1
|
van Rijswijck DMH, Bondt A, Raafat D, Holtfreter S, Wietschel KA, van der Lans SPA, Völker U, Bröker BM, Heck AJR. Persistent IgG1 clones dominate and personalize the plasma antibody repertoire. SCIENCE ADVANCES 2025; 11:eadt7746. [PMID: 40238876 PMCID: PMC12002106 DOI: 10.1126/sciadv.adt7746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 03/11/2025] [Indexed: 04/18/2025]
Abstract
Antibodies play a pivotal role in the immune defense and long-term immunity. Yet, while several studies have highlighted the persistence of antigen-specific antibody responses, it is unclear whether this stems from the continuous production of the same clones or recurrent activation of B cells generating new clones. To examine the stability of the human antibody repertoire, we monitored the concentrations of the most abundant IgG1 clones in plasma samples of 11 healthy donors at nine sampling points over a year. During this year, each donor received three doses of a COVID-19 vaccine. Notwithstanding these vaccinations, the concentrations of the most abundant IgG1 clones remained constant. Given the 2- to 3-week half-life of IgG1 molecules in blood, our data suggest that these clones are associated with long-term immunity and do not undergo somatic hypermutation which would imply short-lived plasma cells. Overall, our data suggest that most of the abundant IgG1 clones in plasma are persistently produced by long-lived plasma cells.
Collapse
Affiliation(s)
- Danique M. H. van Rijswijck
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Padualaan 8, Utrecht 3584 CH, Netherlands
- Netherlands Proteomics Center, Padualaan 8, Utrecht 3584 CH, Netherlands
| | - Albert Bondt
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Padualaan 8, Utrecht 3584 CH, Netherlands
- Netherlands Proteomics Center, Padualaan 8, Utrecht 3584 CH, Netherlands
| | - Dina Raafat
- Institute of Immunology, University Medicine Greifswald, Greifswald, Germany
- Department of Microbiology and Immunology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Silva Holtfreter
- Institute of Immunology, University Medicine Greifswald, Greifswald, Germany
| | - Kilian A. Wietschel
- Institute of Immunology, University Medicine Greifswald, Greifswald, Germany
| | - Sjors P. A. van der Lans
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Uwe Völker
- Department of Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, 17475, Germany
| | - Barbara M. Bröker
- Institute of Immunology, University Medicine Greifswald, Greifswald, Germany
| | - Albert J. R. Heck
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Padualaan 8, Utrecht 3584 CH, Netherlands
- Netherlands Proteomics Center, Padualaan 8, Utrecht 3584 CH, Netherlands
| |
Collapse
|
2
|
Fan L, Qiu Z, Deng Q, Guo T, Rong L. Modeling SARS-CoV-2 Infection Dynamics: Insights into Viral Clearance and Immune Synergy. Bull Math Biol 2025; 87:67. [PMID: 40232610 DOI: 10.1007/s11538-025-01442-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 03/18/2025] [Indexed: 04/16/2025]
Abstract
Understanding the mechanisms of interaction between SARS-CoV-2 infection and the immune system is crucial for developing effective treatment strategies against COVID-19. In this paper, a mathematical model is formulated to investigate the interactions among SARS-CoV-2 infection, cellular immunity, and humoral immunity. Clinical data from eight asymptomatic or mild COVID-19 patients in Munich are used to fit the model, and the dynamics of natural killer (NK) cells, cytotoxic T lymphocytes (CTLs), B cells, and antibodies are further explored using the average of the best-fitting parameter values. Subsequently, the impact of NK cells, CTLs, B cells, and antibodies on SARS-CoV-2 infection is numerically investigated. The results indicate that (i) the synergy of NK cells, CTLs, and antibodies leads to a rapid decrease in the viral load during SARS-CoV-2 infection; (ii) antibodies play a crucial role compared to other immune mechanisms, and enhancing B cell stimulation may be more effective in clearing the virus from the lungs; (iii) in terms of cytotoxic effects, CTLs are stronger and more sustained than NK cells. Furthermore, the existence and local stability of the model's equilibria are fully classified, and complex dynamics of the model are further investigated using bifurcation theory, revealing multistability phenomena, including multiple attractors and periodic solutions. These findings suggest potential uncertainty and diversity in SARS-CoV-2 infection outcomes.
Collapse
Affiliation(s)
- Lele Fan
- School of Mathematics and Statistics, Nanjing University of Science and Technology, Nanjing, 210094, People's Republic of China
| | - Zhipeng Qiu
- School of Mathematics and Statistics, Nanjing University of Science and Technology, Nanjing, 210094, People's Republic of China.
| | - Qi Deng
- Laboratory for Industrial and Applied Mathematics, Department of Mathematics and Statistics, York University, Toronto, ON, M3J1P0, Canada
| | - Ting Guo
- Aliyun School of Big Data, Changzhou University, Changzhou, 213164, People's Republic of China
| | - Libin Rong
- Department of Mathematics, University of Florida, Gainesville, FL, 32611, USA.
| |
Collapse
|
3
|
Raharinirina NA, Gubela N, Börnigen D, Smith MR, Oh DY, Budt M, Mache C, Schillings C, Fuchs S, Dürrwald R, Wolff T, Hölzer M, Paraskevopoulou S, von Kleist M. SARS-CoV-2 evolution on a dynamic immune landscape. Nature 2025; 639:196-204. [PMID: 39880955 PMCID: PMC11882442 DOI: 10.1038/s41586-024-08477-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 12/02/2024] [Indexed: 01/31/2025]
Abstract
Since the onset of the pandemic, many SARS-CoV-2 variants have emerged, exhibiting substantial evolution in the virus' spike protein1, the main target of neutralizing antibodies2. A plausible hypothesis proposes that the virus evolves to evade antibody-mediated neutralization (vaccine- or infection-induced) to maximize its ability to infect an immunologically experienced population1,3. Because viral infection induces neutralizing antibodies, viral evolution may thus navigate on a dynamic immune landscape that is shaped by local infection history. Here we developed a comprehensive mechanistic model, incorporating deep mutational scanning data4,5, antibody pharmacokinetics and regional genomic surveillance data, to predict the variant-specific relative number of susceptible individuals over time. We show that this quantity precisely matched historical variant dynamics, predicted future variant dynamics and explained global differences in variant dynamics. Our work strongly suggests that the ongoing pandemic continues to shape variant-specific population immunity, which determines a variant's ability to transmit, thus defining variant fitness. The model can be applied to any region by utilizing local genomic surveillance data, allows contextualizing risk assessment of variants and provides information for vaccine design.
