151
|
Sievers BL, Gelbart T, Tan GS. A high-throughput SARS-CoV-2 pseudovirus multiplex neutralization assay. STAR Protoc 2022; 3:101835. [PMID: 36595901 PMCID: PMC9579189 DOI: 10.1016/j.xpro.2022.101835] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 09/13/2022] [Accepted: 10/17/2022] [Indexed: 11/05/2022] Open
Abstract
Evaluating the neutralizing antibody titer following SARS-CoV-2 vaccination is essential in defining correlates of protection. We describe an assay that uses single-cycle vesicular stomatitis virus (VSV) pseudoviruses linking a fluorophore with a spike (S) from a variant of concern (VOC). Using two fluorophores linked to two VOC S, respectively, allows us to determine the neutralization titer against two VOCs in a single run. This is a generalizable approach that saves time, samples, and run-to-run variability. For complete details on the use and execution of this protocol, please refer to Sievers et al. (2022).1.
Collapse
Affiliation(s)
| | | | - Gene S. Tan
- J. Craig Venter Institute, La Jolla, CA 92037, USA,Division of Infectious Diseases, Department of Medicine, University of California San Diego, La Jolla, CA 92037, USA,Corresponding author
| |
Collapse
|
152
|
Dobaño C, Ramírez-Morros A, Alonso S, Rubio R, Ruiz-Olalla G, Vidal-Alaball J, Macià D, Catalina QM, Vidal M, Casanovas AF, Prados de la Torre E, Barrios D, Jiménez A, Zanoncello J, Melero NR, Carolis C, Izquierdo L, Aguilar R, Moncunill G, Ruiz-Comellas A. Sustained seropositivity up to 20.5 months after COVID-19. BMC Med 2022; 20:379. [PMID: 36224590 PMCID: PMC9556137 DOI: 10.1186/s12916-022-02570-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 09/15/2022] [Indexed: 01/09/2023] Open
Abstract
This study evaluated the persistence of IgM, IgA, and IgG to SARS-CoV-2 spike and nucleocapsid antigens up to 616 days since the onset of symptoms in a longitudinal cohort of 247 primary health care workers from Barcelona, Spain, followed up since the start of the pandemic. The study also assesses factors affecting antibody levels, including comorbidities and the responses to variants of concern as well as the frequency of reinfections. Despite a gradual and significant decline in antibody levels with time, seropositivity to five SARS-CoV-2 antigens combined was always higher than 90% over the whole study period. In a subset of 23 participants who had not yet been vaccinated by November 2021, seropositivity remained at 95.65% (47.83% IgM, 95.65% IgA, 95.65% IgG). IgG seropositivity against Alpha and Delta predominant variants was comparable to that against the Wuhan variant, while it was lower for Gamma and Beta (minority) variants and for IgA and IgM. Antibody levels at the time point closest to infection were associated with age, smoking, obesity, hospitalization, fever, anosmia/hypogeusia, chest pain, and hypertension in multivariable regression models. Up to 1 year later, just before the massive roll out of vaccination, antibody levels were associated with age, occupation, hospitalization, duration of symptoms, anosmia/hypogeusia, fever, and headache. In addition, tachycardia and cutaneous symptoms associated with slower antibody decay, and oxygen supply with faster antibody decay. Eight reinfections (3.23%) were detected in low responders, which is consistent with a sustained protective role for anti-spike naturally acquired antibodies. Stable persistence of IgG and IgA responses and cross-recognition of the predominant variants circulating in the 2020-2021 period indicate long-lasting and largely variant-transcending humoral immunity in the initial 20.5 months of the pandemic, in the absence of vaccination.
Collapse
Affiliation(s)
- Carlota Dobaño
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Carrer Roselló 153 (CEK building), E-08036, Barcelona, Spain.
- CIBER de Enfermedades Infecciosas, Barcelona, Spain.
| | - Anna Ramírez-Morros
- Unitat de Suport a la Recerca de la Catalunya Central, Fundació Institut Universitari per a la recerca a l'Atenció Primària de Salut Jordi Gol i Gurina, Sant Fruitós de Bages, Spain
| | - Selena Alonso
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Carrer Roselló 153 (CEK building), E-08036, Barcelona, Spain
| | - Rocío Rubio
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Carrer Roselló 153 (CEK building), E-08036, Barcelona, Spain
| | - Gemma Ruiz-Olalla
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Carrer Roselló 153 (CEK building), E-08036, Barcelona, Spain
| | - Josep Vidal-Alaball
- Unitat de Suport a la Recerca de la Catalunya Central, Fundació Institut Universitari per a la recerca a l'Atenció Primària de Salut Jordi Gol i Gurina, Sant Fruitós de Bages, Spain
- Grup de Promoció de la Salut en l'Àmbit Rural (ProSaARu), Institut Català de la Salut, Sant Fruitós de Bages, Spain
- Facultat de Medicina, Universitat de Vic-Universitat Central de Catalunya (UVIC-UCC), Vic, Spain
| | - Dídac Macià
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Carrer Roselló 153 (CEK building), E-08036, Barcelona, Spain
- CIBER de Enfermedades Infecciosas, Barcelona, Spain
| | - Queralt Miró Catalina
- Unitat de Suport a la Recerca de la Catalunya Central, Fundació Institut Universitari per a la recerca a l'Atenció Primària de Salut Jordi Gol i Gurina, Sant Fruitós de Bages, Spain
| | - Marta Vidal
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Carrer Roselló 153 (CEK building), E-08036, Barcelona, Spain
| | - Aina Fuster Casanovas
- Unitat de Suport a la Recerca de la Catalunya Central, Fundació Institut Universitari per a la recerca a l'Atenció Primària de Salut Jordi Gol i Gurina, Sant Fruitós de Bages, Spain
| | - Esther Prados de la Torre
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Carrer Roselló 153 (CEK building), E-08036, Barcelona, Spain
| | - Diana Barrios
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Carrer Roselló 153 (CEK building), E-08036, Barcelona, Spain
| | - Alfons Jiménez
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Carrer Roselló 153 (CEK building), E-08036, Barcelona, Spain
| | - Jasmina Zanoncello
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Carrer Roselló 153 (CEK building), E-08036, Barcelona, Spain
| | - Natalia Rodrigo Melero
- Biomolecular Screening and Protein Technologies Unit, Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Carlo Carolis
- Biomolecular Screening and Protein Technologies Unit, Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Luis Izquierdo
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Carrer Roselló 153 (CEK building), E-08036, Barcelona, Spain
- CIBER de Enfermedades Infecciosas, Barcelona, Spain
| | - Ruth Aguilar
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Carrer Roselló 153 (CEK building), E-08036, Barcelona, Spain
| | - Gemma Moncunill
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Carrer Roselló 153 (CEK building), E-08036, Barcelona, Spain
- CIBER de Enfermedades Infecciosas, Barcelona, Spain
| | - Anna Ruiz-Comellas
- Unitat de Suport a la Recerca de la Catalunya Central, Fundació Institut Universitari per a la recerca a l'Atenció Primària de Salut Jordi Gol i Gurina, Sant Fruitós de Bages, Spain
- Grup de Promoció de la Salut en l'Àmbit Rural (ProSaARu), Institut Català de la Salut, Sant Fruitós de Bages, Spain
- Facultat de Medicina, Universitat de Vic-Universitat Central de Catalunya (UVIC-UCC), Vic, Spain
- Centre d'Atenció Primària (CAP) Sant Joan de Vilatorrada. Gerència Territorial de la Catalunya Central, Institut Català de la Salut, Sant Fruitós de Bages, Spain
| |
Collapse
|
153
|
Kim J, Chang E, Park SY, Lee DW, Kang CK, Choe PG, Kim NJ, Oh MD, Park WB, Lee KH, Im SA. Evaluation of Seropositivity After Standard Doses of Vaccination Against SARS-CoV-2 in Patients With Early Breast Cancer Receiving Adjuvant Treatment. Oncologist 2022; 27:e931-e937. [PMID: 36218350 PMCID: PMC9732225 DOI: 10.1093/oncolo/oyac196] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 08/09/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19) pandemic affected millions of individuals, and patients with cancer are known to be more susceptible. Vaccines against SARS-CoV-2 have been developed and used for patients with cancer, but scarce data are available on their efficacy in patients under active anti-cancer therapies. MATERIALS AND METHODS In this study, we semi-quantitatively measured the titers of the immunoglobulin G against the anti-spike protein subunit 1 of SARS-CoV-2 after vaccination of patients with early breast cancer undergoing concurrent chemotherapy, endocrinal or targeted non-cytotoxic treatments, and no treatments. RESULTS Standard doses of COVID-19 vaccines provided sufficient immune responses in patients with early breast cancer, regardless of the type of anticancer therapies. However, the post-vaccination serum anti-spike antibody titers were significantly lower in the patients under cytotoxic chemotherapy. CONCLUSION Our study emphasizes the importance of the personalized risk stratification and consideration for booster doses in more vulnerable populations.
Collapse
Affiliation(s)
| | | | - Song Yi Park
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Korea
| | - Dae-Won Lee
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Korea
| | - Chang Kyung Kang
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Korea
| | - Pyoeng Gyun Choe
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Korea
| | - Nam Joong Kim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Korea
| | - Myoung-don Oh
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Korea
| | - Wan Beom Park
- Wan Beom Park, MD, PhD, Department of Internal Medicine, Seoul National University Hospital, 101, Daehak-ro, Jongro-gu, Seoul 03080, Republic of Korea. Tel: +82 2 2072 3596; Email
| | - Kyung-Hun Lee
- Corresponding author: Kyung-Hun Lee, MD, PhD, Department of Internal Medicine, Seoul National University Hospital, 101, Daehak-ro, Jongro-gu, Seoul 03080, Republic of Korea. Tel: 82 2 2072 7207; Email
| | - Seock-Ah Im
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Korea,Cancer Research Institute, Seoul National University, Seoul, Korea
| |
Collapse
|
154
|
Ben Houmich T, Tali A, Debbagh F, Lamrani Hanchi A, Soraa N. Seroprevalence of SARS-CoV-2 antibodies in vaccinated healthcare workers in Marrakech (Morocco). Int J Immunopathol Pharmacol 2022; 36:3946320221133697. [PMID: 36215392 PMCID: PMC9558851 DOI: 10.1177/03946320221133697] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
INTRODUCTION The SARS-CoV-2 pandemic has had a considerable impact, causing millions of deaths worldwide, including many healthcare workers (HCWs). The pharmaceutical industry has been working diligently since the start of the pandemic to develop various vaccines to fight the spread of the virus and protect the population. OBJECTIVE To study the seroprevalence of neutralizing anti-SARS-CoV-2 antibodies in vaccinated HCWs at the Mohamed VI University Hospital in Marrakech and to determine the parameters that can influence immune response. METHODS A cross-sectional study of 138 HCWs was performed between October and December 2021 by measuring IgG antibodies directed against the spike antigen of SARS-CoV-2 using an Abbott Architect® SARS-CoV-2 IgG II assay. RESULTS The mean age was 31.42 years, the sex ratio was 2.94 women to each man, and the overall prevalence was 97%. We found 39.5% of the participants had experienced COVID-19 infections pre-vaccination, which decreased to 26.8% after vaccination. Neutralizing antibody titers were dependent on the type of vaccine: they were higher with the Pfizer-BioNTech vaccine, the number of doses (p < .001), and post-vaccine COVID-19 form. The post-vaccine COVID-19 infection rates were lower with the Sinopharm vaccine. CONCLUSION Heterologous vaccination with non-mRNA and mRNA vaccines and the consideration of post-vaccination COVID-19 infection as a booster could help optimize vaccine results while reducing potential side effects.
Collapse
Affiliation(s)
- Taoufik Ben Houmich
- Department of Microbiology,
Mohamed
VI University Hospital, Marrakech,
Morocco,Taoufik Ben Houmich, Department of
Microbiology, Mohamed VI university hospital of Marrakech, hay socoma 1 askjour,
numero 1089, Marrakech, Morocco.
| | - Abdelali Tali
- Department of Microbiology,
Mohamed
VI University Hospital, Marrakech,
Morocco
| | - Fayrouz Debbagh
- Department of Microbiology,
Mohamed
VI University Hospital, Marrakech,
Morocco
| | - Asma Lamrani Hanchi
- Department of Microbiology,
Mohamed
VI University Hospital, Marrakech,
Morocco,Faculty of Medicine and Pharmacy, Cadi Ayyad
University, Marrakech, Morocco
| | - Nabila Soraa
- Department of Microbiology,
Mohamed
VI University Hospital, Marrakech,
Morocco,Faculty of Medicine and Pharmacy, Cadi Ayyad
University, Marrakech, Morocco
| |
Collapse
|
155
|
Predictors of Nonseroconversion to SARS-CoV-2 Vaccination in Kidney Transplant Recipients. Transplant Direct 2022; 8:e1397. [PMID: 36245996 PMCID: PMC9553374 DOI: 10.1097/txd.0000000000001397] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 09/04/2022] [Indexed: 11/27/2022] Open
Abstract
Kidney transplant recipients (KTRs) are still at risk of severe COVID-19 disease after SARS‑CoV‑2 vaccination, especially when they have limited antibody formation. Our aim was to understand the factors that may limit their humoral response.
