1
|
Yan Y, Ito K, Fukuda H, Nojiri S, Urasaki W, Yamamoto T, Horiuchi Y, Hori S, Takahashi K, Naito T, Tabe Y. SARS-CoV-2 seroprevalence among healthcare workers in a highly vaccinated Japanese medical center from 2020-2023. Hum Vaccin Immunother 2024; 20:2337984. [PMID: 38622888 PMCID: PMC11027999 DOI: 10.1080/21645515.2024.2337984] [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: 12/06/2023] [Accepted: 03/28/2024] [Indexed: 04/17/2024] Open
Abstract
Infection-induced SARS-CoV-2 seroprevalence has been studied worldwide. At Juntendo University Hospital (JUH) in Tokyo, Japan, we have consistently conducted serological studies using the blood residue of healthcare workers (HCWs) at annual health examinations since 2020. In this 2023 study (n = 3,594), N-specific seroprevalence (infection-induced) was examined while univariate and multivariate logistic regression analyses were performed to compute ORs of seroprevalence with respect to basic characteristics of participants. We found that the N-specific seroprevalence in 2023 was 54.1%-a jump from 17.7% in 2022, and 1.6% in 2021-with 37.9% as non-PCR-confirmed asymptomatic infection cases. Those younger than 50 (adjusted OR = 1.62; p < .001) and recipients with 4 doses or less of vaccine had a higher risk to be N-positive, ranging from 1.45 times higher for the participants with 4 doses (p < .001) to 4.31 times higher for the participants with 1 dose (p < .001), compared to those with 5 or more doses. Our findings indicate that robust vaccination programs may have helped alleviate symptoms but consequently caused asymptomatic spread in this hospital, especially among younger HCWs. Although having four doses or less was found to be associated with higher risk of infection, the optimal constitution and intervals for effective booster vaccines warrant further investigations.
Collapse
Affiliation(s)
- Yan Yan
- Department of General Medicine, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Kanami Ito
- Department of Safety and Health Promotion, Juntendo University, Tokyo, Japan
| | - Hiroshi Fukuda
- Department of General Medicine, Juntendo University Faculty of Medicine, Tokyo, Japan
- Department of Safety and Health Promotion, Juntendo University, Tokyo, Japan
| | - Shuko Nojiri
- Medical Technology Innovation Center, Juntendo University, Tokyo, Japan
| | - Wataru Urasaki
- Clinical Research and Trial Center, Juntendo University, Tokyo, Japan
- Graduate School of Science and Technology, Tokyo University of Science, Tokyo, Japan
| | - Takamasa Yamamoto
- Department of Clinical Laboratory, Juntendo University Hospital, Tokyo, Japan
| | - Yuki Horiuchi
- Department of Clinical Laboratory Medicine, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Satoshi Hori
- Infection Control Science, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Kazuhisa Takahashi
- Department of Respiratory Medicine, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Toshio Naito
- Department of General Medicine, Juntendo University Faculty of Medicine, Tokyo, Japan
- Department of Safety and Health Promotion, Juntendo University, Tokyo, Japan
| | - Yoko Tabe
- Department of Clinical Laboratory, Juntendo University Hospital, Tokyo, Japan
- Department of Clinical Laboratory Medicine, Juntendo University Faculty of Medicine, Tokyo, Japan
| |
Collapse
|
2
|
Pinto PBA, Timis J, Chuensirikulchai K, Li QH, Lu HH, Maule E, Nguyen M, Alves RPDS, Verma SK, Ana-Sosa-Batiz F, Valentine K, Landeras-Bueno S, Kim K, Hastie K, Saphire EO, Alves A, Elong Ngono A, Shresta S. Co-immunization with spike and nucleocapsid based DNA vaccines for long-term protective immunity against SARS-CoV-2 Omicron. NPJ Vaccines 2024; 9:252. [PMID: 39702529 PMCID: PMC11659323 DOI: 10.1038/s41541-024-01043-3] [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: 03/06/2024] [Accepted: 12/01/2024] [Indexed: 12/21/2024] Open
Abstract
The continuing evolution of SARS-CoV-2 variants challenges the durability of existing spike (S)-based COVID-19 vaccines. We hypothesized that vaccines composed of both S and nucleocapsid (N) antigens would increase the durability of protection by strengthening and broadening cellular immunity compared with S-based vaccines. To test this, we examined the immunogenicity and efficacy of wild-type SARS-CoV-2 S- and N-based DNA vaccines administered individually or together to K18-hACE2 mice. S, N, and S + N vaccines all elicited polyfunctional CD4+ and CD8+ T cell responses and provided short-term cross-protection against Beta and Omicron BA.2 variants, but only co-immunization with S + N vaccines provided long-term protection against Omicron BA.2. Depletion of CD4+ and CD8+ T cells reduced the long-term efficacy, demonstrating a crucial role for T cells in the durability of protection. These findings underscore the potential to enhance long-lived protection against SARS-CoV-2 variants by combining S and N antigens in next-generation COVID-19 vaccines.
Collapse
Affiliation(s)
- Paolla Beatriz Almeida Pinto
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, 92037, USA
- Laboratory of Biotechnology and Physiology of Viral Infections, Oswaldo Cruz Institute, Fiocruz, Rio de Janeiro, 21040-900, Brazil
| | - Julia Timis
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, 92037, USA
| | - Kantinan Chuensirikulchai
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, 92037, USA
- Department of Microbiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Qin Hui Li
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, 92037, USA
| | - Hsueh Han Lu
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, 92037, USA
| | - Erin Maule
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, 92037, USA
| | - Michael Nguyen
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, 92037, USA
| | | | | | | | - Kristen Valentine
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, 92037, USA
| | - Sara Landeras-Bueno
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, 92037, USA
- University Cardenal Herrera-CEU, CEU Universities, Valencia, 46113, Spain
| | - Kenneth Kim
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, 92037, USA
- Microscopy and Histology Core Facility, La Jolla Institute for Immunology, La Jolla, 92037, USA
| | - Kathryn Hastie
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, 92037, USA
| | - Erica Ollmann Saphire
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, 92037, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California San Diego, La Jolla, 92093, USA
| | - Ada Alves
- Laboratory of Biotechnology and Physiology of Viral Infections, Oswaldo Cruz Institute, Fiocruz, Rio de Janeiro, 21040-900, Brazil
| | - Annie Elong Ngono
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, 92037, USA.
| | - Sujan Shresta
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, 92037, USA.
- Department of Pediatrics, Division of Host-Microbe Systems and Therapeutics, University of California San Diego, La Jolla, 92093, USA.
| |
Collapse
|
3
|
El-Maradny YA, Badawy MA, Mohamed KI, Ragab RF, Moharm HM, Abdallah NA, Elgammal EM, Rubio-Casillas A, Uversky VN, Redwan EM. Unraveling the role of the nucleocapsid protein in SARS-CoV-2 pathogenesis: From viral life cycle to vaccine development. Int J Biol Macromol 2024; 279:135201. [PMID: 39216563 DOI: 10.1016/j.ijbiomac.2024.135201] [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: 05/30/2024] [Revised: 08/24/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND The nucleocapsid protein (N protein) is the most abundant protein in SARS-CoV-2. Viral RNA and this protein are bound by electrostatic forces, forming cytoplasmic helical structures known as nucleocapsids. Subsequently, these nucleocapsids interact with the membrane (M) protein, facilitating virus budding into early secretory compartments. SCOPE OF REVIEW Exploring the role of the N protein in the SARS-CoV-2 life cycle, pathogenesis, post-sequelae consequences, and interaction with host immunity has enhanced our understanding of its function and potential strategies for preventing SARS-CoV-2 infection. MAJOR CONCLUSION This review provides an overview of the N protein's involvement in SARS-CoV-2 infectivity, highlighting its crucial role in the virus-host protein interaction and immune system modulation, which in turn influences viral spread. GENERAL SIGNIFICANCE Understanding these aspects identifies the N protein as a promising target for developing effective antiviral treatments and vaccines against SARS-CoV-2.
Collapse
Affiliation(s)
- Yousra A El-Maradny
- Pharmaceutical and Fermentation Industries Development Center, City of Scientific Research and Technological Applications (SRTA-City), New Borg EL-Arab, Alexandria 21934, Egypt; Microbiology and Immunology, Faculty of Pharmacy, Arab Academy for Science, Technology and Maritime Transport (AASTMT), El Alamein 51718, Egypt.
| | - Moustafa A Badawy
- Industrial Microbiology and Applied Chemistry program, Faculty of Science, Alexandria University, Egypt.
| | - Kareem I Mohamed
- Microbiology and Immunology, Faculty of Pharmacy, Arab Academy for Science, Technology and Maritime Transport (AASTMT), El Alamein 51718, Egypt.
| | - Renad F Ragab
- Microbiology and Immunology, Faculty of Pharmacy, Arab Academy for Science, Technology and Maritime Transport (AASTMT), El Alamein 51718, Egypt.
| | - Hamssa M Moharm
- Genetics, Biotechnology Department, Faculty of Agriculture, Alexandria University, Egypt.
| | - Nada A Abdallah
- Medicinal Plants Department, Faculty of Agriculture, Alexandria University, Egypt.
| | - Esraa M Elgammal
- Microbiology and Immunology, Faculty of Pharmacy, Arab Academy for Science, Technology and Maritime Transport (AASTMT), El Alamein 51718, Egypt.
| | - Alberto Rubio-Casillas
- Autlan Regional Hospital, Health Secretariat, Autlan, JAL 48900, Mexico; Biology Laboratory, Autlan Regional Preparatory School, University of Guadalajara, Autlan, JAL 48900, Mexico.
| | - Vladimir N Uversky
- Department of Molecular Medicine and USF Health Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA.
| | - Elrashdy M Redwan
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia; Centre of Excellence in Bionanoscience Research, King Abdulaziz University, Jeddah 21589, Saudi Arabia; Therapeutic and Protective Proteins Laboratory, Protein Research Department, Genetic Engineering and Biotechnology Research Institute, City of Scientific Research and Technological Applications (SRTA-City), New Borg EL-Arab, 21934 Alexandria, Egypt.
| |
Collapse
|
4
|
Vránová L, Poláková I, Vaníková Š, Saláková M, Musil J, Vaníčková M, Vencálek O, Holub M, Bohoněk M, Řezáč D, Dresler J, Tachezy R, Šmahel M. Multiparametric analysis of the specific immune response against SARS-CoV-2. Infect Dis (Lond) 2024; 56:851-869. [PMID: 38805304 DOI: 10.1080/23744235.2024.2358379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 04/24/2024] [Accepted: 05/17/2024] [Indexed: 05/30/2024] Open
Abstract
BACKGROUND SARS-CoV-2, which causes COVID-19, has killed more than 7 million people worldwide. Understanding the development of postinfectious and postvaccination immune responses is necessary for effective treatment and the introduction of appropriate antipandemic measures. OBJECTIVES We analysed humoral and cell-mediated anti-SARS-CoV-2 immune responses to spike (S), nucleocapsid (N), membrane (M), and open reading frame (O) proteins in individuals collected up to 1.5 years after COVID-19 onset and evaluated immune memory. METHODS Peripheral blood mononuclear cells and serum were collected from patients after COVID-19. Sampling was performed in two rounds: 3-6 months after infection and after another year. Most of the patients were vaccinated between samplings. SARS-CoV-2-seronegative donors served as controls. ELISpot assays were used to detect SARS-CoV-2-specific T and B cells using peptide pools (S, NMO) or recombinant proteins (rS, rN), respectively. A CEF peptide pool consisting of selected viral epitopes was applied to assess the antiviral T-cell response. SARS-CoV-2-specific antibodies were detected via ELISA and a surrogate virus neutralisation assay. RESULTS We confirmed that SARS-CoV-2 infection induces the establishment of long-term memory IgG+ B cells and memory T cells. We also found that vaccination enhanced the levels of anti-S memory B and T cells. Multivariate comparison also revealed the benefit of repeated vaccination. Interestingly, the T-cell response to CEF was lower in patients than in controls. CONCLUSION This study supports the importance of repeated vaccination for enhancing immunity and suggests a possible long-term perturbation of the overall antiviral immune response caused by SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Lucie Vránová
- Department of Genetics and Microbiology, Faculty of Science, Charles University, BIOCEV, Vestec, Czech Republic
| | - Ingrid Poláková
- Department of Genetics and Microbiology, Faculty of Science, Charles University, BIOCEV, Vestec, Czech Republic
| | - Šárka Vaníková
- Department of Immunomonitoring and Flow Cytometry, Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | - Martina Saláková
- Department of Genetics and Microbiology, Faculty of Science, Charles University, BIOCEV, Vestec, Czech Republic
| | - Jan Musil
- Department of Immunomonitoring and Flow Cytometry, Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | - Marie Vaníčková
- Department of Genetics and Microbiology, Faculty of Science, Charles University, BIOCEV, Vestec, Czech Republic
| | - Ondřej Vencálek
- Department of Mathematical Analysis and Applications of Mathematics, Faculty of Science, Palacky University in Olomouc, Olomouc, Czech Republic
| | - Michal Holub
- Department of Infectious Diseases, First Faculty of Medicine, Military University Hospital Prague and Charles University, Prague, Czech Republic
| | - Miloš Bohoněk
- Department of Hematology and Blood Transfusion, Military University Hospital Prague, Prague, Czech Republic
- Faculty of Biomedical Engineering, Czech Technical University, Prague, Czech Republic
| | - David Řezáč
- Department of Infectious Diseases, First Faculty of Medicine, Military University Hospital Prague and Charles University, Prague, Czech Republic
| | - Jiří Dresler
- Military Health Institute, Military Medical Agency, Prague, Czech Republic
| | - Ruth Tachezy
- Department of Genetics and Microbiology, Faculty of Science, Charles University, BIOCEV, Vestec, Czech Republic
| | - Michal Šmahel
- Department of Genetics and Microbiology, Faculty of Science, Charles University, BIOCEV, Vestec, Czech Republic
| |
Collapse
|
5
|
Schoefbaenker M, Günther T, Lorentzen EU, Romberg ML, Hennies MT, Neddermeyer R, Müller MM, Mellmann A, Bojarzyn CR, Lenz G, Stelljes M, Hrincius ER, Vollenberg R, Ludwig S, Tepasse PR, Kühn JE. Characterisation of the antibody-mediated selective pressure driving intra-host evolution of SARS-CoV-2 in prolonged infection. PLoS Pathog 2024; 20:e1012624. [PMID: 39405332 PMCID: PMC11508484 DOI: 10.1371/journal.ppat.1012624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 10/25/2024] [Accepted: 09/28/2024] [Indexed: 10/26/2024] Open
Abstract
Neutralising antibodies against the SARS-CoV-2 spike (S) protein are major determinants of protective immunity, though insufficient antibody responses may cause the emergence of escape mutants. We studied the humoral immune response causing intra-host evolution in a B-cell depleted, haemato-oncologic patient experiencing clinically severe, prolonged SARS-CoV-2 infection with a virus of lineage B.1.177.81. Following bamlanivimab treatment at an early stage of infection, the patient developed a bamlanivimab-resistant mutation, S:S494P. After five weeks of apparent genetic stability, the emergence of additional substitutions and deletions within the N-terminal domain (NTD) and the receptor binding domain (RBD) of S was observed. Notably, the composition and frequency of escape mutations changed in a short period with an unprecedented dynamic. The triple mutant S:Delta141-4 E484K S494P became dominant until virus elimination. Routine serology revealed no evidence of an antibody response in the patient. A detailed analysis of the variant-specific immune response by pseudotyped virus neutralisation test, surrogate virus neutralisation test, and immunoglobulin-capture enzyme immunoassay showed that the onset of an IgM-dominated antibody response coincided with the appearance of escape mutations. The formation of neutralising antibodies against S:Delta141-4 E484K S494P correlated with virus elimination. One year later, the patient experienced clinically mild re-infection with Omicron BA.1.18, which was treated with sotrovimab and resulted in an increase in Omicron-reactive antibodies. In conclusion, the onset of an IgM-dominated endogenous immune response in an immunocompromised patient coincided with the appearance of additional mutations in the NTD and RBD of S in a bamlanivimab-resistant virus. Although virus elimination was ultimately achieved, this humoral immune response escaped detection by routine diagnosis and created a situation temporarily favouring the rapid emergence of various antibody escape mutants with known epidemiological relevance.
Collapse
Affiliation(s)
| | - Theresa Günther
- Institute of Virology Muenster, University of Muenster, Muenster, Germany
| | - Eva Ulla Lorentzen
- Institute of Virology Muenster, University of Muenster, Muenster, Germany
| | | | - Marc Tim Hennies
- Institute of Virology Muenster, University of Muenster, Muenster, Germany
| | - Rieke Neddermeyer
- Institute of Virology Muenster, University of Muenster, Muenster, Germany
| | | | - Alexander Mellmann
- Institute of Hygiene, University Hospital Muenster, University of Muenster, Muenster, Germany
| | | | - Georg Lenz
- Department of Medicine A, Haematology, Oncology and Pneumology, University Hospital Muenster, Muenster, Germany
| | - Matthias Stelljes
- Department of Medicine A, Haematology, Oncology and Pneumology, University Hospital Muenster, Muenster, Germany
| | | | - Richard Vollenberg
- Department of Medicine B for Gastroenterology, Hepatology, Endocrinology and Clinical Infectiology, University Hospital Muenster, Muenster, Germany
| | - Stephan Ludwig
- Institute of Virology Muenster, University of Muenster, Muenster, Germany
| | - Phil-Robin Tepasse
- Department of Medicine B for Gastroenterology, Hepatology, Endocrinology and Clinical Infectiology, University Hospital Muenster, Muenster, Germany
| | - Joachim Ewald Kühn
- Institute of Virology Muenster, University of Muenster, Muenster, Germany
| |
Collapse
|
6
|
Wagenhäuser I, Almanzar G, Förg FB, Stein A, Eiter I, Reusch J, Mees J, Herzog A, Vogel U, Frey A, Lâm TT, Schubert-Unkmeir A, Dölken L, Kurzai O, Frantz S, Gabel A, Petri N, Prelog M, Krone M. Heterologous and homologous COVID-19 mRNA vaccination schemes for induction of basic immunity show similar immunogenicity regarding long-term spike-specific cellular immunity in healthcare workers. Vaccine 2024; 42:126132. [PMID: 39034219 DOI: 10.1016/j.vaccine.2024.07.033] [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: 04/04/2024] [Revised: 07/08/2024] [Accepted: 07/09/2024] [Indexed: 07/23/2024]
Abstract
Healthcare workers (HCWs) are recommended to receive at least three spike-antigen exposures to generate basic immunity and to mediate herd protection of vulnerable patients. So far, less attention has been put on the cellular immune response induced by homologous (three BTN162b2mRNA doses) or heterologous (mRNA-1273 as third dose building on two BTN162bmRNA doses) and the immunological impact of breakthrough infections (BTIs). Therefore, in 356 vaccinated HCWs with or without BTIs the Anti-SARS-CoV-2-Spike-IgG concentrations and avidities and B- and T-cell-reactivity against SARS-CoV-2-Spike-S1- and Nucleocapsid-antigens were assessed with Interferon-gamma-ELISpot and by flow-cytometry. HCWs who had hybrid immunity due to BTIs exhibited strong T-cell-reactivity against the Spike-S1-antigen. A lasso regression model revealed a significant reduction in T-cell immune responses among smokers (p < 0.0001), with less significant impact observed for age, sex, heterologous vaccination, body-mass-index, Anti-Nucleocapsid T-cell reactivity, days since last COVID-19-immunization, and Anti-SARS-CoV-2-Spike-IgG. Although subgroup analysis revealed higher Anti-SARS-CoV-2-Spike-IgG after heterologous vaccination, similar cellular reactivity and percentages of Spike-reactive T- and B-cells were found between homologous and heterologous vaccination. Anti-SARS-CoV-2-Spike-IgG concentrations and avidity significantly correlated with activated T-cells. CD4 + and CD8 + responses correlated with each other. A strong long-term cellular immune response should be considered as baseline for recommendations of booster doses in HCWs with prioritization of smokers. HCWs presented significant T-cellular reactivity towards Spike-S1-antigen with particularly strong responses in hybrid immunized HCWs who had BTIs. HCWs without BTI presented similar percentages of Spike-specific B- and T-cells between homologous or heterologous vaccination indicating similar immunogenicity for both mRNA vaccines, BNT162b2mRNA and mRNA-1273.
