence of immune escape mutants. Therefore, a test that can accurately differentiate the protected from the vulnerable is the need of the hour. The plaque reduction neutralization assay is the conventional gold standard test for estimating the titers of neutralizing antibodies that confer protection. However, it has got several drawbacks, which hinder the practical application of this test for wide-scale usage. Hence, various tests have been developed to detect protective immunity against SARS-CoV-2 that directly or indirectly assess the presence of neutralizing antibodies to SARS-CoV-2 in a lower biosafety setting. In this review, the pros and cons of the currently available assays are elaborated in detail and special focus is put on the scope of the novel split nanoluciferase technology for detecting SARS-CoV-2 neutralizing antibodies.
Collapse
Affiliation(s)
| | - Kei Miyakawa
- Department of Microbiology, Yokohama City University School of Medicine, Yokohama 236-0004, Japan
| | - Akihide Ryo
- Department of Microbiology, Yokohama City University School of Medicine, Yokohama 236-0004, Japan
| |
Collapse
|
26
|
Homogeneous surrogate virus neutralization assay to rapidly assess neutralization activity of anti-SARS-CoV-2 antibodies. Nat Commun 2022; 13:3716. [PMID: 35778399 PMCID: PMC9249905 DOI: 10.1038/s41467-022-31300-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 06/13/2022] [Indexed: 12/23/2022] Open
Abstract
The COVID-19 pandemic triggered the development of numerous diagnostic tools to monitor infection and to determine immune response. Although assays to measure binding antibodies against SARS-CoV-2 are widely available, more specific tests measuring neutralization activities of antibodies are immediately needed to quantify the extent and duration of protection that results from infection or vaccination. We previously developed a 'Serological Assay based on a Tri-part split-NanoLuc® (SATiN)' to detect antibodies that bind to the spike (S) protein of SARS-CoV-2. Here, we expand on our previous work and describe a reconfigured version of the SATiN assay, called Neutralization SATiN (Neu-SATiN), which measures neutralization activity of antibodies directly from convalescent or vaccinated sera. The results obtained with our assay and other neutralization assays are comparable but with significantly shorter preparation and run time for Neu-SATiN. As the assay is modular, we further demonstrate that Neu-SATiN enables rapid assessment of the effectiveness of vaccines and level of protection against existing SARS-CoV-2 variants of concern and can therefore be readily adapted for emerging variants.
Collapse
|
27
|
Kawasuji H, Morinaga Y, Tani H, Saga Y, Kaneda M, Murai Y, Ueno A, Miyajima Y, Fukui Y, Nagaoka K, Ono C, Matsuura Y, Niimi H, Yamamoto Y. Effectiveness of the third dose of BNT162b2 vaccine on neutralizing Omicron variant in the Japanese population. J Infect Chemother 2022; 28:1273-1278. [PMID: 35691864 PMCID: PMC9186405 DOI: 10.1016/j.jiac.2022.05.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 04/25/2022] [Accepted: 05/16/2022] [Indexed: 11/26/2022]
Abstract
Introduction The vaccine against SARS-CoV-2 provides humoral immunity to fight COVID-19; however, the acquired immunity gradually declines. Booster vaccination restores reduced humoral immunity; however, its effect on newly emerging variants, such as the Omicron variant, is a concern. As the waves of COVID-19 cases and vaccine programs differ between countries, it is necessary to know the domestic effect of the booster. Methods Serum samples were obtained from healthcare workers (20–69 years old) in the Pfizer BNT162b2 vaccine program at the Toyama University Hospital 6 months after the second dose (6mA2D, n = 648) and 2 weeks after the third dose (2wA3D, n = 565). The anti-SARS-CoV-2 antibody level was measured, and neutralization against the wild-type and variants (Delta and Omicron) was evaluated using pseudotyped viruses. Data on booster-related events were collected using questionnaires. Results The median anti-SARS-CoV-2 antibody was >30.9-fold elevated after the booster (6mA2D, 710.0 U/mL [interquartile range (IQR): 443.0–1068.0 U/mL]; 2wA3D, 21927 U/mL [IQR: 15321.0–>25000.0 U/mL]). Median neutralizing activity using 100-fold sera against wild-type-, Delta-, and Omicron-derived variants was elevated from 84.6%, 36.2%, and 31.2% at 6mA2D to >99.9%, 99.1%, and 94.6% at 2wA3D, respectively. The anti-SARS-CoV-2 antibody levels were significantly elevated in individuals with fever ≥37.5 °C, general fatigue, and myalgia, local swelling, and local hardness. Conclusion The booster effect, especially against the Omicron variant, was observed in the Japanese population. These findings contribute to the precise understanding of the efficacy and side effects of the booster and the promotion of vaccine campaigns.