Collapse
|
156
|
Hvidt AK, Baerends EAM, Søgaard OS, Stærke NB, Raben D, Reekie J, Nielsen H, Johansen IS, Wiese L, Benfield TL, Iversen KK, Mustafa AB, Juhl MR, Petersen KT, Ostrowski SR, Lindvig SO, Rasmussen LD, Schleimann MH, Andersen SD, Juhl AK, Dietz LL, Andreasen SR, Lundgren J, Østergaard L, Tolstrup M. Comparison of vaccine-induced antibody neutralization against SARS-CoV-2 variants of concern following primary and booster doses of COVID-19 vaccines. Front Med (Lausanne) 2022; 9:994160. [PMID: 36262278 PMCID: PMC9574042 DOI: 10.3389/fmed.2022.994160] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 09/07/2022] [Indexed: 12/20/2022] Open
Abstract
The SARS-CoV-2 pandemic has, as of July 2022, infected more than 550 million people and caused over 6 million deaths across the world. COVID-19 vaccines were quickly developed to protect against severe disease, hospitalization and death. In the present study, we performed a direct comparative analysis of four COVID-19 vaccines: BNT162b2 (Pfizer/BioNTech), mRNA-1273 (Moderna), ChAdOx1 (Oxford/AstraZeneca) and Ad26.COV2.S (Johnson & Johnson/Janssen), following primary and booster vaccination. We focused on the vaccine-induced antibody-mediated immune response against multiple SARS-CoV-2 variants: wildtype, B.1.1.7 (Alpha), B.1.351 (Beta), B.1.617.2 (Delta) and B.1.1.529 (Omicron). The analysis included the quantification of total IgG levels against SARS-CoV-2 Spike, as well as the quantification of antibody neutralization titers. Furthermore, the study assessed the high-throughput ACE2 competition assay as a surrogate for the traditional pseudovirus neutralization assay. The results demonstrated marked differences in antibody-mediated immune responses. The lowest Spike-specific IgG levels and antibody neutralization titers were induced by one dose of the Ad26.COV2.S vaccine, intermediate levels by two doses of the BNT162b2 vaccine, and the highest levels by two doses of the mRNA-1273 vaccine or heterologous vaccination of one dose of the ChAdOx1 vaccine and a subsequent mRNA vaccine. The study also demonstrated that accumulation of SARS-CoV-2 Spike protein mutations was accompanied by a marked decline in antibody neutralization capacity, especially for B.1.1.529. Administration of a booster dose was shown to significantly increase Spike-specific IgG levels and antibody neutralization titers, erasing the differences between the vaccine-induced antibody-mediated immune response between the four vaccines. The findings of this study highlight the importance of booster vaccines and the potential inclusion of future heterologous vaccination strategies for broad protection against current and emerging SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Astrid K. Hvidt
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark,Department of Clinical Medicine, Aarhus University, Aarhus, Denmark,*Correspondence: Astrid K. Hvidt,
| | - Eva A. M. Baerends
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark,Department of Clinical Medicine, Aarhus University, Aarhus, Denmark,Eva A. M. Baerends,
| | - Ole S. Søgaard
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark,Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Nina B. Stærke
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark,Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Dorthe Raben
- Center of Excellence for Health, Immunity and Infections, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Joanne Reekie
- Center of Excellence for Health, Immunity and Infections, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Henrik Nielsen
- Department of Infectious Diseases, Aalborg University Hospital, Aalborg, Denmark,Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Isik S. Johansen
- Department of Infectious Diseases, Odense University Hospital, Odense, Denmark,Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Lothar Wiese
- Department of Medicine, Zealand University Hospital, Roskilde, Denmark
| | - Thomas L. Benfield
- Department of Infectious Diseases, Copenhagen University Hospital—Amager and Hvidovre, Hvidovre, Denmark,Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Kasper K. Iversen
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark,Deparment of Cardiology and Emergency Medicine, Herlev Hospital, Herlev, Denmark
| | - Ahmed B. Mustafa
- Department of Infectious Diseases, Copenhagen University Hospital—Amager and Hvidovre, Hvidovre, Denmark,Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Maria R. Juhl
- Department of Infectious Diseases, Aalborg University Hospital, Aalborg, Denmark,Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Kristine T. Petersen
- Department of Infectious Diseases, Aalborg University Hospital, Aalborg, Denmark,Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Sisse R. Ostrowski
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark,Department of Clinical Immunology, Copenhagen University Hospital—Rigshospitalet, Copenhagen, Denmark
| | - Susan O. Lindvig
- Department of Infectious Diseases, Odense University Hospital, Odense, Denmark,Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Line D. Rasmussen
- Department of Infectious Diseases, Odense University Hospital, Odense, Denmark,Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Marianne H. Schleimann
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark,Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Sidsel D. Andersen
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark,Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Anna K. Juhl
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark,Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Lisa L. Dietz
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark,Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Signe R. Andreasen
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark,Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Jens Lundgren
- Center of Excellence for Health, Immunity and Infections, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark,Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark,Department of Infectious Diseases, Copenhagen University Hospital—Rigshospitalet, Copenhagen, Denmark
| | - Lars Østergaard
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark,Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Martin Tolstrup
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark,Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | | |
Collapse
|
157
|
Sharma P, Gupta E, Basu S, Agarwal R, Mishra S, Kale P, Mundeja N, Charan BS, Singh GK, Singh MM. Neutralizing antibody responses to SARS-CoV-2: A population based seroepidemiological analysis. Indian J Med Microbiol 2022; 40:585-587. [PMID: 35953349 PMCID: PMC9359489 DOI: 10.1016/j.ijmmb.2022.07.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 07/15/2022] [Accepted: 07/16/2022] [Indexed: 11/26/2022]
Abstract
This study (August-September 2021) estimated the seroprevalence of SARS-CoV-2 neutralizing antibodies in the general population of Delhi and correlated it with their anti-SARS-CoV-2 IgG levels. Samples were selected by simple random sampling method. The neutralizing capacity was estimated by performing a surrogate virus neutralization test (sVNT) (GenScript), Piscataway, NJ, USA. A total of 2233 (87.1%, 95% C.I. 85.7, 88.3) of the 2564 SARS-CoV-2 IgG seropositive samples had detectable SARS-CoV-2 neutralizing antibodies. In samples with S/CO ≥ 4.00, the neutralizing antibodies ranged from 94.5% to 100%. The SARS-CoV-2 neutralizing antibody seroprevalence strongly correlated with the S/CO range of IgG SARS-CoV-2 (r = 0.62, p = 0.002).
Collapse
Affiliation(s)
- Pragya Sharma
- Department of Community Medicine, Maulana Azad Medical College, New Delhi, India
| | - Ekta Gupta
- Department of Clinical Virology, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Saurav Basu
- Department of Community Medicine, Maulana Azad Medical College, New Delhi, India; Indian Institute of Public Health - Delhi, Public Health Foundation of India, New Delhi, India
| | - Reshu Agarwal
- Department of Clinical Virology, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Suruchi Mishra
- Department of Community Medicine, Maulana Azad Medical College, New Delhi, India.
| | - Pratibha Kale
- Department of Clinical Virology, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Nutan Mundeja
- Directorate General Health Services, Government of NCT, New Delhi, India
| | - B S Charan
- Directorate General Health Services, Government of NCT, New Delhi, India
| | - Gautam Kumar Singh
- Directorate General Health Services, Government of NCT, New Delhi, India
| | - M M Singh
- Department of Community Medicine, Maulana Azad Medical College, New Delhi, India
| |
Collapse
|
158
|
de Boer J, Saade U, Granjon E, Trouillet-Assant S, Saade C, Pottel H, Zrein M. A novel assessment method for COVID-19 humoral immunity duration using serial measurements in naturally infected and vaccinated subjects. PLoS One 2022; 17:e0274553. [PMID: 36173972 PMCID: PMC9521896 DOI: 10.1371/journal.pone.0274553] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 08/31/2022] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Collecting information on sustainability of immune responses after infection or vaccination is crucial to inform medical decision-making and vaccination strategies. Data on how long-lasting antibodies against SARS-COV-2 could provide a humoral and protective immunity and prevent reinfection with SARS-CoV-2 or its variants is particularly valuable. This study presents a novel method to quantitatively measure and monitor the diversity of SARS-CoV-2 specific antibody profiles over time. METHODS Serum samples from two groups were used in this study: Samples from 20 naturally infected subjects (followed for up to 1 year) and samples from 83 subjects vaccinated with one or two doses of the Pfizer BioNtech vaccine (BNT162b2/BNT162b2) (followed for up to 6 months). The Multi-SARS-CoV-2 assay, a multiparameter serology test developed for the serological confirmation of past-infections, was used to determine the reactivity of six different SARS-CoV-2 antigens. For each subject sample, 3 dilutions (1/50, 1/400 and 1/3200) were defined as an optimal set over the six antigens and their respective linear ranges. This allowed accurate quantification of the corresponding six antibodies. Nonlinear mixed-effects modelling was applied to convert intensity readings from 3 determined dilutions to a single quantification value for each antibody. RESULTS Median half-life for the 20 naturally infected vs 74 vaccinated subjects (two doses) was 120 vs 50 days for RBD, 127 vs 53 days for S1 and 187 vs 86 days for S2 antibodies respectively. CONCLUSION The newly proposed method, based on a series of a limited number of dilutions, can convert a conventional qualitative assay into a quantitative assay. This conversion helps define the sustainability of specific immune responses against each relevant viral antigen and can help in defining the protection characteristics after an infection or a vaccination.
Collapse
Affiliation(s)
- Jasper de Boer
- Department of Public Health and Primary Care, KU Leuven Campus Kulak, Kortrijk, Belgium
| | - Ursula Saade
- R&D Department, InfYnity Biomarkers, Lyon, France
| | | | - Sophie Trouillet-Assant
- Virology Laboratory, Institute of Infectious Agents, Laboratory associated with the National Reference Centre for Respiratory Viruses, Civils Hospices of Lyon, Lyon, France
- International Center of Research in Infectiology, Institut National de la Santé et de la Recherche Médicale U1111, Centre National de la Recherche Scientifique UMR5308, Ecole Normale Supérieure de Lyon, Claude Bernard Lyon 1 University, Lyon, France
| | - Carla Saade
- Virology Laboratory, Institute of Infectious Agents, Laboratory associated with the National Reference Centre for Respiratory Viruses, Civils Hospices of Lyon, Lyon, France
| | - Hans Pottel
- Department of Public Health and Primary Care, KU Leuven Campus Kulak, Kortrijk, Belgium
| | - Maan Zrein
- R&D Department, InfYnity Biomarkers, Lyon, France
| | - Covid ser study group
- Virology Laboratory, Institute of Infectious Agents, Laboratory associated with the National Reference Centre for Respiratory Viruses, Civils Hospices of Lyon, Lyon, France
| |
Collapse
|
159
|
Sugiyama A, Kurisu A, Nagashima S, Hando K, Saipova K, Akhmedova S, Abe K, Imada H, Hussain MRA, Ouoba S, E B, Ko K, Akita T, Yamazaki S, Yokozaki M, Tanaka J. Seroepidemiological study of factors affecting anti-spike IgG antibody titers after a two-dose mRNA COVID-19 vaccination in 3744 healthy Japanese volunteers. Sci Rep 2022; 12:16294. [PMID: 36175506 PMCID: PMC9520958 DOI: 10.1038/s41598-022-20747-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 09/19/2022] [Indexed: 02/06/2023] Open
Abstract
Several factors related to anti-spike(S) IgG antibody titers after mRNA COVID-19 vaccination have been elucidated, but the magnitude of the effects of each factor has not been fully understood. This cross-sectional study assessed anti-S and anti-nucleocapsid (N) antibody titers on 3744 healthy volunteers (median age, 36 years; IQR, 24-49 years; females, 59.0%) who received two doses of mRNA-1273 or BNT162b2 vaccine and completed a survey questionnaire. Multiple regression was conducted to identify factors associated with antibody titers. All but one participant tested positive for anti-S antibodies (99.97%). The following factors were independently and significantly associated with high antibody titer: < 3 months from vaccination (ratio of means 4.41); mRNA-1273 vaccine (1.90, vs BNT162b2); anti-N antibody positivity (1.62); age (10's: 1.50, 20's: 1.37, 30's: 1.26, 40's: 1.16, 50's: 1.15, vs ≧60's); female (1.07); immunosuppressive therapy (0.54); current smoking (0.85); and current drinking (0.96). The largest impact on anti-S IgG antibody titers was found in elapsed time after vaccination, followed by vaccine brand, immunosuppressants, previous SARS-CoV-2 infection (anti-N antibody positive), and age. Although the influence of adverse reactions after the vaccine, gender, smoking, and drinking was relatively small, they were independently related factors.