Collapse
Affiliation(s)
- Isabell Wagenhäuser
- Infection Control and Antimicrobial Stewardship Unit, University Hospital Würzburg, Würzburg, 97080, Germany; Department of Internal Medicine I, University Hospital Würzburg, Würzburg, 97080, Germany
| | - Giovanni Almanzar
- Pediatric Rheumatology/Special Immunology / Department of Pediatrics, University Hospital Würzburg, Würzburg, 97080, Germany
| | - Franziska Bernhardine Förg
- Pediatric Rheumatology/Special Immunology / Department of Pediatrics, University Hospital Würzburg, Würzburg, 97080, Germany
| | - Astrid Stein
- Pediatric Rheumatology/Special Immunology / Department of Pediatrics, University Hospital Würzburg, Würzburg, 97080, Germany
| | - Isabella Eiter
- Infection Control and Antimicrobial Stewardship Unit, University Hospital Würzburg, Würzburg, 97080, Germany
| | - Julia Reusch
- Infection Control and Antimicrobial Stewardship Unit, University Hospital Würzburg, Würzburg, 97080, Germany; Department of Internal Medicine I, University Hospital Würzburg, Würzburg, 97080, Germany
| | - Juliane Mees
- Infection Control and Antimicrobial Stewardship Unit, University Hospital Würzburg, Würzburg, 97080, Germany
| | - Anna Herzog
- Department of Internal Medicine I, University Hospital Würzburg, Würzburg, 97080, Germany
| | - Ulrich Vogel
- Infection Control and Antimicrobial Stewardship Unit, University Hospital Würzburg, Würzburg, 97080, Germany; Institute for Hygiene and Microbiology, Julius-Maximilians-Universität Würzburg, Würzburg, 97080, Germany
| | - Anna Frey
- Department of Internal Medicine I, University Hospital Würzburg, Würzburg, 97080, Germany
| | - Thiên-Trí Lâm
- Institute for Hygiene and Microbiology, Julius-Maximilians-Universität Würzburg, Würzburg, 97080, Germany
| | - Alexandra Schubert-Unkmeir
- Institute for Hygiene and Microbiology, Julius-Maximilians-Universität Würzburg, Würzburg, 97080, Germany
| | - Lars Dölken
- Institute for Virology and Immunobiology, Julius-Maximilians-Universität Würzburg, Würzburg, 97080, Germany
| | - Oliver Kurzai
- Institute for Hygiene and Microbiology, Julius-Maximilians-Universität Würzburg, Würzburg, 97080, Germany; Leibniz Institute for Natural Product Research and Infection Biology - Hans-Knoell-Institute, Jena, 07745, Germany
| | - Stefan Frantz
- Department of Internal Medicine I, University Hospital Würzburg, Würzburg, 97080, Germany
| | - Alexander Gabel
- Infection Control and Antimicrobial Stewardship Unit, University Hospital Würzburg, Würzburg, 97080, Germany
| | - Nils Petri
- Department of Internal Medicine I, University Hospital Würzburg, Würzburg, 97080, Germany
| | - Martina Prelog
- Pediatric Rheumatology/Special Immunology / Department of Pediatrics, University Hospital Würzburg, Würzburg, 97080, Germany.
| | - Manuel Krone
- Infection Control and Antimicrobial Stewardship Unit, University Hospital Würzburg, Würzburg, 97080, Germany; Institute for Hygiene and Microbiology, Julius-Maximilians-Universität Würzburg, Würzburg, 97080, Germany
| |
Collapse
|
7
|
Fischer F, Mücke J, Werny L, Gerrer K, Mihatsch L, Zehetmaier S, Riedel I, Geisperger J, Bodenhausen M, Schulte-Hillen L, Hoffmann D, Protzer U, Mautner J, Behrends U, Bauer T, Körber N. Evaluation of novel Epstein-Barr virus-derived antigen formulations for monitoring virus-specific T cells in pediatric patients with infectious mononucleosis. Virol J 2024; 21:139. [PMID: 38877590 PMCID: PMC11179387 DOI: 10.1186/s12985-024-02411-0] [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: 07/19/2023] [Accepted: 06/06/2024] [Indexed: 06/16/2024] Open
Abstract
BACKGROUND Infection with the Epstein-Barr virus (EBV) elicits a complex T-cell response against a broad range of viral proteins. Hence, identifying potential differences in the cellular immune response of patients with different EBV-associated diseases or different courses of the same disorder requires interrogation of a maximum number of EBV antigens. Here, we tested three novel EBV-derived antigen formulations for their ability to reactivate virus-specific T cells ex vivo in patients with EBV-associated infectious mononucleosis (IM). METHODS We comparatively analyzed EBV-specific CD4+ and CD8+ T-cell responses to three EBV-derived antigen formulations in 20 pediatric patients during the early phase of IM: T-activated EBV proteins (BZLF1, EBNA3A) and EBV-like particles (EB-VLP), both able to induce CD4+ and CD8+ T-cell responses ex vivo, as well as an EBV-derived peptide pool (PP) covering 94 well-characterized CD8+ T-cell epitopes. We assessed the specificity, magnitude, kinetics, and functional characteristics of EBV-specific immune responses at two sequential time points (v1 and v2) within the first six weeks after IM symptom onset (Tonset). RESULTS All three tested EBV-derived antigen formulations enabled the detection of EBV-reactive T cells during the early phase of IM without prior T-cell expansion in vitro. EBV-reactive CD4+ and CD8+ T cells were mainly mono-functional (CD4+: mean 64.92%, range 56.15-71.71%; CD8+: mean 58.55%, range 11.79-85.22%) within the first two weeks after symptom onset (v1) with IFN-γ and TNF-secreting cells representing the majority of mono-functional EBV-reactive T cells. By contrast, PP-reactive CD8+ T cells were primarily bi-functional (>60% at v1 and v2), produced IFN-γ and TNF and had more tri-functional than mono-functional components. We observed a moderate correlation between viral load and EBNA3A, EB-VLP, and PP-reactive CD8+ T cells (rs = 0.345, 0.418, and 0.356, respectively) within the first two weeks after Tonset, but no correlation with the number of detectable EBV-reactive CD4+ T cells. CONCLUSIONS All three EBV-derived antigen formulations represent innovative and generic recall antigens suitable for monitoring EBV-specific T-cell responses ex vivo. Their combined use facilitates a thorough analysis of EBV-specific T-cell immunity and allows the identification of functional T-cell signatures linked to disease development and severity.
Collapse
Affiliation(s)
- Franziska Fischer
- Children's Hospital, School of Medicine, Technical University of Munich, Munich, Germany
| | - Johannes Mücke
- Children's Hospital, School of Medicine, Technical University of Munich, Munich, Germany
| | - Louisa Werny
- Children's Hospital, School of Medicine, Technical University of Munich, Munich, Germany
- Institute of Virology, School of Medicine, Technical University of Munich and Helmholtz Munich, Schneckenburgerstr. 8, 81675, Munich, Germany
| | - Katrin Gerrer
- Children's Hospital, School of Medicine, Technical University of Munich, Munich, Germany
| | - Lorenz Mihatsch
- Children's Hospital, School of Medicine, Technical University of Munich, Munich, Germany
| | - Stefanie Zehetmaier
- Children's Hospital, School of Medicine, Technical University of Munich, Munich, Germany
- Research Unit Gene Vectors, Helmholtz Munich, Munich, Germany
| | - Isa Riedel
- Children's Hospital, School of Medicine, Technical University of Munich, Munich, Germany
| | - Jonas Geisperger
- Children's Hospital, School of Medicine, Technical University of Munich, Munich, Germany
| | - Maren Bodenhausen
- Children's Hospital, School of Medicine, Technical University of Munich, Munich, Germany
| | - Lina Schulte-Hillen
- Children's Hospital, School of Medicine, Technical University of Munich, Munich, Germany
| | - Dieter Hoffmann
- Institute of Virology, School of Medicine, Technical University of Munich and Helmholtz Munich, Schneckenburgerstr. 8, 81675, Munich, Germany
| | - Ulrike Protzer
- Institute of Virology, School of Medicine, Technical University of Munich and Helmholtz Munich, Schneckenburgerstr. 8, 81675, Munich, Germany
- German Centre for Infection Research (DZIF), Munich, Germany
| | - Josef Mautner
- Institute of Virology, School of Medicine, Technical University of Munich and Helmholtz Munich, Schneckenburgerstr. 8, 81675, Munich, Germany
- Research Unit Gene Vectors, Helmholtz Munich, Munich, Germany
- German Centre for Infection Research (DZIF), Munich, Germany
| | - Uta Behrends
- Children's Hospital, School of Medicine, Technical University of Munich, Munich, Germany
- Research Unit Gene Vectors, Helmholtz Munich, Munich, Germany
- German Centre for Infection Research (DZIF), Munich, Germany
| | - Tanja Bauer
- Institute of Virology, School of Medicine, Technical University of Munich and Helmholtz Munich, Schneckenburgerstr. 8, 81675, Munich, Germany
- German Centre for Infection Research (DZIF), Munich, Germany
| | - Nina Körber
- Institute of Virology, School of Medicine, Technical University of Munich and Helmholtz Munich, Schneckenburgerstr. 8, 81675, Munich, Germany.
- German Centre for Infection Research (DZIF), Munich, Germany.
| |
Collapse
|
8
|
Nemeth D, Vago H, Tothfalusi L, Ulakcsai Z, Becker D, Szabo Z, Rojkovich B, Gunkl-Toth L, Merkely B, Nagy G. Escalating SARS-CoV-2 specific humoral immune response in rheumatoid arthritis patients and healthy controls. Front Immunol 2024; 15:1397052. [PMID: 38911866 PMCID: PMC11190160 DOI: 10.3389/fimmu.2024.1397052] [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: 03/07/2024] [Accepted: 05/28/2024] [Indexed: 06/25/2024] Open
Abstract
Background Immunocompromised patients are at particular risk of Severe Acute Respiratory Syndrome Corona Virus 2 (SARS-CoV-2) infection and previous findings suggest that the infection or vaccination induced immune response decreases over time. Our main goal was to investigate the SARS-CoV-2-specific immune response in rheumatoid arthritis patients and healthy controls over prolonged time. Methods The SARS-CoV-2-specific humoral immune response was measured by Elecsys Anti-SARS-CoV-2 Spike (S) immunoassay, and antibodies against SARS-CoV-2 nucleocapsid protein (NCP) were also evaluated by Euroimmun enzyme-linked immunosorbent assay (ELISA) test. The SARS-CoV-2-specific T-cell response was detected by an IFN- γ release assay. Results We prospectively enrolled 84 patients diagnosed with rheumatoid arthritis (RA) and 43 healthy controls in our longitudinal study. Our findings demonstrate that RA patients had significantly lower anti-S antibody response and reduced SARS-CoV-2-specific T-cell response compared to healthy controls (p<0.01 for healthy controls, p<0.001 for RA patients). Furthermore, our results present evidence of a notable increase in the SARS-CoV-2-specific humoral immune response during the follow-up period in both study groups (p<0.05 for healthy volunteers, p<0.0001 for RA patients, rank-sum test). Participants who were vaccinated against Coronavirus disease-19 (COVID-19) during the interim period had 2.72 (CI 95%: 1.25-5.95, p<0.05) times higher anti-S levels compared to those who were not vaccinated during this period. Additionally, individuals with a confirmed SARS-CoV-2 infection exhibited 2.1 times higher (CI 95%: 1.31-3.37, p<0.01) anti-S levels compared to those who were not infected during the interim period. It is worth noting that patients treated with targeted therapy had 52% (CI 95%: 0.25-0.94, p<0.05) lower anti-S levels compared to matched patients who did not receive targeted therapy. Concerning the SARS-CoV-2-specific T-cell response, our findings revealed that its level had not changed substantially in the study groups. Conclusion Our present data revealed that the level of SARS-CoV-2-specific humoral immune response is actually higher, and the SARS-CoV-2-specific T-cell response remained at the same level over time in both study groups. This heightened humoral response, the nearly permanent SARS-CoV-2-specific T-cell response and the coexistence of different SARS-CoV-2 variants within the population, might be contributing to the decline in severe COVID-19 cases.
Collapse
Affiliation(s)
- Dora Nemeth
- Department of Rheumatology and Clinical Immunology, Semmelweis University, Budapest, Hungary
- Department of Internal Medicine and Oncology, Semmelweis University, Budapest, Hungary
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
| | - Hajnalka Vago
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
- Department of Sports Medicine, Semmelweis University, Budapest, Hungary
| | - Laszlo Tothfalusi
- Department of Pharmacodynamics, Semmelweis University, Budapest, Hungary
| | | | - David Becker
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Zsofia Szabo
- Department of Laboratory Medicine, Semmelweis University, Budapest, Hungary
| | - Bernadett Rojkovich
- Buda Hospital of the Hospitaller Order of Saint John of God, Budapest, Hungary
| | - Lilla Gunkl-Toth
- Department of Rheumatology and Clinical Immunology, Semmelweis University, Budapest, Hungary
- Department of Internal Medicine and Oncology, Semmelweis University, Budapest, Hungary
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary
- Chronic Pain Research Group, Hungarian Research Network - University of Pécs (HUN-REN-PTE), Pécs, Hungary
| | - Bela Merkely
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
- Department of Sports Medicine, Semmelweis University, Budapest, Hungary
| | - Gyorgy Nagy
- Department of Rheumatology and Clinical Immunology, Semmelweis University, Budapest, Hungary
- Department of Internal Medicine and Oncology, Semmelweis University, Budapest, Hungary
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
- Buda Hospital of the Hospitaller Order of Saint John of God, Budapest, Hungary
| |
Collapse
|
9
|
Inoue W, Kimura Y, Okamoto S, Nogimori T, Sakaguchi-Mikami A, Yamamoto T, Tsunetsugu-Yokota Y. SARS-CoV-2-Specific Immune Responses in Vaccination and Infection during the Pandemic in 2020-2022. Viruses 2024; 16:446. [PMID: 38543812 PMCID: PMC10974545 DOI: 10.3390/v16030446] [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: 02/01/2024] [Revised: 03/10/2024] [Accepted: 03/11/2024] [Indexed: 05/23/2024] Open
Abstract
To gain insight into how immunity develops against SARS-CoV-2 from 2020 to 2022, we analyzed the immune response of a small group of university staff and students who were either infected or vaccinated. We investigated the levels of receptor-binding domain (RBD)-specific and nucleocapsid (N)-specific IgG and IgA antibodies in serum and saliva samples taken early (around 10 days after infection or vaccination) and later (around 1 month later), as well as N-specific T-cell responses. One patient who had been infected in 2020 developed serum RBD and N-specific IgG antibodies, but declined eight months later, then mRNA vaccination in 2021 produced a higher level of anti-RBD IgG than natural infection. In the vaccination of naïve individuals, vaccines induced anti-RBD IgG, but it declined after six months. A third vaccination boosted the IgG level again, albeit to a lower level than after the second. In 2022, when the Omicron variant became dominant, familial transmission occurred among vaccinated people. In infected individuals, the levels of serum anti-RBD IgG antibodies increased later, while anti-N IgG peaked earlier. The N-specific activated T cells expressing IFN γ or CD107a were detected only early. Although SARS-CoV-2-specific salivary IgA was undetectable, two individuals showed a temporary peak in RBD- and N-specific IgA antibodies in their saliva on the second day after infection. Our study, despite having a small sample size, revealed that SARS-CoV-2 infection triggers the expected immune responses against acute viral infections. Moreover, our findings suggest that the temporary mucosal immune responses induced early during infection may provide better protection than the currently available intramuscular vaccines.
Collapse
Affiliation(s)
- Wakana Inoue
- Department of Medical Technology, School of Health Sciences and Graduate School of Medical Technology, Tokyo University of Technology, Tokyo 144-8535, Japan; (W.I.); (Y.K.); (S.O.); (A.S.-M.)
| | - Yuta Kimura
- Department of Medical Technology, School of Health Sciences and Graduate School of Medical Technology, Tokyo University of Technology, Tokyo 144-8535, Japan; (W.I.); (Y.K.); (S.O.); (A.S.-M.)
| | - Shion Okamoto
- Department of Medical Technology, School of Health Sciences and Graduate School of Medical Technology, Tokyo University of Technology, Tokyo 144-8535, Japan; (W.I.); (Y.K.); (S.O.); (A.S.-M.)
| | - Takuto Nogimori
- Laboratory of Precision Immunology, Center for Intractable Diseases and ImmunoGenomics, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka 567-0085, Japan; (T.N.); (T.Y.)
| | - Akane Sakaguchi-Mikami
- Department of Medical Technology, School of Health Sciences and Graduate School of Medical Technology, Tokyo University of Technology, Tokyo 144-8535, Japan; (W.I.); (Y.K.); (S.O.); (A.S.-M.)
| | - Takuya Yamamoto
- Laboratory of Precision Immunology, Center for Intractable Diseases and ImmunoGenomics, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka 567-0085, Japan; (T.N.); (T.Y.)
- Laboratory of Aging and Immune Regulation, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka 565-0871, Japan
- Department of Virology and Immunology, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | - Yasuko Tsunetsugu-Yokota
- Department of Medical Technology, School of Health Sciences and Graduate School of Medical Technology, Tokyo University of Technology, Tokyo 144-8535, Japan; (W.I.); (Y.K.); (S.O.); (A.S.-M.)
- Laboratory of Precision Immunology, Center for Intractable Diseases and ImmunoGenomics, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka 567-0085, Japan; (T.N.); (T.Y.)
- Research Institute, The World New Prosperity (WNP), Tokyo 169-0075, Japan
| |
Collapse
|
10
|
Yan Y, Saito K, Naito T, Ito K, Nojiri S, Horiuchi Y, Deshpande GA, Yokokawa H, Tabe Y. Seroprevalence of SARS-CoV-2 antibodies among Japanese healthcare workers from 2020 to 2022 as assayed by two commercial kits. Sci Rep 2024; 14:3102. [PMID: 38326367 PMCID: PMC10850062 DOI: 10.1038/s41598-024-53656-2] [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: 05/06/2023] [Accepted: 02/03/2024] [Indexed: 02/09/2024] Open
Abstract
Antibody tests are used as surveillance tools for informing health policy making. However, results may vary by type of antibody assay and timing of sample collection following infection. Long-term longitudinal cohort studies on antibody assay seropositivity have remained limited, especially among Asian populations. Using blood samples obtained at health physicals (2020-2022) of healthcare workers (mass vaccinated with mRNA COVID-19 vaccines) at a Japanese medical center, we measured N-specific antibodies using two commercially available systems. Roche Elecsys Anti-SARS-CoV-2 measures total antibodies and Abbott Alinity SARS-CoV-2 IgG measures only IgG. Among 2538 participants, seroprevalence was found to be 16.6% via total antibody assay versus 12.9% by IgG-only (including grayzone) by mid-June 2022. For 219 cases with a previous PCR-confirmed infection, positivity was 97.3% using total antibody assay versus 76.3% using IgG-only assay at the 2022 health physical. Using PCR positive test date as day 0, while the positivity of the total antibody assay was retained for the entire study period (until more than 24-months post-infection), the IgG-only assay's positivity declined after month 4. The Mantel-Haenszel test found a significant difference in the two assays' seropositivity, between stratified groups of "within 3 months" and "4 months or more" from infection (P < 0.001). Our study found significant differences in seropositivity over time of total antibody versus IgG-only assays, suggesting an optimal assay for retaining sensitivity over the entire infection period when designing seroprevalence studies.
Collapse
Affiliation(s)
- Yan Yan
- Department of General Medicine, Faculty of Medicine, Juntendo University, Hongo 2-1-1, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Kaori Saito
- Department of Clinical Laboratory Medicine, Faculty of Medicine, Juntendo University, Tokyo, Japan
| | - Toshio Naito
- Department of General Medicine, Faculty of Medicine, Juntendo University, Hongo 2-1-1, Bunkyo-ku, Tokyo, 113-8421, Japan.
- Department of Safety and Health Promotion, Juntendo University, Tokyo, Japan.
| | - Kanami Ito
- Department of Safety and Health Promotion, Juntendo University, Tokyo, Japan
| | - Shuko Nojiri
- Medical Technology Innovation Center, Juntendo University, Tokyo, Japan
| | - Yuki Horiuchi
- Department of Clinical Laboratory Medicine, Faculty of Medicine, Juntendo University, Tokyo, Japan
| | - Gautam A Deshpande
- Department of General Medicine, Faculty of Medicine, Juntendo University, Hongo 2-1-1, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Hirohide Yokokawa
- Department of General Medicine, Faculty of Medicine, Juntendo University, Hongo 2-1-1, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Yoko Tabe
- Department of Clinical Laboratory Medicine, Faculty of Medicine, Juntendo University, Tokyo, Japan
| |
Collapse
|
11
|
Lapuente D, Winkler TH, Tenbusch M. B-cell and antibody responses to SARS-CoV-2: infection, vaccination, and hybrid immunity. Cell Mol Immunol 2024; 21:144-158. [PMID: 37945737 PMCID: PMC10805925 DOI: 10.1038/s41423-023-01095-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Accepted: 10/13/2023] [Indexed: 11/12/2023] Open
Abstract
The emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in 2019 prompted scientific, medical, and biotech communities to investigate infection- and vaccine-induced immune responses in the context of this pathogen. B-cell and antibody responses are at the center of these investigations, as neutralizing antibodies (nAbs) are an important correlate of protection (COP) from infection and the primary target of SARS-CoV-2 vaccine modalities. In addition to absolute levels, nAb longevity, neutralization breadth, immunoglobulin isotype and subtype composition, and presence at mucosal sites have become important topics for scientists and health policy makers. The recent pandemic was and still is a unique setting in which to study de novo and memory B-cell (MBC) and antibody responses in the dynamic interplay of infection- and vaccine-induced immunity. It also provided an opportunity to explore new vaccine platforms, such as mRNA or adenoviral vector vaccines, in unprecedented cohort sizes. Combined with the technological advances of recent years, this situation has provided detailed mechanistic insights into the development of B-cell and antibody responses but also revealed some unexpected findings. In this review, we summarize the key findings of the last 2.5 years regarding infection- and vaccine-induced B-cell immunity, which we believe are of significant value not only in the context of SARS-CoV-2 but also for future vaccination approaches in endemic and pandemic settings.
Collapse
Affiliation(s)
- Dennis Lapuente
- Institut für klinische und molekulare Virologie, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossgarten 4, 91054, Erlangen, Germany
| | - Thomas H Winkler
- Department of Biology, Division of Genetics, Nikolaus-Fiebiger-Center for Molecular Medicine, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany.
- Medical Immunology Campus Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossplatz 1, 91054, Erlangen, Germany.
| | - Matthias Tenbusch
- Institut für klinische und molekulare Virologie, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossgarten 4, 91054, Erlangen, Germany
- Medical Immunology Campus Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossplatz 1, 91054, Erlangen, Germany
| |
Collapse
|
12
|
Wettengel JM, Strehle K, von Lucke C, Roggendorf H, Jeske SD, Christa C, Zelger O, Haller B, Protzer U, Knolle PA. Improved detection of infection with SARS-CoV-2 Omicron variants of concern in healthcare workers by a second-generation rapid antigen test. Microbiol Spectr 2023; 11:e0176823. [PMID: 37831440 PMCID: PMC10714798 DOI: 10.1128/spectrum.01768-23] [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: 04/27/2023] [Accepted: 09/11/2023] [Indexed: 10/14/2023] Open
Abstract
IMPORTANCE The results from this study demonstrate the usefulness of a second-generation rapid antigen test for early detection of infection with the SARS-CoV-2 Omicron variant of concern (VoC) and reveal a higher sensitivity to detect immune escape Omicron VoCs compared to a first-generation rapid antigen test (89.4% vs 83.7%) in the high-risk group of healthcare workers.
Collapse
Affiliation(s)
- Jochen M. Wettengel
- Institute of Virology, School of Medicine and Health, Technical University of Munich (TUM), München, Germany
- German Center for Infection Research (DZIF), Munich Partner Site, München, Germany
| | - Katharina Strehle
- Institute of Molecular Immunology, School of Medicine and Health, TUM, München, Germany
- Coronavirus Diagnostic Center of the University Hospital München Rechts der Isar, School of Medicine, TUM, München, Germany
| | - Catharina von Lucke
- Coronavirus Diagnostic Center of the University Hospital München Rechts der Isar, School of Medicine, TUM, München, Germany
| | - Hedwig Roggendorf
- Institute of Molecular Immunology, School of Medicine and Health, TUM, München, Germany
- Coronavirus Diagnostic Center of the University Hospital München Rechts der Isar, School of Medicine, TUM, München, Germany
| | - Samuel D. Jeske
- Institute of Virology, School of Medicine and Health, Technical University of Munich (TUM), München, Germany
| | - Catharina Christa
- Institute of Virology, School of Medicine and Health, Technical University of Munich (TUM), München, Germany
| | - Otto Zelger
- Coronavirus Diagnostic Center of the University Hospital München Rechts der Isar, School of Medicine, TUM, München, Germany
| | - Bernhard Haller
- Institute for AI and Informatics in Medicine Statistics, School of Medicine and Health, TUM, München, Germany
| | - Ulrike Protzer
- Institute of Virology, School of Medicine and Health, Technical University of Munich (TUM), München, Germany
- German Center for Infection Research (DZIF), Munich Partner Site, München, Germany
- Institute of Virology, Helmholtz Munich, München, Germany
| | - Percy A. Knolle
- German Center for Infection Research (DZIF), Munich Partner Site, München, Germany
- Institute of Molecular Immunology, School of Medicine and Health, TUM, München, Germany
| |
Collapse
|
13
|
Rak A, Isakova-Sivak I, Rudenko L. Overview of Nucleocapsid-Targeting Vaccines against COVID-19. Vaccines (Basel) 2023; 11:1810. [PMID: 38140214 PMCID: PMC10747980 DOI: 10.3390/vaccines11121810] [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: 11/04/2023] [Revised: 11/30/2023] [Accepted: 12/01/2023] [Indexed: 12/24/2023] Open
Abstract
The new SARS-CoV-2 coronavirus, which emerged in late 2019, is a highly variable causative agent of COVID-19, a contagious respiratory disease with potentially severe complications. Vaccination is considered the most effective measure to prevent the spread and complications of this infection. Spike (S) protein-based vaccines were very successful in preventing COVID-19 caused by the ancestral SARS-CoV-2 strain; however, their efficacy was significantly reduced when coronavirus variants antigenically different from the original strain emerged in circulation. This is due to the high variability of this major viral antigen caused by escape from the immunity caused by the infection or vaccination with spike-targeting vaccines. The nucleocapsid protein (N) is a much more conserved SARS-CoV-2 antigen than the spike protein and has therefore attracted the attention of scientists as a promising target for broad-spectrum vaccine development. Here, we summarized the current data on various N-based COVID-19 vaccines that have been tested in animal challenge models or clinical trials. Despite the high conservatism of the N protein, escape mutations gradually occurring in the N sequence can affect its protective properties. During the three years of the pandemic, at least 12 mutations have arisen in the N sequence, affecting more than 40 known immunogenic T-cell epitopes, so the antigenicity of the N protein of recent SARS-CoV-2 variants may be altered. This fact should be taken into account as a limitation in the development of cross-reactive vaccines based on N-protein.