Collapse
Affiliation(s)
- N Alexia Raharinirina
- Department of Mathematics & Computer Science, Freie Universität Berlin, Berlin, Germany
| | - Nils Gubela
- Department of Mathematics & Computer Science, Freie Universität Berlin, Berlin, Germany
- International Max-Planck Research School for Biology and Computation (IMPRS-BAC), Max-Planck Institute for Molecular Genetics, Berlin, Germany
| | | | | | - Djin-Ye Oh
- Department 1, Robert Koch Institute, Berlin, Germany
| | - Matthias Budt
- Department 1, Robert Koch Institute, Berlin, Germany
| | | | - Claudia Schillings
- Department of Mathematics & Computer Science, Freie Universität Berlin, Berlin, Germany
| | - Stephan Fuchs
- Department MFI, Robert Koch Institute, Berlin, Germany
| | - Ralf Dürrwald
- Department 1, Robert Koch Institute, Berlin, Germany
| | | | - Martin Hölzer
- Department MFI, Robert Koch Institute, Berlin, Germany
| | | | - Max von Kleist
- Department of Mathematics & Computer Science, Freie Universität Berlin, Berlin, Germany.
- Project Groups, Robert Koch Institute, Berlin, Germany.
| |
Collapse
|
4
|
Anderson AR, Strouse JJ, Manwani D, Brandow AM, Vichinsky E, Campbell A, Leavey PJ, Nero A, Ibrahim IF, Field JJ, Baer A, Soto-Calderon H, Vincent L, Zhao Y, Santos JJS, Hensley SE, Mortier N, Lanzkron S, Neuberg D, Abrams CS. COVID-19 mRNA vaccination responses in individuals with sickle cell disease: an ASH RC Sickle Cell Research Network Study. Blood Adv 2024; 8:4549-4553. [PMID: 38991137 PMCID: PMC11399661 DOI: 10.1182/bloodadvances.2024013878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 06/24/2024] [Accepted: 06/27/2024] [Indexed: 07/13/2024] Open
Abstract
ABSTRACT Children and adults with sickle cell disease (SCD) have increases in morbidity and mortality with COVID-19 infections. The American Society of Hematology Research Collaborative Sickle Cell Disease Research Network performed a prospective COVID-19 vaccine study to assess antibody responses and analyze whether messenger RNA (mRNA) vaccination precipitated any adverse effects unique to individuals with SCD. Forty-one participants received 2 doses of the Pfizer-BioNTech vaccine and provided baseline blood samples before vaccination and 2 months after the initial vaccination for analysis of immunoglobulin G (IgG) reactivity against the receptor binding domain (RBD) of the severe acute respiratory syndrome coronavirus 2 spike protein. Six-month IgG reactivity against the viral RBD was also available in 37 patients. Postvaccination reactogenicity was common and similar to the general population. There were no fevers that required inpatient admission. Vaso-occlusive pain within 2 to 3 days of first or second vaccination was reported by 5 participants (12%) including 4 (10%) who sought medical care. Twenty-seven participants (66%) were seropositive at baseline, and all 14 initially seronegative participants (34%) converted to seropositive after vaccination. Overall, mRNA vaccination had a good risk-benefit profile in individuals with SCD. This mRNA vaccine study also marks the first evaluation of vaccine safety and antibody response in very young children with SCD. This trial was registered at www.ClinicalTrials.gov as #NCT05139992.
Collapse
Affiliation(s)
- Alan R. Anderson
- PRISMA Health Comprehensive Sickle Cell Disease Program, Division of Pediatric Hematology-Oncology, University of South Carolina School of Medicine, Greenville, SC
| | - John J. Strouse
- Division of Hematology, Department of Medicine, Duke University School of Medicine, Durham, NC
| | - Deepa Manwani
- Children's Hospital at Montefiore, Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY
| | - Amanda M. Brandow
- Department of Pediatrics, Section of Hematology/Oncology/Bone Marrow Transplantation, Medical College of Wisconsin and Children’s Research Institute of Children’s Wisconsin, Milwaukee, WI
| | - Elliott Vichinsky
- Benioff Children's Hospital Oakland, Department of Pediatrics, UCSF, Oakland, CA
| | - Andrew Campbell
- Department of Pediatrics, Children's National Hospital, George Washington University School of Medicine and Health Sciences, Washington, DC
| | - Patrick J. Leavey
- Simmons Comprehensive Cancer Center, Children's Medical Center, Department of Pediatrics, The University of Texas Southwestern Medical Center, Dallas, TX
| | - Alecia Nero
- Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX
| | - Ibrahim F. Ibrahim
- Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX
| | - Joshua J. Field
- Department of Medicine, Medical College of Wisconsin, Versiti Blood Research Institute, Milwaukee, WI
| | - Amanda Baer
- Department of Medicine, University of Pennsylvania, Philadelphia, PA
| | | | - Lauren Vincent
- Department of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Yan Zhao
- Department of Microbiology, University of Pennsylvania, Philadelphia, PA
| | | | - Scott E. Hensley
- Department of Microbiology, University of Pennsylvania, Philadelphia, PA
| | | | - Sophie Lanzkron
- Division of Hematology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Donna Neuberg
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA
| | - Charles S. Abrams
- Department of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
5
|
Musoles-Cuenca B, Aguiló-Gisbert J, Lorenzo-Bermejo T, Canales R, Ballester B, Romani-Cremaschi U, Martínez-Valverde R, Maiques E, Marteles D, Rueda P, Rubio V, Villanueva-Saz S, Rubio-Guerri C. Molecular and Serological Studies on Potential SARS-CoV-2 Infection among 43 Lemurs under Human Care-Evidence for Past Infection in at Least One Individual. Animals (Basel) 2023; 14:140. [PMID: 38200871 PMCID: PMC10778278 DOI: 10.3390/ani14010140] [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: 11/20/2023] [Revised: 12/24/2023] [Accepted: 12/26/2023] [Indexed: 01/12/2024] Open
Abstract
In the setting of the recent COVID-19 pandemic, transmission of SARS-CoV-2 to animals has been reported in both domestic and wild animals and is a matter of concern. Given the genetic and functional similarities to humans, non-human primates merit particular attention. In the case of lemurs, generally considered endangered, they are believed to be susceptible to SARS-CoV-2 infection. We have conducted a study for evidence of SARS-CoV-2 infection among the 43 lemurs of Mundomar, a zoological park in Benidorm, Spain. They belong to two endangered lemur species, 23 black-and-white ruffed lemurs (Varecia variegata) and 20 ring-tailed lemurs (Lemur catta). Health assessments conducted in 2022 and 2023 included molecular analyses for SARS-CoV-2 RNA of oral and rectal swabs using two different RT-qPCR assays, always with negative results for SARS-CoV-2 in all animals. The assessment also included serological testing for antibodies against the receptor-binding domain (RBD) of the spike protein (S) of SARS-CoV-2, which again yielded negative results in all animals except one black-and-white ruffed lemur, supporting prior infection of that animal with SARS-CoV-2. Our data, while not indicating a high susceptibility of lemurs to SARS-CoV-2 infection, show that they can be infected, adding to the existing information body on potential ways for SARS-CoV-2 virus spreading in zoos, highlighting the need for animal surveillance for the virus.