Collapse
Affiliation(s)
- Aya Sugiyama
- grid.257022.00000 0000 8711 3200Department of Epidemiology, Infectious Disease Control and Prevention, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima, 734-8551 Japan
| | - Akemi Kurisu
- grid.257022.00000 0000 8711 3200Department of Epidemiology, Infectious Disease Control and Prevention, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima, 734-8551 Japan
| | - Shintaro Nagashima
- grid.257022.00000 0000 8711 3200Department of Epidemiology, Infectious Disease Control and Prevention, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima, 734-8551 Japan
| | - Kiyomi Hando
- grid.257022.00000 0000 8711 3200Department of Epidemiology, Infectious Disease Control and Prevention, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima, 734-8551 Japan
| | - Khilola Saipova
- grid.257022.00000 0000 8711 3200Department of Epidemiology, Infectious Disease Control and Prevention, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima, 734-8551 Japan ,grid.444564.30000 0004 0402 7972Department of Clinical Radiology and Oncology, Andijan State Medical Institute, Andijan, Uzbekistan
| | - Sayyora Akhmedova
- grid.257022.00000 0000 8711 3200Department of Epidemiology, Infectious Disease Control and Prevention, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima, 734-8551 Japan ,Department of Cardiorheumatology, Republican Specialized Scientific-Practical Medical Center of Pediatrics, Tashkent, Uzbekistan
| | - Kanon Abe
- grid.257022.00000 0000 8711 3200Department of Epidemiology, Infectious Disease Control and Prevention, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima, 734-8551 Japan
| | - Hirohito Imada
- grid.257022.00000 0000 8711 3200Department of Epidemiology, Infectious Disease Control and Prevention, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima, 734-8551 Japan
| | - Md Razeen Ashraf Hussain
- grid.257022.00000 0000 8711 3200Department of Epidemiology, Infectious Disease Control and Prevention, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima, 734-8551 Japan
| | - Serge Ouoba
- grid.257022.00000 0000 8711 3200Department of Epidemiology, Infectious Disease Control and Prevention, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima, 734-8551 Japan ,grid.457337.10000 0004 0564 0509Unité de Recherche Clinique de Nanoro (URCN), Institut de Recherche en Science de La Santé (IRSS), Nanoro, Burkina Faso
| | - Bunthen E
- grid.257022.00000 0000 8711 3200Department of Epidemiology, Infectious Disease Control and Prevention, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima, 734-8551 Japan ,grid.415732.6Payment Certification Agency (PCA), Ministry of Health, Phnom Penh, Cambodia
| | - Ko Ko
- grid.257022.00000 0000 8711 3200Department of Epidemiology, Infectious Disease Control and Prevention, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima, 734-8551 Japan
| | - Tomoyuki Akita
- grid.257022.00000 0000 8711 3200Department of Epidemiology, Infectious Disease Control and Prevention, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima, 734-8551 Japan
| | - Shinichi Yamazaki
- grid.470097.d0000 0004 0618 7953Division of Clinical Laboratory Medicine, Hiroshima University Hospital, Hiroshima, Japan
| | - Michiya Yokozaki
- grid.470097.d0000 0004 0618 7953Division of Clinical Laboratory Medicine, Hiroshima University Hospital, Hiroshima, Japan
| | - Junko Tanaka
- grid.257022.00000 0000 8711 3200Department of Epidemiology, Infectious Disease Control and Prevention, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima, 734-8551 Japan
| |
Collapse
|
160
|
Williams E, Colson J, Valiathan R, Carreño JM, Krammer F, Hoffer M, Pallikkuth S, Pahwa S, Andrews D. Permissive omicron breakthrough infections in individuals with binding or neutralizing antibodies to ancestral SARS-CoV-2. Vaccine 2022; 40:5868-5872. [PMID: 36088193 PMCID: PMC9424516 DOI: 10.1016/j.vaccine.2022.08.058] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 08/12/2022] [Accepted: 08/24/2022] [Indexed: 12/14/2022]
Abstract
BACKGROUND Breakthrough infection with the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) Omicron variant (B.1.1.529) has occurred in populations with high vaccination rates. METHODS In a longitudinal cohort study, pre-breakthrough infection sera for Omicron breakthroughs (n = 12) were analyzed. Assays utilized include a laboratory-developed solid phase binding assay to recombinant spike protein, a commercial assay to the S1 domain of the spike protein calibrated to the World Health Organization (WHO) standard, and a commercial solid-phase surrogate neutralizing activity (SNA) assay. All assays employed spike protein preparations based on sequences from the Wuhan-Hu-1 strain. RESULTS Pre-breakthrough binding antibody titers ranged from 1:800 to 1:51,200 for the laboratory-developed binding assay, which correlated well and agreed quantitatively with the commercial spike S1 domain WHO calibrated assay. SNA was detected in 10/12 (83%) samples. CONCLUSIONS Neither high binding titers nor SNA were markers of protection from Omicron infection/re-infection.
Collapse
Affiliation(s)
- Erin Williams
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Department of Biomedical Engineering, University of Miami, Miami, FL 33136, USA
| | - Jordan Colson
- Department of Pathology and Laboratory Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Ranjini Valiathan
- Department of Pathology and Laboratory Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Juan Manuel Carreño
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Center for Vaccine Research and Pandemic Preparedness (C-VARPP), Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Pathology, Molecular and Cell-based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Center for Vaccine Research and Pandemic Preparedness (C-VARPP), Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Michael Hoffer
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Department of Neurological Surgery, University of Miami, Miller School of Medicine, Miami, FL 33136, USA
| | - Suresh Pallikkuth
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Savita Pahwa
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| | - David Andrews
- Department of Pathology and Laboratory Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| |
Collapse
|
161
|
Preventing SARS-CoV-2 Infection Using Anti-spike Nanobody-IFN-β Conjugated Exosomes. Pharm Res 2022; 40:927-935. [PMID: 36163411 PMCID: PMC9512977 DOI: 10.1007/s11095-022-03400-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Accepted: 09/18/2022] [Indexed: 12/03/2022]
Abstract
Purpose To inhibit the transmission of SARS-CoV-2, we developed engineered exosomes that were conjugated with anti-spike nanobodies and type I interferon β (IFN-β). We evaluated the efficacy and potency of nanobody-IFN-β conjugated exosomes to treatment of SARS-CoV-2 infection. Methods Milk fat globule epidermal growth factor 8 (MFG-E8) is a glycoprotein that binds to phosphatidylserine (PS) exposed on the exosomes. We generated nanobody-IFN-β conjugated exosomes by fusing an anti-spike nanobody and IFN-β with MFG-E8. We used the SARS-CoV-2 pseudovirus with the spike of the D614G mutant that encodes ZsGreen to mimic the infection process of the SARS-CoV-2. The SARS-CoV-2 pseudovirus was infected with angiotensin-converting enzyme-2 (ACE2) expressing adenocarcinomic human alveolar basal epithelial cells (A549) or ACE2 expressing HEK-blue IFNα/β cells in the presence of nanobody-IFN-β conjugated exosomes. By assessing the expression of ZsGreen in target cells and the upregulation of interferon-stimulated genes (ISGs) in infected cells, we evaluated the anti-viral effects of nanobody-IFN-β conjugated exosomes. Results We confirmed the anti-spike nanobody and IFN-β expressions on the exosomes. Exosomes conjugated with nanobody-hIFN-β inhibited the interaction between the spike protein and ACE2, thereby inhibiting the infection of host cells with SARS-CoV-2 pseudovirus. At the same time, IFN-β was selectively delivered to SARS-CoV-2 infected cells, resulting in the upregulation of ISGs expression. Conclusion Exosomes conjugated with nanobody-IFN-β may provide potential benefits in the treatment of COVID-19 because of the cooperative anti-viral effects of the anti-spike nanobody and the IFN-β. Supplementary Information The online version contains supplementary material available at 10.1007/s11095-022-03400-0.
Collapse
|
162
|
Vallejo A, Vizcarra P, Martín-Hondarza A, Gómez-Maldonado S, Haemmerle J, Velasco H, Casado JL. Impact of SARS-CoV-2-specific memory B cells on the immune response after mRNA-based Comirnaty vaccine in seronegative health care workers. Front Microbiol 2022; 13:1002748. [PMID: 36212856 PMCID: PMC9535088 DOI: 10.3389/fmicb.2022.1002748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 09/08/2022] [Indexed: 12/04/2022] Open
Abstract
Purpose To analyze the impact of SARS-COV-2-specific memory B cells (MBC) on the immune response after two doses of mRNA-based Comirnaty COVID-19 vaccine in seronegative health care workers. This study is seeking a rationale for boosting vaccines. Methods Longitudinal study including 31 seronegative health care workers with undetectable specific MBCs (IgG−MBC− group), 24 seronegative with detectable specific MBCs (IgG−MBC+ group), and 24 seropositive with detectable specific MBCs (IgG+MBC+ group). The level of antibodies that inhibit ACE2-RBD interaction, and anti-Spike IgG, IgA, and IgM antibodies was quantified by ELISA. In addition, specific memory B and T cells were quantified by flow cytometry. Results The level of specific MBCs, and isotypes, in the IgG−MBC− group was lower compared to that found in IgG−MBC+ (p = 0.0001) and IgG+MBC+ (p < 0.0001) groups, respectively. ACE2-RBD neutralizing antibodies and anti-S IgG antibodies were at lower levels in the IgG−MBC−group after the vaccine. Specific MBCs directly correlated with specific CD4+ T cells (although not significant, p = 0.065), while no correlation was found with specific CD8+ T cells (p = 0.156) after the vaccine. In parallel, ACE2-RBD neutralizing antibodies only positively correlated with specific CD4+ T cells (p = 0.034). Conclusion IgG−MBC− individuals showed the worst humoral and cellular responses, both in frequency and magnitude, after vaccination. Individuals whose antibodies wane and become undetectable after a given period of time post vaccination and show no specific MBCs are less protected and hence are good candidates for boosting vaccine. On the other hand, seronegative individuals with specific MBC showed faster and higher responses compared to the IgG−MBC− group.
Collapse
Affiliation(s)
- Alejandro Vallejo
- Laboratory of Immunovirology, Department of Infectious Diseases, Ramon y Cajal Institute for Health Investigation (IRyCIS), University Hospital Ramón y Cajal, Madrid, Spain
- *Correspondence: Alejandro Vallejo,
| | - Pilar Vizcarra
- Department of Infectious Diseases, Ramon y Cajal Institute for Health Investigation (IRyCIS), University Hospital Ramón y Cajal, Madrid, Spain
| | - Adrián Martín-Hondarza
- Laboratory of Immunovirology, Department of Infectious Diseases, Ramon y Cajal Institute for Health Investigation (IRyCIS), University Hospital Ramón y Cajal, Madrid, Spain
| | - Sandra Gómez-Maldonado
- Department of Infectious Diseases, Ramon y Cajal Institute for Health Investigation (IRyCIS), University Hospital Ramón y Cajal, Madrid, Spain
| | - Johannes Haemmerle
- Department of Prevention of Occupational Risks, University Hospital Ramón y Cajal, Madrid, Spain
| | - Héctor Velasco
- Laboratory of Immunovirology, Department of Infectious Diseases, Ramon y Cajal Institute for Health Investigation (IRyCIS), University Hospital Ramón y Cajal, Madrid, Spain
| | - José L. Casado
- Department of Infectious Diseases, Ramon y Cajal Institute for Health Investigation (IRyCIS), University Hospital Ramón y Cajal, Madrid, Spain
- José L. Casado,
| |
Collapse
|
163
|
Hartley GE, Edwards ESJ, O’Hehir RE, van Zelm MC. New insights into human immune memory from SARS-CoV-2 infection and vaccination. Allergy 2022; 77:3553-3566. [PMID: 36048132 PMCID: PMC9538469 DOI: 10.1111/all.15502] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 08/13/2022] [Accepted: 08/29/2022] [Indexed: 01/28/2023]
Abstract
Since early 2020, the world has been embroiled in an ongoing viral pandemic with SARS-CoV-2 and emerging variants resulting in mass morbidity and an estimated 6 million deaths globally. The scientific community pivoted rapidly, providing unique and innovative means to identify infected individuals, technologies to evaluate immune responses to infection and vaccination, and new therapeutic strategies to treat infected individuals. Never before has immunology been so critically at the forefront of combatting a global pandemic. It has now become evident that not just antibody responses, but formation and durability of immune memory cells following vaccination are associated with protection against severe disease from SARS-CoV-2 infection. Furthermore, the emergence of variants of concern (VoC) highlight the need for immunological markers to quantify the protective capacity of Wuhan-based vaccines. Thus, harnessing and modulating the immune response is key to successful vaccination and treatment of disease. We here review the latest knowledge about immune memory generation and durability following natural infection and vaccination, and provide insights into the attributes of immune memory that may protect from emerging variants.
Collapse
Affiliation(s)
- Gemma E. Hartley
- Allergy and Clinical Immunology Laboratory, Department of Immunology and Pathology, Central Clinical SchoolMonash UniversityMelbourneVictoriaAustralia
| | - Emily S. J. Edwards
- Allergy and Clinical Immunology Laboratory, Department of Immunology and Pathology, Central Clinical SchoolMonash UniversityMelbourneVictoriaAustralia
| | - Robyn E. O’Hehir
- Allergy and Clinical Immunology Laboratory, Department of Immunology and Pathology, Central Clinical SchoolMonash UniversityMelbourneVictoriaAustralia,Allergy, Asthma and Clinical Immunology ServiceAlfred HospitalMelbourneVictoriaAustralia
| | - Menno C. van Zelm
- Allergy and Clinical Immunology Laboratory, Department of Immunology and Pathology, Central Clinical SchoolMonash UniversityMelbourneVictoriaAustralia,Allergy, Asthma and Clinical Immunology ServiceAlfred HospitalMelbourneVictoriaAustralia
| |
Collapse
|
164
|
El-Menyar A, Khan NA, Mekkodathil A, Rizoli S, Consunji R, Elmenyar E, Galwankar S, Al-Thani H. A quick scoping review of the first year of vaccination against the COVID-19 pandemic: Do we need more shots or time? Medicine (Baltimore) 2022; 101:e30609. [PMID: 36123868 PMCID: PMC9477714 DOI: 10.1097/md.0000000000030609] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 08/16/2022] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND The emergence of new severe acute respiratory syndrome coronavirus 2 variants, along with the waning of vaccine-induced immunity, has increased breakthrough infections and urged booster jabs and debates. In the short term, the administration of booster doses has been reported to be safe and enhance severe acute respiratory syndrome coronavirus 2-specific neutralizing antibody levels. However, the effects of these doses on the pandemic trajectory and herd immunity are unclear. There is insufficient evidence that a third booster shot of the coronavirus disease 2019 (COVID-19) vaccine maintains longer immunity and covers new viral variants. The lack of sufficient evidence, combined with the fact that millions of people have not yet received 1 or 2 jabs of the COVID-19 vaccine, has raised concerns regarding the call for booster vaccinations. METHODS We conducted a quick scoping review to explore the literature on the need for a booster COVID-19 vaccination from January 1, 2021, to April 30, 2022. RESULTS Sixty-one relevant publications were identified, of which 17 were related to waning immunity after 2 doses of the vaccine among the general population or healthcare workers, 19 were related to the third or booster dose of vaccination after the second dose among the general population or healthcare workers, and 25 were related to booster dose among immunocompromised patient. CONCLUSIONS Initially, the need for a booster dose was equivocal; however, several studies demonstrated the benefit of the booster dose over time. Adequate scientific information is required regarding the administration of booster doses to the general population as well as the high-risk individuals.