Collapse
Affiliation(s)
- Alexandra Rak
- Department of Virology, Institute of Experimental Medicine, St. Petersburg 197022, Russia; (I.I.-S.); (L.R.)
| | | | | |
Collapse
|
14
|
Platen L, Liao BH, Tellenbach M, Cheng CC, Holzmann-Littig C, Christa C, Dächert C, Kappler V, Bester R, Werz ML, Schönhals E, Platen E, Eggerer P, Tréguer L, Küchle C, Schmaderer C, Heemann U, Keppler OT, Renders L, Braunisch MC, Protzer U. Longitudinal SARS-CoV-2 neutralization of Omicron BA.1, BA.5 and BQ.1.1 after four vaccinations and the impact of breakthrough infections in haemodialysis patients. Clin Kidney J 2023; 16:2447-2460. [PMID: 38046025 PMCID: PMC10689143 DOI: 10.1093/ckj/sfad147] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Indexed: 12/05/2023] Open
Abstract
Background Individuals on haemodialysis (HD) are more vulnerable to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection than the general population due to end-stage kidney disease-induced immunosuppression. Methods A total of 26 HD patients experiencing SARS-CoV-2 infection after a third vaccination were matched 1:1 with 26 of 92 SARS-CoV-2-naïve patients by age, sex, dialysis vintage and immunosuppressive drugs receiving a fourth vaccination with a messenger RNA-based vaccine. A competitive surrogate neutralization assay was used to monitor vaccination success. To determine infection neutralization titres, Vero-E6 cells were infected with SARS-CoV-2 variants of concern (VoCs), Omicron sublineage BA.1, BA.5 and BQ.1.1. The 50% inhibitory concentration (IC50, serum dilution factor 1:x) was determined before, 4 weeks after and 6 months after the fourth vaccination. Results A total of 52 HD patients received four coronavirus disease 2019 (COVID-19) vaccinations and were followed up for a median of 6.3 months. Patient characteristics did not differ between the matched cohorts. Patients without a SARS-CoV-2 infection had a significant reduction of real virus neutralization capacity for all Omicron sublineages after 6 months (P < .001 each). Those patients with a virus infection did not experience a reduction in real virus neutralization capacity after 6 months. Compared with the other Omicron VoC, the BQ.1.1 sublineage had the lowest virus neutralization capacity. Conclusions SARS-CoV-2-naïve HD patients had significantly decreased virus neutralization capacity 6 months after the fourth vaccination, whereas patients with a SARS-CoV-2 infection had no change in neutralization capacity. This was independent of age, sex, dialysis vintage and immunosuppression. Therefore, in infection-naïve HD patients a fifth COVID-19 vaccination might be reasonable 6 months after the fourth vaccination.
Collapse
Affiliation(s)
- Louise Platen
- Department of Nephrology, University Hospital rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany
| | - Bo-Hung Liao
- Institute of Virology, Technical University of Munich, School of Medicine, Munich, Germany
| | - Myriam Tellenbach
- Department of Nephrology, University Hospital rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany
| | - Cho-Chin Cheng
- Institute of Virology, Technical University of Munich, School of Medicine, Munich, Germany
| | - Christopher Holzmann-Littig
- Institute of Virology, Technical University of Munich, School of Medicine, Munich, Germany
- TUM Medical Education Center, Technical University of Munich, School of Medicine, Munich, Germany
| | - Catharina Christa
- Institute of Virology, Technical University of Munich, School of Medicine, Munich, Germany
| | - Christopher Dächert
- Max von Pettenkofer Institute & Gene Center, Virology, Ludwig Maximilian University of Munich, Munich, Germany
| | - Verena Kappler
- Department of Nephrology, University Hospital rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany
| | - Romina Bester
- Institute of Virology, Technical University of Munich, School of Medicine, Munich, Germany
| | - Maia Lucia Werz
- Department of Nephrology, University Hospital rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany
| | - Emely Schönhals
- Department of Nephrology, University Hospital rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany
| | - Eva Platen
- Kidney Center Eifel Dialyse, Mechernich, Germany
| | - Peter Eggerer
- KfH Kidney Center Harlaching, Munich-Harlaching, Germany
| | - Laëtitia Tréguer
- German Center for Infection Research (DZIF), Partner Site, Munich, Germany
| | - Claudius Küchle
- Department of Nephrology, University Hospital rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany
| | - Christoph Schmaderer
- Department of Nephrology, University Hospital rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany
| | - Uwe Heemann
- Department of Nephrology, University Hospital rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany
- German Center for Infection Research (DZIF), Partner Site, Munich, Germany
| | - Oliver T Keppler
- Max von Pettenkofer Institute & Gene Center, Virology, Ludwig Maximilian University of Munich, Munich, Germany
- German Center for Infection Research (DZIF), Partner Site, Munich, Germany
| | - Lutz Renders
- Department of Nephrology, University Hospital rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany
- KfH Kidney Center, Traunstein, Germany
| | - Matthias Christoph Braunisch
- Department of Nephrology, University Hospital rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany
| | - Ulrike Protzer
- Institute of Virology, Technical University of Munich, School of Medicine, Munich, Germany
- German Center for Infection Research (DZIF), Partner Site, Munich, Germany
- Institute of Virology, Helmholtz Munich, Munich, Germany
| |
Collapse
|
15
|
Wang M, Zhou B, Fan Q, Zhou X, Liao X, Lin J, Ma Z, Dong J, Wang H, Ge X, Ju B, Zhang Z. Omicron variants escape the persistent SARS-CoV-2-specific antibody response in 2-year COVID-19 convalescents regardless of vaccination. Emerg Microbes Infect 2023; 12:2151381. [PMID: 36444724 PMCID: PMC9809350 DOI: 10.1080/22221751.2022.2151381] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
With the ongoing COVID-19 pandemic and the emergence of various SARS-CoV-2 variants, a comprehensive evaluation of long-term efficacy of antibody response in convalescent individuals is urgently needed. Several longitudinal studies had reported the antibody dynamics after SARS-CoV-2 acute infection, but the follow-up was mostly limited to 1 year or 18 months at the maximum. In this study, we investigated the durability, potency, and susceptibility to immune evasion of SARS-CoV-2-specific antibody in COVID-19 convalescents for 2 years after discharge. These results showed the persistent antibody-dependent immunity could protect against the WT and Delta variant to some extent. However, the Omicron variants (BA.1, BA.2, and BA.4/5) largely escaped this preexisting immunity in recovered individuals. Furthermore, we revealed that inactivated vaccines (BBIBP-CorV, CoronaVac, or KCONVAC) could improve the plasma neutralization and help to maintain the broadly neutralizing antibodies at a certain level. Notably, with the time-dependent decline of antibody, 1-dose or 2-dose vaccination strategy seemed not to be enough to provide immune protection against the emerging variants. Overall, these results facilitated our understanding of SARS-CoV-2-induced antibody memory, contributing to the development of immunization strategy against SARS-CoV-2 variants for such a large number of COVID-19 survivors.
Collapse
Affiliation(s)
- Miao Wang
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, People’s Republic of China
| | - Bing Zhou
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, People’s Republic of China
| | - Qing Fan
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, People’s Republic of China
| | - Xinrong Zhou
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, People’s Republic of China
| | - Xuejiao Liao
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, People’s Republic of China
| | - Jingyan Lin
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, People’s Republic of China
| | - Zhenghua Ma
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, People’s Republic of China
| | - Jingke Dong
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, People’s Republic of China
| | - Haiyan Wang
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, People’s Republic of China
| | - Xiangyang Ge
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, People’s Republic of China
| | - Bin Ju
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, People’s Republic of China,Guangdong Key Laboratory for Anti-infection Drug Quality Evaluation, Shenzhen, People’s Republic of China, Bin Ju Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong518112, People’s Republic of China; Guangdong Key Laboratory for Anti-infection Drug Quality Evaluation, Shenzhen, Guangdong518112, People’s Republic of China; Zheng Zhang Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong518112, People’s Republic of China; Guangdong Key Laboratory for Anti-infection Drug Quality Evaluation, Shenzhen, Guangdong518112, People’s Republic of China; Shenzhen Research Center for Communicable Disease Diagnosis and Treatment of Chinese Academy of Medical Science, Shenzhen, Guangdong518112, People’s Republic of China
| | - Zheng Zhang
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, People’s Republic of China,Guangdong Key Laboratory for Anti-infection Drug Quality Evaluation, Shenzhen, People’s Republic of China,Shenzhen Research Center for Communicable Disease Diagnosis and Treatment of Chinese Academy of Medical Science, Shenzhen, People’s Republic of China, Bin Ju Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong518112, People’s Republic of China; Guangdong Key Laboratory for Anti-infection Drug Quality Evaluation, Shenzhen, Guangdong518112, People’s Republic of China; Zheng Zhang Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong518112, People’s Republic of China; Guangdong Key Laboratory for Anti-infection Drug Quality Evaluation, Shenzhen, Guangdong518112, People’s Republic of China; Shenzhen Research Center for Communicable Disease Diagnosis and Treatment of Chinese Academy of Medical Science, Shenzhen, Guangdong518112, People’s Republic of China
| |
Collapse
|
16
|
Yu H, Guan F, Miller H, Lei J, Liu C. The role of SARS-CoV-2 nucleocapsid protein in antiviral immunity and vaccine development. Emerg Microbes Infect 2023; 12:e2164219. [PMID: 36583642 PMCID: PMC9980416 DOI: 10.1080/22221751.2022.2164219] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
ABSTRACTThe coronavirus disease 2019 (COVID-19) has caused enormous health risks and global economic disruption. This disease is caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The SARS-CoV-2 nucleocapsid protein is a structural protein involved in viral replication and assembly. There is accumulating evidence indicating that the nucleocapsid protein is multi-functional, playing a key role in the pathogenesis of COVID-19 and antiviral immunity against SARS-CoV-2. Here, we summarize its potential application in the prevention of COVID-19, which is based on its role in inflammation, cell death, antiviral innate immunity, and antiviral adaptive immunity.
Collapse
Affiliation(s)
- Haiyun Yu
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Fei Guan
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Heather Miller
- Laboratory of Intracellular Parasites, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Jiahui Lei
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Chaohong Liu
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China, Chaohong Liu
| |
Collapse
|
17
|
Karkanitsa M, Li Y, Valenti S, Spathies J, Kelly S, Hunsberger S, Yee L, Croker JA, Wang J, Alfonso AL, Faust M, Mehalko J, Drew M, Denson JP, Putman Z, Fathi P, Ngo TB, Siripong N, Baus HA, Petersen B, Ford EW, Sundaresan V, Josyula A, Han A, Giurgea LT, Rosas LA, Bean R, Athota R, Czajkowski L, Klumpp-Thomas C, Cervantes-Medina A, Gouzoulis M, Reed S, Graubard B, Hall MD, Kalish H, Esposito D, Kimberly RP, Reis S, Sadtler K, Memoli MJ. Dynamics of SARS-CoV-2 Seroprevalence in a Large US population Over a Period of 12 Months. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.10.20.23297329. [PMID: 37904956 PMCID: PMC10614993 DOI: 10.1101/2023.10.20.23297329] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Due to a combination of asymptomatic or undiagnosed infections, the proportion of the United States population infected with SARS-CoV-2 was unclear from the beginning of the pandemic. We previously established a platform to screen for SARS-CoV-2 positivity across a representative proportion of the US population, from which we reported that almost 17 million Americans were estimated to have had undocumented infections in the Spring of 2020. Since then, vaccine rollout and prevalence of different SARS-CoV-2 variants have further altered seropositivity trends within the United States population. To explore the longitudinal impacts of the pandemic and vaccine responses on seropositivity, we re-enrolled participants from our baseline study in a 6- and 12- month follow-up study to develop a longitudinal antibody profile capable of representing seropositivity within the United States during a critical period just prior to and during the initiation of vaccine rollout. Initial measurements showed that, since July 2020, seropositivity elevated within this population from 4.8% at baseline to 36.2% and 89.3% at 6 and 12 months, respectively. We also evaluated nucleocapsid seropositivity and compared to spike seropositivity to identify trends in infection versus vaccination relative to baseline. These data serve as a window into a critical timeframe within the COVID-19 pandemic response and serve as a resource that could be used in subsequent respiratory illness outbreaks.
Collapse
Affiliation(s)
- Maria Karkanitsa
- Section on Immunoengineering, National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda MD 20894
| | - Yan Li
- Joint Program in Survey Methodology, Department of Epidemiology and Biostatistics, University of Maryland College Park, College Park, MD 20742
| | - Shannon Valenti
- Clinical and Translational Science Institute (CTSI), University of Pittsburgh, Pittsburgh, PA 15213
| | - Jacquelyn Spathies
- Trans-NIH Shared Resource on Biomedical Engineering and Physical Science (BEPS), NIBIB, NIH, Bethesda MD 20894
| | - Sophie Kelly
- Trans-NIH Shared Resource on Biomedical Engineering and Physical Science (BEPS), NIBIB, NIH, Bethesda MD 20894
| | - Sally Hunsberger
- Biostatistics Research Branch, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, MD 20894
| | - Laura Yee
- Division of Cancer Treatment and Diagnosis, National Cancer Institute (NCI), NIH, MD 20894
| | - Jennifer A. Croker
- Center for Clinical and Translational Science, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jing Wang
- Clinical Monitoring Research Program Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD 21702
| | - Andrea Lucia Alfonso
- Section on Immunoengineering, National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda MD 20894
| | - Mondreakest Faust
- Section on Immunoengineering, National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda MD 20894
| | - Jennifer Mehalko
- Protein Expression Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD, 21702
| | - Matthew Drew
- Protein Expression Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD, 21702
| | - John-Paul Denson
- Protein Expression Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD, 21702
| | - Zoe Putman
- Protein Expression Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD, 21702
| | - Parinaz Fathi
- Section on Immunoengineering, National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda MD 20894
| | - Tran B. Ngo
- Section on Immunoengineering, National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda MD 20894
| | - Nalyn Siripong
- Clinical and Translational Science Institute (CTSI), University of Pittsburgh, Pittsburgh, PA 15213
| | - Holly Ann Baus
- Laboratory of Immunoregulation, NIAID, NIH, Bethesda MD 20894
| | - Brian Petersen
- Clinical and Translational Science Institute (CTSI), University of Pittsburgh, Pittsburgh, PA 15213
| | - Eric W. Ford
- Department of Health Care Organization, and Policy, School of Public Health, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Vanathi Sundaresan
- Section on Immunoengineering, National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda MD 20894
| | - Aditya Josyula
- Section on Immunoengineering, National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda MD 20894
| | - Alison Han
- LID Clinical Studies Unit, Laboratory of Infectious Diseases, NIAID, NIH, Bethesda, MD 20894
| | - Luca T. Giurgea
- LID Clinical Studies Unit, Laboratory of Infectious Diseases, NIAID, NIH, Bethesda, MD 20894
| | - Luz Angela Rosas
- LID Clinical Studies Unit, Laboratory of Infectious Diseases, NIAID, NIH, Bethesda, MD 20894
| | - Rachel Bean
- LID Clinical Studies Unit, Laboratory of Infectious Diseases, NIAID, NIH, Bethesda, MD 20894
| | - Rani Athota
- LID Clinical Studies Unit, Laboratory of Infectious Diseases, NIAID, NIH, Bethesda, MD 20894
| | - Lindsay Czajkowski
- LID Clinical Studies Unit, Laboratory of Infectious Diseases, NIAID, NIH, Bethesda, MD 20894
| | - Carleen Klumpp-Thomas
- National Center for Advancing Translational Sciences (NCATS), NIH, Rockville, MD 20850
| | | | - Monica Gouzoulis
- LID Clinical Studies Unit, Laboratory of Infectious Diseases, NIAID, NIH, Bethesda, MD 20894
| | - Susan Reed
- LID Clinical Studies Unit, Laboratory of Infectious Diseases, NIAID, NIH, Bethesda, MD 20894
| | - Barry Graubard
- Division of Cancer Epidemiology & Genetics, Biostatistics Branch, NCI, NIH, Bethesda, MD 20894
| | - Matthew D. Hall
- National Center for Advancing Translational Sciences (NCATS), NIH, Rockville, MD 20850
| | - Heather Kalish
- Trans-NIH Shared Resource on Biomedical Engineering and Physical Science (BEPS), NIBIB, NIH, Bethesda MD 20894
| | - Dominic Esposito
- Protein Expression Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD, 21702
| | - Robert P. Kimberly
- Center for Clinical and Translational Science, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Steven Reis
- Clinical and Translational Science Institute (CTSI), University of Pittsburgh, Pittsburgh, PA 15213
| | - Kaitlyn Sadtler
- Section on Immunoengineering, National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda MD 20894
| | - Matthew J Memoli
- LID Clinical Studies Unit, Laboratory of Infectious Diseases, NIAID, NIH, Bethesda, MD 20894
| |
Collapse
|
18
|
Farnsworth CW, O’Neil CA, Dalton C, McDonald D, Vogt L, Hock K, Arter O, Wallace MA, Muenks C, Amor M, Alvarado K, Peacock K, Jolani K, Fraser VJ, Burnham CAD, Babcock HM, Budge PJ, Kwon JH. Association between SARS-CoV-2 Symptoms, Ct Values, and Serological Response in Vaccinated and Unvaccinated Healthcare Personnel. J Appl Lab Med 2023; 8:871-886. [PMID: 37478837 PMCID: PMC10482509 DOI: 10.1093/jalm/jfad042] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 05/15/2023] [Indexed: 07/23/2023]
Abstract
BACKGROUND SARS-CoV-2 vaccines are effective at reducing symptomatic and asymptomatic COVID-19. Limited studies have compared symptoms, threshold cycle (Ct) values from reverse transcription (RT)-PCR testing, and serological testing results between previously vaccinated vs unvaccinated populations with SARS-CoV-2 infection. METHODS Healthcare personnel (HCP) with a positive SARS-CoV-2 RT-PCR test within the previous 14 to 28 days completed surveys including questions about demographics, medical conditions, social factors, and symptoms of COVID-19. Ct values were observed, and serological testing was performed for anti-nucleocapsid (anti-N) and anti-Spike (anti-S) antibodies at enrollment and 40 to 90 days later. Serological results were compared to HCP with no known SARS-CoV-2 infection and negative anti-N testing. RESULTS There were 104 unvaccinated/not fully vaccinated and 77 vaccinated HCP with 2 doses of an mRNA vaccine at time of infection. No differences in type or duration of symptoms were reported (P = 0.45). The median (interquartile range [IQR]) Ct was 21.4 (17.6-24.6) and 21.5 (18.1-24.6) for the unvaccinated and vaccinated HCP, respectively. Higher anti-N IgG was observed in unvaccinated HCP (5.08 S/CO, 3.08-6.92) than vaccinated (3.61 signal to cutoff ratio [S/CO], 2.16-5.05). Anti-S IgG was highest among vaccinated HCP with infection (34 285 aribitrary units [AU]/mL, 17 672-61 775), followed by vaccinated HCP with no prior infection (1452 AU/mL, 791-2943), then unvaccinated HCP with infection (829 AU/mL, 290-1555). Anti-S IgG decreased 1.56% (0.9%-1.79%) per day in unvaccinated and 0.38% (0.03%-0.94%) in vaccinated HCP. CONCLUSIONS Vaccinated HCP infected with SARS-CoV-2 reported comparable symptoms and had similar Ct values relative to unvaccinated. However, vaccinated HCP had increased and prolonged anti-S and decreased anti-N response relative to unvaccinated.
Collapse
Affiliation(s)
- Christopher W Farnsworth
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, United States
| | - Caroline A O’Neil
- Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, MO, United States
| | - Claire Dalton
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, United States
| | - David McDonald
- Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, MO, United States
| | - Lucy Vogt
- Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, MO, United States
| | - Karl Hock
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, United States
| | - Olivia Arter
- Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, MO, United States
| | - Meghan A Wallace
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, United States
| | - Carol Muenks
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, United States
| | - Mostafa Amor
- Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, MO, United States
| | - Kelly Alvarado
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, United States
| | - Kate Peacock
- Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, MO, United States
| | - Kevin Jolani
- Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, MO, United States
| | - Victoria J Fraser
- Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, MO, United States
| | - Carey-Ann D Burnham
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, United States
| | - Hilary M Babcock
- Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, MO, United States
| | - Phillip J Budge
- Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, MO, United States
| | - Jennie H Kwon
- Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, MO, United States
| |
Collapse
|
19
|
Mohamed Mohamed K, Álvarez-Hernández MP, Jiménez García C, Guevara-Hoyer K, Freites D, Martínez Prada C, Pérez-Sancristóbal I, Fernández Gutiérrez B, Mato Chaín G, Rodero M, Rodríguez de la Peña A, Mulero T, Bravo C, Toledano E, Culebras López E, Mediero Valeros B, Pérez Segura P, Sánchez-Ramón S, Candelas Rodríguez G. Specific Cellular and Humoral Response after the Third Dose of Anti-SARS-CoV-2 RNA Vaccine in Patients with Immune-Mediated Rheumatic Diseases on Immunosuppressive Therapy. Biomedicines 2023; 11:2418. [PMID: 37760858 PMCID: PMC10525269 DOI: 10.3390/biomedicines11092418] [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: 06/11/2023] [Revised: 07/31/2023] [Accepted: 08/23/2023] [Indexed: 09/29/2023] Open
Abstract
OBJECTIVE Data on cellular and humoral immunogenicity after the third dose of anti-SARS-CoV-2 vaccines in patients with immune-mediated rheumatic diseases (IMRDs) are scarce. Herein, we evaluated the adaptive immune response in IMRD patients treated with different immunosuppressive therapies (conventional synthetic disease-modifying antirheumatic drugs [csDMARDs], biological disease-modifying antirheumatic drugs [bDMARDs], and targeted synthetic disease-modifying antirheumatic drugs [tsDMARDs]) after the booster of the anti-SARS-CoV-2 vaccine to determine whether any drug reduced the vaccine's response. METHODS A single-center prospective study was conducted, including patients presenting with IMRD and healthy controls (HC). Specific anti-SARS-CoV-2 interferon-gamma (IFN-γ) production was evaluated between 8-12 weeks after the third dose of the SARS-CoV-2 vaccine. In addition, anti-Spike IgG antibody titers were also measured. RESULTS Samples were obtained from 79 IMRD patients (51 women, 28 men; mean age 57 ± 11.3 years old): 43 rheumatoid arthritis, 10 psoriatic arthritis, 14 ankylosing spondylitis, 10 undifferentiated spondyloarthritis, and 2 inflammatory bowel disease-associated spondyloarthritis (IBD-SpA). In total, 31 HC (mean age 50.9 ± 13.1 years old, 67.7% women) were included in the study. Post-vaccine results displayed positive T-cell immune responses in 68 out of 79 (86.1%) IMRD patients (82.3% of those without prior COVID-19). All HC and IMRDs patients had an antibody response against the SARS-CoV-2 receptor-binding domain; however, the HC response was significantly higher (median of 18,048 AU/mL) than in IMRDs patients (median of 6590.3 AU/mL, p < 0.001). MTX and leflunomide were associated with lower titers of IgG and IFN-γ responses. Among bDMARDs, adalimumab, etanercept, and guselkumab are associated with reduced cellular responses. CONCLUSION Our preliminary data show that the majority of our IMRD patients develop cellular and humoral responses after the SARS-CoV-2 booster vaccination, emphasizing the relevance of vaccination in this group. However, the magnitude of specific responses was dependent on the immunosuppressive therapy administered. Specific vaccination protocols and personalized decisions about boosters are essential for these patients.