Collapse
Affiliation(s)
- Beatriz Musoles-Cuenca
- Department of Biomedical Sciences, Faculty of Health Sciences, Universidad Cardenal Herrera-CEU, 46113 Valencia, Spain; (B.M.-C.); (T.L.-B.); (B.B.); (E.M.)
| | - Jordi Aguiló-Gisbert
- Servicio de Análisis, Investigación, Gestión de Animales Silvestres (SAIGAS), Veterinary Faculty, Universidad Cardenal Herrera-CEU, 46113 Valencia, Spain;
| | - Teresa Lorenzo-Bermejo
- Department of Biomedical Sciences, Faculty of Health Sciences, Universidad Cardenal Herrera-CEU, 46113 Valencia, Spain; (B.M.-C.); (T.L.-B.); (B.B.); (E.M.)
| | - Rocío Canales
- Veterinary Department, Mundomar Benidorm, 03503 Alicante, Spain; (R.C.); (U.R.-C.)
| | - Beatriz Ballester
- Department of Biomedical Sciences, Faculty of Health Sciences, Universidad Cardenal Herrera-CEU, 46113 Valencia, Spain; (B.M.-C.); (T.L.-B.); (B.B.); (E.M.)
| | | | | | - Elisa Maiques
- Department of Biomedical Sciences, Faculty of Health Sciences, Universidad Cardenal Herrera-CEU, 46113 Valencia, Spain; (B.M.-C.); (T.L.-B.); (B.B.); (E.M.)
| | - Diana Marteles
- Clinical Immunology Laboratory, Veterinary Faculty, University of Zaragoza, 50013 Zaragoza, Spain; (D.M.); (P.R.)
| | - Pablo Rueda
- Clinical Immunology Laboratory, Veterinary Faculty, University of Zaragoza, 50013 Zaragoza, Spain; (D.M.); (P.R.)
| | - Vicente Rubio
- Department of Genomics and Proteomics, Instituto de Biomedicina de Valencia del Consejo Superior de Investigaciones Científicas (IBV-CSIC), 46010 Valencia, Spain
- Group 739, IBV-CSIC, Centre for Biomedical Network Research, Instituto de Salud Carlos III (CIBERER-ISCIII), 46010 Valencia, Spain
| | - Sergio Villanueva-Saz
- Clinical Immunology Laboratory, Veterinary Faculty, University of Zaragoza, 50013 Zaragoza, Spain; (D.M.); (P.R.)
| | - Consuelo Rubio-Guerri
- Department of Pharmacy, Faculty of Health Sciences, Universidad Cardenal Herrera-CEU, 46113 Valencia, Spain
| |
Collapse
|
6
|
Milligan EC, Olstad K, Williams CA, Mallory M, Cano P, Cross KA, Munt JE, Garrido C, Lindesmith L, Watanabe J, Usachenko JL, Hopkins L, Immareddy R, Shaan Lakshmanappa Y, Elizaldi SR, Roh JW, Sammak RL, Pollard RE, Yee JL, Herbek S, Scobey T, Miehlke D, Fouda G, Ferrari G, Gao H, Shen X, Kozlowski PA, Montefiori D, Hudgens MG, Edwards DK, Carfi A, Corbett KS, Graham BS, Fox CB, Tomai M, Iyer SS, Baric R, Reader R, Dittmer DP, Van Rompay KKA, Permar SR, De Paris K. Infant rhesus macaques immunized against SARS-CoV-2 are protected against heterologous virus challenge 1 year later. Sci Transl Med 2023; 15:eadd6383. [PMID: 36454813 PMCID: PMC9765459 DOI: 10.1126/scitranslmed.add6383] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
The U.S. Food and Drug Administration only gave emergency use authorization of the BNT162b2 and mRNA-1273 SARS-CoV-2 vaccines for infants 6 months and older in June 2022. Yet questions regarding the durability of vaccine efficacy, especially against emerging variants, in this age group remain. We demonstrated previously that a two-dose regimen of stabilized prefusion Washington SARS-CoV-2 S-2P spike (S) protein encoded by mRNA encapsulated in lipid nanoparticles (mRNA-LNP) or purified S-2P mixed with 3M-052, a synthetic Toll-like receptor (TLR) 7/8 agonist, in a squalene emulsion (Protein+3M-052-SE) was safe and immunogenic in infant rhesus macaques. Here, we demonstrate that broadly neutralizing and spike-binding antibodies against variants of concern (VOCs), as well as T cell responses, persisted for 12 months. At 1 year, corresponding to human toddler age, we challenged vaccinated rhesus macaques and age-matched nonvaccinated controls intranasally and intratracheally with a high dose of heterologous SARS-CoV-2 B.1.617.2 (Delta). Seven of eight control rhesus macaques exhibited severe interstitial pneumonia and high virus replication in the upper and lower respiratory tract. In contrast, vaccinated rhesus macaques had faster viral clearance with mild to no pneumonia. Neutralizing and binding antibody responses to the B.1.617.2 variant at the day of challenge correlated with lung pathology and reduced virus replication. Overall, the Protein+3M-052-SE vaccine provided superior protection to the mRNA-LNP vaccine, emphasizing opportunities for optimization of current vaccine platforms. The observed efficacy of both vaccines 1 year after vaccination supports the implementation of an early-life SARS-CoV-2 vaccine.