Collapse
Affiliation(s)
- Ayman El-Menyar
- Department of Surgery, Trauma and Vascular Surgery Clinical Research, Hamad Medical Corporation, Qatar
- Clinical Medicine, Weill Cornell Medical College, Doha, Qatar
| | - Naushad Ahmad Khan
- Department of Surgery, Trauma and Vascular Surgery Clinical Research, Hamad Medical Corporation, Qatar
| | - Ahammed Mekkodathil
- Department of Surgery, Trauma and Vascular Surgery Clinical Research, Hamad Medical Corporation, Qatar
| | - Sandro Rizoli
- Department of Surgery, Trauma Surgery, Hamad Medical Corporation, Qatar
| | - Rafael Consunji
- Department of Surgery, Trauma Surgery, Hamad Medical Corporation, Qatar
| | | | - Sagar Galwankar
- Department of Emergency Medicine, Sarasota Memorial Hospital, Sarasota, FL
| | - Hassan Al-Thani
- Department of Surgery, Trauma Surgery, Hamad Medical Corporation, Qatar
| |
Collapse
|
165
|
Wei X, Rong N, Liu J. Prospects of animal models and their application in studies on adaptive immunity to SARS-CoV-2. Front Immunol 2022; 13:993754. [PMID: 36189203 PMCID: PMC9523127 DOI: 10.3389/fimmu.2022.993754] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 09/05/2022] [Indexed: 01/08/2023] Open
Abstract
The adaptive immune response induced by SARS-CoV-2 plays a key role in the antiviral process and can protect the body from the threat of infection for a certain period of time. However, owing to the limitations of clinical studies, the antiviral mechanisms, protective thresholds, and persistence of the immune memory of adaptive immune responses remain unclear. This review summarizes existing research models for SARS-CoV-2 and elaborates on the advantages of animal models in simulating the clinical symptoms of COVID-19 in humans. In addition, we systematically summarize the research progress on the SARS-CoV-2 adaptive immune response and the remaining key issues, as well as the application and prospects of animal models in this field. This paper provides direction for in-depth analysis of the anti-SARS-CoV-2 mechanism of the adaptive immune response and lays the foundation for the development and application of vaccines and drugs.
Collapse
Affiliation(s)
- Xiaohui Wei
- National Health Commission Key Laboratory of Human Disease Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China
| | | | - Jiangning Liu
- National Health Commission Key Laboratory of Human Disease Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China
| |
Collapse
|
166
|
Waning of SARS-CoV-2 Seropositivity among Healthy Young Adults over Seven Months. Vaccines (Basel) 2022; 10:vaccines10091532. [PMID: 36146610 PMCID: PMC9505545 DOI: 10.3390/vaccines10091532] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 09/03/2022] [Accepted: 09/09/2022] [Indexed: 01/19/2023] Open
Abstract
Background: We conducted a longitudinal study to estimate immunity produced in response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection among university students over seven months. Methods: All participants were attending a public university and resided in Pitt County, North Carolina. University students enrolled weekly for 10 weeks between 26 August 2020 and 28 October 2020, resulting in 136 young adults completing at least one study visit by 17 November 2020. Enrolled students completed an online survey and nasal swab collection at two-week intervals and monthly blood collection between 26 August 2020 and 31 March 2021. Results: Amongst 695 serum samples tested during follow-up, the prevalence of a positive result for anti-nucleocapsid antibodies (N-IgG) was 9.78%. In 22 students with more than one positive N-IgG serum sample, 68.1% of the group lost persistence of N-IgG below the positive threshold over 140 days. Anti-spike IgG antibodies were significantly higher among 11 vaccinated compared to 10 unvaccinated. Conclusions: In healthy young adults, N-IgG wanes below the detectable threshold within five months. S-IgG titer remained consistently elevated months after infection, and significantly increased after vaccination.
Collapse
|
167
|
Methodology to estimate natural- and vaccine-induced antibodies to SARS-CoV-2 in a large geographic region. PLoS One 2022; 17:e0273694. [PMID: 36084125 PMCID: PMC9462720 DOI: 10.1371/journal.pone.0273694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Accepted: 08/14/2022] [Indexed: 11/19/2022] Open
Abstract
Accurate estimates of natural and/or vaccine-induced antibodies to SARS-CoV-2 are difficult to obtain. Although model-based estimates of seroprevalence have been proposed, they require inputting unknown parameters including viral reproduction number, longevity of immune response, and other dynamic factors. In contrast to a model-based approach, the current study presents a data-driven detailed statistical procedure for estimating total seroprevalence (defined as antibodies from natural infection or from full vaccination) in a region using prospectively collected serological data and state-level vaccination data. Specifically, we conducted a longitudinal statewide serological survey with 88,605 participants 5 years or older with 3 prospective blood draws beginning September 30, 2020. Along with state vaccination data, as of October 31, 2021, the estimated percentage of those 5 years or older with naturally occurring antibodies to SARS-CoV-2 in Texas is 35.0% (95% CI = (33.1%, 36.9%)). This is 3× higher than, state-confirmed COVID-19 cases (11.83%) for all ages. The percentage with naturally occurring or vaccine-induced antibodies (total seroprevalence) is 77.42%. This methodology is integral to pandemic preparedness as accurate estimates of seroprevalence can inform policy-making decisions relevant to SARS-CoV-2.
Collapse
|
168
|
Meyrath M, Szpakowska M, Plesseria JM, Domingues O, Langlet J, Weber B, Krüger R, Ollert M, Chevigné A. Nanoluciferase-based cell fusion assay for rapid and high-throughput assessment of SARS-CoV-2-neutralizing antibodies in patient samples. Methods Enzymol 2022; 675:351-381. [PMID: 36220277 PMCID: PMC9459433 DOI: 10.1016/bs.mie.2022.07.015] [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] [Indexed: 11/19/2022]
Abstract
After more than two years, COVID-19 still represents a global health burden of unprecedented extent and assessing the degree of immunity of individuals against SARS-CoV-2 remains a challenge. Virus neutralization assays represent the gold standard for assessing antibody-mediated protection against SARS-CoV-2 in sera from recovered and/or vaccinated individuals. Neutralizing antibodies block the interaction of viral spike protein with human angiotensin-converting enzyme 2 (ACE2) receptor in vitro and prevent viral entry into host cells. Classical viral neutralization assays using full replication-competent viruses are restricted to specific biosafety level 3-certified laboratories, limiting their utility for routine and large-scale applications. We developed therefore a cell-fusion-based assay building on the interaction between viral spike and ACE2 receptor expressed on two different cell lines, substantially reducing biosafety risks associated with classical viral neutralization assays. This chapter describes this simple, sensitive, safe and cost-effective approach for rapid and high-throughput evaluation of SARS-CoV-2 neutralizing antibodies relying on high-affinity NanoLuc® luciferase complementation technology (HiBiT). When applied to a variety of standards and patient samples, this method yields highly reproducible results in 96-well, as well as in 384-well format. The use of novel NanoLuc® substrates with increased signal stability like Nano-Glo® Endurazine™ furthermore allows for high flexibility in assay set-up and full automatization of all reading processes. Lastly, the assay is suitable to evaluate the neutralizing capacity of sera against the existing spike variants, and potentially variants that will emerge in the future.
Collapse
Affiliation(s)
- Max Meyrath
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg; Laboratoires Réunis Luxembourg, Z.A.C. Laangwiss, Junglinster, Luxembourg
| | - Martyna Szpakowska
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg
| | - Jean-Marc Plesseria
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg
| | - Olivia Domingues
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg
| | - Jérémie Langlet
- Business Development Office, Luxembourg Institute of Health (LIH), Strassen, Luxembourg
| | - Bernard Weber
- Laboratoires Réunis Luxembourg, Z.A.C. Laangwiss, Junglinster, Luxembourg
| | - Rejko Krüger
- Transversal Translational Medicine (TTM), Luxembourg Institute of Health (LIH), Strassen, Luxembourg; Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-Belval, Luxembourg; Centre Hospitalier de Luxembourg (CHL), Luxembourg, Luxembourg
| | - Markus Ollert
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg; Department of Dermatology and Allergy Center, Odense Research Center for Anaphylaxis, Odense University Hospital, University of Southern Denmark, Odense, Denmark
| | - Andy Chevigné
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg.
| |
Collapse
|
169
|
Pavel-Tanasa M, Constantinescu D, Cianga CM, Anisie E, Mereuta AI, Tuchilus CG, Cianga P. Adipokines, and not vitamin D, associate with antibody immune responses following dual BNT162b2 vaccination within individuals younger than 60 years. Front Immunol 2022; 13:1000006. [PMID: 36119038 PMCID: PMC9481237 DOI: 10.3389/fimmu.2022.1000006] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 08/17/2022] [Indexed: 11/24/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) led to a global health outbreak known as the COVID-19 pandemic which has been lasting since March 2020. Vaccine became accessible to people only at the beginning of 2021 which greatly helped reducing the mortality rate and severity of COVID-19 infection afterwards. The efficacy of vaccines was not fully known and studies documenting the immune responses following vaccination are continuing to emerge. Recent evidence indicate that natural infection prior vaccination may improve the antibody and cellular immune responses, while little is known about the factors influencing those processes. Here we investigated the antibody responses following BNT162b2 vaccination in relation to previous-infection status and age, and searched for possible biomarkers associated with the observed changes in immune responses. We found that the previous-infection status caused at least 8-times increase in the antibody titres, effect that was weaker in people over 60 years old and unaltered by the vitamin D serum levels. Furthermore, we identified adiponectin to positively associate with antibody responses and negatively correlate with pro-inflammatory molecules (MCP-1, factor D, CRP, PAI-1), especially in previously-infected individuals.
Collapse
Affiliation(s)
- Mariana Pavel-Tanasa
- Department of Immunology, Faculty of Medicine, Grigore T. Popa University of Medicine and Pharmacy, Iasi, Romania
- Laboratory of Immunology, St. Spiridon County Clinical Emergency Hospital Iasi, Iasi, Romania
- *Correspondence: Mariana Pavel-Tanasa,
| | - Daniela Constantinescu
- Department of Immunology, Faculty of Medicine, Grigore T. Popa University of Medicine and Pharmacy, Iasi, Romania
- Laboratory of Immunology, St. Spiridon County Clinical Emergency Hospital Iasi, Iasi, Romania
| | - Corina Maria Cianga
- Department of Immunology, Faculty of Medicine, Grigore T. Popa University of Medicine and Pharmacy, Iasi, Romania
- Laboratory of Immunology, St. Spiridon County Clinical Emergency Hospital Iasi, Iasi, Romania
| | - Ecaterina Anisie
- Laboratory of Immunology, St. Spiridon County Clinical Emergency Hospital Iasi, Iasi, Romania
| | - Ana Irina Mereuta
- Medical Analysis Laboratory, St. Spiridon County Clinical Emergency Hospital Iasi, Iasi, Romania
| | - Cristina Gabriela Tuchilus
- Medical Analysis Laboratory, St. Spiridon County Clinical Emergency Hospital Iasi, Iasi, Romania
- Department of Preventive Medicine and Interdisciplinarity (Microbiology), Faculty of Medicine, Grigore T. Popa University of Medicine and Pharmacy, Iasi, Romania
| | - Petru Cianga
- Department of Immunology, Faculty of Medicine, Grigore T. Popa University of Medicine and Pharmacy, Iasi, Romania
- Laboratory of Immunology, St. Spiridon County Clinical Emergency Hospital Iasi, Iasi, Romania
| |
Collapse
|
170
|
Shaul AA, Itzhaki Ben Zadok O, Ben-Avraham B, Yaari V, Barsheshet A, Levi A, Ben Zvi H, Eliakim Raz N, Abed G, Abuhazira M, Abu Akel M, Mats I, Barac YD, Aravot D, Kornowski R, Ben-Gal T. Improved immunogenicity following the third dose of BNT162b2 mRNA vaccine in heart transplant recipients. Eur J Cardiothorac Surg 2022; 62:ezac145. [PMID: 35244690 PMCID: PMC9383557 DOI: 10.1093/ejcts/ezac145] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 02/06/2022] [Accepted: 02/21/2022] [Indexed: 01/07/2023] Open
Abstract
OBJECTIVES The immunogenicity of two-dose severe acute respiratory syndrome coronavirus 2 vaccine is lower among heart transplant (HTx) recipients, compared with the general population. Our aim was to assess the immunogenicity of a third-dose vaccine in HTx recipients. METHODS This is a prospective cohort study of HTx recipients who received a third dose of the BNT162b2 vaccine. Immunogenicity was assessed by serum levels of anti-spike immunoglobulin G (S-IgG), taken at baseline and 14-28 days after the third dose. Titres above 50 U/ml were interpreted positive. RESULTS We Included 42 HTx recipients at a median age of 65 years [interquartile range (IQR) 58-70]. At baseline, the median of 27 days (IQR 13-42) before the third dose and the median titre of the whole group was 18 U/ml (IQR 4-130). Only 14 patients (33%) were S-IgG seropositive. After the third dose, the proportion of seropositive patients increased significantly to 57% (P = 0.05) and the median titre increased significantly to 633 U/ml (IQR 7-6104, P < 0.0001). Younger age at HTx (OR per 1-year decrease 1.07, P = 0.05), low tacrolimus serum level (OR per 1-unit decrease 2.28, P = 0.02), mammalian target of rapamycin use (OR 13.3, P = 0.003), lack of oral steroids use (OR 4.17, P = 0.04) and lack of calcineurin inhibitor use (71% of responders vs 100% non-responders received calcineurin inhibitors, P = 0.01) were predictors of seropositive result after the third dose. However, no significant association was detected following adjustment for baseline S-IgG titre. CONCLUSIONS Third-dose booster of BNT162b2 vaccine significantly increased immunogenicity among HTx recipients who previously received a two-dose vaccine.