Collapse
Affiliation(s)
- Hitoshi Kawasuji
- Department of Clinical Infectious Diseases, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Yoshitomo Morinaga
- Department of Microbiology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan; Clinical and Research Center for Infectious Diseases, Toyama University Hospital, Toyama, Japan.
| | - Hideki Tani
- Department of Microbiology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan; Department of Virology, Toyama Institute of Health, Toyama, Japan
| | - Yumiko Saga
- Department of Microbiology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan; Department of Virology, Toyama Institute of Health, Toyama, Japan
| | - Makito Kaneda
- Department of Clinical Infectious Diseases, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Yushi Murai
- Department of Clinical Infectious Diseases, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Akitoshi Ueno
- Department of Clinical Infectious Diseases, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Yuki Miyajima
- Department of Clinical Infectious Diseases, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Yasutaka Fukui
- Department of Clinical Infectious Diseases, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Kentaro Nagaoka
- Department of Clinical Infectious Diseases, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan; Clinical and Research Center for Infectious Diseases, Toyama University Hospital, Toyama, Japan
| | - Chikako Ono
- Laboratory of Virus Control, Center for Infectious Disease Education and Research (CiDER), Osaka University, Osaka, Japan; Laboratory of Virus Control, Research Institute for Microbial Diseases (RIMD), Osaka University, Osaka, Japan
| | - Yoshiharu Matsuura
- Laboratory of Virus Control, Center for Infectious Disease Education and Research (CiDER), Osaka University, Osaka, Japan; Laboratory of Virus Control, Research Institute for Microbial Diseases (RIMD), Osaka University, Osaka, Japan
| | - Hideki Niimi
- Clinical and Research Center for Infectious Diseases, Toyama University Hospital, Toyama, Japan; Department of Clinical Laboratory and Molecular Pathology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Yoshihiro Yamamoto
- Department of Clinical Infectious Diseases, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan; Clinical and Research Center for Infectious Diseases, Toyama University Hospital, Toyama, Japan
| |
Collapse
|
28
|
Wang Y, Tang CY, Wan XF. Antigenic characterization of influenza and SARS-CoV-2 viruses. Anal Bioanal Chem 2022; 414:2841-2881. [PMID: 34905077 PMCID: PMC8669429 DOI: 10.1007/s00216-021-03806-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 11/21/2021] [Accepted: 11/24/2021] [Indexed: 12/24/2022]
Abstract
Antigenic characterization of emerging and re-emerging viruses is necessary for the prevention of and response to outbreaks, evaluation of infection mechanisms, understanding of virus evolution, and selection of strains for vaccine development. Primary analytic methods, including enzyme-linked immunosorbent/lectin assays, hemagglutination inhibition, neuraminidase inhibition, micro-neutralization assays, and antigenic cartography, have been widely used in the field of influenza research. These techniques have been improved upon over time for increased analytical capacity, and some have been mobilized for the rapid characterization of the SARS-CoV-2 virus as well as its variants, facilitating the development of highly effective vaccines within 1 year of the initially reported outbreak. While great strides have been made for evaluating the antigenic properties of these viruses, multiple challenges prevent efficient vaccine strain selection and accurate assessment. For influenza, these barriers include the requirement for a large virus quantity to perform the assays, more than what can typically be provided by the clinical samples alone, cell- or egg-adapted mutations that can cause antigenic mismatch between the vaccine strain and circulating viruses, and up to a 6-month duration of vaccine development after vaccine strain selection, which allows viruses to continue evolving with potential for antigenic drift and, thus, antigenic mismatch between the vaccine strain and the emerging epidemic strain. SARS-CoV-2 characterization has faced similar challenges with the additional barrier of the need for facilities with high biosafety levels due to its infectious nature. In this study, we review the primary analytic methods used for antigenic characterization of influenza and SARS-CoV-2 and discuss the barriers of these methods and current developments for addressing these challenges.
Collapse
Affiliation(s)
- Yang Wang
- MU Center for Influenza and Emerging Infectious Diseases (CIEID), University of Missouri, Columbia, MO, USA
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO, USA
- Bond Life Sciences Center, University of Missouri, Columbia, MO, USA
| | - Cynthia Y Tang
- MU Center for Influenza and Emerging Infectious Diseases (CIEID), University of Missouri, Columbia, MO, USA
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO, USA
- Bond Life Sciences Center, University of Missouri, Columbia, MO, USA
- Institute for Data Science and Informatics, University of Missouri, Columbia, MO, USA
| | - Xiu-Feng Wan
- MU Center for Influenza and Emerging Infectious Diseases (CIEID), University of Missouri, Columbia, MO, USA.
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO, USA.
- Bond Life Sciences Center, University of Missouri, Columbia, MO, USA.
- Institute for Data Science and Informatics, University of Missouri, Columbia, MO, USA.