Collapse
Affiliation(s)
- Kauzar Mohamed Mohamed
- Department of Immunology, IML and IdISSC, Hospital Clínico San Carlos, Calle Profesor Martín Lagos, S/N, 28040 Madrid, Spain
| | - María Paula Álvarez-Hernández
- Rheumatology Department, Hospital Universitario Clínico San Carlos, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Carlos Jiménez García
- Department of Immunology, IML and IdISSC, Hospital Clínico San Carlos, Calle Profesor Martín Lagos, S/N, 28040 Madrid, Spain
| | - Kissy Guevara-Hoyer
- Department of Immunology, IML and IdISSC, Hospital Clínico San Carlos, Calle Profesor Martín Lagos, S/N, 28040 Madrid, Spain
| | - Dalifer Freites
- Rheumatology Department, Hospital Universitario Clínico San Carlos, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Cristina Martínez Prada
- Rheumatology Department, Hospital Universitario Clínico San Carlos, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Inés Pérez-Sancristóbal
- Rheumatology Department, Hospital Universitario Clínico San Carlos, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Benjamín Fernández Gutiérrez
- Rheumatology Department, Hospital Universitario Clínico San Carlos, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Gloria Mato Chaín
- Unidad de Vacunación del Adulto, Servicio de Medicina Preventiva, Hospital Clínico San Carlos, 28040 Madrid, Spain
| | - Maria Rodero
- Rheumatology Department, Hospital Universitario Clínico San Carlos, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Antonia Rodríguez de la Peña
- Department of Immunology, IML and IdISSC, Hospital Clínico San Carlos, Calle Profesor Martín Lagos, S/N, 28040 Madrid, Spain
| | - Teresa Mulero
- Rheumatology Department, Hospital Universitario Clínico San Carlos, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Cecilia Bravo
- Rheumatology Department, Hospital Universitario Clínico San Carlos, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Esther Toledano
- Rheumatology Department, Hospital Universitario Clínico San Carlos, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Esther Culebras López
- Department of Microbiology, IML and IdISSC, Hospital Clínico San Carlos, 28040 Madrid, Spain
| | - Beatriz Mediero Valeros
- Department of Immunology, IML and IdISSC, Hospital Clínico San Carlos, Calle Profesor Martín Lagos, S/N, 28040 Madrid, Spain
| | - Pedro Pérez Segura
- Department of Medical Oncology, Hospital Clinico San Carlos, IdISSC, Calle Profesor Martín Lagos, 28040 Madrid, Spain
| | - Silvia Sánchez-Ramón
- Department of Immunology, IML and IdISSC, Hospital Clínico San Carlos, Calle Profesor Martín Lagos, S/N, 28040 Madrid, Spain
| | - Gloria Candelas Rodríguez
- Rheumatology Department, Hospital Universitario Clínico San Carlos, Universidad Complutense de Madrid, 28040 Madrid, Spain
| |
Collapse
|
20
|
Föh B, Schnoor M, Balck A, Waldeck F, Käding N, Borsche M, Rupp J, Katalinic A, Klein C. Transition to endemic: 2-year SARS-CoV-2 surveillance follow-up of the ELISA cohort. ERJ Open Res 2023; 9:00746-2022. [PMID: 37362882 PMCID: PMC10257863 DOI: 10.1183/23120541.00746-2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 05/25/2023] [Indexed: 06/28/2023] Open
Abstract
The 2-year SARS-CoV-2 surveillance follow-up of the ELISA cohort shows the successful transition from COVID-19 pandemic to endemic, confirms occupational risk factors in healthcare and identifies household risk factors in a high-incidence period https://bit.ly/43x8q6i.
Collapse
Affiliation(s)
- Bandik Föh
- Department of Medicine I, University Hospital Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
- These authors contributed equally
| | - Maike Schnoor
- Institute of Social Medicine and Epidemiology, University of Lübeck, Lübeck, Germany
- These authors contributed equally
| | - Alexander Balck
- Institute of Neurogenetics, University of Lübeck and University Hospital Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
- Department of Neurology, University of Lübeck, Lübeck, Germany
- These authors contributed equally
| | - Frederike Waldeck
- Department of Infectious Diseases and Microbiology, University of Lübeck and University Hospital Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Nadja Käding
- Department of Infectious Diseases and Microbiology, University of Lübeck and University Hospital Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Max Borsche
- Institute of Neurogenetics, University of Lübeck and University Hospital Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
- Department of Neurology, University of Lübeck, Lübeck, Germany
| | - Jan Rupp
- Department of Infectious Diseases and Microbiology, University of Lübeck and University Hospital Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Alexander Katalinic
- Institute of Social Medicine and Epidemiology, University of Lübeck, Lübeck, Germany
| | - Christine Klein
- Institute of Neurogenetics, University of Lübeck and University Hospital Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
- Department of Neurology, University of Lübeck, Lübeck, Germany
| |
Collapse
|
21
|
Boumar I, Deliorman M, Sukumar P, Qasaimeh MA. Spike- and nucleocapsid-based gold colloid assay toward the development of an adhesive bandage for rapid SARS-CoV-2 immune response detection and screening. MICROSYSTEMS & NANOENGINEERING 2023; 9:82. [PMID: 37351273 PMCID: PMC10281977 DOI: 10.1038/s41378-023-00554-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 05/02/2023] [Accepted: 05/14/2023] [Indexed: 06/24/2023]
Abstract
Immunoglobulin M (IgM) and immunoglobulin G (IgG) antibodies are important biomarkers used for the diagnosis and screening of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections in both symptomatic and asymptomatic individuals. These antibodies are highly specific to the spike (S) and nucleocapsid (N) proteins of the SARS-CoV-2 virus. This paper outlines the development steps of a novel hybrid (vertical-lateral-vertical) flow assay in the form of a finger-stick point-of-care device, similar to an adhesive bandage, designed for the timely detection and screening of IgM and IgG immune responses to SARS-CoV-2 infections. The assay, comprising a vertically stacked plasma/serum separation membrane, conjugate pad, and detection (readout) zone, utilizes gold nanoparticles (AuNPs) conjugated with SARS-CoV-2 S and N proteins to effectively capture IgM and IgG antibodies from a pinprick (~15 µL) of blood in just one step and provides results of no immune IgM-/IgG-, early immune IgM+/IgG-, active immune IgM+/IgG+ or immune IgM-/IgG+ in a short amount of time (minutes). The adhesive bandage-like construction is an example of the design of rapid, low-cost, disposable, and easy-to-use tests for large-scale detection and screening in households. Furthermore, the bandage can be easily adjusted and optimized to detect different viral infections as they arise by simply selecting appropriate antigens related to pandemics and outbreaks.
Collapse
Affiliation(s)
- Imen Boumar
- Division of Engineering, New York University Abu Dhabi (NYUAD), Abu Dhabi, UAE
| | | | - Pavithra Sukumar
- Division of Engineering, New York University Abu Dhabi (NYUAD), Abu Dhabi, UAE
| | - Mohammad A. Qasaimeh
- Division of Engineering, New York University Abu Dhabi (NYUAD), Abu Dhabi, UAE
- NYU Tandon School of Engineering, New York University, New York, USA
| |
Collapse
|
22
|
Tiezzi C, Vecchi A, Rossi M, Cavazzini D, Bolchi A, Laccabue D, Doselli S, Penna A, Sacchelli L, Brillo F, Meschi T, Ticinesi A, Nouvenne A, Donofrio G, Zanelli P, Benecchi M, Giuliodori S, Fisicaro P, Montali I, Ceccatelli Berti C, Reverberi V, Montali A, Urbani S, Pedrazzi G, Missale G, Telenti A, Corti D, Ottonello S, Ferrari C, Boni C. Natural heteroclitic-like peptides are generated by SARS-CoV-2 mutations. iScience 2023; 26:106940. [PMID: 37275517 PMCID: PMC10200277 DOI: 10.1016/j.isci.2023.106940] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 04/13/2023] [Accepted: 05/18/2023] [Indexed: 06/07/2023] Open
Abstract
Humoral immunity is sensitive to evasion by SARS-CoV-2 mutants, but CD8 T cells seem to be more resistant to mutational inactivation. By a systematic analysis of 30 spike variant peptides containing the most relevant VOC and VOI mutations that have accumulated overtime, we show that in vaccinated and convalescent subjects, mutated epitopes can have not only a neutral or inhibitory effect on CD8 T cell recognition but can also enhance or generate de novo CD8 T cell responses. The emergence of these mutated T cell function enhancing epitopes likely reflects an epiphenomenon of SARS-CoV-2 evolution driven by antibody evasion and increased virus transmissibility. In a subset of individuals with weak and narrowly focused CD8 T cell responses selection of these heteroclitic-like epitopes may bear clinical relevance by improving antiviral protection. The functional enhancing effect of these peptides is also worth of consideration for the future development of new generation, more potent COVID-19 vaccines.
Collapse
Affiliation(s)
- Camilla Tiezzi
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Andrea Vecchi
- Laboratory of Viral Immunopathology, Unit of Infectious Diseases and Hepatology, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy
| | - Marzia Rossi
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Davide Cavazzini
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
| | - Angelo Bolchi
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
- Interdepartmental Center Biopharmanet-Tec, University of Parma, Parma, Italy
| | - Diletta Laccabue
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Sara Doselli
- Laboratory of Viral Immunopathology, Unit of Infectious Diseases and Hepatology, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy
| | - Amalia Penna
- Laboratory of Viral Immunopathology, Unit of Infectious Diseases and Hepatology, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy
| | - Luca Sacchelli
- Laboratory of Viral Immunopathology, Unit of Infectious Diseases and Hepatology, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy
| | - Federica Brillo
- Laboratory of Viral Immunopathology, Unit of Infectious Diseases and Hepatology, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy
| | - Tiziana Meschi
- Department of Medicine and Surgery, University of Parma, Parma, Italy
- Geriatric-Rehabilitation Department, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy
| | - Andrea Ticinesi
- Department of Medicine and Surgery, University of Parma, Parma, Italy
- Geriatric-Rehabilitation Department, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy
| | - Antonio Nouvenne
- Geriatric-Rehabilitation Department, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy
| | - Gaetano Donofrio
- Department of Veterinary Science, University of Parma, Parma, Italy
| | - Paola Zanelli
- Unità di Immunogenetica dei Trapianti, Azienda Ospedaliero Universitaria di Parma, Parma, Italy
| | - Magda Benecchi
- Unità di Immunogenetica dei Trapianti, Azienda Ospedaliero Universitaria di Parma, Parma, Italy
| | - Silvia Giuliodori
- Unità di Immunogenetica dei Trapianti, Azienda Ospedaliero Universitaria di Parma, Parma, Italy
| | - Paola Fisicaro
- Laboratory of Viral Immunopathology, Unit of Infectious Diseases and Hepatology, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy
| | - Ilaria Montali
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | | | - Valentina Reverberi
- Laboratory of Viral Immunopathology, Unit of Infectious Diseases and Hepatology, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy
| | - Anna Montali
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Simona Urbani
- UO Immunoematologia e Medicina Trasfusionale, Dipartimento Diagnostico, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy
| | - Giuseppe Pedrazzi
- Department of Neuroscience - Biophysics and Medical Physics Unit, University of Parma, Parma, Italy
| | - Gabriele Missale
- Laboratory of Viral Immunopathology, Unit of Infectious Diseases and Hepatology, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | | | - Davide Corti
- Humabs Biomed SA, a subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | - Simone Ottonello
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
- Interdepartmental Center Biopharmanet-Tec, University of Parma, Parma, Italy
| | - Carlo Ferrari
- Laboratory of Viral Immunopathology, Unit of Infectious Diseases and Hepatology, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Carolina Boni
- Laboratory of Viral Immunopathology, Unit of Infectious Diseases and Hepatology, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy
| |
Collapse
|
23
|
Bunse T, Koerber N, Wintersteller H, Schneider J, Graf A, Radonic A, Thuermer A, von Kleist M, Blum H, Spinner CD, Bauer T, Knolle PA, Protzer U, Schulte EC. T-Cell-Dominated Immune Response Resolves Protracted SARS-CoV-2 Infection in the Absence of Neutralizing Antibodies in an Immunocompromised Individual. Microorganisms 2023; 11:1562. [PMID: 37375064 DOI: 10.3390/microorganisms11061562] [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: 05/03/2023] [Revised: 06/01/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
Immunocompromised individuals are at higher risk of developing protracted and severe COVID-19, and understanding individual disease courses and SARS-CoV-2 immune responses in these individuals is of the utmost importance. For more than two years, we followed an immunocompromised individual with a protracted SARS-CoV-2 infection that was eventually cleared in the absence of a humoral neutralizing SARS-CoV-2 antibody response. By conducting an in-depth examination of this individual's immune response and comparing it to a large cohort of convalescents who spontaneously cleared a SARS-CoV-2 infection, we shed light on the interplay between B- and T-cell immunity and how they interact in clearing SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Till Bunse
- Institute of Virology, School of Medicine, Technical University of Munich, 81675 Munich, Germany
- Institute of Virology, Helmholtz Munich, Trogerstrasse 30, 81675 Munich, Germany
| | - Nina Koerber
- Institute of Virology, School of Medicine, Technical University of Munich, 81675 Munich, Germany
- Institute of Virology, Helmholtz Munich, Trogerstrasse 30, 81675 Munich, Germany
| | - Hannah Wintersteller
- Institute of Molecular Immunology and Experimental Oncology, School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Jochen Schneider
- Department of Internal Medicine II, University Hospital Rechts der Isar, School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Alexander Graf
- Laboratory for Functional Genome Analysis, Gene Center, LMU Munich, 81377 Munich, Germany
| | - Aleksandar Radonic
- Method Development, Research Infrastructure & IT (MFI), Robert-Koch Institute (RKI), 13353 Berlin, Germany
| | - Andrea Thuermer
- Method Development, Research Infrastructure & IT (MFI), Robert-Koch Institute (RKI), 13353 Berlin, Germany
| | - Max von Kleist
- Department of Mathematics and Computer Science, Freie Universität (FU) Berlin, 14195 Berlin, Germany
- Project Groups, Robert-Koch Institute (RKI), 13353 Berlin, Germany
| | - Helmut Blum
- Laboratory for Functional Genome Analysis, Gene Center, LMU Munich, 81377 Munich, Germany
| | - Christoph D Spinner
- Department of Internal Medicine II, University Hospital Rechts der Isar, School of Medicine, Technical University of Munich, 81675 Munich, Germany
- German Center for Infection Research (DZIF), Munich Partner Site, 81675 Munich, Germany
| | - Tanja Bauer
- Institute of Virology, School of Medicine, Technical University of Munich, 81675 Munich, Germany
- Institute of Virology, Helmholtz Munich, Trogerstrasse 30, 81675 Munich, Germany
- German Center for Infection Research (DZIF), Munich Partner Site, 81675 Munich, Germany
| | - Percy A Knolle
- Institute of Molecular Immunology and Experimental Oncology, School of Medicine, Technical University of Munich, 81675 Munich, Germany
- German Center for Infection Research (DZIF), Munich Partner Site, 81675 Munich, Germany
| | - Ulrike Protzer
- Institute of Virology, School of Medicine, Technical University of Munich, 81675 Munich, Germany
- Institute of Virology, Helmholtz Munich, Trogerstrasse 30, 81675 Munich, Germany
- German Center for Infection Research (DZIF), Munich Partner Site, 81675 Munich, Germany
| | - Eva C Schulte
- Institute of Virology, School of Medicine, Technical University of Munich, 81675 Munich, Germany
- Institute of Virology, Helmholtz Munich, Trogerstrasse 30, 81675 Munich, Germany
- Department of Psychiatry, University Hospital, LMU Munich, 80336 Munich, Germany
- Institute of Psychiatric Phenomics and Genomics, University Hospital, LMU Munich, 80336 Munich, Germany
| |
Collapse
|
24
|
Braeye T, van Loenhout JAF, Brondeel R, Stouten V, Hubin P, Billuart M, Chung PYJ, Vandromme M, Wyndham-Thomas C, Blot K, Catteau L. COVID-19 vaccine effectiveness against symptomatic infection and hospitalisation in Belgium, July 2021 to May 2022. Euro Surveill 2023; 28:2200768. [PMID: 37382885 PMCID: PMC10311948 DOI: 10.2807/1560-7917.es.2023.28.26.2200768] [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: 09/14/2022] [Accepted: 02/15/2023] [Indexed: 06/30/2023] Open
Abstract
BackgroundThe Belgian COVID-19 vaccination campaign aimed to reduce disease spread and severity.AimWe estimated SARS-CoV-2 variant-specific vaccine effectiveness against symptomatic infection (VEi) and hospitalisation (VEh), given time since vaccination and prior infection.MethodsNationwide healthcare records from July 2021 to May 2022 on testing and vaccination were combined with a clinical hospital survey. We used a test-negative design and proportional hazard regression to estimate VEi and VEh, controlling for prior infection, time since vaccination, age, sex, residence and calendar week of sampling.ResultsWe included 1,932,546 symptomatic individuals, of whom 734,115 tested positive. VEi against Delta waned from an initial estimate of 80% (95% confidence interval (CI): 80-81) to 55% (95% CI: 54-55) 100-150 days after the primary vaccination course. Booster vaccination increased initial VEi to 85% (95% CI: 84-85). Against Omicron, an initial VEi of 33% (95% CI: 30-36) waned to 17% (95% CI: 15-18), while booster vaccination increased VEi to 50% (95% CI: 49-50), which waned to 20% (95% CI: 19-21) 100-150 days after vaccination. Initial VEh for booster vaccination decreased from 96% (95% CI: 95-96) against Delta to 87% (95% CI: 86-89) against Omicron. VEh against Omicron waned to 73% (95% CI: 71-75) 100-150 days after booster vaccination. While recent prior infections conferred higher protection, infections occurring before 2021 remained associated with significant risk reduction against symptomatic infection. Vaccination and prior infection outperformed vaccination or prior infection only.ConclusionWe report waning and a significant decrease in VEi and VEh from Delta to Omicron-dominant periods. Booster vaccination and prior infection attenuated these effects.
Collapse
Affiliation(s)
- Toon Braeye
- Department of Epidemiology and public health, Sciensano, Brussels, Belgium
| | | | - Ruben Brondeel
- Department of Epidemiology and public health, Sciensano, Brussels, Belgium
| | - Veerle Stouten
- Department of Epidemiology and public health, Sciensano, Brussels, Belgium
| | - Pierre Hubin
- Department of Epidemiology and public health, Sciensano, Brussels, Belgium
| | - Matthieu Billuart
- Department of Epidemiology and public health, Sciensano, Brussels, Belgium
| | | | - Mathil Vandromme
- Department of Epidemiology and public health, Sciensano, Brussels, Belgium
| | | | - Koen Blot
- Department of Epidemiology and public health, Sciensano, Brussels, Belgium
| | - Lucy Catteau
- Department of Epidemiology and public health, Sciensano, Brussels, Belgium
| |
Collapse
|
25
|
Westphal T, Mader M, Karsten H, Cords L, Knapp M, Schulte S, Hermanussen L, Peine S, Ditt V, Grifoni A, Addo MM, Huber S, Sette A, Lütgehetmann M, Pischke S, Kwok WW, Sidney J, Schulze zur Wiesch J. Evidence for broad cross-reactivity of the SARS-CoV-2 NSP12-directed CD4 + T-cell response with pre-primed responses directed against common cold coronaviruses. Front Immunol 2023; 14:1182504. [PMID: 37215095 PMCID: PMC10196118 DOI: 10.3389/fimmu.2023.1182504] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 04/06/2023] [Indexed: 05/24/2023] Open
Abstract
Introduction The nonstructural protein 12 (NSP12) of the severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2) has a high sequence identity with common cold coronaviruses (CCC). Methods Here, we comprehensively assessed the breadth and specificity of the NSP12-specific T-cell response after in vitro T-cell expansion with 185 overlapping 15-mer peptides covering the entire SARS-CoV-2 NSP12 at single-peptide resolution in a cohort of 27 coronavirus disease 2019 (COVID-19) patients. Samples of nine uninfected seronegative individuals, as well as five pre-pandemic controls, were also examined to assess potential cross-reactivity with CCCs. Results Surprisingly, there was a comparable breadth of individual NSP12 peptide-specific CD4+ T-cell responses between COVID-19 patients (mean: 12.82 responses; range: 0-25) and seronegative controls including pre-pandemic samples (mean: 12.71 responses; range: 0-21). However, the NSP12-specific T-cell responses detected in acute COVID-19 patients were on average of a higher magnitude. The most frequently detected CD4+ T-cell peptide specificities in COVID-19 patients were aa236-250 (37%) and aa246-260 (44%), whereas the peptide specificities aa686-700 (50%) and aa741-755 (36%), were the most frequently detected in seronegative controls. In CCC-specific peptide-expanded T-cell cultures of seronegative individuals, the corresponding SARS-CoV-2 NSP12 peptide specificities also elicited responses in vitro. However, the NSP12 peptide-specific CD4+ T-cell response repertoire only partially overlapped in patients analyzed longitudinally before and after a SARS-CoV-2 infection. Discussion The results of the current study indicate the presence of pre-primed, cross-reactive CCC-specific T-cell responses targeting conserved regions of SARS-CoV-2, but they also underline the complexity of the analysis and the limited understanding of the role of the SARS-CoV-2 specific T-cell response and cross-reactivity with the CCCs.