Collapse
Affiliation(s)
- Emma C Milligan
- Department of Microbiology and Immunology, Children's Research Institute, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Katherine Olstad
- California National Primate Research Center, University of California at Davis, Davis, CA 95616, USA
| | - Caitlin A Williams
- Department of Pediatrics, Weill Cornell Medical College, New York, NY 10065, USA
| | - Michael Mallory
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Patricio Cano
- Lineberger Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Kaitlyn A Cross
- Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jennifer E Munt
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Carolina Garrido
- Center for Immunology and Infectious Diseases, University of California at Davis, Davis, CA 95616, USA
| | - Lisa Lindesmith
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jennifer Watanabe
- California National Primate Research Center, University of California at Davis, Davis, CA 95616, USA
| | - Jodie L Usachenko
- California National Primate Research Center, University of California at Davis, Davis, CA 95616, USA
| | - Lincoln Hopkins
- California National Primate Research Center, University of California at Davis, Davis, CA 95616, USA
| | - Ramya Immareddy
- California National Primate Research Center, University of California at Davis, Davis, CA 95616, USA
| | | | - Sonny R Elizaldi
- Center for Immunology and Infectious Diseases, University of California at Davis, Davis, CA 95616, USA.,Graduate Group in Immunology, University of California at Davis, Davis, CA 95616, USA
| | - Jamin W Roh
- Center for Immunology and Infectious Diseases, University of California at Davis, Davis, CA 95616, USA.,Graduate Group in Immunology, University of California at Davis, Davis, CA 95616, USA
| | - Rebecca L Sammak
- California National Primate Research Center, University of California at Davis, Davis, CA 95616, USA
| | - Rachel E Pollard
- School of Veterinary Medicine, University of California at Davis, Davis, CA 95616, USA
| | - JoAnn L Yee
- California National Primate Research Center, University of California at Davis, Davis, CA 95616, USA
| | - Savannah Herbek
- Department of Pediatrics, Weill Cornell Medical College, New York, NY 10065, USA
| | - Trevor Scobey
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Dieter Miehlke
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC 27710, USA.,Department of Surgery, Duke University School of Medicine, Durham, NC 27710, USA.,Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Genevieve Fouda
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC 27710, USA.,Department of Surgery, Duke University School of Medicine, Durham, NC 27710, USA.,Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Guido Ferrari
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC 27710, USA.,Department of Surgery, Duke University School of Medicine, Durham, NC 27710, USA.,Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Hongmei Gao
- Department of Surgery, Duke University School of Medicine, Durham, NC 27710, USA
| | - Xiaoying Shen
- Department of Surgery, Duke University School of Medicine, Durham, NC 27710, USA
| | - Pamela A Kozlowski
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - David Montefiori
- Department of Surgery, Duke University School of Medicine, Durham, NC 27710, USA
| | - Michael G Hudgens
- Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | | | | | - Kizzmekia S Corbett
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Barney S Graham
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20852, USA
| | - Christopher B Fox
- Access to Advanced Health Institute, Seattle, WA 98102, USA.,Department of Global Health, University of Washington, Seattle, WA 98105, USA
| | - Mark Tomai
- 3M Corporate Research Materials Laboratory, Saint Paul, MN 55144, USA
| | - Smita S Iyer
- California National Primate Research Center, University of California at Davis, Davis, CA 95616, USA.,Center for Immunology and Infectious Diseases, University of California at Davis, Davis, CA 95616, USA
| | - Ralph Baric
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Rachel Reader
- California National Primate Research Center, University of California at Davis, Davis, CA 95616, USA
| | - Dirk P Dittmer
- Department of Microbiology and Immunology, Children's Research Institute, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.,Department of Pediatrics, Weill Cornell Medical College, New York, NY 10065, USA
| | - Koen K A Van Rompay
- California National Primate Research Center, University of California at Davis, Davis, CA 95616, USA.,Department of Pathology, Microbiology and Immunology, University of California at Davis, Davis, CA 95616, USA
| | - Sallie R Permar
- Department of Pediatrics, Weill Cornell Medical College, New York, NY 10065, USA
| | - Kristina De Paris
- Department of Microbiology and Immunology, Children's Research Institute, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
7
|
Korosec CS, Farhang-Sardroodi S, Dick DW, Gholami S, Ghaemi MS, Moyles IR, Craig M, Ooi HK, Heffernan JM. Long-term durability of immune responses to the BNT162b2 and mRNA-1273 vaccines based on dosage, age and sex. Sci Rep 2022; 12:21232. [PMID: 36481777 PMCID: PMC9732004 DOI: 10.1038/s41598-022-25134-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Accepted: 11/25/2022] [Indexed: 12/13/2022] Open
Abstract
The lipid nanoparticle (LNP)-formulated mRNA vaccines BNT162b2 and mRNA-1273 are a widely adopted multi vaccination public health strategy to manage the COVID-19 pandemic. Clinical trial data has described the immunogenicity of the vaccine, albeit within a limited study time frame. Here, we use a within-host mathematical model for LNP-formulated mRNA vaccines, informed by available clinical trial data from 2020 to September 2021, to project a longer term understanding of immunity as a function of vaccine type, dosage amount, age, and sex. We estimate that two standard doses of either mRNA-1273 or BNT162b2, with dosage times separated by the company-mandated intervals, results in individuals losing more than 99% humoral immunity relative to peak immunity by 8 months following the second dose. We predict that within an 8 month period following dose two (corresponding to the original CDC time-frame for administration of a third dose), there exists a period of time longer than 1 month where an individual has lost more than 99% humoral immunity relative to peak immunity, regardless of which vaccine was administered. We further find that age has a strong influence in maintaining humoral immunity; by 8 months following dose two we predict that individuals aged 18-55 have a four-fold humoral advantage compared to aged 56-70 and 70+ individuals. We find that sex has little effect on the immune response and long-term IgG counts. Finally, we find that humoral immunity generated from two low doses of mRNA-1273 decays at a substantially slower rate relative to peak immunity gained compared to two standard doses of either mRNA-1273 or BNT162b2. Our predictions highlight the importance of the recommended third booster dose in order to maintain elevated levels of antibodies.