- Department of Electrical Engineering & Computer Science, College of Engineering, University of Missouri, Columbia, MO, USA.
| |
Collapse
|
29
|
Mahalingam G, Rachamalla HK, Arjunan P, Periyasami Y, M S, Thangavel S, Mohankumar KM, Moorthy M, Velayudhan SR, Srivastava A, Marepally S. Optimization of SARS-CoV-2 Pseudovirion Production in Lentivirus Backbone With a Novel Liposomal System. Front Pharmacol 2022; 13:840727. [PMID: 35401169 PMCID: PMC8990231 DOI: 10.3389/fphar.2022.840727] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 02/21/2022] [Indexed: 01/11/2023] Open
Abstract
Due to the fast mutating nature of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the development of novel therapeutics, vaccines, and evaluating the efficacies of existing one’s against the mutated strains is critical for containing the virus. Pseudotyped SARS-CoV-2 viruses are proven to be instrumental in evaluating the efficiencies of therapeutics, owing to their ease in application and safety when compared to handling the live virus. However, a comprehensive protocol that includes selecting transfection reagents, validating different packaging systems for high-throughput screening of neutralizing antibodies, is still a requisite. To this end, we designed and synthesized amide linker-based cationic lipids with varying hydrophilic head groups from dimethyl (Lipo-DME) to methyl, ethylhydroxyl (Lipo-MeOH), and diethylhydroxyl (Lipo-DOH) keeping the hydrophobic tail, stearic acid, as constant. Among the liposomal formulations of these lipids, Lipo-DOH was found to be superior in delivering plasmids and demonstrated comparable transfection efficiencies with commercial standard Lipofectamine 3000. We further used Lipo-DOH for lentivirus and SARS-CoV-2 pseudovirion preparation. For comparing different lentivirus packaging systems, we optimized conditions using Addgene and BEI systems and found that the BEI lenti plasmid system was found to be efficient in making lentiviruses using Lipo-DOH. Using the optimized transfection reagent and the lentivirus system, we developed a robust protocol for the generation of SARS-CoV-2 pseudovirions and characterized their infectivity in human ACE2 expressing HEK-293T cells and neutralizing properties in IgG against spike protein of SARS-CoV-2 positive human sera from individuals recovered from COVID-19.
Collapse
Affiliation(s)
- Gokulnath Mahalingam
- Centre for Stem Cell Research (CSCR) (a Unit of InStem, Bengaluru), CMC Campus, Vellore, India
| | | | - Porkizhi Arjunan
- Centre for Stem Cell Research (CSCR) (a Unit of InStem, Bengaluru), CMC Campus, Vellore, India
| | - Yogapriya Periyasami
- Centre for Stem Cell Research (CSCR) (a Unit of InStem, Bengaluru), CMC Campus, Vellore, India
| | - Salma M
- Centre for Stem Cell Research (CSCR) (a Unit of InStem, Bengaluru), CMC Campus, Vellore, India
| | | | | | - Mahesh Moorthy
- Department of Clinical Virology, Christian Medical College, Vellore, India
| | - Shaji R. Velayudhan
- Centre for Stem Cell Research (CSCR) (a Unit of InStem, Bengaluru), CMC Campus, Vellore, India
| | - Alok Srivastava
- Centre for Stem Cell Research (CSCR) (a Unit of InStem, Bengaluru), CMC Campus, Vellore, India
| | - Srujan Marepally
- Centre for Stem Cell Research (CSCR) (a Unit of InStem, Bengaluru), CMC Campus, Vellore, India
- *Correspondence: Srujan Marepally,
| |
Collapse
|
30
|
Kovalenko AO, Ryabchevskaya EM, Evtushenko EA, Manukhova TI, Kondakova OA, Ivanov PA, Arkhipenko MV, Gushchin VA, Nikitin NA, Karpova OV. Vaccine Candidate Against COVID-19 Based on Structurally Modified Plant Virus as an Adjuvant. Front Microbiol 2022; 13:845316. [PMID: 35295298 PMCID: PMC8919459 DOI: 10.3389/fmicb.2022.845316] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 02/04/2022] [Indexed: 12/24/2022] Open
Abstract
A recombinant vaccine candidate has been developed based on the major coronaviruses’ antigen (S protein) fragments and a novel adjuvant—spherical particles (SPs) formed during tobacco mosaic virus thermal remodeling. The receptor-binding domain and the highly conserved antigenic fragments of the S2 protein subunit were chosen for the design of recombinant coronavirus antigens. The set of three antigens (Co1, CoF, and PE) was developed and used to create a vaccine candidate composed of antigens and SPs (SPs + 3AG). Recognition of SPs + 3AG compositions by commercially available antibodies against spike proteins of SARS-CoV and SARS-CoV-2 was confirmed. The immunogenicity testing of these compositions in a mouse model showed that SPs improved immune response to the CoF and PE antigens. Total IgG titers against both proteins were 9–16 times higher than those to SPs. Neutralizing activity against SARS-CoV-2 in serum samples collected from hamsters immunized with the SPs + 3AG was demonstrated.