Collapse
Affiliation(s)
- Aviv Avraham Shaul
- Department of Cardiology, Rabin Medical Center, Petach Tikva, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Osnat Itzhaki Ben Zadok
- Department of Cardiology, Rabin Medical Center, Petach Tikva, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Binyamin Ben-Avraham
- Department of Cardiology, Rabin Medical Center, Petach Tikva, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Vicky Yaari
- Department of Cardiology, Rabin Medical Center, Petach Tikva, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Alon Barsheshet
- Department of Cardiology, Rabin Medical Center, Petach Tikva, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Amos Levi
- Department of Cardiology, Rabin Medical Center, Petach Tikva, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Haim Ben Zvi
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Microbiology Laboratory, Rabin Medical Center, Petach Tikva, Israel
| | - Noa Eliakim Raz
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Department of Infectious Diseases, Rabin Medical Center, Petach Tikva, Israel
| | - Galia Abed
- Department of Cardiology, Rabin Medical Center, Petach Tikva, Israel
| | - Miriam Abuhazira
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Department of Cardio-Thoracic Surgery, Rabin Medical Center, Petach Tikva, Israel
| | - Mahmood Abu Akel
- Department of Cardiology, Rabin Medical Center, Petach Tikva, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Israel Mats
- Department of Cardiology, Rabin Medical Center, Petach Tikva, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yaron D Barac
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Department of Cardio-Thoracic Surgery, Rabin Medical Center, Petach Tikva, Israel
| | - Dan Aravot
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Department of Cardio-Thoracic Surgery, Rabin Medical Center, Petach Tikva, Israel
| | - Ran Kornowski
- Department of Cardiology, Rabin Medical Center, Petach Tikva, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Tuvia Ben-Gal
- Department of Cardiology, Rabin Medical Center, Petach Tikva, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
171
|
Meyer-Arndt L, Braun J, Fauchere F, Vanshylla K, Loyal L, Henze L, Kruse B, Dingeldey M, Jürchott K, Mangold M, Maraj A, Braginets A, Böttcher C, Nitsche A, de la Rosa K, Ratswohl C, Sawitzki B, Holenya P, Reimer U, Sander LE, Klein F, Paul F, Bellmann-Strobl J, Thiel A, Giesecke-Thiel C. SARS-CoV-2 mRNA vaccinations fail to elicit humoral and cellular immune responses in patients with multiple sclerosis receiving fingolimod. J Neurol Neurosurg Psychiatry 2022; 93:960-971. [PMID: 35835468 PMCID: PMC9380499 DOI: 10.1136/jnnp-2022-329395] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 06/27/2022] [Indexed: 11/16/2022]
Abstract
BACKGROUND SARS-CoV-2 mRNA vaccination of healthy individuals is highly immunogenic and protective against severe COVID-19. However, there are limited data on how disease-modifying therapies (DMTs) alter SARS-CoV-2 mRNA vaccine immunogenicity in patients with autoimmune diseases. METHODS As part of a prospective cohort study, we investigated the induction, stability and boosting of vaccine-specific antibodies, B cells and T cells in patients with multiple sclerosis (MS) on different DMTs after homologous primary, secondary and booster SARS-CoV-2 mRNA vaccinations. Of 126 patients with MS analysed, 105 received either anti-CD20-based B cell depletion (aCD20-BCD), fingolimod, interferon-β, dimethyl fumarate, glatiramer acetate, teriflunomide or natalizumab, and 21 were untreated MS patients for comparison. RESULTS In contrast to all other MS patients, and even after booster, most aCD20-BCD- and fingolimod-treated patients showed no to markedly reduced anti-S1 IgG, serum neutralising activity and a lack of receptor binding domain-specific and S2-specific B cells. Patients receiving fingolimod additionally lacked spike-reactive CD4+ T cell responses. The duration of fingolimod treatment, rather than peripheral blood B and T cell counts prior to vaccination, determined whether a humoral immune response was elicited. CONCLUSIONS The lack of immunogenicity under long-term fingolimod treatment demonstrates that functional immune responses require not only immune cells themselves, but also access of these cells to the site of inoculation and their unimpeded movement. The absence of humoral and T cell responses suggests that fingolimod-treated patients with MS are at risk for severe SARS-CoV-2 infections despite booster vaccinations, which is highly relevant for clinical decision-making and adapted protective measures, particularly considering additional recently approved sphingosine-1-phosphate receptor antagonists for MS treatment.
Collapse
Affiliation(s)
- Lil Meyer-Arndt
- Regenerative Immunology and Aging, BIH Immunomics, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- NeuroCure Clinical Research Center, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Department of Neurology with Experimental Neurology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Julian Braun
- Regenerative Immunology and Aging, BIH Immunomics, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Si-M / "Der Simulierte Mensch" a science framework of Technische Universität Berlin and Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Florent Fauchere
- Regenerative Immunology and Aging, BIH Immunomics, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Si-M / "Der Simulierte Mensch" a science framework of Technische Universität Berlin and Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Kanika Vanshylla
- Laboratory of Experimental Immunology, Institute of Virology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Lucie Loyal
- Regenerative Immunology and Aging, BIH Immunomics, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Si-M / "Der Simulierte Mensch" a science framework of Technische Universität Berlin and Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Larissa Henze
- Regenerative Immunology and Aging, BIH Immunomics, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Si-M / "Der Simulierte Mensch" a science framework of Technische Universität Berlin and Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Beate Kruse
- Regenerative Immunology and Aging, BIH Immunomics, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Si-M / "Der Simulierte Mensch" a science framework of Technische Universität Berlin and Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Manuela Dingeldey
- Regenerative Immunology and Aging, BIH Immunomics, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Si-M / "Der Simulierte Mensch" a science framework of Technische Universität Berlin and Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Karsten Jürchott
- Regenerative Immunology and Aging, BIH Immunomics, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Si-M / "Der Simulierte Mensch" a science framework of Technische Universität Berlin and Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Maike Mangold
- Regenerative Immunology and Aging, BIH Immunomics, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Ardit Maraj
- Regenerative Immunology and Aging, BIH Immunomics, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Andre Braginets
- Regenerative Immunology and Aging, BIH Immunomics, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Chotima Böttcher
- Department of Neuropsychiatry, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Experimental and Clinical Research Center, a cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité - Universitätsmedizin Berlin, Berlin, Germany
- Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | | | - Kathrin de la Rosa
- Department of Cancer and Immunology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Christoph Ratswohl
- Department of Cancer and Immunology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Birgit Sawitzki
- Berlin Institute of Health (BIH), Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Institute for Medical Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | | | - Ulf Reimer
- JPT Peptide Technologies, Berlin, Germany
| | - Leif E Sander
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Florian Klein
- Laboratory of Experimental Immunology, Institute of Virology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- German Center for Infection Research (DZIF), Partner site Bonn-Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Friedemann Paul
- NeuroCure Clinical Research Center, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Experimental and Clinical Research Center, a cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité - Universitätsmedizin Berlin, Berlin, Germany
- Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Judith Bellmann-Strobl
- NeuroCure Clinical Research Center, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Experimental and Clinical Research Center, a cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité - Universitätsmedizin Berlin, Berlin, Germany
- Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Andreas Thiel
- Regenerative Immunology and Aging, BIH Immunomics, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Si-M / "Der Simulierte Mensch" a science framework of Technische Universität Berlin and Charité - Universitätsmedizin Berlin, Berlin, Germany
| | | |
Collapse
|
172
|
Abstract
The COVID-19 pandemic has caused an unprecedented health crisis and economic burden worldwide. Its etiological agent SARS-CoV-2, a new virus in the coronavirus family, has infected hundreds of millions of people worldwide. SARS-CoV-2 has evolved over the past 2 years to increase its transmissibility as well as to evade the immunity established by previous infection and vaccination. Nevertheless, strong immune responses can be elicited by viral infection and vaccination, which have proved to be protective against the emergence of variants, particularly with respect to hospitalization or severe disease. Here, we review our current understanding of how the virus enters the host cell and how our immune system is able to defend against cell entry and infection. Neutralizing antibodies are a major component of our immune defense and have been extensively studied for SARS-CoV-2 and its variants. Structures of these neutralizing antibodies have provided valuable insights into epitopes that are protective against the original ancestral virus and the variants that have emerged. The molecular characterization of neutralizing epitopes as well as epitope conservation and resistance are important for design of next-generation vaccines and antibody therapeutics.
Collapse
Affiliation(s)
- Hejun Liu
- Department of Integrative Structural and Computational BiologyThe Scripps Research InstituteLa JollaCaliforniaUSA
| | - Ian A. Wilson
- Department of Integrative Structural and Computational BiologyThe Scripps Research InstituteLa JollaCaliforniaUSA
- The Skaggs Institute for Chemical BiologyThe Scripps Research InstituteLa JollaCaliforniaUSA
| |
Collapse
|
173
|
Ozonoff A, Schaenman J, Jayavelu ND, Milliren CE, Calfee CS, Cairns CB, Kraft M, Baden LR, Shaw AC, Krammer F, van Bakel H, Esserman DA, Liu S, Sesma AF, Simon V, Hafler DA, Montgomery RR, Kleinstein SH, Levy O, Bime C, Haddad EK, Erle DJ, Pulendran B, Nadeau KC, Davis MM, Hough CL, Messer WB, Higuita NIA, Metcalf JP, Atkinson MA, Brakenridge SC, Corry D, Kheradmand F, Ehrlich LI, Melamed E, McComsey GA, Sekaly R, Diray-Arce J, Peters B, Augustine AD, Reed EF, Altman MC, Becker PM, Rouphael N, Ozonoff A, Schaenman J, Jayavelu ND, Milliren CE, Calfee CS, Cairns CB, Kraft M, Baden LR, Shaw AC, Krammer F, van Bakel H, Esserman DA, Liu S, Sesma AF, Simon V, Hafler DA, Montgomery RR, Kleinstein SH, Levy O, Bime C, Haddad EK, Erle DJ, Pulendran B, Nadeau KC, Davis MM, Hough CL, Messer WB, Higuita NIA, Metcalf JP, Atkinson MA, Brakenridge SC, Corry D, Kheradmand F, Ehrlich LI, Melamed E, McComsey GA, Sekaly R, Diray-Arce J, Peters B, Augustine AD, Reed EF, McEnaney K, Barton B, Lentucci C, Saluvan M, Chang AC, Hoch A, Albert M, Shaheen T, Kho AT, Thomas S, Chen J, Murphy MD, Cooney M, Presnell S, Fragiadakis GK, et alOzonoff A, Schaenman J, Jayavelu ND, Milliren CE, Calfee CS, Cairns CB, Kraft M, Baden LR, Shaw AC, Krammer F, van Bakel H, Esserman DA, Liu S, Sesma AF, Simon V, Hafler DA, Montgomery RR, Kleinstein SH, Levy O, Bime C, Haddad EK, Erle DJ, Pulendran B, Nadeau KC, Davis MM, Hough CL, Messer WB, Higuita NIA, Metcalf JP, Atkinson MA, Brakenridge SC, Corry D, Kheradmand F, Ehrlich LI, Melamed E, McComsey GA, Sekaly R, Diray-Arce J, Peters B, Augustine AD, Reed EF, Altman MC, Becker PM, Rouphael N, Ozonoff A, Schaenman J, Jayavelu ND, Milliren CE, Calfee CS, Cairns CB, Kraft M, Baden LR, Shaw AC, Krammer F, van Bakel H, Esserman DA, Liu S, Sesma AF, Simon V, Hafler DA, Montgomery RR, Kleinstein SH, Levy O, Bime C, Haddad EK, Erle DJ, Pulendran B, Nadeau KC, Davis MM, Hough CL, Messer WB, Higuita NIA, Metcalf JP, Atkinson MA, Brakenridge SC, Corry D, Kheradmand F, Ehrlich LI, Melamed E, McComsey GA, Sekaly R, Diray-Arce J, Peters B, Augustine AD, Reed EF, McEnaney K, Barton B, Lentucci C, Saluvan M, Chang AC, Hoch A, Albert M, Shaheen T, Kho AT, Thomas S, Chen J, Murphy MD, Cooney M, Presnell S, Fragiadakis GK, Patel R, Guan L, Gygi J, Pawar S, Brito A, Khalil Z, Maguire C, Fourati S, Overton JA, Vita R, Westendorf K, Salehi-Rad R, Leligdowicz A, Matthay MA, Singer JP, Kangelaris KN, Hendrickson CM, Krummel MF, Langelier CR, Woodruff PG, Powell DL, Kim JN, Simmons B, Goonewardene IM, Smith CM, Martens M, Mosier J, Kimura H, Sherman AC, Walsh SR, Issa NC, Dela Cruz C, Farhadian S, Iwasaki A, Ko AI, Chinthrajah S, Ahuja N, Rogers AJ, Artandi