Collapse
Affiliation(s)
- Tim Westphal
- Infectious Diseases Unit I, Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- German Center for Infection Research Deutsches Zentrum für Infektionsforschung (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| | - Maria Mader
- Infectious Diseases Unit I, Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Hendrik Karsten
- Infectious Diseases Unit I, Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Leon Cords
- Infectious Diseases Unit I, Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Maximilian Knapp
- Infectious Diseases Unit I, Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sophia Schulte
- Infectious Diseases Unit I, Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lennart Hermanussen
- Infectious Diseases Unit I, Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sven Peine
- Institute of Transfusion Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Vanessa Ditt
- Institute of Transfusion Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Alba Grifoni
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA, United States
| | - Marylyn Martina Addo
- Infectious Diseases Unit I, Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- German Center for Infection Research Deutsches Zentrum für Infektionsforschung (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
- Department for Clinical Immunology of Infectious Diseases, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- Institute of Infection Research and Vaccine Development, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Samuel Huber
- Infectious Diseases Unit I, Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA, United States
| | - Marc Lütgehetmann
- German Center for Infection Research Deutsches Zentrum für Infektionsforschung (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
- Institute of Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sven Pischke
- Infectious Diseases Unit I, Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- German Center for Infection Research Deutsches Zentrum für Infektionsforschung (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| | - William W. Kwok
- Benaroya Research Institute at Virginia Mason, Seattle, WA, United States
| | - John Sidney
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA, United States
| | - Julian Schulze zur Wiesch
- Infectious Diseases Unit I, Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- German Center for Infection Research Deutsches Zentrum für Infektionsforschung (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| |
Collapse
|
26
|
Körber N, Holzmann-Littig C, Wilkens G, Liao BH, Werz ML, Platen L, Cheng CC, Tellenbach M, Kappler V, Lehner V, Mijočević H, Christa C, Assfalg V, Heemann U, Schmaderer C, Protzer U, Braunisch MC, Bauer T, Renders L. Comparable cellular and humoral immunity upon homologous and heterologous COVID-19 vaccination regimens in kidney transplant recipients. Front Immunol 2023; 14:1172477. [PMID: 37063863 PMCID: PMC10102365 DOI: 10.3389/fimmu.2023.1172477] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 03/21/2023] [Indexed: 04/03/2023] Open
Abstract
BackgroundKidney transplant recipients (KTRs) are at high risk for a severe course of coronavirus disease 2019 (COVID-19); thus, effective vaccination is critical. However, the achievement of protective immunogenicity is hampered by immunosuppressive therapies. We assessed cellular and humoral immunity and breakthrough infection rates in KTRs vaccinated with homologous and heterologous COVID-19 vaccination regimens.MethodWe performed a comparative in-depth analysis of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)–specific T-cell responses using multiplex Fluorospot assays and SARS-CoV-2-specific neutralizing antibodies (NAbs) between three-times homologously (n = 18) and heterologously (n = 8) vaccinated KTRs.ResultsWe detected SARS-CoV-2-reactive T cells in 100% of KTRs upon third vaccination, with comparable frequencies, T-cell expression profiles, and relative interferon γ and interleukin 2 production per single cell between homologously and heterologously vaccinated KTRs. SARS-CoV-2-specific NAb positivity rates were significantly higher in heterologously (87.5%) compared to homologously vaccinated (50.0%) KTRs (P < 0.0001), whereas the magnitudes of NAb titers were comparable between both subcohorts after third vaccination. SARS-CoV-2 breakthrough infections occurred in equal numbers in homologously (38.9%) and heterologously (37.5%) vaccinated KTRs with mild-to-moderate courses of COVID-19.ConclusionOur data support a more comprehensive assessment of not only humoral but also cellular SARS-CoV-2-specific immunity in KTRs to provide an in-depth understanding about the COVID-19 vaccine–induced immune response in a transplant setting.
Collapse
Affiliation(s)
- Nina Körber
- Institute of Virology, Helmholtz Zentrum München, Munich, Germany
- *Correspondence: Nina Körber,
| | - Christopher Holzmann-Littig
- Department of Nephrology, Technical University of Munich, School of Medicine, Klinikum Rechts der Isar, Munich, Germany
- Technical University of Munich (TUM) Medical Education Center, School of Medicine, Technical University of Munich, Munich, Germany
| | - Gesa Wilkens
- Institute of Virology, Helmholtz Zentrum München, Munich, Germany
| | - Bo-Hung Liao
- Institute of Virology, Technical University of Munich, School of Medicine, Munich, Germany
| | - Maia L. Werz
- Department of Nephrology, Technical University of Munich, School of Medicine, Klinikum Rechts der Isar, Munich, Germany
| | - Louise Platen
- Department of Nephrology, Technical University of Munich, School of Medicine, Klinikum Rechts der Isar, Munich, Germany
| | - Cho-Chin Cheng
- Institute of Virology, Technical University of Munich, School of Medicine, Munich, Germany
| | - Myriam Tellenbach
- Department of Nephrology, Technical University of Munich, School of Medicine, Klinikum Rechts der Isar, Munich, Germany
| | - Verena Kappler
- Department of Nephrology, Technical University of Munich, School of Medicine, Klinikum Rechts der Isar, Munich, Germany
| | - Viktor Lehner
- Department of Nephrology, Technical University of Munich, School of Medicine, Klinikum Rechts der Isar, Munich, Germany
| | - Hrvoje Mijočević
- Institute of Virology, Technical University of Munich, School of Medicine, Munich, Germany
| | - Catharina Christa
- Institute of Virology, Technical University of Munich, School of Medicine, Munich, Germany
| | - Volker Assfalg
- Department of Surgery, Technical University of Munich, School of Medicine, Klinikum Rechts der Isar, Munich, Germany
| | - Uwe Heemann
- Department of Nephrology, Technical University of Munich, School of Medicine, Klinikum Rechts der Isar, Munich, Germany
| | - Christoph Schmaderer
- Department of Nephrology, Technical University of Munich, School of Medicine, Klinikum Rechts der Isar, Munich, Germany
| | - Ulrike Protzer
- Institute of Virology, Helmholtz Zentrum München, Munich, Germany
- Institute of Virology, Technical University of Munich, School of Medicine, Munich, Germany
- German Center for Infection Research (DZIF), partner site Munich, Munich, Germany
| | - Matthias C. Braunisch
- Department of Nephrology, Technical University of Munich, School of Medicine, Klinikum Rechts der Isar, Munich, Germany
| | - Tanja Bauer
- Institute of Virology, Helmholtz Zentrum München, Munich, Germany
- German Center for Infection Research (DZIF), partner site Munich, Munich, Germany
| | - Lutz Renders
- Department of Nephrology, Technical University of Munich, School of Medicine, Klinikum Rechts der Isar, Munich, Germany
- German Center for Infection Research (DZIF), partner site Munich, Munich, Germany
| |
Collapse
|
27
|
Mohamed KM, Guevara-Hoyer K, García CJ, Bravo LG, Jiménez-Huete A, de la Peña AR, Valeros BM, Velázquez CC, López EC, Cabello N, Estrada V, Corbí ÁL, Fernández-Arquero M, Ocaña A, Delgado-Iribarren A, Martínez-Novillo M, Bolaños E, Anguita E, Peña A, Benavente C, Benítez Fuentes JD, Pérez Segura P, Sánchez-Ramón S. Specific Cellular and Humoral Immune Responses to the Neoantigen RBD of SARS-CoV-2 in Patients with Primary and Secondary Immunodeficiency and Healthy Donors. Biomedicines 2023; 11:biomedicines11041042. [PMID: 37189660 DOI: 10.3390/biomedicines11041042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/14/2023] [Accepted: 03/24/2023] [Indexed: 03/30/2023] Open
Abstract
Patients with antibody deficiency disorders, such as primary immunodeficiency (PID) or secondary immunodeficiency (SID) to B-cell lymphoproliferative disorder (B-CLPD), are two groups vulnerable to developing the severe or chronic form of coronavirus disease caused by SARS-CoV-2 (COVID-19). The data on adaptive immune responses against SARS-CoV-2 are well described in healthy donors, but still limited in patients with antibody deficiency of a different cause. Herein, we analyzed spike-specific IFN-γ and anti-spike IgG antibody responses at 3 to 6 months after exposure to SARS-CoV-2 derived from vaccination and/or infection in two cohorts of immunodeficient patients (PID vs. SID) compared to healthy controls (HCs). Pre-vaccine anti-SARS-CoV-2 cellular responses before vaccine administration were measured in 10 PID patients. Baseline cellular responses were detectable in 4 out of 10 PID patients who had COVID-19 prior to vaccination, perceiving an increase in cellular responses after two-dose vaccination (p < 0.001). Adequate specific cellular responses were observed in 18 out of 20 (90%) PID patients, in 14 out of 20 (70%) SID patients and in 74 out of 81 (96%) HCs after vaccination (and natural infection in some cases). Specific IFN-γ response was significantly higher in HC with respect to PID (1908.5 mUI/mL vs. 1694.1 mUI/mL; p = 0.005). Whereas all SID and HC patients mounted a specific humoral immune response, only 80% of PID patients showed positive anti-SARS-CoV-2 IgG. The titer of anti-SARS-CoV-2 IgG was significantly lower in SID compared with HC patients (p = 0.040), without significant differences between PID and HC patients (p = 0.123) and between PID and SID patients (p =0.683). High proportions of PID and SID patients showed adequate specific cellular responses to receptor binding domain (RBD) neoantigen, with a divergence between the two arms of the adaptive immune response in PID and SID patients. We also focused on the correlation of protection of positive SARS-CoV-2 cellular response to omicron exposure: 27 out of 81 (33.3%) HCs referred COVID-19 detected by PCR or antigen test, 24 with a mild course, 1 with moderate symptoms and the remaining 2 with bilateral pneumonia that were treated in an outpatient basis. Our results might support the relevance of these immunological studies to determine the correlation of protection with severe disease and for deciding the need for additional boosters on a personalized basis. Follow-up studies are required to evaluate the duration and variability in the immune response to COVID-19 vaccination or infection.
Collapse
Affiliation(s)
- Kauzar Mohamed Mohamed
- Department of Immunology, Laboratory Medicine Institute (IML) and Fundación para la Investigación Biomédica del Hospital Clínico San Carlos (IdISSC), Hospital Clínico San Carlos, Calle Profesor Martín Lagos SN, 28040 Madrid, Spain
| | - Kissy Guevara-Hoyer
- Department of Immunology, Laboratory Medicine Institute (IML) and Fundación para la Investigación Biomédica del Hospital Clínico San Carlos (IdISSC), Hospital Clínico San Carlos, Calle Profesor Martín Lagos SN, 28040 Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Complutense University, 28040 Madrid, Spain
| | - Carlos Jiménez García
- Department of Immunology, Laboratory Medicine Institute (IML) and Fundación para la Investigación Biomédica del Hospital Clínico San Carlos (IdISSC), Hospital Clínico San Carlos, Calle Profesor Martín Lagos SN, 28040 Madrid, Spain
| | - Laura García Bravo
- Department of Immunology, Laboratory Medicine Institute (IML) and Fundación para la Investigación Biomédica del Hospital Clínico San Carlos (IdISSC), Hospital Clínico San Carlos, Calle Profesor Martín Lagos SN, 28040 Madrid, Spain
| | | | - Antonia Rodríguez de la Peña
- Department of Immunology, Laboratory Medicine Institute (IML) and Fundación para la Investigación Biomédica del Hospital Clínico San Carlos (IdISSC), Hospital Clínico San Carlos, Calle Profesor Martín Lagos SN, 28040 Madrid, Spain
| | - Beatriz Mediero Valeros
- Department of Immunology, Laboratory Medicine Institute (IML) and Fundación para la Investigación Biomédica del Hospital Clínico San Carlos (IdISSC), Hospital Clínico San Carlos, Calle Profesor Martín Lagos SN, 28040 Madrid, Spain
| | - Cristina Cañizares Velázquez
- Department of Immunology, Laboratory Medicine Institute (IML) and Fundación para la Investigación Biomédica del Hospital Clínico San Carlos (IdISSC), Hospital Clínico San Carlos, Calle Profesor Martín Lagos SN, 28040 Madrid, Spain
| | - Esther Culebras López
- Department of Microbiology, IML and IdISSC, Hospital Clínico San Carlos, 28040 Madrid, Spain
| | - Noemí Cabello
- Unit of Infectious Diseases, Department of Internal Medicine, Hospital Clínico San Carlos, Calle Profesor Martín Lagos SN, 28040 Madrid, Spain
| | - Vicente Estrada
- Unit of Infectious Diseases, Department of Internal Medicine, Hospital Clínico San Carlos, Calle Profesor Martín Lagos SN, 28040 Madrid, Spain
| | - Ángel L Corbí
- Centro de Investigaciones Biológicas (CSIC), C./Ramiro de Maeztu, 9, 28040 Madrid, Spain
| | - Miguel Fernández-Arquero
- Department of Immunology, Laboratory Medicine Institute (IML) and Fundación para la Investigación Biomédica del Hospital Clínico San Carlos (IdISSC), Hospital Clínico San Carlos, Calle Profesor Martín Lagos SN, 28040 Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Complutense University, 28040 Madrid, Spain
| | - Alberto Ocaña
- Department of Microbiology, IML and IdISSC, Hospital Clínico San Carlos, 28040 Madrid, Spain
| | | | - Mercedes Martínez-Novillo
- Clinical Analysis Department, Laboratory Medicine Institute (IML) and Fundación para la Investigación Biomédica del Hospital Clínico San Carlos (IdISSC), Hospital Clínico San Carlos, Calle Profesor Martín Lagos SN, 28040 Madrid, Spain
| | - Estefanía Bolaños
- Department of Hematology, Hospital Clínico San Carlos, IML, IdISSC, Calle Profesor Martín Lagos SN, 28040 Madrid, Spain
| | - Eduardo Anguita
- Department of Hematology, Hospital Clínico San Carlos, IML, IdISSC, Calle Profesor Martín Lagos SN, 28040 Madrid, Spain
| | - Ascensión Peña
- Department of Hematology, Hospital Clínico San Carlos, IML, IdISSC, Calle Profesor Martín Lagos SN, 28040 Madrid, Spain
| | - Celina Benavente
- Department of Hematology, Hospital Clínico San Carlos, IML, IdISSC, Calle Profesor Martín Lagos SN, 28040 Madrid, Spain
| | - Javier David Benítez Fuentes
- Department of Medical Oncology, Hospital Clínico San Carlos, Calle Profesor Martín Lagos SN, 28040 Madrid, Spain
| | - Pedro Pérez Segura
- Department of Medical Oncology, Hospital Clínico San Carlos, Calle Profesor Martín Lagos SN, 28040 Madrid, Spain
| | - Silvia Sánchez-Ramón
- Department of Immunology, Laboratory Medicine Institute (IML) and Fundación para la Investigación Biomédica del Hospital Clínico San Carlos (IdISSC), Hospital Clínico San Carlos, Calle Profesor Martín Lagos SN, 28040 Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Complutense University, 28040 Madrid, Spain
- Department of Clinical Immunology, Hospital Universitario Clínico San Carlos and IdISSC, Calle Profesor Martín Lagos SN, 28040 Madrid, Spain
| |
Collapse
|
28
|
Kanamori R, Yan Y, Ito K, Fukuda H, Hori S, Yamamoto T, Igawa G, Saito K, Horiuchi Y, Nojiri S, Nishizaki Y, Tabe Y, Takahashi K, Naito T. Increased SARS-CoV-2 seroprevalence and spread of infection without awareness among healthcare workers through 2020-2022 in a Japanese medical center. Sci Rep 2023; 13:4941. [PMID: 36973531 PMCID: PMC10040914 DOI: 10.1038/s41598-023-32193-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 03/23/2023] [Indexed: 03/29/2023] Open
Abstract
Despite Japan's high vaccination coverage, daily numbers of new COVID-19 cases have been high. However, studies on the seroprevalence among Japanese people and the causative factors for rapid spread have remained limited. In this study, we aimed to examine the seroprevalence and associated factors in healthcare workers (HCWs) of a medical center in Tokyo using blood samples drawn at annual check-ups from 2020 to 2022. We found that of the 3,788 HCWs in 2022 (by mid-June), 669 were seropositive for N-specific antibodies (tested by Roche Elecsys Anti-SARS-CoV-2 assay); the seroprevalence surged from 0.3% in 2020 and 1.6% in 2021 to 17.7% in 2022. Notably, our study found 325 (48.6%; 325/669) cases were infected without awareness. Among those with a previously PCR-confirmed SARS-CoV-2 infection during the past three years, 79.0% (282/357) were found after January 2022, after the Omicron variant was first detected in Tokyo at the end of 2021. This study indicates the fast spread of the SARS-CoV-2 among HCWs during the Omicron surge in Japan. The high percentage of infection without awareness may be a key driving factor causing rapid person-to-person transmission, as shown in this medical center with high vaccination coverage and strict infection control measures.
Collapse
Affiliation(s)
- Rie Kanamori
- Department of General Medicine, Juntendo University Faculty of Medicine, Hongo 2-1-2, Bunkyo-Ku, Tokyo, 113-8421, Japan
| | - Yan Yan
- Department of General Medicine, Juntendo University Faculty of Medicine, Hongo 2-1-2, Bunkyo-Ku, Tokyo, 113-8421, Japan
| | - Kanami Ito
- Department of Safety and Health Promotion, Juntendo University, Tokyo, Japan
| | - Hiroshi Fukuda
- Department of General Medicine, Juntendo University Faculty of Medicine, Hongo 2-1-2, Bunkyo-Ku, Tokyo, 113-8421, Japan
- Department of Safety and Health Promotion, Juntendo University, Tokyo, Japan
| | - Satoshi Hori
- Infection Control Science, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Takamasa Yamamoto
- Department of Clinical Laboratory, Juntendo University Hospital, Tokyo, Japan
| | - Gene Igawa
- Department of Clinical Laboratory, Juntendo University Hospital, Tokyo, Japan
| | - Kaori Saito
- Department of Clinical Laboratory Medicine, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Yuki Horiuchi
- Department of Clinical Laboratory Medicine, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Shuko Nojiri
- Medical Technology Innovation Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yuji Nishizaki
- Department of General Medicine, Juntendo University Faculty of Medicine, Hongo 2-1-2, Bunkyo-Ku, Tokyo, 113-8421, Japan
- Division of Medical Education, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Yoko Tabe
- Department of Clinical Laboratory Medicine, Juntendo University Faculty of Medicine, Tokyo, Japan
- Department of Research Support Utilizing Bioresource Bank, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Kazuhisa Takahashi
- Department of Research Support Utilizing Bioresource Bank, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Department of Respiratory Medicine, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Toshio Naito
- Department of General Medicine, Juntendo University Faculty of Medicine, Hongo 2-1-2, Bunkyo-Ku, Tokyo, 113-8421, Japan.
- Department of Safety and Health Promotion, Juntendo University, Tokyo, Japan.
- Department of Research Support Utilizing Bioresource Bank, Juntendo University Graduate School of Medicine, Tokyo, Japan.
| |
Collapse
|
29
|
Aguilar R, Li X, Crowell CS, Burrell T, Vidal M, Rubio R, Jiménez A, Hernández-Luis P, Hofmann D, Mijočević H, Jeske S, Christa C, D'Ippolito E, Lingor P, Knolle PA, Roggendorf H, Priller A, Yazici S, Carolis C, Mayor A, Schreiner P, Poppert H, Beyer H, Schambeck SE, Izquierdo L, Tortajada M, Angulo A, Soutschek E, Engel P, Garcia-Basteiro A, Busch DH, Moncunill G, Protzer U, Dobaño C, Gerhard M. RBD-Based ELISA and Luminex Predict Anti-SARS-CoV-2 Surrogate-Neutralizing Activity in Two Longitudinal Cohorts of German and Spanish Health Care Workers. Microbiol Spectr 2023; 11:e0316522. [PMID: 36622140 PMCID: PMC9927417 DOI: 10.1128/spectrum.03165-22] [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: 08/12/2022] [Accepted: 12/04/2022] [Indexed: 01/10/2023] Open
Abstract
The ability of antibodies to neutralize severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is an important correlate of protection. For routine evaluation of protection, however, a simple and cost-efficient anti-SARS-CoV-2 serological assay predictive of serum neutralizing activity is needed. We analyzed clinical epidemiological data and blood samples from two cohorts of health care workers in Barcelona and Munich to compare several immunological readouts for evaluating antibody levels that could be surrogates of neutralizing activity. We measured IgG levels against SARS-CoV-2 spike protein (S), its S2 subunit, the S1 receptor binding domain (RBD), and the full length and C terminus of nucleocapsid (N) protein by Luminex, and against RBD by enzyme-linked immunosorbent assay (ELISA), and assessed those as predictors of plasma surrogate-neutralizing activity measured by a flow cytometry assay. In addition, we determined the clinical and demographic factors affecting plasma surrogate-neutralizing capacity. Both cohorts showed a high positive correlation between IgG levels to S antigen, especially to RBD, and the levels of plasma surrogate-neutralizing activity, suggesting RBD IgG as a good correlate of plasma neutralizing activity. Symptomatic infection, with symptoms such as loss of taste, dyspnea, rigors, fever and fatigue, was positively associated with anti-RBD IgG positivity by ELISA and Luminex, and with plasma surrogate-neutralizing activity. Our serological assays allow for the prediction of serum neutralization activity without the cost, hazards, time, and expertise needed for surrogate or conventional neutralization assays. Once a cutoff is established, these relatively simple high-throughput antibody assays will provide a fast and cost-effective method of assessing levels of protection from SARS-CoV-2 infection. IMPORTANCE Neutralizing antibody titers are the best correlate of protection against SARS-CoV-2. However, current tests to measure plasma or serum neutralizing activity do not allow high-throughput screening at the population level. Serological tests could be an alternative if they are proved to be good predictors of plasma neutralizing activity. In this study, we analyzed the SARS-CoV-2 serological profiles of two cohorts of health care workers by applying Luminex and ELISA in-house serological assays. Correlations of both serological tests were assessed between them and with a flow cytometry assay to determine plasma surrogate-neutralizing activity. Both assays showed a high positive correlation between IgG levels to S antigens, especially RBD, and the levels of plasma surrogate-neutralizing activity. This result suggests IgG to RBD as a good correlate of plasma surrogate-neutralizing activity and indicates that serology of IgG to RBD could be used to assess levels of protection from SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Ruth Aguilar
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Xue Li
- Institute of Medical Microbiology, Immunology, and Hygiene, School of Medicine, Technical University of Munich (TUM), Munich, Germany
| | - Claudia S. Crowell
- Institute of Medical Microbiology, Immunology, and Hygiene, School of Medicine, Technical University of Munich (TUM), Munich, Germany
| | - Teresa Burrell
- Institute of Medical Microbiology, Immunology, and Hygiene, School of Medicine, Technical University of Munich (TUM), Munich, Germany
| | - Marta Vidal
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Rocio Rubio
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Alfons Jiménez
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Barcelona, Catalonia, Spain
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
| | - Pablo Hernández-Luis
- Immunology Unit, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universitat de Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Dieter Hofmann
- Institute of Virology, School of Medicine, Technical University of Munich, Munich, Germany
- German Center for Infection Research (DZIF), Munich, Germany
| | - Hrvoje Mijočević
- Institute of Virology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Samuel Jeske
- Institute of Virology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Catharina Christa
- Institute of Virology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Elvira D'Ippolito
- Institute of Medical Microbiology, Immunology, and Hygiene, School of Medicine, Technical University of Munich (TUM), Munich, Germany
| | - Paul Lingor
- Klinikum rechts der Isar, Department of Neurology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Percy A. Knolle
- German Center for Infection Research (DZIF), Munich, Germany
- Klinikum rechts der Isar, Institute of Molecular Immunology and Experimental Oncology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Hedwig Roggendorf
- Klinikum rechts der Isar, Institute of Molecular Immunology and Experimental Oncology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Alina Priller
- Klinikum rechts der Isar, Institute of Molecular Immunology and Experimental Oncology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Sarah Yazici
- Klinikum rechts der Isar, Institute of Molecular Immunology and Experimental Oncology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Carlo Carolis
- Biomolecular Screening and Protein Technologies Unit, Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Alfredo Mayor
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Barcelona, Catalonia, Spain
| | | | | | | | - Sophia E. Schambeck
- Institute of Medical Microbiology, Immunology, and Hygiene, School of Medicine, Technical University of Munich (TUM), Munich, Germany
- Helios Klinikum München West, Munich, Germany
| | - Luis Izquierdo
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Barcelona, Catalonia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Barcelona, Spain
| | - Marta Tortajada
- Occupational Health Department, Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
| | - Ana Angulo
- Immunology Unit, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universitat de Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | | | - Pablo Engel
- Immunology Unit, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universitat de Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Alberto Garcia-Basteiro
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Barcelona, Catalonia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Barcelona, Spain
- Department of Preventive Medicine and Epidemiology, Hospital Clinic, Universitat de Barcelona, Barcelona, Spain
| | - Dirk H. Busch
- Institute of Medical Microbiology, Immunology, and Hygiene, School of Medicine, Technical University of Munich (TUM), Munich, Germany
- German Center for Infection Research (DZIF), Munich, Germany
| | - Gemma Moncunill
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Barcelona, Catalonia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Barcelona, Spain
| | - Ulrike Protzer
- Institute of Virology, School of Medicine, Technical University of Munich, Munich, Germany
- German Center for Infection Research (DZIF), Munich, Germany
| | - Carlota Dobaño
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Barcelona, Catalonia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Barcelona, Spain
| | - Markus Gerhard
- Institute of Medical Microbiology, Immunology, and Hygiene, School of Medicine, Technical University of Munich (TUM), Munich, Germany
- German Center for Infection Research (DZIF), Munich, Germany
| |
Collapse
|
30
|
Hybrid immunity in immunocompromised patients with CLL after SARS-CoV-2 infection followed by booster mRNA vaccination. Blood 2022; 140:2403-2407. [PMID: 36150168 PMCID: PMC9512527 DOI: 10.1182/blood.2022016815] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 09/03/2022] [Indexed: 01/21/2023] Open
|
31
|
Cardoso AR, Alves JF, Frasco MF, Piloto AM, Serrano V, Mateus D, Sebastião AI, Matos AM, Carmo A, Cruz T, Fortunato E, Sales MGF. An ultra-sensitive electrochemical biosensor using the Spike protein for capturing antibodies against SARS-CoV-2 in point-of-care. Mater Today Bio 2022; 16:100354. [PMID: 35847374 PMCID: PMC9270181 DOI: 10.1016/j.mtbio.2022.100354] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 07/02/2022] [Accepted: 07/05/2022] [Indexed: 11/30/2022] Open
Abstract
This work presents an innovative ultra-sensitive biosensor having the Spike protein on carbon-based screen-printed electrodes (SPEs), for monitoring in point-of-care antibodies against SARS-CoV-2, a very important tool for epidemiological monitoring of COVID-19 infection and establishing vaccination schemes. In an innovative and simple approach, a highly conductive support is combined with the direct adsorption of Spike protein to enable an extensive antibody capture. The high conductivity was ensured by using carboxylated carbon nanotubes on the carbon electrode, by means of a simple and quick approach, which also increased the surface area. These were then modified with EDC/NHS chemistry to produce an amine layer and undergo Spike protein adsorption, to generate a stable layer capable of capturing the antibodies against SARS-CoV-2 in serum with great sensitivity. Electrochemical impedance spectroscopy was used to evaluate the analytical performance of this biosensor in serum. It displayed a linear response between 1.0 pg/mL and 10 ng/mL, with a detection limit of ∼0.7 pg/mL. The analysis of human positive sera containing antibody in a wide range of concentrations yielded accurate data, correlating well with the reference method. It also offered the unique ability of discriminating antibody concentrations in sera below 2.3 μg/mL, the lowest value detected by the commercial method. In addition, a proof-of-concept study was performed by labelling anti-IgG antibodies with quantum dots to explore a new electrochemical readout based on the signal generated upon binding to the anti-S protein antibodies recognised on the surface of the biosensor. Overall, the alternative serologic assay presented is a promising tool for assessing protective immunity to SARS-CoV-2 and a potential guide for revaccination. An ultra-sensitive biosensor for detection of low levels of antibodies against SARS-CoV-2. Highly conductive substrate with adsorbed protein S and point-of-care capability. Application to human sera samples and good correlation with commercial method. Electrochemical impedance readings with an iron-based redox probe. Alternative electrochemical impedance readings with anti-IgG labelled with quantum dots.