Collapse
Affiliation(s)
- Chapin S Korosec
- Modelling Infection and Immunity Lab, Mathematics and Statistics, York University, 4700 Keele St, Toronto, ON, M3J 1P3, Canada.
- Centre for Disease Modelling, Mathematics and Statistics, York University, 4700 Keele St, Toronto, ON, M3J 1P3, Canada.
| | - Suzan Farhang-Sardroodi
- Centre for Disease Modelling, Mathematics and Statistics, York University, 4700 Keele St, Toronto, ON, M3J 1P3, Canada
- Department of Mathematics, University of Manitoba, 186 Dysart Road, Winnipeg, MB, R3T 2N2, Canada
| | - David W Dick
- Modelling Infection and Immunity Lab, Mathematics and Statistics, York University, 4700 Keele St, Toronto, ON, M3J 1P3, Canada
- Centre for Disease Modelling, Mathematics and Statistics, York University, 4700 Keele St, Toronto, ON, M3J 1P3, Canada
| | - Sameneh Gholami
- Modelling Infection and Immunity Lab, Mathematics and Statistics, York University, 4700 Keele St, Toronto, ON, M3J 1P3, Canada
- Centre for Disease Modelling, Mathematics and Statistics, York University, 4700 Keele St, Toronto, ON, M3J 1P3, Canada
| | - Mohammad Sajjad Ghaemi
- Digital Technologies Research Centre, National Research Council Canada, 222 College Street, Toronto, ON, M5T 3J1, Canada
| | - Iain R Moyles
- Modelling Infection and Immunity Lab, Mathematics and Statistics, York University, 4700 Keele St, Toronto, ON, M3J 1P3, Canada
- Centre for Disease Modelling, Mathematics and Statistics, York University, 4700 Keele St, Toronto, ON, M3J 1P3, Canada
| | - Morgan Craig
- Department of Mathematics and Statistics, Université de Montréal & Sainte-Justine University Hospital Research Centre, 3175, ch. Côte Sainte-Catherine, Montréal, QC, H3T 1C5, Canada
| | - Hsu Kiang Ooi
- Digital Technologies Research Centre, National Research Council Canada, 222 College Street, Toronto, ON, M5T 3J1, Canada
| | - Jane M Heffernan
- Modelling Infection and Immunity Lab, Mathematics and Statistics, York University, 4700 Keele St, Toronto, ON, M3J 1P3, Canada.
- Centre for Disease Modelling, Mathematics and Statistics, York University, 4700 Keele St, Toronto, ON, M3J 1P3, Canada.
| |
Collapse
|
8
|
Schmetzer C, Vogt E, Stellar L, Godonou ET, Liphardt AM, Muehlensiepen F, Vuillerme N, Hueber AJ, Kleyer A, Krönke G, Schett G, Simon D, Knitza J. Self-collection of capillary blood and saliva to determine COVID-19 vaccine immunogenicity in patients with immune-mediated inflammatory diseases and health professionals. Front Public Health 2022; 10:994770. [PMID: 36311633 PMCID: PMC9616117 DOI: 10.3389/fpubh.2022.994770] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 09/20/2022] [Indexed: 01/26/2023] Open
Abstract
Introduction Being able to independently determine vaccine induced antibody responses by minimal-invasive methods is of great interest to enable a flexible and effective vaccination strategy. This study aimed to evaluate (1) the accuracy, feasibility, usability and acceptability of capillary blood and saliva self-sampling to determine SARS-CoV-2 antibody responses in patients with immune-mediated inflammatory diseases (IMIDs) and health professionals (HP). Methods IMID patients and HP having received two doses of SARS-CoV-2 vaccines, self-collected capillary blood (Tasso+) and saliva samples. Capillary samples were considered interchangeable with venous blood if three criteria were met: Spearman's correlation coefficient (r) > 0.8, non-significant Wilcoxon signed-rank test (i.e., p > 0.05), and a small bias or 95% of tests within 10% difference through Bland-Altman. Participants completed a survey to investigate self-sampling usability (system usability scale; SUS) and acceptability (net promoter score; NPS). Study personnel monitored correct self-sampling completion and recorded protocol deviations. Results 60 participants (30 IMID patients and 30 HP) were analyzed. We observed interchangeability for capillary samples with an accuracy of 98.3/100% for Anti-SARS-CoV-2 IgG/IgA antibodies, respectively. Fifty-eight capillary blood samples and all 60 saliva samples were successfully collected within the first attempt. Usability of both self-sampling procedures was rated as excellent, with significantly higher saliva ratings (p < 0.001). Capillary self-sampling was perceived as significantly (p < 0.001) less painful compared to traditional venous blood collection. Participants reported a NPS for capillary and saliva self-sampling of +68% and +63%, respectively. The majority of both groups (73%) preferred capillary self-sampling over professional venous blood collection. Conclusion Our results indicate that capillary self-sampling is accurate, feasible and preferred over conventional venous blood collection. Implementation could enable easy access, flexible vaccination monitoring, potentially leading to a better protection of vulnerable patient groups. Self-collection of saliva is feasible and safe however more work is needed to determine its application in clinical practice.