Collapse
Affiliation(s)
- Angelina O Kovalenko
- Department of Virology, Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | | | - Ekaterina A Evtushenko
- Department of Virology, Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Tatiana I Manukhova
- Department of Virology, Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Olga A Kondakova
- Department of Virology, Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Peter A Ivanov
- Department of Virology, Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Marina V Arkhipenko
- Department of Virology, Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Vladimir A Gushchin
- Department of Virology, Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia.,N.F. Gamaleya National Research Center for Epidemiology and Microbiology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Nikolai A Nikitin
- Department of Virology, Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Olga V Karpova
- Department of Virology, Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| |
Collapse
|
31
|
Portilho AI, Gimenes Lima G, De Gaspari E. Enzyme-Linked Immunosorbent Assay: An Adaptable Methodology to Study SARS-CoV-2 Humoral and Cellular Immune Responses. J Clin Med 2022; 11:1503. [PMID: 35329828 PMCID: PMC8948777 DOI: 10.3390/jcm11061503] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 01/19/2022] [Accepted: 01/27/2022] [Indexed: 02/07/2023] Open
Abstract
The Enzyme-Linked Immunosorbent Assay is a versatile technique, which can be used for several applications. It has enormously contributed to the study of infectious diseases. This review highlights how this methodology supported the science conducted in COVID-19 pandemics, allowing scientists to better understand the immune response against SARS-CoV-2. ELISA can be modified to assess the functionality of antibodies, as avidity and neutralization, respectively by the standardization of avidity-ELISA and surrogate-neutralization methods. Cellular immunity can also be studied using this assay. Products secreted by cells, like proteins and cytokines, can be studied by ELISA or its derivative Enzyme-linked immunospot (ELISpot) assay. ELISA and ELISA-based methods aided the area of immunology against infectious diseases and is still relevant, for example, as a promising approach to study the differences between natural and vaccine-induced immune responses against SARS-CoV-2.
Collapse
Affiliation(s)
- Amanda Izeli Portilho
- Immunology Center, Adolfo Lutz Institute, Sao Paulo 01246-902, SP, Brazil; (A.I.P.); (G.G.L.)
- Graduate Program Interunits in Biotechnology, University of Sao Paulo, Sao Paulo 05508-900, SP, Brazil
| | - Gabrielle Gimenes Lima
- Immunology Center, Adolfo Lutz Institute, Sao Paulo 01246-902, SP, Brazil; (A.I.P.); (G.G.L.)
- Graduate Program Interunits in Biotechnology, University of Sao Paulo, Sao Paulo 05508-900, SP, Brazil
| | - Elizabeth De Gaspari
- Immunology Center, Adolfo Lutz Institute, Sao Paulo 01246-902, SP, Brazil; (A.I.P.); (G.G.L.)
- Graduate Program Interunits in Biotechnology, University of Sao Paulo, Sao Paulo 05508-900, SP, Brazil
| |
Collapse
|
32
|
Lippi G, Adeli K, Plebani M. Commercial immunoassays for detection of anti-SARS-CoV-2 spike and RBD antibodies: urgent call for validation against new and highly mutated variants. Clin Chem Lab Med 2022; 60:338-342. [PMID: 34911171 DOI: 10.1515/cclm-2021-1287] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 12/10/2021] [Indexed: 02/06/2023]
Abstract
Measuring the level of protection conferred by anti-SARS-CoV-2 (trimeric) spike or RBD (receptor binding domain) antibodies (especially total and IgG) is a suitable and reliable approach for predicting biological protection against the risk of infection and severe coronavirus disease 2019 (COVID-19) illness. Nonetheless, SARS-CoV-2 has undergone a broad process of recombination since the identification of the prototype lineage in 2019, introducing a huge number of mutations in its genome and generating a vast array of variants of interest (VoI) and concern (VoC). Many of such variants developed several mutations in spike protein and RBD, with the new Omicron (B.1.1.529) clade displaying over 30 changes, 15 of which concentrated in the RBD. Besides their impact on virus biology, as well as on the risk of detection failure with some molecular techniques (i.e., S gene dropout), recent evidence suggests that these mutations may also jeopardize the reliability of currently available commercial immunoassays for detecting anti-SARS-CoV-2 antibodies. The antigen (either spike or RBD) and epitopes of the prototype SARS-CoV-2 coated in some immunoassays may no longer reflect the sequence of circulating variants. On the other hand, anti-SARS-CoV-2 antibodies elicited by highly mutated SARS-CoV-2 variants may no longer be efficiently recognized by the currently available commercial immunoassays. Therefore, beside the compelling need to regularly re-evaluate and revalidate all commercially available immunoassays against live virus neutralization assays based on emerging VoCs or VoIs, diagnostic companies may also consider to redevelop their methods, replacing former SARS-CoV-2 antigens and epitopes with those of the new variants.