Collapse
Affiliation(s)
- Ana R Cardoso
- BioMark@UC/CEB - LABBELS, Faculty of Sciences and Technology, University of Coimbra, Coimbra, Portugal.,BioMark@ISEP/ CEB - LABBELS, School of Engineering, Polytechnic Institute of Porto, Porto, Portugal.,CENIMAT
- i3N, Department of Materials Science, School of Science and Technology, NOVA University of Lisbon and CEMOP/UNINOVA, Campus de Caparica, 2829-516, Caparica, Portugal
| | - João Frederico Alves
- BioMark@UC/CEB - LABBELS, Faculty of Sciences and Technology, University of Coimbra, Coimbra, Portugal
| | - Manuela F Frasco
- BioMark@UC/CEB - LABBELS, Faculty of Sciences and Technology, University of Coimbra, Coimbra, Portugal
| | - Ana Margarida Piloto
- BioMark@ISEP/ CEB - LABBELS, School of Engineering, Polytechnic Institute of Porto, Porto, Portugal
| | - Verónica Serrano
- BioMark@UC/CEB - LABBELS, Faculty of Sciences and Technology, University of Coimbra, Coimbra, Portugal
| | - Daniela Mateus
- BioMark@UC/CEB - LABBELS, Faculty of Sciences and Technology, University of Coimbra, Coimbra, Portugal.,Center for Neuroscience and Cell Biology, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| | - Ana Isabel Sebastião
- Center for Neuroscience and Cell Biology, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| | - Ana Miguel Matos
- Chemical Engineering Processes and Forest Products Research Center, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| | - Anália Carmo
- Clinical Pathology Department, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Teresa Cruz
- Center for Neuroscience and Cell Biology, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| | - Elvira Fortunato
- CENIMAT
- i3N, Department of Materials Science, School of Science and Technology, NOVA University of Lisbon and CEMOP/UNINOVA, Campus de Caparica, 2829-516, Caparica, Portugal
| | - M Goreti F Sales
- BioMark@UC/CEB - LABBELS, Faculty of Sciences and Technology, University of Coimbra, Coimbra, Portugal.,BioMark@ISEP/ CEB - LABBELS, School of Engineering, Polytechnic Institute of Porto, Porto, Portugal
| |
Collapse
|
32
|
Bispo ECI, Silva-Carvalho AÉ, Sousa MRR, Neves FDAR, Carvalho JL, Arganaraz ER, Saldanha-Araujo F. Differential peripheral blood mononuclear cell reactivity against SARS-CoV-2 proteins in naïve and previously infected subjects following COVID-19 vaccination. CLINICAL IMMUNOLOGY COMMUNICATIONS 2022; 2:172-176. [PMID: 38013967 PMCID: PMC9714124 DOI: 10.1016/j.clicom.2022.11.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 11/29/2022] [Accepted: 11/30/2022] [Indexed: 12/03/2022]
Abstract
The decline in vaccine efficacy and the risk of reinfection by SARS-CoV-2 make new studies important to better characterize the immune response against the virus and its components. Here, we investigated the pattern of activation of T-cells and the expression of inflammatory factors by PBMCs obtained from naïve and previously infected subjects following COVID-19 vaccination, after PBMCs stimulation with S1, RBD, and N-RBD SARS-CoV-2 proteins. PBMCs showed low levels of ACE2 and TMPRSS2 transcripts, which were not modulated by the exposure of these cells to SARS-CoV-2 proteins. Compared to S1 and RBD, N-RBD stimulation showed a greater ability to stimulate T-cell reactivity, according to CD25 and CD69 markers. Interestingly, T-cell reactivity was more pronounced in vaccinated subjects with prior SARS-CoV-2 infection than in vaccinated donors who never had been diagnosed with COVID-19. Finally, N-RBD stimulation promoted greater expression of IL-6 and IFN-γ in PBMCs, which reinforces the greater immunogenic potential of this protein in the vaccinated subjects. These data suggest that PBMCs from previously infected and vaccinated subjects are more reactive than those derived from just vaccinated donors. Moreover, the N-RBD together viral proteins showed a greater stimulatory capacity than S1 and RBD viral proteins.
Collapse
Affiliation(s)
- Elizabete Cristina Iseke Bispo
- Laboratório de Hematologia e Células-Tronco, Faculdade de Ciências da Saúde, Universidade de Brasília, Campus Darcy Ribeiro, Av. L2 Norte, Brasília, DF 70.910-900, Brasil
| | - Amandda Évelin Silva-Carvalho
- Laboratório de Hematologia e Células-Tronco, Faculdade de Ciências da Saúde, Universidade de Brasília, Campus Darcy Ribeiro, Av. L2 Norte, Brasília, DF 70.910-900, Brasil
| | - Marielly Reis Resende Sousa
- Laboratório de Hematologia e Células-Tronco, Faculdade de Ciências da Saúde, Universidade de Brasília, Campus Darcy Ribeiro, Av. L2 Norte, Brasília, DF 70.910-900, Brasil
| | - Francisco de Assis Rocha Neves
- Laboratório de Farmacologia Molecular, Faculdade de Ciências da Saúde, Universidade de Brasília, Av. L2 Norte, Brasília, DF 70.910-900, Brasil
| | - Juliana Lott Carvalho
- Laboratório Interdisciplinar de Biociências, Faculdade de Medicina, Universidade de Brasília, Av. L2 Norte, Brasília, DF 70.910-900, Brasil
| | - Enrique Roberto Arganaraz
- Laboratório de Virologia Molecular, Faculdade de Ciências da Saúde, Universidade de Brasília, Av. L2 Norte, Brasília, DF 70.910-900, Brasil
| | - Felipe Saldanha-Araujo
- Laboratório de Hematologia e Células-Tronco, Faculdade de Ciências da Saúde, Universidade de Brasília, Campus Darcy Ribeiro, Av. L2 Norte, Brasília, DF 70.910-900, Brasil
| |
Collapse
|
33
|
Antibody efficacy of inactivated vaccine boosters (CoronaVac) against Omicron variant from a 15-month follow-up study. J Infect 2022; 85:e119-e121. [PMID: 35760302 PMCID: PMC9233874 DOI: 10.1016/j.jinf.2022.06.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 06/15/2022] [Accepted: 06/18/2022] [Indexed: 01/06/2023]
|
34
|
Karachaliou M, Moncunill G, Espinosa A, Castaño-Vinyals G, Rubio R, Vidal M, Jiménez A, Prados E, Carreras A, Cortés B, Blay N, Bañuls M, Pleguezuelos V, Melero NR, Serra P, Parras D, Izquierdo L, Santamaría P, Carolis C, Papantoniou K, Goldberg X, Aguilar R, Garcia-Aymerich J, de Cid R, Kogevinas M, Dobaño C. SARS-CoV-2 infection, vaccination, and antibody response trajectories in adults: a cohort study in Catalonia. BMC Med 2022; 20:347. [PMID: 36109713 PMCID: PMC9479347 DOI: 10.1186/s12916-022-02547-2] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 09/01/2022] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND Heterogeneity of the population in relation to infection, COVID-19 vaccination, and host characteristics is likely reflected in the underlying SARS-CoV-2 antibody responses. METHODS We measured IgM, IgA, and IgG levels against SARS-CoV-2 spike and nucleocapsid antigens in 1076 adults of a cohort study in Catalonia between June and November 2020 and a second time between May and July 2021. Questionnaire data and electronic health records on vaccination and COVID-19 testing were available in both periods. Data on several lifestyle, health-related, and sociodemographic characteristics were also available. RESULTS Antibody seroreversion occurred in 35.8% of the 64 participants non-vaccinated and infected almost a year ago and was related to asymptomatic infection, age above 60 years, and smoking. Moreover, the analysis on kinetics revealed that among all responses, IgG RBD, IgA RBD, and IgG S2 decreased less within 1 year after infection. Among vaccinated, 2.1% did not present antibodies at the time of testing and approximately 1% had breakthrough infections post-vaccination. In the post-vaccination era, IgM responses and those against nucleoprotein were much less prevalent. In previously infected individuals, vaccination boosted the immune response and there was a slight but statistically significant increase in responses after a 2nd compared to the 1st dose. Infected vaccinated participants had superior antibody levels across time compared to naïve-vaccinated people. mRNA vaccines and, particularly the Spikevax, induced higher antibodies after 1st and 2nd doses compared to Vaxzevria or Janssen COVID-19 vaccines. In multivariable regression analyses, antibody responses after vaccination were predicted by the type of vaccine, infection age, sex, smoking, and mental and cardiovascular diseases. CONCLUSIONS Our data support that infected people would benefit from vaccination. Results also indicate that hybrid immunity results in superior antibody responses and infection-naïve people would need a booster dose earlier than previously infected people. Mental diseases are associated with less efficient responses to vaccination.
Collapse
Affiliation(s)
- Marianna Karachaliou
- Barcelona Institute for Global Health (ISGlobal), Doctor Aiguader, 88, 08003, Barcelona, Spain.
| | - Gemma Moncunill
- Barcelona Institute for Global Health (ISGlobal), Doctor Aiguader, 88, 08003, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Barcelona, Spain
| | - Ana Espinosa
- Barcelona Institute for Global Health (ISGlobal), Doctor Aiguader, 88, 08003, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), 08036, Madrid, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- Hospital del Mar Medical Research Institute (IMIM), 08003, Barcelona, Spain
| | - Gemma Castaño-Vinyals
- Barcelona Institute for Global Health (ISGlobal), Doctor Aiguader, 88, 08003, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), 08036, Madrid, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- Hospital del Mar Medical Research Institute (IMIM), 08003, Barcelona, Spain
| | - Rocío Rubio
- Barcelona Institute for Global Health (ISGlobal), Doctor Aiguader, 88, 08003, Barcelona, Spain
| | - Marta Vidal
- Barcelona Institute for Global Health (ISGlobal), Doctor Aiguader, 88, 08003, Barcelona, Spain
| | - Alfons Jiménez
- Barcelona Institute for Global Health (ISGlobal), Doctor Aiguader, 88, 08003, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), 08036, Madrid, Spain
| | - Esther Prados
- Barcelona Institute for Global Health (ISGlobal), Doctor Aiguader, 88, 08003, Barcelona, Spain
| | - Anna Carreras
- Genomes for Life-GCAT lab. Institute for Health Science Research Germans Trias i Pujol (IGTP), Badalona, Spain
| | - Beatriz Cortés
- Genomes for Life-GCAT lab. Institute for Health Science Research Germans Trias i Pujol (IGTP), Badalona, Spain
| | - Natàlia Blay
- Genomes for Life-GCAT lab. Institute for Health Science Research Germans Trias i Pujol (IGTP), Badalona, Spain
| | - Marc Bañuls
- Barcelona Institute for Global Health (ISGlobal), Doctor Aiguader, 88, 08003, Barcelona, Spain
| | | | | | - Pau Serra
- Institut d'Investigacions Biomèdiques August Pi Sunyer (IDIBAPS), Barcelona, Spain
| | - Daniel Parras
- Institut d'Investigacions Biomèdiques August Pi Sunyer (IDIBAPS), Barcelona, Spain
| | - Luis Izquierdo
- Barcelona Institute for Global Health (ISGlobal), Doctor Aiguader, 88, 08003, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Barcelona, Spain
| | - Pere Santamaría
- Institut d'Investigacions Biomèdiques August Pi Sunyer (IDIBAPS), Barcelona, Spain
- Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Carlo Carolis
- Centre for Genomic Regulation (CRG), Barcelona, Spain
| | - Kyriaki Papantoniou
- Department of Epidemiology, Center for Public Health, Medical University of Vienna, Vienna, Austria
| | - Ximena Goldberg
- Barcelona Institute for Global Health (ISGlobal), Doctor Aiguader, 88, 08003, Barcelona, Spain
| | - Ruth Aguilar
- Barcelona Institute for Global Health (ISGlobal), Doctor Aiguader, 88, 08003, Barcelona, Spain
| | - Judith Garcia-Aymerich
- Barcelona Institute for Global Health (ISGlobal), Doctor Aiguader, 88, 08003, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), 08036, Madrid, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- Hospital del Mar Medical Research Institute (IMIM), 08003, Barcelona, Spain
| | - Rafael de Cid
- Genomes for Life-GCAT lab. Institute for Health Science Research Germans Trias i Pujol (IGTP), Badalona, Spain
| | - Manolis Kogevinas
- Barcelona Institute for Global Health (ISGlobal), Doctor Aiguader, 88, 08003, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), 08036, Madrid, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- Hospital del Mar Medical Research Institute (IMIM), 08003, Barcelona, Spain
| | - Carlota Dobaño
- Barcelona Institute for Global Health (ISGlobal), Doctor Aiguader, 88, 08003, Barcelona, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Barcelona, Spain.
- Barcelona Institute for Global Health (ISGlobal), Carrer Rosello 132, 08036, Barcelona, Spain.
| |
Collapse
|
35
|
Cross-Reactivity of SARS-CoV-2 Nucleocapsid-Binding Antibodies and Its Implication for COVID-19 Serology Tests. Viruses 2022; 14:v14092041. [PMID: 36146847 PMCID: PMC9502088 DOI: 10.3390/v14092041] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 09/09/2022] [Accepted: 09/12/2022] [Indexed: 11/17/2022] Open
Abstract
The emergence of the new coronavirus SARS-CoV-2 in late 2019 led to the global pandemic COVID-19, causing a profound socioeconomic crisis. Adequate diagnostic tools need to be developed to control the ongoing spread of infection. Virus-specific humoral immunity in COVID-19 patients and those vaccinated with specific vaccines has been characterized in numerous studies, mainly using Spike protein-based serology tests. However, Spike protein and specifically its receptor-binding domain (RBD) are mutation-prone, suggesting the reduced sensitivity of the validated serology tests in detecting antibodies raised to variants of concern (VOC). The viral nucleocapsid (N) protein is more conserved compared to Spike, but little is known about cross-reactivity of the N-specific antibodies between the ancestral B.1 virus and different VOCs. Here, we generated recombinant N phosphoproteins from different SARS-CoV-2 strains and analyzed the magnitude of N-specific antibodies in COVID-19 convalescent sera using an in-house N-based ELISA test system. We found a strong positive correlation in the magnitude of anti-N (B.1) antibodies and antibodies specific to various VOCs in COVID-19-recovered patients, suggesting that the N-binding antibodies are highly cross-reactive, and the most immunogenic epitopes within this protein are not under selective pressure. Overall, our study suggests that the RBD-based serology tests should be timely updated to reflect the constantly evolving nature of the SARS-CoV-2 Spike protein, whereas the validated N-based test systems can be used for the analysis of sera from COVID-19 patients regardless of the strain that caused the infection.
Collapse
|
36
|
Brlić PK, Pavletić M, Lerga M, Krstanović F, Matešić MP, Miklić K, Malić S, Mikša L, Pajcur M, Peruč D, Schubert M, Bertoglio F, Arapović J, Protić A, Šustić A, Milošević M, Šain LČ, Jonjić S, Lisnić VJ, Brizić I. SARS-CoV-2 Spike and Nucleocapsid Antibody Response in Vaccinated Croatian Healthcare Workers and Infected Hospitalized Patients: A Single Center Cohort Study. Viruses 2022; 14:1966. [PMID: 36146773 PMCID: PMC9503044 DOI: 10.3390/v14091966] [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: 08/12/2022] [Revised: 08/31/2022] [Accepted: 09/01/2022] [Indexed: 11/17/2022] Open
Abstract
Studies assessing the dynamics and duration of antibody responses following SARS-CoV-2 infection or vaccination are an invaluable tool for vaccination schedule planning, assessment of risk groups and management of pandemics. In this study, we developed and employed ELISA assays to analyze the humoral responses to Nucleocapsid and Spike proteins in vaccinated health-care workers (HCW) and critically ill COVID-19 patients. Sera of more than 1000 HCWs and critically ill patients from the Clinical Hospital Center Rijeka were tested across a one-year period, encompassing the spread of major SARS-CoV-2 variants of concern (VOCs). We observed 97% of seroconversion in HCW cohort as well as sustained anti-Spike antibody response in vaccinees for more than 6 months. In contrast, the infection-induced anti-Nucleocapsid response was waning significantly in a six-month period. Furthermore, a substantial decrease in vaccinees' anti-Spike antibodies binding to Spike protein of Omicron VOC was also observed. Critically ill COVID-19 patients had higher levels of anti-Spike and anti-Nucleocapsid antibodies compared to HCWs. No significant differences in anti-Spike and anti-Nucleocapsid antibody levels between the critically ill COVID-19 patients that were on non-invasive oxygen supplementation and those on invasive ventilation support were observed. However, stronger anti-Spike, but not anti-Nucleocapsid, antibody response correlated with a better disease outcome in the cohort of patients on invasive ventilation support. Altogether, our results contribute to the growing pool of data on humoral responses to SARS-CoV-2 infection and vaccination.
Collapse
Affiliation(s)
- Paola Kučan Brlić
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia
| | - Martina Pavletić
- Emergency Department, Clinical Hospital Center Rijeka, 51000 Rijeka, Croatia
| | - Mate Lerga
- Emergency Department, Clinical Hospital Center Rijeka, 51000 Rijeka, Croatia
| | - Fran Krstanović
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia
| | - Marina Pribanić Matešić
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia
| | - Karmela Miklić
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia
| | - Suzana Malić
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia
| | - Leonarda Mikša
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia
| | - Maja Pajcur
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia
| | - Dolores Peruč
- Department of Microbiology and Parasitology, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia
| | - Maren Schubert
- Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Technische Universität Braunschweig, Spielmannstr. 7, 38106 Braunschweig, Germany
| | - Federico Bertoglio
- Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Technische Universität Braunschweig, Spielmannstr. 7, 38106 Braunschweig, Germany
| | - Jurica Arapović
- Faculty of Medicine, University of Mostar, Bijeli Brijeg b.b., 88000 Mostar, Bosnia and Herzegovina
| | - Alen Protić
- Department of Anesthesiology, Reanimation, Intensive Care and Emergency Medicine, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| | - Alan Šustić
- Department of Anesthesiology, Reanimation, Intensive Care and Emergency Medicine, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
- Department of Clinical Medical Science II, Faculty of Health Studies, University of Rijeka, 51000 Rijeka, Croatia
| | - Marko Milošević
- Department of Anesthesiology, Reanimation, Intensive Care and Emergency Medicine, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| | - Luka Čičin Šain
- Helmholtz Center for Infection Research, Department of Viral Immunology, 38124 Braunschweig, Germany
- German Centre for Infection Research (DZIF), Partner Site Hannover/Braunschweig, 38124 Braunschweig, Germany
- Centre for Individualised Infection Medicine (CiiM), Joint Venture of Helmholtz Centre for Infection Research and Hannover Medical School, 30625 Hannover, Germany
| | - Stipan Jonjić
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia
| | - Vanda Juranić Lisnić
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia
| | - Ilija Brizić
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia
| |
Collapse
|
37
|
Garlantezec R, Tadié E, Heslan C, Gary-Bobo P, Oumari S, Saade A, Sitruk A, Tattevin P, Thibault V, Paris C. SARS-CoV-2 seroprevalence and antibodies persistence among health care workers after the first COVID-19 wave in nine hospitals in Western France. Infect Dis Now 2022; 52:447-452. [PMID: 36108975 PMCID: PMC9467933 DOI: 10.1016/j.idnow.2022.09.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 07/05/2022] [Accepted: 09/06/2022] [Indexed: 11/15/2022]
Abstract
Objectives To estimate the SARS-CoV-2 IgG seroprevalence rate in healthcare workers (HCWs) from Western France after the first 2020 wave, its determinants and the kinetics of total SARS-CoV-2 antibodies. Patients and methods Overall, 9,453 HCWs responded to a self-questionnaire and underwent a lateral flow immunoassay to assess SARS-CoV-2 IgG presence. For 72 HCWs who tested positive, total anti-nucleocapsid antibodies were assessed at day 0, 30, and 90. Results SARS-CoV-2 IgG seroprevalence rate was 1.06 % [0.86 %–1.27 %]. Factors associated with IgG presence were gender, performing upper respiratory tract samples, contact with HCWs or household members diagnosed with COVID-19. Total antibodies decreased between day 0 and day 90, with anosmia or ageusia, and were higher in HCWs older than 50 years. Conclusion We reported a low prevalence rate of IgG and identified several risk factors associated with its presence and persistence of total antibodies. Additional studies are needed to confirm these observations.