Collapse
Affiliation(s)
- Caroline Schmetzer
- Department of Internal Medicine 3 – Rheumatology and Immunology Friedrich-Alexander University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany,Deutsches Zentrum für Immuntherapie, Friedrich-Alexander University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | | | | | - Elie-Tino Godonou
- Department of Internal Medicine 3 – Rheumatology and Immunology Friedrich-Alexander University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany,Deutsches Zentrum für Immuntherapie, Friedrich-Alexander University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Anna-Maria Liphardt
- Department of Internal Medicine 3 – Rheumatology and Immunology Friedrich-Alexander University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany,Deutsches Zentrum für Immuntherapie, Friedrich-Alexander University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Felix Muehlensiepen
- Centre for Health Services Research Brandenburg, Brandenburg Medical School, Neuruppin, Germany,Faculty of Health Sciences Brandenburg, Brandenburg Medical School, Neuruppin, Germany,Université Grenoble Alpes, AGEIS, Grenoble, France
| | - Nicolas Vuillerme
- Université Grenoble Alpes, AGEIS, Grenoble, France,Institut Universitaire de France, Paris, France,LabCom Telecom4Health, Orange Labs and Univ. Grenoble Alpes, CNRS, Inria, Grenoble INP-UGA, Grenoble, France
| | - Axel J. Hueber
- Department of Internal Medicine 3 – Rheumatology and Immunology Friedrich-Alexander University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany,Division of Rheumatology, Klinikum Nürnberg, Paracelsus Medical University, Nürnberg, Germany
| | - Arnd Kleyer
- Department of Internal Medicine 3 – Rheumatology and Immunology Friedrich-Alexander University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany,Deutsches Zentrum für Immuntherapie, Friedrich-Alexander University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Gerhard Krönke
- Department of Internal Medicine 3 – Rheumatology and Immunology Friedrich-Alexander University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany,Deutsches Zentrum für Immuntherapie, Friedrich-Alexander University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Georg Schett
- Department of Internal Medicine 3 – Rheumatology and Immunology Friedrich-Alexander University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany,Deutsches Zentrum für Immuntherapie, Friedrich-Alexander University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - David Simon
- Department of Internal Medicine 3 – Rheumatology and Immunology Friedrich-Alexander University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany,Deutsches Zentrum für Immuntherapie, Friedrich-Alexander University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Johannes Knitza
- Department of Internal Medicine 3 – Rheumatology and Immunology Friedrich-Alexander University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany,Deutsches Zentrum für Immuntherapie, Friedrich-Alexander University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany,Université Grenoble Alpes, AGEIS, Grenoble, France,*Correspondence: Johannes Knitza
| |
Collapse
|
9
|
Fernandes RS, de Oliveira Silva J, Gomes KB, Azevedo RB, Townsend DM, de Paula Sabino A, Branco de Barros AL. Recent advances in point of care testing for COVID-19 detection. Biomed Pharmacother 2022; 153:113538. [PMID: 36076617 PMCID: PMC9371983 DOI: 10.1016/j.biopha.2022.113538] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/03/2022] [Accepted: 08/04/2022] [Indexed: 12/23/2022] Open
Abstract
The World Health Organizations declaration of the COVID-19 pandemic was a milestone for the scientific community. The high transmission rate and the huge number of deaths, along with the lack of knowledge about the virus and the evolution of the disease, stimulated a relentless search for diagnostic tests, treatments, and vaccines. The main challenges were the differential diagnosis of COVID-19 and the development of specific, rapid, and sensitive tests that could reach all people. RT-PCR remains the gold standard for diagnosing COVID-19. However, new methods, such as other molecular techniques and immunoassays emerged. Also, the need for accessible tests with quick results boosted the development of point of care tests (POCT) that are fast, and automated, with high precision and accuracy. This assay reduces the dependence on laboratory conditions and mass testing of the population, dispersing the pressure regarding screening and detection. This review summarizes the advances in the diagnostic field since the pandemic started, emphasizing various laboratory techniques for detecting COVID-19. We reviewed the main existing diagnostic methods, as well as POCT under development, starting with RT-PCR detection, but also exploring other nucleic acid techniques, such as digital PCR, loop-mediated isothermal amplification-based assay (RT-LAMP), clustered regularly interspaced short palindromic repeats (CRISPR), and next-generation sequencing (NGS), and immunoassay tests, and nanoparticle-based biosensors, developed as portable instruments for the rapid standard diagnosis of COVID-19.
Collapse
|
10
|
Menges D, Zens KD, Ballouz T, Caduff N, Llanas-Cornejo D, Aschmann HE, Domenghino A, Pellaton C, Perreau M, Fenwick C, Pantaleo G, Kahlert CR, Münz C, Puhan MA, Fehr JS. Heterogenous humoral and cellular immune responses with distinct trajectories post-SARS-CoV-2 infection in a population-based cohort. Nat Commun 2022; 13:4855. [PMID: 35982045 PMCID: PMC9386650 DOI: 10.1038/s41467-022-32573-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Accepted: 08/06/2022] [Indexed: 12/14/2022] Open
Abstract
To better understand the development of SARS-CoV-2-specific immunity over time, a detailed evaluation of humoral and cellular responses is required. Here, we characterize anti-Spike (S) IgA and IgG in a representative population-based cohort of 431 SARS-CoV-2-infected individuals up to 217 days after diagnosis, demonstrating that 85% develop and maintain anti-S responses. In a subsample of 64 participants, we further assess anti-Nucleocapsid (N) IgG, neutralizing antibody activity, and T cell responses to Membrane (M), N, and S proteins. In contrast to S-specific antibody responses, anti-N IgG levels decline substantially over time and neutralizing activity toward Delta and Omicron variants is low to non-existent within just weeks of Wildtype SARS-CoV-2 infection. Virus-specific T cells are detectable in most participants, albeit more variable than antibody responses. Cluster analyses of the co-evolution of antibody and T cell responses within individuals identify five distinct trajectories characterized by specific immune patterns and clinical factors. These findings demonstrate the relevant heterogeneity in humoral and cellular immunity to SARS-CoV-2 while also identifying consistent patterns where antibody and T cell responses may work in a compensatory manner to provide protection. The persistence of the immune response to SARS-CoV-2 after recovery from infection is an indicator for subsequent protection against infection. Here the authors follow recovered patients and measure antibody and T cell responses and find that these two parts of the immune response may have different longevity.