Collapse
Affiliation(s)
- Giuseppe Lippi
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Clinical Biochemistry and School of Medicine, University of Verona, Verona, Italy
- IFCC Task Force on COVID-19, Milano, Italy
| | - Khosrow Adeli
- IFCC Task Force on COVID-19, Milano, Italy
- Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Mario Plebani
- Department of Medicine-DIMED, University of Padova, Padova, Italy
| |
Collapse
|
33
|
Padoan A, Cosma C, Galla L, Basso D, Plebani M. Two rapid SARS-CoV-2 disposable devices for semi-quantitative S-RBD antibody levels determination compared with CLIA and ELISA assays at different protective thresholds. Clin Chim Acta 2022; 529:104-108. [PMID: 35202620 PMCID: PMC8858771 DOI: 10.1016/j.cca.2022.02.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 02/17/2022] [Accepted: 02/17/2022] [Indexed: 11/17/2022]
Abstract
BACKGROUND AND AIMS Performance of two disposable devices for identifying subjects with low anti-SARS-CoV-2 protection was compared with that of automated enzyme-linked immunosorbent (ELISA) and chemiluminescent (CLIA) assay. MATERIALS AND METHODS In July 2021, 123 healthcare workers (HCW), twice vaccinated by BNT162b2/Comirnaty mRNA (BioNTech-Pfizer), underwent Ab iRapid COVID-19 Quant "Neutralizing" Self-test (iRapid Self-test) and "Neutralizing" Professional-use (iRapid pro) (DIESSE, Diagnostica Senese, Siena, Italy). Simultaneously, serum Ab were determined by Maglumi 2000 plus (anti S-RBD CLIA assay, Snibe Diagnostics, Shenzhen, China) and SARS-CoV-2 "Neutralizing" Ab Chorus ELISA (DIESSE, Siena, Italy). Results were evaluated against two "protective-thresholds", 90 kBAU/L and 506 kBAU/L. RESULTS HCW mean age, 46.2 (±12.6) years; 26 (20.5%), males, 101 (79.5%), females. The mean time interval (and standard deviation) between the first vaccine dose and Ab determination was 129.5 (±36.4) days and was neither gender (p = 0.879) nor age (p = 0.341) related. With Maglumi, 114 (89.7%) and 43 (33.8%) HCW presented Ab ≥ 90 kBAU/L and Ab ≥ 506 kBAU/L, respectively; with Chorus, 96 (75.6%) presented Ab values ≥506 kBAU/L. CLIA and ELISA agreement was 56.7%. At 90 kBAU/L, iRapid self-test and Pro sensitivities were 98.2% (95% CI: 92.7-99.8), specificity 69.2% (95% CI: 38.6-90.9%) and 76.9% (46.2-95%), respectively. At 506 kBAU/L, iRapid sensitivities were 58.1-91.6%, and specificities, 89-96.6%. On evaluating Ab at <4 and ≥4 months, protective titers had decreased. CONCLUSIONS iRapid semi-quantitative devices had very good overall agreements of 95.1% and 95.9% for detecting individuals with low anti-SARS-CoV-2 protection.
Collapse
Affiliation(s)
- Andrea Padoan
- Department of Medicine-DIMED, University of Padova, Italy; Department of Laboratory Medicine, University-Hospital of Padova, Italy
| | - Chiara Cosma
- Department of Laboratory Medicine, University-Hospital of Padova, Italy
| | - Luisa Galla
- Department of Laboratory Medicine, University-Hospital of Padova, Italy
| | - Daniela Basso
- Department of Medicine-DIMED, University of Padova, Italy; Department of Laboratory Medicine, University-Hospital of Padova, Italy
| | - Mario Plebani
- Department of Medicine-DIMED, University of Padova, Italy; Department of Laboratory Medicine, University-Hospital of Padova, Italy.
| |
Collapse
|
34
|
Kitchin D, Richardson SI, van der Mescht MA, Motlou T, Mzindle N, Moyo-Gwete T, Makhado Z, Ayres F, Manamela NP, Spencer H, Lambson B, Oosthuysen B, Kaldine H, du Pisanie M, Mennen M, Skelem S, Williams N, Ntusi NA, Burgers WA, Gray GG, Bekker LG, Boswell MT, Rossouw TM, Ueckermann V, Moore PL. Ad26.COV2.S breakthrough infections induce high titers of neutralizing antibodies against Omicron and other SARS-CoV-2 variants of concern. Cell Rep Med 2022; 3:100535. [PMID: 35474744 PMCID: PMC8828412 DOI: 10.1016/j.xcrm.2022.100535] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 01/19/2022] [Accepted: 01/27/2022] [Indexed: 01/20/2023]
Abstract
The Janssen (Johnson & Johnson) Ad26.COV2.S non-replicating viral vector vaccine has been widely deployed for COVID-19 vaccination programs in resource-limited settings. Here we confirm that neutralizing and binding antibody responses to Ad26.COV2.S vaccination are stable for 6 months post-vaccination, when tested against multiple SARS-CoV-2 variants. Secondly, using longitudinal samples from individuals who experienced clinically mild breakthrough infections 4 to 5 months after vaccination, we show dramatically boosted binding antibodies, Fc effector function, and neutralization. These high titer responses are of similar magnitude to humoral immune responses measured in convalescent donors who had been hospitalized with severe illness, and are cross-reactive against diverse SARS-CoV-2 variants, including the neutralization-resistant Omicron (B.1.1.529) variant that currently dominates global infections, as well as SARS-CoV-1. These data have implications for population immunity in areas where the Ad26.COV2.S vaccine has been widely deployed, but where ongoing infections continue to occur at high levels.