Collapse
Affiliation(s)
- R Garlantezec
- CHU de Rennes, Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000 Rennes, France.
| | - E Tadié
- CHU de Rennes, F-35000 Rennes, France
| | - C Heslan
- CHU de Rennes, F-35000 Rennes, France
| | | | - S Oumari
- CHU de Rennes, F-35000 Rennes, France
| | - A Saade
- CHU de Rennes, Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000 Rennes, France
| | - A Sitruk
- CHU de Rennes, Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000 Rennes, France
| | - P Tattevin
- CHU de Rennes, Univ Rennes, INSERM U1230, IFR140, F-35033 Rennes, France
| | - V Thibault
- CHU de Rennes, Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000 Rennes, France
| | - C Paris
- CHU de Rennes, Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000 Rennes, France
| |
Collapse
|
38
|
Cheng CC, Platen L, Christa C, Tellenbach M, Kappler V, Bester R, Liao BH, Holzmann-Littig C, Werz M, Schönhals E, Platen E, Eggerer P, Tréguer L, Küchle C, Schmaderer C, Heemann U, Renders L, Protzer U, Braunisch MC. Improved SARS-CoV-2 Neutralization of Delta and Omicron BA.1 Variants of Concern after Fourth Vaccination in Hemodialysis Patients. Vaccines (Basel) 2022; 10:vaccines10081328. [PMID: 36016216 PMCID: PMC9415993 DOI: 10.3390/vaccines10081328] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/11/2022] [Accepted: 08/13/2022] [Indexed: 12/21/2022] Open
Abstract
Hemodialysis patients are exposed to a markedly increased risk when infected with SARS-CoV-2. To date, it is unclear if hemodialysis patients benefit from four vaccinations. A total of 142 hemodialysis patients received four COVID-19 vaccinations until March 2022. RDB binding antibody titers were determined in a competitive surrogate neutralization assay. Vero-E6 cells were infected with SARS-CoV-2 variants of concern (VoC), Delta (B.1.617.2), or Omicron (B.1.1.529, sub-lineage BA.1) to determine serum infection neutralization capacity. Four weeks after the fourth vaccination, serum infection neutralization capacity significantly increased from a 50% inhibitory concentration (IC50, serum dilution factor 1:x) of 247.0 (46.3−1560.8) to 2560.0 (1174.0−2560.0) for the Delta VoC, and from 37.5 (20.0−198.8) to 668.5 (182.2−2560.0) for the Omicron VoC (each p < 0.001) compared to four months after the third vaccination. A significant increase in the neutralization capacity was even observed for patients with high antibody titers after three vaccinations (p < 0.001). Ten patients with SARS-CoV-2 breakthrough infection after the first blood sampling had by trend lower prior neutralization capacity for Omicron (p = 0.051). Our findings suggest that hemodialysis patients benefit from a fourth vaccination in particular in the light of the highly infectious SARS-CoV-2 Omicron-variants. A routinely applied four-time vaccination seems to broaden immunity against variants and would be recommended in hemodialysis patients.
Collapse
Affiliation(s)
- Cho-Chin Cheng
- Institute of Virology, School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Louise Platen
- Department of Nephrology, School of Medicine, Technical University of Munich, Klinikum Rechts der Isar, 81675 Munich, Germany
| | - Catharina Christa
- Institute of Virology, School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Myriam Tellenbach
- Department of Nephrology, School of Medicine, Technical University of Munich, Klinikum Rechts der Isar, 81675 Munich, Germany
| | - Verena Kappler
- Department of Nephrology, School of Medicine, Technical University of Munich, Klinikum Rechts der Isar, 81675 Munich, Germany
| | - Romina Bester
- Institute of Virology, School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Bo-Hung Liao
- Institute of Virology, School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Christopher Holzmann-Littig
- Department of Nephrology, School of Medicine, Technical University of Munich, Klinikum Rechts der Isar, 81675 Munich, Germany
- TUM Medical Education Center, School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Maia Werz
- Department of Nephrology, School of Medicine, Technical University of Munich, Klinikum Rechts der Isar, 81675 Munich, Germany
| | - Emely Schönhals
- Department of Nephrology, School of Medicine, Technical University of Munich, Klinikum Rechts der Isar, 81675 Munich, Germany
| | - Eva Platen
- Kidney Center Eifel Dialyse, 53894 Mechernich, Germany
| | - Peter Eggerer
- KfH Kidney Center Harlaching, Munich-Harlaching, 81545 Munich, Germany
| | | | - Claudius Küchle
- Department of Nephrology, School of Medicine, Technical University of Munich, Klinikum Rechts der Isar, 81675 Munich, Germany
| | - Christoph Schmaderer
- Department of Nephrology, School of Medicine, Technical University of Munich, Klinikum Rechts der Isar, 81675 Munich, Germany
| | - Uwe Heemann
- Department of Nephrology, School of Medicine, Technical University of Munich, Klinikum Rechts der Isar, 81675 Munich, Germany
| | - Lutz Renders
- Department of Nephrology, School of Medicine, Technical University of Munich, Klinikum Rechts der Isar, 81675 Munich, Germany
- German Center for Infection Research (DZIF), Partner Site, 81675 Munich, Germany
| | - Ulrike Protzer
- Institute of Virology, School of Medicine, Technical University of Munich, 81675 Munich, Germany
- German Center for Infection Research (DZIF), Partner Site, 81675 Munich, Germany
- Institute of Virology, Helmholtz Munich, 85764 Munich, Germany
- Correspondence: (U.P.); (M.C.B.); Tel.: +0049-(0)-89-4140-6863 (U.P.); +0049-(0)-89-4140-2231 (M.C.B.); Fax: +0049-(0)-89-4140-6823 (U.P.); +0049-(0)-89-4140-7734 (M.C.B.)
| | - Matthias Christoph Braunisch
- Department of Nephrology, School of Medicine, Technical University of Munich, Klinikum Rechts der Isar, 81675 Munich, Germany
- Correspondence: (U.P.); (M.C.B.); Tel.: +0049-(0)-89-4140-6863 (U.P.); +0049-(0)-89-4140-2231 (M.C.B.); Fax: +0049-(0)-89-4140-6823 (U.P.); +0049-(0)-89-4140-7734 (M.C.B.)
| |
Collapse
|
39
|
Karsten H, Cords L, Westphal T, Knapp M, Brehm TT, Hermanussen L, Omansen TF, Schmiedel S, Woost R, Ditt V, Peine S, Lütgehetmann M, Huber S, Ackermann C, Wittner M, Addo MM, Sette A, Sidney J, Schulze zur Wiesch J. High-resolution analysis of individual spike peptide-specific CD4 + T-cell responses in vaccine recipients and COVID-19 patients. Clin Transl Immunology 2022; 11:e1410. [PMID: 35957961 PMCID: PMC9363231 DOI: 10.1002/cti2.1410] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 07/06/2022] [Accepted: 07/20/2022] [Indexed: 12/03/2022] Open
Abstract
Objectives Potential differences in the breadth, distribution and magnitude of CD4+ T-cell responses directed against the SARS-CoV-2 spike glycoprotein between vaccinees, COVID-19 patients and subjects who experienced both ways of immunisation have not been comprehensively compared on a peptide level. Methods Following virus-specific in vitro cultivation, we determined the T-cell responses directed against 253 individual overlapping 15-mer peptides covering the entire SARS-CoV-2 spike glycoprotein using IFN-γ ELISpot and intracellular cytokine staining. In vitro HLA binding was determined for selected peptides. Results We mapped 955 single peptide-specific CD4+ T-cell responses in a cohort of COVID-19 patients (n = 8), uninfected vaccinees (n = 16) and individuals who experienced both infection and vaccination (n = 11). Patients and vaccinees (two-time and three-time vaccinees alike) had a comparable number of CD4+ T-cell responses (median 26 vs. 29, P = 0.7289). Most of these specificities were conserved in B.1.1.529 and the BA.4 and BA.5 sublineages. The highest magnitude of these in vitro IFN-γ CD4+ T-cell responses was observed in COVID-19 patients (median 0.35%), and three-time vaccinees showed a higher magnitude than two-time vaccinees (median 0.091% vs. 0.175%, P < 0.0001). Twelve peptide specificities were each detected in at least 40% of subjects. In vitro HLA binding showed promiscuous presentation by DRB1 molecules for several peptides. Conclusion Both SARS-CoV-2 infection and vaccination prime broadly directed T-cell responses directed against the SARS-CoV-2 spike glycoprotein. This comprehensive high-resolution analysis of spike peptide specificities will be a useful resource for further investigation of spike-specific T-cell responses.
Collapse
Affiliation(s)
- Hendrik Karsten
- Infectious Diseases Unit, 1. Department of MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Leon Cords
- Infectious Diseases Unit, 1. Department of MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Tim Westphal
- Infectious Diseases Unit, 1. Department of MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
- German Center for Infection Research (DZIF)Partner Site Hamburg‐Lübeck‐Borstel‐RiemsHamburgGermany
| | - Maximilian Knapp
- Infectious Diseases Unit, 1. Department of MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Thomas Theo Brehm
- Infectious Diseases Unit, 1. Department of MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
- German Center for Infection Research (DZIF)Partner Site Hamburg‐Lübeck‐Borstel‐RiemsHamburgGermany
| | - Lennart Hermanussen
- Infectious Diseases Unit, 1. Department of MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Till Frederik Omansen
- Infectious Diseases Unit, 1. Department of MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
- Department of Tropical MedicineBernhard Nocht Institute for Tropical MedicineHamburgGermany
| | - Stefan Schmiedel
- Infectious Diseases Unit, 1. Department of MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Robin Woost
- Infectious Diseases Unit, 1. Department of MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Vanessa Ditt
- Institute of Transfusion MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Sven Peine
- Institute of Transfusion MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Marc Lütgehetmann
- German Center for Infection Research (DZIF)Partner Site Hamburg‐Lübeck‐Borstel‐RiemsHamburgGermany
- Institute of Medical Microbiology, Virology and HygieneUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Samuel Huber
- Infectious Diseases Unit, 1. Department of MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Christin Ackermann
- Infectious Diseases Unit, 1. Department of MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Melanie Wittner
- Infectious Diseases Unit, 1. Department of MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
- German Center for Infection Research (DZIF)Partner Site Hamburg‐Lübeck‐Borstel‐RiemsHamburgGermany
| | - Marylyn Martina Addo
- Infectious Diseases Unit, 1. Department of MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
- German Center for Infection Research (DZIF)Partner Site Hamburg‐Lübeck‐Borstel‐RiemsHamburgGermany
- Department of Tropical MedicineBernhard Nocht Institute for Tropical MedicineHamburgGermany
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine ResearchLa Jolla Institute for Immunology (LJI)La JollaCAUSA
| | - John Sidney
- Center for Infectious Disease and Vaccine ResearchLa Jolla Institute for Immunology (LJI)La JollaCAUSA
| | - Julian Schulze zur Wiesch
- Infectious Diseases Unit, 1. Department of MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
- German Center for Infection Research (DZIF)Partner Site Hamburg‐Lübeck‐Borstel‐RiemsHamburgGermany
| |
Collapse
|
40
|
Garcia L, Woudenberg T, Rosado J, Dyer AH, Donnadieu F, Planas D, Bruel T, Schwartz O, Prazuck T, Velay A, Fafi-Kremer S, Batten I, Reddy C, Connolly E, McElheron M, Kennelly SP, Bourke NM, White MT, Pelleau S. Kinetics of the SARS-CoV-2 Antibody Avidity Response Following Infection and Vaccination. Viruses 2022; 14:1491. [PMID: 35891471 PMCID: PMC9321390 DOI: 10.3390/v14071491] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 06/29/2022] [Accepted: 07/06/2022] [Indexed: 12/04/2022] Open
Abstract
Serological assays capable of measuring antibody responses induced by previous infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) have been critical tools in the response to the COVID-19 pandemic. In this study, we use bead-based multiplex assays to measure IgG and IgA antibodies and IgG avidity to five SARS-CoV-2 antigens (Spike (S), receptor-binding domain (RBD), Nucleocapsid (N), S subunit 2, and Membrane-Envelope fusion (ME)). These assays were performed in several cohorts of healthcare workers and nursing home residents, who were followed for up to eleven months after SARS-CoV-2 infection or up to six months after vaccination. Our results show distinct kinetic patterns of antibody quantity (IgG and IgA) and avidity. While IgG and IgA antibody levels waned over time, with IgA antibody levels waning more rapidly, avidity increased with time after infection or vaccination. These contrasting kinetic patterns allow for the estimation of time since previous SARS-CoV-2 infection. Including avidity measurements in addition to antibody levels in a classification algorithm for estimating time since infection led to a substantial improvement in accuracy, from 62% to 78%. The inclusion of antibody avidity in panels of serological assays can yield valuable information for improving serosurveillance during SARS-CoV-2 epidemics.
Collapse
Affiliation(s)
- Laura Garcia
- Infectious Diseases Epidemiology and Analytics Unit, Department of Global Health, Institut Pasteur, Université Paris Cité, 75015 Paris, France; (L.G.); (T.W.); (J.R.); (F.D.)
| | - Tom Woudenberg
- Infectious Diseases Epidemiology and Analytics Unit, Department of Global Health, Institut Pasteur, Université Paris Cité, 75015 Paris, France; (L.G.); (T.W.); (J.R.); (F.D.)
| | - Jason Rosado
- Infectious Diseases Epidemiology and Analytics Unit, Department of Global Health, Institut Pasteur, Université Paris Cité, 75015 Paris, France; (L.G.); (T.W.); (J.R.); (F.D.)
| | - Adam H. Dyer
- Tallaght University Hospital, Tallaght, D24 NR0A Dublin, Ireland; (A.H.D.); (S.P.K.)
- Department of Medical Gerontology, School of Medicine, Trinity College Dublin, D02 PN40 Dublin, Ireland; (I.B.); (C.R.); (E.C.); (M.M.); (N.M.B.)
| | - Françoise Donnadieu
- Infectious Diseases Epidemiology and Analytics Unit, Department of Global Health, Institut Pasteur, Université Paris Cité, 75015 Paris, France; (L.G.); (T.W.); (J.R.); (F.D.)
| | - Delphine Planas
- Virus & Immunity Unit, Department of Virology, Institut Pasteur, Université Paris Cité, 75015 Paris, France; (D.P.); (T.B.); (O.S.)
| | - Timothée Bruel
- Virus & Immunity Unit, Department of Virology, Institut Pasteur, Université Paris Cité, 75015 Paris, France; (D.P.); (T.B.); (O.S.)
| | - Olivier Schwartz
- Virus & Immunity Unit, Department of Virology, Institut Pasteur, Université Paris Cité, 75015 Paris, France; (D.P.); (T.B.); (O.S.)
| | - Thierry Prazuck
- CHR d’Orléans, Service de Maladies Infectieuses, 45100 Orléans, France;
| | - Aurélie Velay
- CHU de Strasbourg, Laboratoire de Virologie, CEDEX, 67091 Strasbourg, France; (A.V.); (S.F.-K.)
- Unité Mixte de Recherche Scientifique Immuno-Rhumathologie Moléculaire (IRM UMR-S) 1109, Strasbourg University, Institut National de la Santé et de la Recherche Médicale (INSERM), CEDEX, 67084 Strasbourg, France
| | - Samira Fafi-Kremer
- CHU de Strasbourg, Laboratoire de Virologie, CEDEX, 67091 Strasbourg, France; (A.V.); (S.F.-K.)
- Unité Mixte de Recherche Scientifique Immuno-Rhumathologie Moléculaire (IRM UMR-S) 1109, Strasbourg University, Institut National de la Santé et de la Recherche Médicale (INSERM), CEDEX, 67084 Strasbourg, France
| | - Isabella Batten
- Department of Medical Gerontology, School of Medicine, Trinity College Dublin, D02 PN40 Dublin, Ireland; (I.B.); (C.R.); (E.C.); (M.M.); (N.M.B.)
| | - Conor Reddy
- Department of Medical Gerontology, School of Medicine, Trinity College Dublin, D02 PN40 Dublin, Ireland; (I.B.); (C.R.); (E.C.); (M.M.); (N.M.B.)
| | - Emma Connolly
- Department of Medical Gerontology, School of Medicine, Trinity College Dublin, D02 PN40 Dublin, Ireland; (I.B.); (C.R.); (E.C.); (M.M.); (N.M.B.)
| | - Matt McElheron
- Department of Medical Gerontology, School of Medicine, Trinity College Dublin, D02 PN40 Dublin, Ireland; (I.B.); (C.R.); (E.C.); (M.M.); (N.M.B.)
| | - Sean P. Kennelly
- Tallaght University Hospital, Tallaght, D24 NR0A Dublin, Ireland; (A.H.D.); (S.P.K.)
- Department of Medical Gerontology, School of Medicine, Trinity College Dublin, D02 PN40 Dublin, Ireland; (I.B.); (C.R.); (E.C.); (M.M.); (N.M.B.)
| | - Nollaig M. Bourke
- Department of Medical Gerontology, School of Medicine, Trinity College Dublin, D02 PN40 Dublin, Ireland; (I.B.); (C.R.); (E.C.); (M.M.); (N.M.B.)
| | - Michael T. White
- Infectious Diseases Epidemiology and Analytics Unit, Department of Global Health, Institut Pasteur, Université Paris Cité, 75015 Paris, France; (L.G.); (T.W.); (J.R.); (F.D.)
| | - Stéphane Pelleau
- Infectious Diseases Epidemiology and Analytics Unit, Department of Global Health, Institut Pasteur, Université Paris Cité, 75015 Paris, France; (L.G.); (T.W.); (J.R.); (F.D.)
| |
Collapse
|
41
|
Shen AR, Jin XX, Tang TT, Ding Y, Liu XT, Zhong X, Wu YD, Han XL, Zhao GY, Shen CL, Lv LL, Liu BC. Exosomal Vaccine Loading T Cell Epitope Peptides of SARS-CoV-2 Induces Robust CD8+ T Cell Response in HLA-A Transgenic Mice. Int J Nanomedicine 2022; 17:3325-3341. [PMID: 35937077 PMCID: PMC9346304 DOI: 10.2147/ijn.s367494] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 07/16/2022] [Indexed: 11/24/2022] Open
Abstract
Purpose Current vaccines for the SARS-CoV-2 virus mainly induce neutralizing antibodies but overlook the T cell responses. This study aims to generate an exosomal vaccine carrying T cell epitope peptides of SARS-CoV-2 for the induction of CD8+ T cell response. Methods Thirty-one peptides presented by HLA-A0201 molecule were conjugated to the DMPE-PEG-NHS molecules, and mixed with DSPE-PEG to form the peptide-PEG-lipid micelles, then fused with exosomes to generate the exosomal vaccine, followed by purification using size-exclusion chromatography and validation by Western blotting, liquid nuclear magnetic resonance (NMR) test and transmission electron microscopy. Furthermore, the exosomal vaccine was mixed with Poly (I:C) adjuvant and subcutaneously administered for three times into the hybrid mice of HLA-A0201/DR1 transgenic mice with wild-type mice. Then, the epitope-specific T cell responses were detected by ex vivo ELISPOT assay and intracellular cytokine staining. Results The exosomal vaccine was purified from the Peak 2 fraction of FPLC and injected into the hybrid mice for three times. The IFN-γ spot forming units and the frequencies of IFN-γ+/CD8+ T cells were 10–82-fold and 13–65-fold, respectively, higher in the exosomal vaccine group compared to the Poly (I:C) control group, without visible organ toxicity. In comparison with the peptides cocktail vaccine generated in our recent work, the exosomal vaccine induced significantly stronger T cell response. Conclusion Exosomal vaccine loading T cell epitope peptides of SARS-CoV-2 virus was initially generated without pre-modification for both peptides and exosomes, and elicited robust CD8+ T cell response in HLA-A transgenic mice.
Collapse
Affiliation(s)
- An-Ran Shen
- Institute of Nephrology, Zhongda Hospital, Medical School of Southeast University, Nanjing, 210009, People’s Republic of China
| | - Xiao-Xiao Jin
- Department of Microbiology and Immunology, Medical School of Southeast University, Nanjing, 210009, People’s Republic of China
| | - Tao-Tao Tang
- Institute of Nephrology, Zhongda Hospital, Medical School of Southeast University, Nanjing, 210009, People’s Republic of China
| | - Yan Ding
- Department of Microbiology and Immunology, Medical School of Southeast University, Nanjing, 210009, People’s Republic of China
| | - Xiao-Tao Liu
- Department of Microbiology and Immunology, Medical School of Southeast University, Nanjing, 210009, People’s Republic of China
| | - Xin Zhong
- Institute of Nephrology, Zhongda Hospital, Medical School of Southeast University, Nanjing, 210009, People’s Republic of China
| | - Yan-Dan Wu
- Department of Microbiology and Immunology, Medical School of Southeast University, Nanjing, 210009, People’s Republic of China
| | - Xue-Lian Han
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, 100071, People’s Republic of China
| | - Guang-Yu Zhao
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, 100071, People’s Republic of China
| | - Chuan-Lai Shen
- Department of Microbiology and Immunology, Medical School of Southeast University, Nanjing, 210009, People’s Republic of China
| | - Lin-Li Lv
- Institute of Nephrology, Zhongda Hospital, Medical School of Southeast University, Nanjing, 210009, People’s Republic of China
- Correspondence: Lin-Li Lv; Bi-Cheng Liu, Institute of Nephrology, Zhongda Hospital, Medical School of Southeast University, Nanjing, 210009, People’s Republic of China, Tel +862583272512, Email ;
| | - Bi-Cheng Liu
- Institute of Nephrology, Zhongda Hospital, Medical School of Southeast University, Nanjing, 210009, People’s Republic of China
| |
Collapse
|
42
|
Paschold L, Klee B, Gottschick C, Willscher E, Diexer S, Schultheiß C, Simnica D, Sedding D, Girndt M, Gekle M, Mikolajczyk R, Binder M. Rapid Hypermutation B Cell Trajectory Recruits Previously Primed B Cells Upon Third SARS-Cov-2 mRNA Vaccination. Front Immunol 2022; 13:876306. [PMID: 35615365 PMCID: PMC9126551 DOI: 10.3389/fimmu.2022.876306] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 04/14/2022] [Indexed: 12/14/2022] Open
Abstract
The COVID-19 pandemic shows that vaccination strategies building on an ancestral viral strain need to be optimized for the control of potentially emerging viral variants. Therefore, aiming at strong B cell somatic hypermutation to increase antibody affinity to the ancestral strain - not only at high antibody titers - is a priority when utilizing vaccines that are not targeted at individual variants since high affinity may offer some flexibility to compensate for strain-individual mutations. Here, we developed a next-generation sequencing based SARS-CoV-2 B cell tracking protocol to rapidly determine the level of immunoglobulin somatic hypermutation at distinct points during the immunization period. The percentage of somatically hypermutated B cells in the SARS-CoV-2 specific repertoire was low after the primary vaccination series, evolved further over months and increased steeply after boosting. The third vaccination mobilized not only naïve, but also antigen-experienced B cell clones into further rapid somatic hypermutation trajectories indicating increased affinity. Together, the strongly mutated post-booster repertoires and antibodies deriving from this may explain why the third, but not the primary vaccination series, offers some protection against immune-escape variants such as Omicron B.1.1.529.