Collapse
Affiliation(s)
- Dominik Menges
- Epidemiology, Biostatistics and Prevention Institute (EBPI), University of Zurich (UZH), Zurich, Switzerland
| | - Kyra D Zens
- Epidemiology, Biostatistics and Prevention Institute (EBPI), University of Zurich (UZH), Zurich, Switzerland.,Institute for Experimental Immunology, University of Zurich (UZH), Zurich, Switzerland
| | - Tala Ballouz
- Epidemiology, Biostatistics and Prevention Institute (EBPI), University of Zurich (UZH), Zurich, Switzerland
| | - Nicole Caduff
- Epidemiology, Biostatistics and Prevention Institute (EBPI), University of Zurich (UZH), Zurich, Switzerland.,Institute for Experimental Immunology, University of Zurich (UZH), Zurich, Switzerland
| | - Daniel Llanas-Cornejo
- Epidemiology, Biostatistics and Prevention Institute (EBPI), University of Zurich (UZH), Zurich, Switzerland
| | - Hélène E Aschmann
- Epidemiology, Biostatistics and Prevention Institute (EBPI), University of Zurich (UZH), Zurich, Switzerland.,Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, USA
| | - Anja Domenghino
- Epidemiology, Biostatistics and Prevention Institute (EBPI), University of Zurich (UZH), Zurich, Switzerland.,Department of Visceral and Transplantation Surgery, University Hospital Zurich (USZ), University of Zurich (UZH), Zurich, Switzerland
| | - Céline Pellaton
- Service of Immunology and Allergy, Lausanne University Hospital (CHUV), University of Lausanne (UNIL), Lausanne, Switzerland
| | - Matthieu Perreau
- Service of Immunology and Allergy, Lausanne University Hospital (CHUV), University of Lausanne (UNIL), Lausanne, Switzerland
| | - Craig Fenwick
- Service of Immunology and Allergy, Lausanne University Hospital (CHUV), University of Lausanne (UNIL), Lausanne, Switzerland
| | - Giuseppe Pantaleo
- Service of Immunology and Allergy, Lausanne University Hospital (CHUV), University of Lausanne (UNIL), Lausanne, Switzerland
| | - Christian R Kahlert
- Division of Infectious Diseases and Hospital Epidemiology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland.,Division of Infectious Diseases and Hospital Epidemiology, Children's Hospital of Eastern Switzerland, St. Gallen, Switzerland
| | - Christian Münz
- Institute for Experimental Immunology, University of Zurich (UZH), Zurich, Switzerland
| | - Milo A Puhan
- Epidemiology, Biostatistics and Prevention Institute (EBPI), University of Zurich (UZH), Zurich, Switzerland.
| | - Jan S Fehr
- Epidemiology, Biostatistics and Prevention Institute (EBPI), University of Zurich (UZH), Zurich, Switzerland
| |
Collapse
|
11
|
Schmidt AE, Vogel P, Chastain CA, Barnes T, Roth NJ, Simon TL. Analysis of 52 240 source plasma donors of convalescent COVID-19 plasma: Sex, ethnicity, and age association with initial antibody levels and rate of dissipation. J Clin Apher 2022; 37:449-459. [PMID: 35815776 PMCID: PMC9350246 DOI: 10.1002/jca.21998] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 06/22/2022] [Accepted: 06/22/2022] [Indexed: 11/11/2022]
Abstract
Background COVID‐19 convalescent plasma (CCP) was approved under emergency authorization to treat critically ill patients with COVID‐19 in the United States in 2020. We explored the demographics of donors contributing plasma for a hyperimmune, plasma‐derived therapy to evaluate factors that may be associated with anti‐SARS‐CoV‐2 antibody response variability and, subsequently, antibody titers. Study Design An electronic search of CCP donors was performed across 282 US plasma donation centers. Donations were screened for nucleocapsid protein‐binding‐IgG using the Abbott SARS‐CoV‐2 IgG assay. Results Overall, 52 240 donors donated 418 046 units of CCP. Donors were of various ethnicities: 43% Caucasian, 34% Hispanic, 17% African American, 2% Native American, 1% Asian, and 3% other. Females had higher initial mean anti‐SARS‐CoV‐2 antibody titers but an overall faster rate of decline (P < .0001). Initial antibody titers increased with age: individuals aged 55 to 66 years had elevated anti‐SARS‐CoV‐2 titers for longer periods compared with other ages (P = .0004). African American donors had the lowest initial antibody titers but a slower rate of decline (P < .0001), while Caucasian (P = .0088) and Hispanic (P = .0193) groups had the fastest rates of decline. Most donor antibody levels decreased below the inclusion criteria (≥1.50) within 30 to 100 days of first donation, but donation frequency did not appear to be associated with rate of decline. Conclusion Several factors may be associated with anti‐SARS‐CoV‐2 antibody response including donor age and sex. Evaluating these factors during development of future hyperimmune globulin products may help generation of therapies with optimal efficacy.
Collapse
|
12
|
Niedrist T, Drexler C, Torreiter PP, Matejka J, Strahlhofer-Augsten M, Kral S, Riegler S, Gülly C, Zurl C, Kriegl L, Krause R, Berghold A, Steinmetz I, Schlenke P, Herrmann M. Longitudinal comparison of automated SARS-CoV-2 serology assays in assessing virus neutralization capacity in COVID-19 convalescent sera. Arch Pathol Lab Med 2022; 146:538-546. [PMID: 35085385 DOI: 10.5858/arpa.2021-0604-sa] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/24/2022] [Indexed: 11/06/2022]
Abstract
CONTEXT.– Serological tests on automated immunology analyzers are increasingly used to monitor the acquired immunity against SARS-CoV-2. The heterogeneity of assays raises concerns about their diagnostic performance and comparability. OBJECTIVE.– To test sera from formerly infected individuals for SARS-Cov-2 antibodies utilizing six automated serology assays and a pseudoneutralization test (PNT). DESIGN.– Six SARS-CoV-2 serology assays were utilized to assess 954 samples collected during a 12 months period from 315 COVID-19 convalescents. The tests determined either antibodies against the viral nucleocapsid (anti-NC) or spike protein (anti-S). Two assays did not distinguish between antibody classes whereas the others selectively measured immunoglubulins G (IgG) antibodies. PNT was used to detect the presence of neutralizing antibodies. RESULTS.– Comparison of qualitative results showed only slight to moderate concordance between the assays (Cohen's kappa < 0.57). Significant correlations (P < .001) were observed between the antibody titers from all quantitative assays. However, titer changes were not detected equally. A total anti-S assay measured an increase in 128 out of 172 cases (74%) of a suitable subset, whereas all IgG anti-S tests reported decreases in at least 118 (69%). Regarding the PNT results, diagnostic sensitivities ≥89% were achieved with PPVs ≥93%. In contrast, specificity changed substantially over time varying from 20 to 100%. CONCLUSIONS.– Comparability of serological SARS-CoV-2 antibody tests is rather poor. Due to different diagnostic specificities, the tested assays were not equally capable of capturing changes in antibody titers. However, with thoroughly validated cut-offs, IgG-selective anti-S assays are a reliable surrogate test for SARS-CoV-2 neutralizing antibodies in former COVID-19 patients.