Collapse
Affiliation(s)
- Dale Kitchin
- National Institute for Communicable Diseases (NICD) of the National Health Laboratory Service (NHLS), Johannesburg, South Africa,SAMRC Antibody Immunity Research Unit, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Simone I. Richardson
- National Institute for Communicable Diseases (NICD) of the National Health Laboratory Service (NHLS), Johannesburg, South Africa,SAMRC Antibody Immunity Research Unit, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Mieke A. van der Mescht
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Thopisang Motlou
- National Institute for Communicable Diseases (NICD) of the National Health Laboratory Service (NHLS), Johannesburg, South Africa,SAMRC Antibody Immunity Research Unit, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Nonkululeko Mzindle
- National Institute for Communicable Diseases (NICD) of the National Health Laboratory Service (NHLS), Johannesburg, South Africa,SAMRC Antibody Immunity Research Unit, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Thandeka Moyo-Gwete
- National Institute for Communicable Diseases (NICD) of the National Health Laboratory Service (NHLS), Johannesburg, South Africa,SAMRC Antibody Immunity Research Unit, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Zanele Makhado
- National Institute for Communicable Diseases (NICD) of the National Health Laboratory Service (NHLS), Johannesburg, South Africa,SAMRC Antibody Immunity Research Unit, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Frances Ayres
- National Institute for Communicable Diseases (NICD) of the National Health Laboratory Service (NHLS), Johannesburg, South Africa,SAMRC Antibody Immunity Research Unit, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Nelia P. Manamela
- National Institute for Communicable Diseases (NICD) of the National Health Laboratory Service (NHLS), Johannesburg, South Africa,SAMRC Antibody Immunity Research Unit, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Holly Spencer
- National Institute for Communicable Diseases (NICD) of the National Health Laboratory Service (NHLS), Johannesburg, South Africa,SAMRC Antibody Immunity Research Unit, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Bronwen Lambson
- National Institute for Communicable Diseases (NICD) of the National Health Laboratory Service (NHLS), Johannesburg, South Africa,SAMRC Antibody Immunity Research Unit, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Brent Oosthuysen
- National Institute for Communicable Diseases (NICD) of the National Health Laboratory Service (NHLS), Johannesburg, South Africa,SAMRC Antibody Immunity Research Unit, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Haajira Kaldine
- National Institute for Communicable Diseases (NICD) of the National Health Laboratory Service (NHLS), Johannesburg, South Africa,SAMRC Antibody Immunity Research Unit, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Marizane du Pisanie
- Division for Infectious Diseases, Department of Internal Medicine, Steve Biko Academic Hospital and University of Pretoria, Pretoria, South Africa
| | - Mathilda Mennen
- Cape Heart Institute, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Sango Skelem
- Cape Heart Institute, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Noleen Williams
- Cape Heart Institute, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Ntobeko A.B. Ntusi
- Cape Heart Institute, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa,Department of Medicine, University of Cape Town and Groote Schuur Hospital, Cape Town, South Africa,Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Wendy A. Burgers
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa,Division of Medical Virology, Department of Pathology, University of Cape Town, Cape Town, South Africa,Wellcome Centre for Infectious Diseases Research in Africa, University of Cape Town, Cape Town, South Africa
| | - Glenda G. Gray
- The South African Medical Research Council, Tygerberg, South Africa
| | - Linda-Gail Bekker
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa,The Desmond Tutu HIV Centre, University of Cape Town, Cape Town, South Africa
| | - Michael T. Boswell
- Division for Infectious Diseases, Department of Internal Medicine, Steve Biko Academic Hospital and University of Pretoria, Pretoria, South Africa
| | - Theresa M. Rossouw
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Veronica Ueckermann
- Division for Infectious Diseases, Department of Internal Medicine, Steve Biko Academic Hospital and University of Pretoria, Pretoria, South Africa
| | - Penny L. Moore
- National Institute for Communicable Diseases (NICD) of the National Health Laboratory Service (NHLS), Johannesburg, South Africa,SAMRC Antibody Immunity Research Unit, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa,Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa,Centre for the AIDS Programme of Research in South Africa, Durban, South Africa,Corresponding author
| |
Collapse
|
35
|
Septisetyani EP, Prasetyaningrum PW, Anam K, Santoso A. SARS-CoV-2 Antibody Neutralization Assay Platforms Based on Epitopes Sources: Live Virus, Pseudovirus, and Recombinant S Glycoprotein RBD. Immune Netw 2022; 21:e39. [PMID: 35036026 PMCID: PMC8733193 DOI: 10.4110/in.2021.21.e39] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 11/03/2021] [Accepted: 11/08/2021] [Indexed: 12/15/2022] Open
Abstract
The high virulent severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus that emerged in China at the end of 2019 has generated novel coronavirus disease, coronavirus disease 2019 (COVID-19), causing a pandemic worldwide. Every country has made great efforts to struggle against SARS-CoV-2 infection, including massive vaccination, immunological patients’ surveillance, and the utilization of convalescence plasma for COVID-19 therapy. These efforts are associated with the attempts to increase the titers of SARS-CoV-2 neutralizing Abs (nAbs) generated either after infection or vaccination that represent the body’s immune status. As there is no standard therapy for COVID-19 yet, virus eradication will mainly depend on these nAbs contents in the body. Therefore, serological nAbs neutralization assays become a requirement for researchers and clinicians to measure nAbs titers. Different platforms have been developed to evaluate nAbs titers utilizing various epitopes sources, including neutralization assays based on the live virus, pseudovirus, and neutralization assays utilizing recombinant SARS-CoV-2 S glycoprotein receptor binding site, receptor-binding domain. As a standard neutralization assay, the plaque reduction neutralization test (PRNT) requires isolation and propagation of live pathogenic SARS-CoV-2 virus conducted in a BSL-3 containment. Hence, other surrogate neutralization assays relevant to the PRNT play important alternatives that offer better safety besides facilitating high throughput analyses. This review discusses the current neutralization assay platforms used to evaluate nAbs, their techniques, advantages, and limitations.