Collapse
Affiliation(s)
- Lisa Paschold
- Department of Internal Medicine IV, Oncology/Hematology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Bianca Klee
- Institute for Medical Epidemiology, Biometrics and Informatics (IMEBI), Interdisciplinary Center for Health Sciences, Medical School of the Martin-Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Cornelia Gottschick
- Institute for Medical Epidemiology, Biometrics and Informatics (IMEBI), Interdisciplinary Center for Health Sciences, Medical School of the Martin-Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Edith Willscher
- Department of Internal Medicine IV, Oncology/Hematology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Sophie Diexer
- Institute for Medical Epidemiology, Biometrics and Informatics (IMEBI), Interdisciplinary Center for Health Sciences, Medical School of the Martin-Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Christoph Schultheiß
- Department of Internal Medicine IV, Oncology/Hematology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Donjete Simnica
- Department of Internal Medicine IV, Oncology/Hematology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Daniel Sedding
- Mid-German Heart Center, Department of Cardiology and Intensive Care Medicine, University Hospital, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Matthias Girndt
- Department of Internal Medicine II, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Michael Gekle
- Julius Bernstein-Institute of Physiology, Faculty of Medicine, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Rafael Mikolajczyk
- Institute for Medical Epidemiology, Biometrics and Informatics (IMEBI), Interdisciplinary Center for Health Sciences, Medical School of the Martin-Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Mascha Binder
- Department of Internal Medicine IV, Oncology/Hematology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| |
Collapse
|
43
|
Mortality in Severe Antibody Deficiencies Patients during the First Two Years of the COVID-19 Pandemic: Vaccination and Monoclonal Antibodies Efficacy. Biomedicines 2022; 10:biomedicines10051026. [PMID: 35625763 PMCID: PMC9138935 DOI: 10.3390/biomedicines10051026] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 04/20/2022] [Accepted: 04/28/2022] [Indexed: 02/04/2023] Open
Abstract
Patients with severely impaired antibody responses represent a group at-risk in the SARS-CoV-2 pandemic due to the lack of Spike-specific neutralizing antibodies. The main objective of this paper was to assess, by a longitudinal prospective study, COVID-19 infection and mortality rates, and disease severity in the first two years of the pandemic in a cohort of 471 Primary Antibody Defects adult patients. As secondary endpoints, we compared SARS-CoV-2 annual mortality rate to that observed over a 10-year follow-up in the same cohort, and we assessed the impact of interventions done in the second year, vaccination and anti-SARS-CoV-2 monoclonal antibodies administration on the disease outcome. Forty-one and 84 patients were infected during the first and the second year, respectively. Despite a higher infection and reinfection rate, and a higher COVID-19-related mortality rate compared to the Italian population, the pandemic did not modify the annual mortality rate for any cause in our cohort compared to that registered over the last ten years in the same cohort. PADs patients who died from COVID-19 had an underlying end-stage lung disease. We showed a beneficial effect of MoAbs administration on the likelihood of hospitalization and development of severe disease. In conclusion, COVID-19 did not cause excess mortality in Severe Antibody Deficiencies.
Collapse
|
44
|
Zhang B, Tian J, Zhang Q, Xie Y, Wang K, Qiu S, Lu K, Liu Y. Comparing the Nucleocapsid Proteins of Human Coronaviruses: Structure, Immunoregulation, Vaccine, and Targeted Drug. Front Mol Biosci 2022; 9:761173. [PMID: 35573742 PMCID: PMC9099148 DOI: 10.3389/fmolb.2022.761173] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 03/28/2022] [Indexed: 01/08/2023] Open
Abstract
The seven pathogenic human coronaviruses (HCoVs) include HCoV-229E, HCoV-OC43, HCoV-NL63, and HCoV-HKU1, which usually cause mild upper respiratory tract diseases, and SARS-CoV, MERS-CoV, and SARS-CoV-2, which cause a severe acute respiratory syndrome. The nucleocapsid (N) protein, as the dominant structural protein from coronaviruses that bind to the genomic RNA, participates in various vital activities after virus invasion and will probably become a promising target of antiviral drug design. Therefore, a comprehensive literature review of human coronavirus’ pathogenic mechanism and therapeutic strategies is necessary for the control of the pandemic. Here, we give a systematic summary of the structures, immunoregulation, and potential vaccines and targeted drugs of the HCoVs N protein. First, we provide a general introduction to the fundamental structures and molecular function of N protein. Next, we outline the N protein mediated immune regulation and pathogenesis mechanism. Finally, we comprehensively summarize the development of potential N protein-targeted drugs and candidate vaccines to treat coronavirus disease 2019 (COVID-19). We believe this review provides insight into the virulence and transmission of SARS-CoV-2 as well as support for further study on epidemic control of COVID-19.
Collapse
Affiliation(s)
- Bo Zhang
- College of Basic Medicine, Zunyi Medical University, Zunyi, China
- *Correspondence: Yang Liu, ; Keyu Lu, ; Bo Zhang,
| | - Junjie Tian
- College of Basic Medicine, Zunyi Medical University, Zunyi, China
| | - Qintao Zhang
- College of Basic Medicine, Zunyi Medical University, Zunyi, China
| | - Yan Xie
- School of Public Health, Zunyi Medical University, Zunyi, China
| | - Kejia Wang
- Key Laboratory of Plant Resource Conservation and Germplasm Innovation in Mountainous Region (Ministry of Education), College of Life Sciences/Institute of Agro-bioengineering, Guizhou University, Guiyang, China
| | - Shuyi Qiu
- Key Laboratory of Plant Resource Conservation and Germplasm Innovation in Mountainous Region (Ministry of Education), College of Life Sciences/Institute of Agro-bioengineering, Guizhou University, Guiyang, China
| | - Keyu Lu
- College of Basic Medicine, Zunyi Medical University, Zunyi, China
- *Correspondence: Yang Liu, ; Keyu Lu, ; Bo Zhang,
| | - Yang Liu
- School of Public Health, Zunyi Medical University, Zunyi, China
- Key Laboratory of Plant Resource Conservation and Germplasm Innovation in Mountainous Region (Ministry of Education), College of Life Sciences/Institute of Agro-bioengineering, Guizhou University, Guiyang, China
- *Correspondence: Yang Liu, ; Keyu Lu, ; Bo Zhang,
| |
Collapse
|
45
|
Smirnov VS, Lyalina LV, Milichkina AM, Khamitova IV, Zueva EV, Ivanov VA, Zaguzov VS, Totolian AA. Longitudinal Randomized Cohort Study of SARS-CoV-2 Antibody Seroprevalence in the St. Petersburg Population. Viruses 2022; 14:913. [PMID: 35632653 PMCID: PMC9146723 DOI: 10.3390/v14050913] [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] [Received: 03/09/2022] [Revised: 04/17/2022] [Accepted: 04/25/2022] [Indexed: 02/01/2023] Open
Abstract
Introduction. Since the detection of the first COVID-19 patient, 2 years have passed, during which more than 287,862,000 people have fallen ill globally, of which about 1.9% died. The implementation of SARS-CoV-2 control programs required efforts from almost all countries. An important direction in the fight against COVID-19 has been the formation of herd immunity, the main tool for managing the pandemic. Study goal. The aim of the study was to assess the seroprevalence of antibodies (Abs) to SARS-CoV-2 nucleocapsid (Nc) and receptor binding domain (RBD) in the St. Petersburg population during the COVID-19 pandemic. Materials and methods. A longitudinal cohort randomized monitoring study of Ab seroprevalence (SARS-CoV-2 Nc, RBD) was organized and conducted according to a unified methodology developed by Rospotrebnadzor with the participation of the St. Petersburg Pasteur Institute. For this purpose, a cohort was formed of 1000 volunteers who participated in all five stages of seromonitoring. The cohort was divided into seven age groups: 1−17; 18−29; 30−39; 40−49; 50−59; 60−69; 70; and older (70+) years. Seropositivity levels (Nc, RBD) were assessed by quantitative and qualitative enzyme immunoassays. During the second year of monitoring, some volunteers were vaccinated with the GamCOVIDVac (84%) or EpiVacCorona (11.6%) vaccines approved in Russia. Statistical processing was carried out using Excel 2010. Confidence intervals for shares and percentages (95% CI) were calculated using the method of A. Wald and J. Wolfowitz with adjustment (A. Agresti, B.A. Coull). The statistical significance of differences was calculated by z-test, using the appropriate online calculator (p < 0.05) unless indicated. Results. There was a trend toward an increase in Nc seropositivity in stages 1−3 of seromonitoring, with a decrease in stages 4−5 among children and adults. The share of RBD seropositive steadily increased during all five stages of seromonitoring. The most frequent finding was low anti-RBD Abs levels (22.6−220 BAU/mL). High Ab levels were recorded statistically significantly less frequently. Asymptomatic forms were observed in 84−88% of SARS-CoV-2 seropositive volunteers. By the fifth stage of monitoring, this indicator significantly decreased to 69.8% (95% CI: 66.1−73.4). The monitoring revealed a statistically significant increase in anti-RBD Abs alongside a statistically significant decrease in the proportion of Nc seropositives. This dynamic was especially characteristic of persons vaccinated with GamCOVIDVac. Conclusion. Prior to the use of specific vaccines, a seroprevalence of anti-Nc Abs was noted. After the introduction of the GamCOVIDVac vaccine in adults, a decrease in the level of anti-Nc Abs was noted due to an increase in the proportion of RBD seropositive persons.
Collapse
Affiliation(s)
- Vyacheslav S. Smirnov
- Saint Petersburg Pasteur Institute, 197101 St. Petersburg, Russia; (L.V.L.); (A.M.M.); (I.V.K.); (E.V.Z.); (V.A.I.); (V.S.Z.); (A.A.T.)
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Fuentes-Villalobos F, Garrido JL, Medina MA, Zambrano N, Ross N, Bravo F, Gaete-Argel A, Oyarzún-Arrau A, Amanat F, Soto-Rifo R, Valiente-Echeverría F, Ocampo R, Esveile C, Ferreira L, Cabrera J, Torres V, Rioseco ML, Riquelme R, Barría S, Alvarez R, Pinos Y, Krammer F, Calvo M, Barria MI. Sustained Antibody-Dependent NK Cell Functions in Mild COVID-19 Outpatients During Convalescence. Front Immunol 2022; 13:796481. [PMID: 35197972 PMCID: PMC8859986 DOI: 10.3389/fimmu.2022.796481] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 01/14/2022] [Indexed: 01/10/2023] Open
Abstract
The coronavirus disease 2019 (COVID19) pandemic has left researchers scrambling to identify the humoral immune correlates of protection from COVID-19. To date, the antibody mediated correlates of virus neutralization have been extensively studied. However, the extent that non-neutralizing functions contribute to anti-viral responses are ill defined. In this study, we profiled the anti-spike antibody subtype/subclass responses, along with neutralization and antibody-dependent natural killer cell functions in 83 blood samples collected between 4 and 201 days post-symptoms onset from a cohort of COVID-19 outpatients. We observed heterogeneous humoral responses against the acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike protein. Overall, anti-spike profiles were characterized by a rapid rise of IgA and sustained IgG titers. In addition, strong antibody-mediated natural killer effector responses correlated with milder disease and being female. While higher neutralization profiles were observed in males along with increased severity. These results give an insight into the underlying function of antibodies beyond neutralization and suggest that antibody-mediated natural killer cell activity is a key function of the humoral response against the SARS-CoV-2 spike protein.
Collapse
Affiliation(s)
| | - Jose L Garrido
- Ichor Biologics LLC, New York, NY, United States.,Facultad de Medicina y Ciencia, Universidad San Sebastián, Puerto Montt, Chile
| | - Matías A Medina
- Department of Microbiology, Faculty of Biological Science, Universidad de Concepción, Concepción, Chile
| | - Nicole Zambrano
- Department of Microbiology, Faculty of Biological Science, Universidad de Concepción, Concepción, Chile
| | - Natalia Ross
- Department of Microbiology, Faculty of Biological Science, Universidad de Concepción, Concepción, Chile
| | - Felipe Bravo
- Department of Microbiology, Faculty of Biological Science, Universidad de Concepción, Concepción, Chile
| | - Aracelly Gaete-Argel
- Laboratory of Molecular and Cellular Virology, Virology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Aarón Oyarzún-Arrau
- Laboratory of Molecular and Cellular Virology, Virology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Fatima Amanat
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Ricardo Soto-Rifo
- Laboratory of Molecular and Cellular Virology, Virology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Fernando Valiente-Echeverría
- Laboratory of Molecular and Cellular Virology, Virology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | | | | | - Leonila Ferreira
- Hospital Clínico Regional Dr. Guillermo Grant Benavente, Concepción, Chile
| | | | - Vivianne Torres
- Institute of Medicine, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile
| | - Maria L Rioseco
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Puerto Montt, Chile.,Hospital Puerto Montt Dr. Eduardo Schütz Schroeder, Puerto Montt, Chile
| | - Raúl Riquelme
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Puerto Montt, Chile.,Hospital Puerto Montt Dr. Eduardo Schütz Schroeder, Puerto Montt, Chile
| | - Sebastián Barría
- Hospital Puerto Montt Dr. Eduardo Schütz Schroeder, Puerto Montt, Chile
| | - Raymond Alvarez
- Ichor Biologics LLC, New York, NY, United States.,Division of Infectious Diseases, Department of Medicine, Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | | | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Mario Calvo
- Institute of Medicine, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile
| | - Maria I Barria
- Department of Microbiology, Faculty of Biological Science, Universidad de Concepción, Concepción, Chile.,Facultad de Medicina y Ciencia, Universidad San Sebastián, Puerto Montt, Chile
| | | |
Collapse
|
47
|
Wratil PR, Stern M, Priller A, Willmann A, Almanzar G, Vogel E, Feuerherd M, Cheng CC, Yazici S, Christa C, Jeske S, Lupoli G, Vogt T, Albanese M, Mejías-Pérez E, Bauernfried S, Graf N, Mijocevic H, Vu M, Tinnefeld K, Wettengel J, Hoffmann D, Muenchhoff M, Daechert C, Mairhofer H, Krebs S, Fingerle V, Graf A, Steininger P, Blum H, Hornung V, Liebl B, Überla K, Prelog M, Knolle P, Keppler OT, Protzer U. Three exposures to the spike protein of SARS-CoV-2 by either infection or vaccination elicit superior neutralizing immunity to all variants of concern. Nat Med 2022; 28:496-503. [PMID: 35090165 DOI: 10.1038/s41591-022-01715-4] [Citation(s) in RCA: 169] [Impact Index Per Article: 56.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 01/25/2022] [Indexed: 11/09/2022]
Abstract
Infection-neutralizing antibody responses after SARS-CoV-2 infection or COVID-19 vaccination are an essential component of antiviral immunity. Antibody-mediated protection is challenged by the emergence of SARS-CoV-2 variants of concern (VoCs) with immune escape properties, such as omicron (B.1.1.529) that is rapidly spreading worldwide. Here, we report neutralizing antibody dynamics in a longitudinal cohort of COVID-19 convalescent and infection-naive individuals vaccinated with mRNA BNT162b2 by quantifying anti-SARS-CoV-2-spike antibodies and determining their avidity and neutralization capacity in serum. Using live-virus neutralization assays, we show that a superior infection-neutralizing capacity against all VoCs, including omicron, developed after either two vaccinations in convalescents or after a third vaccination or breakthrough infection of twice-vaccinated, naive individuals. These three consecutive spike antigen exposures resulted in an increasing neutralization capacity per anti-spike antibody unit and were paralleled by stepwise increases in antibody avidity. We conclude that an infection-plus-vaccination-induced hybrid immunity or a triple immunization can induce high-quality antibodies with superior neutralization capacity against VoCs, including omicron.
Collapse
Affiliation(s)
- Paul R Wratil
- Max von Pettenkofer Institute and Gene Center, Virology, National Reference Center for Retroviruses, Faculty of Medicine, LMU München, Munich, Germany.,German Centre for Infection Research (DZIF), Partner Site, Munich, Germany
| | - Marcel Stern
- Max von Pettenkofer Institute and Gene Center, Virology, National Reference Center for Retroviruses, Faculty of Medicine, LMU München, Munich, Germany
| | - Alina Priller
- Institute of Molecular Immunology and Experimental Oncology, University Hospital rechts der Isar, Technical University of Munich (TUM), School of Medicine, Munich, Germany
| | - Annika Willmann
- Institute of Virology, Helmholtz Center Munich, Technical University of Munich (TUM), School of Medicine, Munich, Germany
| | - Giovanni Almanzar
- Pediatric Rheumatology / Special Immunology, Pediatrics Department, University Hospital Würzburg, Würzburg, Germany
| | - Emanuel Vogel
- Institute of Virology, Helmholtz Center Munich, Technical University of Munich (TUM), School of Medicine, Munich, Germany
| | - Martin Feuerherd
- Institute of Virology, Helmholtz Center Munich, Technical University of Munich (TUM), School of Medicine, Munich, Germany
| | - Cho-Chin Cheng
- Institute of Virology, Helmholtz Center Munich, Technical University of Munich (TUM), School of Medicine, Munich, Germany
| | - Sarah Yazici
- Institute of Molecular Immunology and Experimental Oncology, University Hospital rechts der Isar, Technical University of Munich (TUM), School of Medicine, Munich, Germany
| | - Catharina Christa
- Institute of Virology, Helmholtz Center Munich, Technical University of Munich (TUM), School of Medicine, Munich, Germany
| | - Samuel Jeske
- Institute of Virology, Helmholtz Center Munich, Technical University of Munich (TUM), School of Medicine, Munich, Germany
| | - Gaia Lupoli
- Max von Pettenkofer Institute and Gene Center, Virology, National Reference Center for Retroviruses, Faculty of Medicine, LMU München, Munich, Germany
| | - Tim Vogt
- Pediatric Rheumatology / Special Immunology, Pediatrics Department, University Hospital Würzburg, Würzburg, Germany
| | - Manuel Albanese
- Max von Pettenkofer Institute and Gene Center, Virology, National Reference Center for Retroviruses, Faculty of Medicine, LMU München, Munich, Germany.,National Institute of Molecular Genetics (INGM), Milano, Italy
| | - Ernesto Mejías-Pérez
- Max von Pettenkofer Institute and Gene Center, Virology, National Reference Center for Retroviruses, Faculty of Medicine, LMU München, Munich, Germany
| | - Stefan Bauernfried
- Gene Center and Department of Biochemistry, LMU München, Munich, Germany
| | - Natalia Graf
- Institute of Virology, Helmholtz Center Munich, Technical University of Munich (TUM), School of Medicine, Munich, Germany
| | - Hrvoje Mijocevic
- Institute of Virology, Helmholtz Center Munich, Technical University of Munich (TUM), School of Medicine, Munich, Germany
| | - Martin Vu
- Institute of Virology, Helmholtz Center Munich, Technical University of Munich (TUM), School of Medicine, Munich, Germany
| | - Kathrin Tinnefeld
- Institute of Virology, Helmholtz Center Munich, Technical University of Munich (TUM), School of Medicine, Munich, Germany
| | - Jochen Wettengel
- German Centre for Infection Research (DZIF), Partner Site, Munich, Germany.,Institute of Virology, Helmholtz Center Munich, Technical University of Munich (TUM), School of Medicine, Munich, Germany
| | - Dieter Hoffmann
- German Centre for Infection Research (DZIF), Partner Site, Munich, Germany.,Institute of Virology, Helmholtz Center Munich, Technical University of Munich (TUM), School of Medicine, Munich, Germany
| | - Maximilian Muenchhoff
- Max von Pettenkofer Institute and Gene Center, Virology, National Reference Center for Retroviruses, Faculty of Medicine, LMU München, Munich, Germany.,German Centre for Infection Research (DZIF), Partner Site, Munich, Germany
| | - Christopher Daechert
- Max von Pettenkofer Institute and Gene Center, Virology, National Reference Center for Retroviruses, Faculty of Medicine, LMU München, Munich, Germany
| | - Helga Mairhofer
- Max von Pettenkofer Institute and Gene Center, Virology, National Reference Center for Retroviruses, Faculty of Medicine, LMU München, Munich, Germany
| | - Stefan Krebs
- Laboratory for Functional Genome Analysis, Gene Center, LMU München, Munich, Germany
| | - Volker Fingerle
- Bavarian Health and Food Safety Authority (LGL (LGL), Oberschleißheim, Germany
| | - Alexander Graf
- Laboratory for Functional Genome Analysis, Gene Center, LMU München, Munich, Germany
| | - Philipp Steininger
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Helmut Blum
- Laboratory for Functional Genome Analysis, Gene Center, LMU München, Munich, Germany
| | - Veit Hornung
- Gene Center and Department of Biochemistry, LMU München, Munich, Germany
| | - Bernhard Liebl
- Bavarian Health and Food Safety Authority (LGL (LGL), Oberschleißheim, Germany
| | - Klaus Überla
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Martina Prelog
- Pediatric Rheumatology / Special Immunology, Pediatrics Department, University Hospital Würzburg, Würzburg, Germany
| | - Percy Knolle
- Institute of Molecular Immunology and Experimental Oncology, University Hospital rechts der Isar, Technical University of Munich (TUM), School of Medicine, Munich, Germany
| | - Oliver T Keppler
- Max von Pettenkofer Institute and Gene Center, Virology, National Reference Center for Retroviruses, Faculty of Medicine, LMU München, Munich, Germany. .,German Centre for Infection Research (DZIF), Partner Site, Munich, Germany.
| | - Ulrike Protzer
- German Centre for Infection Research (DZIF), Partner Site, Munich, Germany. .,Institute of Virology, Helmholtz Center Munich, Technical University of Munich (TUM), School of Medicine, Munich, Germany.
| |
Collapse
|