Collapse
Affiliation(s)
- Tobias Niedrist
- Clinical Institute of Medical and Chemical Laboratory Diagnostics (Niedrist, Herrmann), Medical University of Graz, Graz, Austria
| | - Camilla Drexler
- Department of Blood Group Serology and Transfusion Medicine (Drexler, Torreiter, Schlenke), Medical University of Graz, Graz, Austria
| | - Patrick Paul Torreiter
- Department of Blood Group Serology and Transfusion Medicine (Drexler, Torreiter, Schlenke), Medical University of Graz, Graz, Austria
| | - Julia Matejka
- Biobank Graz (Matejka, Strahlhofer-Augsten, Kral, Riegler, Gülly), Medical University of Graz, Graz, Austria
| | - Manuela Strahlhofer-Augsten
- Biobank Graz (Matejka, Strahlhofer-Augsten, Kral, Riegler, Gülly), Medical University of Graz, Graz, Austria
| | - Sabrina Kral
- Biobank Graz (Matejka, Strahlhofer-Augsten, Kral, Riegler, Gülly), Medical University of Graz, Graz, Austria
| | - Skaiste Riegler
- Biobank Graz (Matejka, Strahlhofer-Augsten, Kral, Riegler, Gülly), Medical University of Graz, Graz, Austria
| | - Christian Gülly
- Biobank Graz (Matejka, Strahlhofer-Augsten, Kral, Riegler, Gülly), Medical University of Graz, Graz, Austria.,Center for Medical Research (Gülly), Medical University of Graz, Graz, Austria
| | - Christoph Zurl
- Division of Infectious Diseases in the Department of Internal Medicine (Zurl, Kriegl, Krause), Medical University of Graz, Graz, Austria.,Division of General Paediatrics in the Department of Paediatrics and Adolescents Medicine (Zurl), Medical University of Graz, Graz, Austria
| | - Lisa Kriegl
- Division of Infectious Diseases in the Department of Internal Medicine (Zurl, Kriegl, Krause), Medical University of Graz, Graz, Austria
| | - Robert Krause
- Division of Infectious Diseases in the Department of Internal Medicine (Zurl, Kriegl, Krause), Medical University of Graz, Graz, Austria.,BioTechMed Graz, Graz, Austria (Krause)
| | - Andrea Berghold
- Institute for Medical Informatics, Statistics and Documentation (Berghold), Medical University of Graz, Graz, Austria
| | - Ivo Steinmetz
- Diagnostic and Research Institute of Hygiene, Microbiology and Environmental Medicine (Steinmetz), Medical University of Graz, Graz, Austria
| | - Peter Schlenke
- Department of Blood Group Serology and Transfusion Medicine (Drexler, Torreiter, Schlenke), Medical University of Graz, Graz, Austria
| | - Markus Herrmann
- Clinical Institute of Medical and Chemical Laboratory Diagnostics (Niedrist, Herrmann), Medical University of Graz, Graz, Austria
| |
Collapse
|
13
|
Stinca S, Barnes TW, Vogel P, Meyers W, Schulte-Pelkum J, Filchtinski D, Steller L, Hauser T, Manni S, Gardiner DF, Popik S, Roth NJ, Schuetz P. Modelling the concentration of anti-SARS-CoV-2 immunoglobulin G in intravenous immunoglobulin product batches. PLoS One 2021; 16:e0259731. [PMID: 34843493 PMCID: PMC8629175 DOI: 10.1371/journal.pone.0259731] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 10/24/2021] [Indexed: 11/18/2022] Open
Abstract
Background Plasma-derived intravenous immunoglobulin (IVIg) products contain a dynamic spectrum of immunoglobulin (Ig) G reactivities reflective of the donor population from which they are derived. We sought to model the concentration of anti-severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) IgG which could be expected in future plasma pool and final-product batches of CSL Behring’s immunoglobulin product Privigen. Study design and methods Data was extracted from accessible databases, including the incidence of coronavirus disease 2019 and SARS-CoV-2 vaccination status, antibody titre in convalescent and vaccinated groups and antibody half-life. Together, these parameters were used to create an integrated mathematical model that could be used to predict anti-SARS-CoV-2 antibody levels in future IVIg preparations. Results We predict that anti-SARS-CoV-2 IgG concentration will peak in batches produced in mid-October 2021, containing levels in the vicinity of 190-fold that of the mean convalescent (unvaccinated) plasma concentration. An elevated concentration (approximately 35-fold convalescent plasma) is anticipated to be retained in batches produced well into 2022. Measurement of several Privigen batches using the Phadia™ EliA™ SARS-CoV-2-Sp1 IgG binding assay confirmed the early phase of this model. Conclusion The work presented in this paper may have important implications for physicians and patients who use Privigen for indicated diseases.
Collapse
Affiliation(s)
- Sara Stinca
- Department of Bioanalytical Sciences, Plasma Product Development, Research & Development, CSL Behring AG, Bern, Switzerland
| | - Thomas W. Barnes
- Department of Bioanalytical Sciences, Plasma Product Development, Research & Development, CSL Behring AG, Bern, Switzerland
| | - Peter Vogel
- Global Digital Core, Plasma Product Development, Research & Development, CSL Behring Innovation GmbH, Marburg, Germany
| | - Wilfried Meyers
- Global Digital Core, Plasma Product Development, Research & Development, CSL Behring Innovation GmbH, Marburg, Germany
| | | | - Daniel Filchtinski
- Assay Design, Thermo Fisher Scientific ImmunoDiagnostics Phadia GmbH, Freiburg, Germany
| | - Laura Steller
- Assay Design, Thermo Fisher Scientific ImmunoDiagnostics Phadia GmbH, Freiburg, Germany
| | - Thomas Hauser
- Department of Bioanalytical Sciences, Plasma Product Development, Research & Development, CSL Behring AG, Bern, Switzerland
| | - Sandro Manni
- Department of Bioanalytical Sciences, Plasma Product Development, Research & Development, CSL Behring AG, Bern, Switzerland
| | - David F. Gardiner
- Immunology, CSL Behring, King of Prussia, Pennsylvania, United States of America
| | - Sharon Popik
- Immunology, CSL Behring, King of Prussia, Pennsylvania, United States of America
| | - Nathan J. Roth
- Department of Bioanalytical Sciences, Plasma Product Development, Research & Development, CSL Behring AG, Bern, Switzerland
| | - Patrick Schuetz
- Department of Bioanalytical Sciences, Plasma Product Development, Research & Development, CSL Behring AG, Bern, Switzerland
- * E-mail:
| |
Collapse
|