Collapse
Affiliation(s)
- Endah Puji Septisetyani
- Research Center for Biotechnology, National Research and Innovation Agency, Bogor, West Java, Indonesia
| | | | - Khairul Anam
- Research Center for Biotechnology, National Research and Innovation Agency, Bogor, West Java, Indonesia
| | - Adi Santoso
- Research Center for Biotechnology, National Research and Innovation Agency, Bogor, West Java, Indonesia
| |
Collapse
|
36
|
Lippi G, Plebani M. Not all SARS-CoV-2 IgG and neutralizing antibody assays are created equal. Clin Chim Acta 2021; 526:81-82. [PMID: 34958752 PMCID: PMC8702590 DOI: 10.1016/j.cca.2021.12.020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 12/21/2021] [Indexed: 12/22/2022]
Affiliation(s)
- Giuseppe Lippi
- Section of Clinical Biochemistry and School of Medicine, University of Verona, Verona, Italy.
| | - Mario Plebani
- Department of Laboratory Medicine, Padua University School of Medicine, Padua, Italy
| |
Collapse
|
37
|
Padoan A, Cosma C, Bonfante F, Della Rocca F, Barbaro F, Santarossa C, Dall'Olmo L, Pagliari M, Bortolami A, Cattelan A, Cianci V, Basso D, Plebani M. Neutralizing antibody titers six months after Comirnaty vaccination: kinetics and comparison with SARS-CoV-2 immunoassays. Clin Chem Lab Med 2021; 60:456-463. [PMID: 34911170 DOI: 10.1515/cclm-2021-1247] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 12/03/2021] [Indexed: 11/15/2022]
Abstract
OBJECTIVES mRNA vaccines, including Comirnaty (BNT162b2 mRNA, BioNTech-Pfizer), elicit high IgG and neutralizing antibody (NAb) responses after the second dose, but the progressive decrease in serum antibodies against SARS-CoV-2 following vaccination have raised questions concerning long-term immunity, decreased antibody levels being associated with breakthrough infections after vaccination, prompting the consideration of booster doses. METHODS A total number of 189 Padua University-Hospital healthcare workers (HCW) who had received a second vaccine dose were asked to collect serum samples for determining Ab at 12 (t12) and 28 (t28) days, and 6 months (t6m) after their first Comirnaty/BNT162b2 inoculation. Ab titers were measured with plaque reduction neutralization test (PRNT), and three chemiluminescent immunoassays, targeting the receptor binding domain (RBD), the trimeric Spike protein (trimeric-S), and surrogate viral neutralization tests (sVNT). RESULTS The median percentages (interquartile range) for decrease in antibodies values 6 months after the first dose were 86.8% (67.1-92.8%) for S-RBD IgG, 82% (58.6-89.3%) for trimeric-S, 70.4% (34.5-86.4%) for VNT-Nab, 75% (50-87.5%) for PRNT50 and 75% (50-93.7%) for PRNT90. At 6 months, neither PRNT titers nor VNT-Nab and S-RBD IgG bAb levels correlated with age (p=0.078) or gender (p=0.938), while they were correlated with previous infection (p<0.001). CONCLUSIONS After 6 months, a method-independent reduction of around 90% in anti-SARS-CoV-2 antibodies was detected, while no significant differences were found between values of males and females aged between 24 and 65 years without compromised health status. Further efforts to improve analytical harmonization and standardization are needed.