M, Siegel SA, Lu Z, Drevets DA, Brown BR, Anderson ML, Guirgis FW, Thyagarajan RV, Rousseau JF, Wylie D, Busch J, Gandhi S, Triplett TA, Yendewa G, Giddings O, Anderson EJ, Mehta AK, Sevransky JE, Khor B, Rahman A, Stadlbauer D, Dutta J, Xie H, Kim-Schulze S, Gonzalez-Reiche AS, van de Guchte A, Farrugia K, Khan Z, Maecker HT, Elashoff D, Brook J, Ramires-Sanchez E, Llamas M, Rivera A, Perdomo C, Ward DC, Magyar CE, Fulcher JA, Abe-Jones Y, Asthana S, Beagle A, Bhide S, Carrillo SA, Chak S, Fragiadakis GK, Ghale R, Gonzalez A, Jauregui A, Jones N, Lea T, Lee D, Lota R, Milush J, Nguyen V, Pierce L, Prasad PA, Rao A, Samad B, Shaw C, Sigman A, Sinha P, Ward A, Willmore A, Zhan J, Rashid S, Rodriguez N, Tang K, Altamirano LT, Betancourt L, Curiel C, Sutter N, Paz MT, Tietje-Ulrich G, Leroux C, Connors J, Bernui M, Kutzler MA, Edwards C, Lee E, Lin E, Croen B, Semenza NC, Rogowski B, Melnyk N, Woloszczuk K, Cusimano G, Bell MR, Furukawa S, McLin R, Marrero P, Sheidy J, Tegos GP, Nagle C, Mege N, Ulring K, Seyfert-Margolis V, Conway M, Francisco D, Molzahn A, Erickson H, Wilson CC, Schunk R, Sierra B, Hughes T, Smolen K, Desjardins M, van Haren S, Mitre X, Cauley J, Li X, Tong A, Evans B, Montesano C, Licona JH, Krauss J, Chang JBP, Izaguirre N, Chaudhary O, Coppi A, Fournier J, Mohanty S, Muenker MC, Nelson A, Raddassi K, Rainone M, Ruff WE, Salahuddin S, Schulz WL, Vijayakumar P, Wang H, Wunder Jr. E, Young HP, Zhao Y, Saksena M, Altman D, Kojic E, Srivastava K, Eaker LQ, Bermúdez-González MC, Beach KF, Sominsky LA, Azad AR, Carreño JM, Singh G, Raskin A, Tcheou J, Bielak D, Kawabata H, Mulder LCF, Kleiner G, Lee AS, Do ED, Fernandes A, Manohar M, Hagan T, Blish CA, Din HN, Roque J, Yang S, Brunton A, Sullivan PE, Strnad M, Lyski ZL, Coulter FJ, Booth JL, Sinko LA, Moldawer LL, Borresen B, Roth-Manning B, Song LZ, Nelson E, Lewis-Smith M, Smith J, Tipan PG, Siles N, Bazzi S, Geltman J, Hurley K, Gabriele G, Sieg S, Vaysman T, Bristow L, Hussaini L, Hellmeister K, Samaha H, Cheng A, Spainhour C, Scherer EM, Johnson B, Bechnak A, Ciric CR, Hewitt L, Carter E, Mcnair N, Panganiban B, Huerta C, Usher J, Ribeiro SP, Altman MC, Becker PM, Rouphael N. Phenotypes of disease severity in a cohort of hospitalized COVID-19 patients: Results from the IMPACC study. EBioMedicine 2022; 83:104208. [PMID: 35952496 PMCID: PMC9359694 DOI: 10.1016/j.ebiom.2022.104208] [Show More Authors] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 07/11/2022] [Accepted: 07/25/2022] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Better understanding of the association between characteristics of patients hospitalized with coronavirus disease 2019 (COVID-19) and outcome is needed to further improve upon patient management. METHODS Immunophenotyping Assessment in a COVID-19 Cohort (IMPACC) is a prospective, observational study of 1164 patients from 20 hospitals across the United States. Disease severity was assessed using a 7-point ordinal scale based on degree of respiratory illness. Patients were prospectively surveyed for 1 year after discharge for post-acute sequalae of COVID-19 (PASC) through quarterly surveys. Demographics, comorbidities, radiographic findings, clinical laboratory values, SARS-CoV-2 PCR and serology were captured over a 28-day period. Multivariable logistic regression was performed. FINDINGS The median age was 59 years (interquartile range [IQR] 20); 711 (61%) were men; overall mortality was 14%, and 228 (20%) required invasive mechanical ventilation. Unsupervised clustering of ordinal score over time revealed distinct disease course trajectories. Risk factors associated with prolonged hospitalization or death by day 28 included age ≥ 65 years (odds ratio [OR], 2.01; 95% CI 1.28-3.17), Hispanic ethnicity (OR, 1.71; 95% CI 1.13-2.57), elevated baseline creatinine (OR 2.80; 95% CI 1.63- 4.80) or troponin (OR 1.89; 95% 1.03-3.47), baseline lymphopenia (OR 2.19; 95% CI 1.61-2.97), presence of infiltrate by chest imaging (OR 3.16; 95% CI 1.96-5.10), and high SARS-CoV2 viral load (OR 1.53; 95% CI 1.17-2.00). Fatal cases had the lowest ratio of SARS-CoV-2 antibody to viral load levels compared to other trajectories over time (p=0.001). 589 survivors (51%) completed at least one survey at follow-up with 305 (52%) having at least one symptom consistent with PASC, most commonly dyspnea (56% among symptomatic patients). Female sex was the only associated risk factor for PASC. INTERPRETATION Integration of PCR cycle threshold, and antibody values with demographics, comorbidities, and laboratory/radiographic findings identified risk factors for 28-day outcome severity, though only female sex was associated with PASC. Longitudinal clinical phenotyping offers important insights, and provides a framework for immunophenotyping for acute and long COVID-19. FUNDING NIH.
Collapse
Affiliation(s)
- Al Ozonoff
- Clinical & Data Coordinating Center (CDCC); Precision Vaccines Program, Boston Children's Hospital, Boston, MA, United States
| | - Joanna Schaenman
- David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA, United States
| | | | - Carly E. Milliren
- Clinical & Data Coordinating Center (CDCC); Precision Vaccines Program, Boston Children's Hospital, Boston, MA, United States
| | - Carolyn S. Calfee
- University of California San Francisco School of Medicine, San Francisco, CA, United States
| | - Charles B. Cairns
- Drexel University/Tower Health Hospital, Philadelphia, PA, United States
| | - Monica Kraft
- University of Arizona, Tucson, AZ, United States
| | - Lindsey R. Baden
- Boston Clinical Site: Precision Vaccines Program, Boston Children's Hospital, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA, United States
| | - Albert C. Shaw
- Yale School of Medicine, and Yale School of Public Health, New Haven, CT, United States
| | - Florian Krammer
- Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Harm van Bakel
- Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Denise A. Esserman
- Yale School of Medicine, and Yale School of Public Health, New Haven, CT, United States
| | - Shanshan Liu
- Clinical & Data Coordinating Center (CDCC); Precision Vaccines Program, Boston Children's Hospital, Boston, MA, United States
| | | | - Viviana Simon
- Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - David A. Hafler
- Yale School of Medicine, and Yale School of Public Health, New Haven, CT, United States
| | - Ruth R. Montgomery
- Yale School of Medicine, and Yale School of Public Health, New Haven, CT, United States
| | - Steven H. Kleinstein
- Yale School of Medicine, and Yale School of Public Health, New Haven, CT, United States
| | - Ofer Levy
- Boston Clinical Site: Precision Vaccines Program, Boston Children's Hospital, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA, United States
| | | | - Elias K. Haddad
- Drexel University/Tower Health Hospital, Philadelphia, PA, United States
| | - David J. Erle
- University of California San Francisco School of Medicine, San Francisco, CA, United States
| | | | | | | | | | | | | | - Jordan P. Metcalf
- Oklahoma University Health Sciences Center, Oklahoma, OK, United States
| | - Mark A. Atkinson
- University of Florida, Gainesville and University of South Florida, Tampa, FL, United States
| | - Scott C. Brakenridge
- University of Florida, Gainesville and University of South Florida, Tampa, FL, United States
| | - David Corry
- Baylor College of Medicine, and the Center for Translational Research on Inflammatory Diseases, Michael E. DeBakey, Houston, TX, United States
| | - Farrah Kheradmand
- Baylor College of Medicine, and the Center for Translational Research on Inflammatory Diseases, Michael E. DeBakey, Houston, TX, United States
| | | | - Esther Melamed
- The University of Texas at Austin, Austin, TX, United States
| | | | - Rafick Sekaly
- Case Western Reserve University, Cleveland, OH, United States
| | - Joann Diray-Arce
- Clinical & Data Coordinating Center (CDCC); Precision Vaccines Program, Boston Children's Hospital, Boston, MA, United States
| | - Bjoern Peters
- La Jolla Institute for Immunology, La Jolla, CA, United States
| | - Alison D. Augustine
- National Institute of Allergy and Infectious Diseases/National Institutes of Health, Bethesda, MD, United States
| | - Elaine F. Reed
- David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA, United States
| | | | - Patrice M. Becker
- National Institute of Allergy and Infectious Diseases/National Institutes of Health, Bethesda, MD, United States
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
174
|
Jansen EB, Orvold SN, Swan CL, Yourkowski A, Thivierge BM, Francis ME, Ge A, Rioux M, Darbellay J, Howland JG, Kelvin AA. After the virus has cleared-Can preclinical models be employed for Long COVID research? PLoS Pathog 2022; 18:e1010741. [PMID: 36070309 PMCID: PMC9451097 DOI: 10.1371/journal.ppat.1010741] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Severe Acute Respiratory Syndrome Coronavirus (SARS-CoV-2) can cause the life-threatening acute respiratory disease called COVID-19 (Coronavirus Disease 2019) as well as debilitating multiorgan dysfunction that persists after the initial viral phase has resolved. Long COVID or Post-Acute Sequelae of COVID-19 (PASC) is manifested by a variety of symptoms, including fatigue, dyspnea, arthralgia, myalgia, heart palpitations, and memory issues sometimes affecting between 30% and 75% of recovering COVID-19 patients. However, little is known about the mechanisms causing Long COVID and there are no widely accepted treatments or therapeutics. After introducing the clinical aspects of acute COVID-19 and Long COVID in humans, we summarize the work in animals (mice, Syrian hamsters, ferrets, and nonhuman primates (NHPs)) to model human COVID-19. The virology, pathology, immune responses, and multiorgan involvement are explored. Additionally, any studies investigating time points longer than 14 days post infection (pi) are highlighted for insight into possible long-term disease characteristics. Finally, we discuss how the models can be leveraged for treatment evaluation, including pharmacological agents that are currently in human clinical trials for treating Long COVID. The establishment of a recognized Long COVID preclinical model representing the human condition would allow the identification of mechanisms causing disease as well as serve as a vehicle for evaluating potential therapeutics.
Collapse
Affiliation(s)
- Ethan B. Jansen
- Vaccine and Infectious Disease Organization VIDO, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
- Department of Biochemistry, Microbiology, and Immunology, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Spencer N. Orvold
- Department of Anatomy, Physiology, and Pharmacology, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Cynthia L. Swan
- Vaccine and Infectious Disease Organization VIDO, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Anthony Yourkowski
- Vaccine and Infectious Disease Organization VIDO, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
- Department of Biochemistry, Microbiology, and Immunology, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Brittany M. Thivierge
- Vaccine and Infectious Disease Organization VIDO, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Magen E. Francis
- Vaccine and Infectious Disease Organization VIDO, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
- Department of Biochemistry, Microbiology, and Immunology, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Anni Ge
- Department of Microbiology and Immunology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Melissa Rioux
- Department of Microbiology and Immunology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Joseph Darbellay
- Vaccine and Infectious Disease Organization VIDO, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - John G. Howland
- Department of Anatomy, Physiology, and Pharmacology, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Alyson A. Kelvin
- Vaccine and Infectious Disease Organization VIDO, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| |
Collapse
|
175
|
Bhiman JN, Moore PL. Leveraging South African HIV research to define SARS-CoV-2 immunity triggered by sequential variants of concern. Immunol Rev 2022; 310:61-75. [PMID: 35599324 PMCID: PMC9349367 DOI: 10.1111/imr.13086] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the virus that causes coronavirus disease 2019 (COVID-19), has shifted our paradigms about B cell immunity and the goals of vaccination for respiratory viruses. The development of population immunity, through responses directed to highly immunogenic regions of this virus, has been a strong driving force in the emergence of progressively mutated variants. This review highlights how the strength of the existing global virology and immunology networks built for HIV vaccine research enabled rapid adaptation of techniques, assays, and skill sets, to expeditiously respond to the SARS-CoV-2 pandemic. Allying real-time genomic surveillance to immunological platforms enabled the characterization of immune responses elicited by infection with distinct variants, in sequential epidemic waves, as well as studies of vaccination and hybrid immunity (combination of infection- and vaccination-induced immunity). These studies have shown that consecutive variants of concern have steadily diminished the ability of vaccines to prevent infection, but that increasing levels of hybrid immunity result in higher frequencies of cross-reactive responses. Ultimately, this rapid pivot from HIV to SARS-CoV-2 enabled a depth of understanding of the SARS-CoV-2 antigenic vulnerabilities as population immunity expanded and diversified, providing key insights for future responses to the SARS-CoV-2 pandemic.