Collapse
Affiliation(s)
- Andrea Padoan
- Department of Medicine-DIMED, University of Padova, Padova, Italy.,Department of Laboratory Medicine, University-Hospital of Padova, Padova, Italy
| | - Chiara Cosma
- Department of Laboratory Medicine, University-Hospital of Padova, Padova, Italy
| | - Francesco Bonfante
- Laboratory of Experimental Animal Models, Division of Comparative Biomedical Sciences, Istituto Zooprofilattico Sperimentale delle Venezie, Legnaro, Italy
| | | | - Francesco Barbaro
- Infective and Tropical Disease Unit, Padua University Hospital, Padova, Italy
| | | | - Luigi Dall'Olmo
- Department of Surgical Oncological and Gastroenterological Sciences - DISCOG, University of Padova, Padova, Italy.,Veneto Institute of Oncology, IOV-IRCCS, Padova, Italy
| | - Matteo Pagliari
- Laboratory of Experimental Animal Models, Division of Comparative Biomedical Sciences, Istituto Zooprofilattico Sperimentale delle Venezie, Legnaro, Italy
| | - Alessio Bortolami
- Laboratory of Experimental Animal Models, Division of Comparative Biomedical Sciences, Istituto Zooprofilattico Sperimentale delle Venezie, Legnaro, Italy
| | - Annamaria Cattelan
- Infective and Tropical Disease Unit, Padua University Hospital, Padova, Italy
| | - Vito Cianci
- Emergency Department, Padua University Hospital, Padova, Italy
| | - Daniela Basso
- Department of Medicine-DIMED, University of Padova, Padova, Italy.,Department of Laboratory Medicine, University-Hospital of Padova, Padova, Italy
| | - Mario Plebani
- Department of Medicine-DIMED, University of Padova, Padova, Italy.,Department of Laboratory Medicine, University-Hospital of Padova, Padova, Italy
| |
Collapse
|
38
|
Padoan A, Cosma C, Bonfante F, Rocca FD, Barbaro F, Santarossa C, Dall'Olmo L, Pagliari M, Bortolami A, Cattelan A, Cianci V, Basso D, Plebani M. SARS-CoV-2 neutralizing antibodies after one or two doses of Comirnaty (BNT162b2, BioNTech/Pfizer): Kinetics and comparison with chemiluminescent assays. Clin Chim Acta 2021; 523:446-453. [PMID: 34755646 PMCID: PMC8553360 DOI: 10.1016/j.cca.2021.10.028] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 10/26/2021] [Accepted: 10/26/2021] [Indexed: 12/16/2022]
Abstract
BACKGROUND Studies evaluating neutralizing antibody (NAb) after BNT162b2 vaccine are scarce. We therefore compared NAb using the plaque reduction neutralization test (PRNT) in vaccinated subjects, with those from five chemiluminescent (CLIA) assays, two targeting ACE and S-RBD interaction. METHODS Sera from 174 completely Comirnaty/BNT162b2 vaccinated healthcare workers (HCW) were evaluated at t12 and t28. NAb titers at low (PRNT50) or high (PRNT90) stringency were compared with: Liaison SARS-CoV-2 Trimeric-S IgG, Elecsys S-RBD Ab, Maglumi SARS-CoV-2 S-RBD IgG and SARS-CoV-2 Nab; iFlash 2019-nCoV NAb. RESULTS Neither PRNT50 nor PRNT90 correlated with age (range, 24-65 years); no significant differences were found for gender. PRNT50 and PRNT90 seropositive titers (≥1:20) were 43 (24.7%) and 15 (8.6%) at t12 and 167 (95.9%) and 149 (85.6%) at t28. CLIA results at t28 were uncorrelated with age, apart from Elecsys S-RBD Ab (r = -0.164, p = 0.046). Gender differences were found for Maglumi SARS-CoV-2 S-RBD IgG (p = 0.037) and Maglumi NAb (p = 0.046). Considering PRNT50 at thresholds of 1:20 (or 1:40) and 1:160 (or 1:320), corresponding to different immune protective levels, CLIA cut-offs have been identified. CONCLUSIONS Comirnaty/BNT162b2 elicits strong NAb production, especially 28 days after first inoculum. Differences in correlation between Nab titers and circulating antibodies measured by 5 immunoassays have been found, being stronger the correlation for Maglumi Nab.
Collapse
Affiliation(s)
- Andrea Padoan
- Department of Medicine-DIMED, University of Padua, Italy; Department of Laboratory Medicine, University-Hospital of Padua, Italy
| | - Chiara Cosma
- Department of Laboratory Medicine, University-Hospital of Padua, Italy
| | - Francesco Bonfante
- Laboratory of Experimental Animal Models, Division of Comparative Biomedical Sciences, Istituto Zooprofilattico Sperimentale delle Venezie, Legnaro, Italy
| | | | - Francesco Barbaro
- Infective and Tropical Disease Unit, Padua University Hospital, Padua, Italy
| | | | - Luigi Dall'Olmo
- Department of Surgical Oncological and Gastroenterological Sciences - DISCOG, University of Padua, Italy; Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Matteo Pagliari
- Laboratory of Experimental Animal Models, Division of Comparative Biomedical Sciences, Istituto Zooprofilattico Sperimentale delle Venezie, Legnaro, Italy
| | - Alessio Bortolami
- Laboratory of Experimental Animal Models, Division of Comparative Biomedical Sciences, Istituto Zooprofilattico Sperimentale delle Venezie, Legnaro, Italy
| | - Annamaria Cattelan
- Infective and Tropical Disease Unit, Padua University Hospital, Padua, Italy
| | - Vito Cianci
- Emergency Department, Padua University Hospital, Padua, Italy
| | - Daniela Basso
- Department of Medicine-DIMED, University of Padua, Italy; Department of Laboratory Medicine, University-Hospital of Padua, Italy
| | - Mario Plebani
- Department of Medicine-DIMED, University of Padua, Italy; Department of Laboratory Medicine, University-Hospital of Padua, Italy.
| |
Collapse
|