Collapse
Affiliation(s)
- Jinal N Bhiman
- National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg, South Africa
- SAMRC Antibody Immunity Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg, South Africa
| | - Penny L Moore
- National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg, South Africa
- SAMRC Antibody Immunity Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg, South Africa
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
- Centre for the AIDS Programme of Research in South Africa, Durban, South Africa
| |
Collapse
|
176
|
Goldblatt D, Alter G, Crotty S, Plotkin SA. Correlates of protection against SARS-CoV-2 infection and COVID-19 disease. Immunol Rev 2022; 310:6-26. [PMID: 35661178 PMCID: PMC9348242 DOI: 10.1111/imr.13091] [Citation(s) in RCA: 198] [Impact Index Per Article: 66.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Antibodies against epitopes in S1 give the most accurate CoP against infection by the SARS-CoV-2 coronavirus. Measurement of those antibodies by neutralization or binding assays both have predictive value, with binding antibody titers giving the highest statistical correlation. However, the protective functions of antibodies are multiple. Antibodies with multiple functions other than neutralization influence efficacy. The role of cellular responses can be discerned with respect to CD4+ T cells and their augmentation of antibodies, and with respect to CD8+ cells with regard to control of viral replication, particularly in the presence of insufficient antibody. More information is needed on mucosal responses.
Collapse
Affiliation(s)
- David Goldblatt
- Great Ormond Street Institute of Child HealthUniversity College LondonLondonUK
| | - Galit Alter
- Massachusetts General HospitalRagon Institute of MGH, MIT and HarvardCambridgeMassachusettsUSA
| | - Shane Crotty
- Center for Infectious Disease and Vaccine ResearchLa Jolla Institute for Immunology (LJI)La JollaCaliforniaUSA
- Department of Medicine, Division of Infectious Diseases and Global Public HealthUniversity of California San Diego (UCSD)La JollaCaliforniaUSA
| | | |
Collapse
|
177
|
Sette A, Crotty S. Immunological memory to SARS-CoV-2 infection and COVID-19 vaccines. Immunol Rev 2022; 310:27-46. [PMID: 35733376 PMCID: PMC9349657 DOI: 10.1111/imr.13089] [Citation(s) in RCA: 177] [Impact Index Per Article: 59.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 05/04/2022] [Indexed: 12/12/2022]
Abstract
Immunological memory is the basis of protective immunity provided by vaccines and previous infections. Immunological memory can develop from multiple branches of the adaptive immune system, including CD4 T cells, CD8 T cells, B cells, and long-lasting antibody responses. Extraordinary progress has been made in understanding memory to SARS-CoV-2 infection and COVID-19 vaccines, addressing development; quantitative and qualitative features of different cellular and anatomical compartments; and durability of each cellular component and antibodies. Given the sophistication of the measurements; the size of the human studies; the use of longitudinal samples and cross-sectional studies; and head-to-head comparisons between infection and vaccines or between multiple vaccines, the understanding of immune memory for 1 year to SARS-CoV-2 infection and vaccines already supersedes that of any other acute infectious disease. This knowledge may help inform public policies regarding COVID-19 and COVID-19 vaccines, as well as the scientific development of future vaccines against SARS-CoV-2 and other diseases.
Collapse
Affiliation(s)
- Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA 92037, USA
| | - Shane Crotty
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA 92037, USA
| |
Collapse
|
178
|
Dimeglio C, Herin F, Da-Silva I, Gernigon C, Porcheron M, Chapuy-Regaud S, Izopet J. Decreased Efficiency of Neutralizing Antibodies from Previously Infected or Vaccinated Individuals against the B.1.617.2 (Delta) SARS-CoV-2 Variant. Microbiol Spectr 2022; 10:e0270621. [PMID: 35867411 PMCID: PMC9430143 DOI: 10.1128/spectrum.02706-21] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 05/16/2022] [Indexed: 12/13/2022] Open
Abstract
The neutralizing antibody response is a key component of adaptive immunity and a primary protection against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. The increased transmissibility of the SARS-CoV-2 Delta variant and its capacity to cause more severe disease could be linked to a significant reduction in neutralizing antibodies generated during a previous infection or vaccination. We analyzed blood samples from 162 unvaccinated health care workers (HCWs) collected 1 to 3 months postinfection and from 263 vaccinated health care workers 1 month after the last injection. We have compared the neutralizing antibody titers obtained using two virus strains, B.1.160 and B.1.617.2 (Delta variant). Binding antibody concentrations were measured by an immunoassay. The median neutralizing antibody titer against the B.1.160 strain was 128 (interquartile range [IQR], 16 to 256) and 32 (IQR, 8 to 128) against the Delta variant. To obtain a neutralizing antibody titer of 32 or 64, a binding antibody concentration of 182 binding antibody units (BAU)/mL (IQR, 81 to 974) was required with the strain B.1.160, while a concentration of 2,595 BAU/mL (IQR, 1,176 to 5,353) was required with the Delta variant. Our data indicate that antibodies neutralize the SARS-CoV-2 Delta variant 4 times less efficiently than they neutralize an earlier strain. Half of the HCWs had decreased protection from 94% to 76.8% or less for the same total antibody concentration. But neutralization might be correlated with other immune responses. The contributions of other responses, such as those of the T cell and B cell systems, to protection require further investigation. IMPORTANCE Recent studies showed that the neutralizing antibody titer is an important contributor to protection against SARS-CoV-2. With the emergence of new variants, the question arises of maintaining the neutralizing capacities of vaccines and/or of a past infection. We had protective data associated with total antibody concentrations and neutralizing antibody titers for a B.1.160 strain. We showed that to maintain the same levels of protection and, therefore, the same levels of neutralizing antibodies, a total antibody concentration 8.5 times greater is required with the Delta strain. (This study has been registered at ClinicalTrials.gov under registration no. NCT04385108.).
Collapse
Affiliation(s)
- Chloé Dimeglio
- CHU Toulouse, Hôpital Purpan, Virology Laboratory, Toulouse, France
- INSERM UMR1291 – CNRS UMR5051, Toulouse Institute for Infectious and Inflammatory Diseases (INFINITy), Toulouse, France
| | - Fabrice Herin
- Occupational Diseases Department, Toulouse University Hospital, Toulouse, France
- UMR1295, unité mixte INSERM - Université Toulouse III Paul Sabatier, Centre for Epidemiology and Research in Population Health Unit (CERPOP), Toulouse, France
| | | | - Caroline Gernigon
- Occupational Diseases Department, Toulouse University Hospital, Toulouse, France
- UMR1295, unité mixte INSERM - Université Toulouse III Paul Sabatier, Centre for Epidemiology and Research in Population Health Unit (CERPOP), Toulouse, France
| | - Marion Porcheron
- CHU Toulouse, Hôpital Purpan, Virology Laboratory, Toulouse, France
| | - Sabine Chapuy-Regaud
- CHU Toulouse, Hôpital Purpan, Virology Laboratory, Toulouse, France
- INSERM UMR1291 – CNRS UMR5051, Toulouse Institute for Infectious and Inflammatory Diseases (INFINITy), Toulouse, France
| | - Jacques Izopet
- CHU Toulouse, Hôpital Purpan, Virology Laboratory, Toulouse, France
- INSERM UMR1291 – CNRS UMR5051, Toulouse Institute for Infectious and Inflammatory Diseases (INFINITy), Toulouse, France
| |
Collapse
|
179
|
Protective Immunity of COVID-19 Vaccination with ChAdOx1 nCoV-19 Following Previous SARS-CoV-2 Infection: A Humoral and Cellular Investigation. Viruses 2022; 14:v14091916. [PMID: 36146723 PMCID: PMC9504152 DOI: 10.3390/v14091916] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 08/20/2022] [Accepted: 08/24/2022] [Indexed: 11/17/2022] Open
Abstract
Infections caused by SARS-CoV-2 induce a severe acute respiratory syndrome called COVID-19 and have led to more than six million deaths worldwide. Vaccination is the most effective preventative measure, and cellular and humoral immunity is crucial to developing individual protection. Here, we aim to investigate hybrid immunity against SARS-CoV-2 triggered by the ChAadOx1 nCoV-19 vaccine in a Brazilian cohort. We investigated the immune response from ChAadOx1 nCoV-19 vaccination in naïve (noCOVID-19) and previously infected individuals (COVID-19) by analyzing levels of D-dimers, total IgG, neutralizing antibodies (Nabs), IFN-γ (interferon-γ) secretion, and immunophenotyping of memory lymphocytes. No significant differences in D-dimer levels were observed 7 or 15 days after vaccination (DAV). All vaccinated individuals presented higher levels of total IgG or Nabs with a positive correlation (R = 0.88). Individuals in the COVID-19 group showed higher levels of antibody and memory B cells, with a faster antibody response starting at 7 DAV compared to noCOVID-19 at 15 DAV. Further, ChAadOx1 nCoV-19 vaccination led to enhanced IFN-γ production (15 DAV) and an increase in activated T CD4+ naïve cells in noCOVID-19 individuals in contrast with COVID-19 individuals. Hence, our data support that hybrid immunity triggered by ChAadOx1 nCoV-19 vaccination is associated with enhanced humoral response, together with a balanced cellular response.
Collapse
|
180
|
Differential persistence of neutralizing antibody against SARS-CoV-2 in post immunized Bangladeshi population. Sci Rep 2022; 12:14681. [PMID: 36038600 PMCID: PMC9421641 DOI: 10.1038/s41598-022-18302-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 08/09/2022] [Indexed: 11/09/2022] Open
Abstract
Development of effective vaccines have been immensely welcomed by the world to prevent the transmission of SARS-CoV-2. However, the duration and clinical implications of antibody-mediated natural immunity in SARS-CoV-2 have not been adequately elucidated alongside some other immune system transforming factors. In a cohort study, we measured NAb titer following the 2nd immunization dosage of the CoviShield (AZD1222) vaccine. The enzyme-linked immunoassay was used to look for SARS-CoV-2—specific NAb. We measured NAb at 30 days after the 2nd dosage of immunization and > 96% titer was detected in 42.9% of subjects, but only 5.1% of subjects retained the same level after 180 days. The median NAb titer dropped significantly, from 92% at 30 days to 58% at 180 days (p < 0.001). Besides, there were significant differences observed in NAb titer after 180 days by age, sex, COVID-19 infection, tobacco use, and asthma patients. However, SARS-CoV-2 infection along with two dosages of immunization upheld NAb titer (p < 0.001) even at the end of the study period.
Collapse
|
181
|
A Pilot Study to Examine If Dietary Habits Can Affect Symptomology in Mild Pre-Vaccination COVID-19 Cases. BIOLOGY 2022; 11:biology11091274. [PMID: 36138753 PMCID: PMC9495586 DOI: 10.3390/biology11091274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 08/17/2022] [Accepted: 08/25/2022] [Indexed: 11/16/2022]
Abstract
The heterogeneity of the severity of symptoms of COVID-19 experienced by the young and healthy individuals is poorly understood. The present study was undertaken to mainly examine whether the respective diets and the type of symptoms experienced by patients are predictive of having long COVID-19. Disease severity was assessed with a symptomatology questionnaire and used to group 55 participants in asymptomatic (AS), mild symptoms (S) and long COVID (LC). We found that experiencing a higher number of symptoms as well as fatigue were predictors of developing LC whereas those who experienced rhinorrhea were less likely to develop LC. Blood samples were also taken to measure vitamin D [25(OH)D] concentrations and duration of spike IgG antibodies. In this study, serum 25(OH)D was not significantly different between 3 symptom groups with median (IQR) ng/mL levels of 22.0 (12.3) in the AS, 22.3 (7.5) in S, and 24.9 (9.4) in the LC group (p ≥ 0.05). The duration of IgG antibody response was found to vary greatly, with some individuals showing raised IgG over a year after infection. To examine whether dietary factors can influence the severity of symptoms, diet was analysed using 4–7-day food diaries as well as a Food Frequency Questionnaire (FFQ). Some nutrients such as vitamin E, polyunsaturated fatty acids, fibre, and iron were associated with lower severity of COVID-19. Lower intake of vitamin E was associated with having LC with a median (IQR) intake of 6.2 mg (3.8) seen in LC vs. 8.6 mg (7.2) in the AS group (p = 0.047). This pilot study has highlighted a few differences in the number and type of symptoms experienced by the young non-hospitalised individuals with mild and long COVID-19 and identified a few dietary components for their potential protective role against long COVID-19, however, the findings need to be confirmed with further large scale studies.
Collapse
|
182
|
André S, Azarias da Silva M, Picard M, Alleaume-Buteau A, Kundura L, Cezar R, Soudaramourty C, André SC, Mendes-Frias A, Carvalho A, Capela C, Pedrosa J, Gil Castro A, Loubet P, Sotto A, Muller L, Lefrant JY, Roger C, Claret PG, Duvnjak S, Tran TA, Zghidi-Abouzid O, Nioche P, Silvestre R, Corbeau P, Mammano F, Estaquier J. Low quantity and quality of anti-spike humoral response is linked to CD4 T-cell apoptosis in COVID-19 patients. Cell Death Dis 2022; 13:741. [PMID: 36030261 PMCID: PMC9419645 DOI: 10.1038/s41419-022-05190-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 08/11/2022] [Accepted: 08/15/2022] [Indexed: 01/21/2023]
Abstract
In addition to an inflammatory reaction, Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2)-infected patients present lymphopenia, which we recently reported as being related to abnormal programmed cell death. As an efficient humoral response requires CD4 T-cell help, we hypothesized that the propensity of CD4 T cells to die may impact the quantity and quality of the humoral response in acutely infected individuals. In addition to specific immunoglobulins (Ig)A, IgM, and IgG against SARS-CoV-2 nucleocapsid (N), membrane (M), and spike (S1) proteins, we assessed the quality of IgG response by measuring the avidity index. Because the S protein represents the main target for neutralization and antibody-dependent cellular cytotoxicity responses, we also analyzed anti-S-specific IgG using S-transfected cells (S-Flow). Our results demonstrated that most COVID-19 patients have a predominant IgA anti-N humoral response during the early phase of infection. This specific humoral response preceded the anti-S1 in time and magnitude. The avidity index of anti-S1 IgG was low in acutely infected individuals compared to convalescent patients. We showed that the percentage of apoptotic CD4 T cells is inversely correlated with the levels of specific IgG antibodies. These lower levels were also correlated positively with plasma levels of CXCL10, a marker of disease severity, and soluble Fas ligand that contributes to T-cell death. Finally, we found lower S-Flow responses in patients with higher CD4 T-cell apoptosis. Altogether, these results demonstrate that individuals with high levels of CD4 T-cell apoptosis and CXCL10 have a poor ability to build an efficient anti-S response. Consequently, preventing CD4 T-cell death might be a strategy for improving humoral response during the acute phase, thereby reducing COVID-19 pathogenicity.
Collapse
Affiliation(s)
- Sonia André
- grid.508487.60000 0004 7885 7602Université Paris Cité, INSERM U1124, F-75006 Paris, France
| | - Marne Azarias da Silva
- grid.508487.60000 0004 7885 7602Université Paris Cité, INSERM U1124, F-75006 Paris, France
| | - Morgane Picard
- grid.508487.60000 0004 7885 7602Université Paris Cité, INSERM U1124, F-75006 Paris, France
| | - Aurélie Alleaume-Buteau
- grid.508487.60000 0004 7885 7602Université Paris Cité, INSERM U1124, F-75006 Paris, France ,grid.508487.60000 0004 7885 7602Structural and Molecular Analysis Platform, BioMedTech Facilities INSERM US36-CNRS UMS2009, Université Paris Cité, Paris, France
| | - Lucy Kundura
- grid.411165.60000 0004 0593 8241Laboratoire d’Immunologie, CHU de Nîmes, Nîmes, France
| | - Renaud Cezar
- grid.411165.60000 0004 0593 8241Laboratoire d’Immunologie, CHU de Nîmes, Nîmes, France
| | | | - Santa Cruz André
- grid.10328.380000 0001 2159 175XLife and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal ,grid.10328.380000 0001 2159 175XICVS/3B’s—PT Government Associate Laboratory, Braga/Guimarães, Portugal ,Department of Internal Medicine, Hospital of Braga, Braga, Portugal ,grid.512329.eClinical Academic Center-Braga, Braga, Portugal
| | - Ana Mendes-Frias
- Department of Internal Medicine, Hospital of Braga, Braga, Portugal ,grid.512329.eClinical Academic Center-Braga, Braga, Portugal
| | - Alexandre Carvalho
- grid.10328.380000 0001 2159 175XLife and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal ,grid.10328.380000 0001 2159 175XICVS/3B’s—PT Government Associate Laboratory, Braga/Guimarães, Portugal ,Department of Internal Medicine, Hospital of Braga, Braga, Portugal ,grid.512329.eClinical Academic Center-Braga, Braga, Portugal
| | - Carlos Capela
- grid.10328.380000 0001 2159 175XLife and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal ,grid.10328.380000 0001 2159 175XICVS/3B’s—PT Government Associate Laboratory, Braga/Guimarães, Portugal ,Department of Internal Medicine, Hospital of Braga, Braga, Portugal ,grid.512329.eClinical Academic Center-Braga, Braga, Portugal
| | - Jorge Pedrosa
- grid.10328.380000 0001 2159 175XLife and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal ,grid.10328.380000 0001 2159 175XICVS/3B’s—PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - António Gil Castro
- grid.10328.380000 0001 2159 175XLife and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal ,grid.10328.380000 0001 2159 175XICVS/3B’s—PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Paul Loubet
- grid.411165.60000 0004 0593 8241Service des Maladies Infectieuses et Tropicales, CHU de Nîmes, Nîmes, France
| | - Albert Sotto
- grid.411165.60000 0004 0593 8241Service des Maladies Infectieuses et Tropicales, CHU de Nîmes, Nîmes, France
| | - Laurent Muller
- grid.411165.60000 0004 0593 8241Service de Réanimation Chirugicale, CHU de Nîmes, Nîmes, France
| | - Jean-Yves Lefrant
- grid.411165.60000 0004 0593 8241Service de Réanimation Chirugicale, CHU de Nîmes, Nîmes, France
| | - Claire Roger
- grid.411165.60000 0004 0593 8241Service de Réanimation Chirugicale, CHU de Nîmes, Nîmes, France
| | - Pierre-Géraud Claret
- grid.411165.60000 0004 0593 8241Urgences Médico-Chirugicales Hospitalisation, CHU de Nîmes, Nîmes, France
| | - Sandra Duvnjak
- grid.411165.60000 0004 0593 8241Service de Gérontologie et Prévention du Vieillissement, CHU de Nîmes, Nîmes, France
| | - Tu-Anh Tran
- grid.411165.60000 0004 0593 8241Service de Pédiatrie, CHU de Nîmes, Nîmes, France
| | | | - Pierre Nioche
- grid.508487.60000 0004 7885 7602Université Paris Cité, INSERM U1124, F-75006 Paris, France ,grid.508487.60000 0004 7885 7602Structural and Molecular Analysis Platform, BioMedTech Facilities INSERM US36-CNRS UMS2009, Université Paris Cité, Paris, France
| | - Ricardo Silvestre
- grid.10328.380000 0001 2159 175XLife and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal ,grid.10328.380000 0001 2159 175XICVS/3B’s—PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Pierre Corbeau
- grid.411165.60000 0004 0593 8241Laboratoire d’Immunologie, CHU de Nîmes, Nîmes, France ,grid.121334.60000 0001 2097 0141Institut de Génétique Humaine UMR9002 CNRS-Université de Montpellier, Montpellier, France
| | - Fabrizio Mammano
- grid.508487.60000 0004 7885 7602Université Paris Cité, INSERM U1124, F-75006 Paris, France ,INSERM U1259 MAVIVH, Université de Tours, Tours, France
| | - Jérôme Estaquier
- grid.508487.60000 0004 7885 7602Université Paris Cité, INSERM U1124, F-75006 Paris, France ,CHU de Québec—Université Laval Research Center, Québec City, QC Canada
| |
Collapse
|
183
|
Strahm C, Seneghini M, Güsewell S, Egger T, Leal-Neto O, Brucher A, Lemmenmeier E, Meier Kleeb D, Möller JC, Rieder P, Ruetti M, Rutz R, Schmid HR, Stocker R, Vuichard-Gysin D, Wiggli B, Besold U, Kuster SP, McGeer A, Risch L, Friedl A, Schlegel M, Schmid D, Vernazza P, Kahlert CR, Kohler P. Symptoms Compatible With Long Coronavirus Disease (COVID) in Healthcare Workers With and Without Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) Infection-Results of a Prospective Multicenter Cohort. Clin Infect Dis 2022; 75:e1011-e1019. [PMID: 35090015 PMCID: PMC9383387 DOI: 10.1093/cid/ciac054] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND The burden of long-term symptoms (ie, long COVID) in patients after mild COVID-19 is debated. Within a cohort of healthcare workers (HCWs), frequency and risk factors for symptoms compatible with long COVID are assessed. METHODS Participants answered baseline (August/September 2020) and weekly questionnaires on SARS-CoV-2 nasopharyngeal swab (NPS) results and acute disease symptoms. In January 2021, SARS-CoV-2 serology was performed; in March, symptoms compatible with long COVID (including psychometric scores) were asked and compared between HCWs with positive NPS, seropositive HCWs without positive NPS (presumable asymptomatic/pauci-symptomatic infections), and negative controls. The effect of time since diagnosis and quantitative anti-spike protein antibodies (anti-S) was evaluated. Poisson regression was used to identify risk factors for symptom occurrence. RESULTS Of 3334 HCWs (median, 41 years; 80% female), 556 (17%) had a positive NPS and 228 (7%) were only seropositive. HCWs with positive NPS more frequently reported ≥1 symptom compared with controls (73% vs 52%, P < .001); seropositive HCWs without positive NPS did not score higher than controls (58% vs 52%, P = .13), although impaired taste/olfaction (16% vs 6%, P < .001) and hair loss (17% vs 10%, P = .004) were more common. Exhaustion/burnout was reported by 24% of negative controls. Many symptoms remained elevated in those diagnosed >6 months ago; anti-S titers correlated with high symptom scores. Acute viral symptoms in weekly questionnaires best predicted long-COVID symptoms. Physical activity at baseline was negatively associated with neurocognitive impairment and fatigue scores. CONCLUSIONS Seropositive HCWs without positive NPS are only mildly affected by long COVID. Exhaustion/burnout is common, even in noninfected HCWs. Physical activity might be protective against neurocognitive impairment/fatigue symptoms after COVID-19.
Collapse
Affiliation(s)
- Carol Strahm
- Correspondence: C. Strahm, Division of Infectious Diseases and Hospital Epidemiology, Cantonal Hospital St Gallen, Rorschacherstrasse 95, 9011 St Gallen, Switzerland ()
| | - Marco Seneghini
- Division of Infectious Diseases and Hospital Epidemiology, Cantonal Hospital St Gallen, St Gallen, Switzerland
| | - Sabine Güsewell
- Division of Infectious Diseases and Hospital Epidemiology, Cantonal Hospital St Gallen, St Gallen, Switzerland
| | - Thomas Egger
- Division of Infectious Diseases and Hospital Epidemiology, Cantonal Hospital St Gallen, St Gallen, Switzerland
| | - Onicio Leal-Neto
- Epitrack, Recife, Brazil
- Department of Economics, University of Zurich, Zurich, Switzerland
| | - Angela Brucher
- Psychiatry Services of the Canton of St Gallen (South), St Gallen, Switzerland
| | - Eva Lemmenmeier
- Clienia Littenheid AG, Private Clinic for Psychiatry and Psychotherapy, Littenheid, Switzerland
| | | | - J Carsten Möller
- Center for Neurological Rehabilitation, Zihlschlacht, Switzerland
| | | | - Markus Ruetti
- Fuerstenland Toggenburg Hospital Group, Wil, Switzerland
| | - Remus Rutz
- Division of Infectious Diseases and Hospital Epidemiology, Cantonal Hospital St Gallen, St Gallen, Switzerland
| | | | | | - Danielle Vuichard-Gysin
- Division of Infectious Diseases and Hospital Epidemiology, Thurgau Hospital Group, Muensterlingen, Switzerland
| | - Benedikt Wiggli
- Division of Infectious Diseases and Hospital Epidemiology, Kantonsspital Baden, Baden, Switzerland
| | | | - Stefan P Kuster
- Division of Infectious Diseases and Hospital Epidemiology, Cantonal Hospital St Gallen, St Gallen, Switzerland
- Federal Office of Public Health, Bern, Switzerland
| | | | - Lorenz Risch
- Labormedizinisches Zentrum Dr Risch Ostschweiz AG, Buchs, Switzerland
- Private Universität im Fürstentum Liechtenstein, Triesen, Liechtenstein
- Center of Laboratory Medicine, Institute of Clinical Chemistry, University of Bern, Inselspital, Bern, Switzerland
| | - Andrée Friedl
- Division of Infectious Diseases and Hospital Epidemiology, Kantonsspital Baden, Baden, Switzerland
| | - Matthias Schlegel
- Division of Infectious Diseases and Hospital Epidemiology, Cantonal Hospital St Gallen, St Gallen, Switzerland
| | - Dagmar Schmid
- Clinic for Psychosomatic and Consultation Psychiatry, Cantonal Hospital St Gallen, St Gallen, Switzerland
| | - Pietro Vernazza
- Division of Infectious Diseases and Hospital Epidemiology, Cantonal Hospital St Gallen, St Gallen, Switzerland
| | | | | |
Collapse
|
184
|
Menges D, Zens KD, Ballouz T, Caduff N, Llanas-Cornejo D, Aschmann HE, Domenghino A, Pellaton C, Perreau M, Fenwick C, Pantaleo G, Kahlert CR, Münz C, Puhan MA, Fehr JS. Heterogenous humoral and cellular immune responses with distinct trajectories post-SARS-CoV-2 infection in a population-based cohort. Nat Commun 2022; 13:4855. [PMID: 35982045 PMCID: PMC9386650 DOI: 10.1038/s41467-022-32573-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Accepted: 08/06/2022] [Indexed: 12/14/2022] Open
Abstract
To better understand the development of SARS-CoV-2-specific immunity over time, a detailed evaluation of humoral and cellular responses is required. Here, we characterize anti-Spike (S) IgA and IgG in a representative population-based cohort of 431 SARS-CoV-2-infected individuals up to 217 days after diagnosis, demonstrating that 85% develop and maintain anti-S responses. In a subsample of 64 participants, we further assess anti-Nucleocapsid (N) IgG, neutralizing antibody activity, and T cell responses to Membrane (M), N, and S proteins. In contrast to S-specific antibody responses, anti-N IgG levels decline substantially over time and neutralizing activity toward Delta and Omicron variants is low to non-existent within just weeks of Wildtype SARS-CoV-2 infection. Virus-specific T cells are detectable in most participants, albeit more variable than antibody responses. Cluster analyses of the co-evolution of antibody and T cell responses within individuals identify five distinct trajectories characterized by specific immune patterns and clinical factors. These findings demonstrate the relevant heterogeneity in humoral and cellular immunity to SARS-CoV-2 while also identifying consistent patterns where antibody and T cell responses may work in a compensatory manner to provide protection. The persistence of the immune response to SARS-CoV-2 after recovery from infection is an indicator for subsequent protection against infection. Here the authors follow recovered patients and measure antibody and T cell responses and find that these two parts of the immune response may have different longevity.
Collapse
Affiliation(s)
- Dominik Menges
- Epidemiology, Biostatistics and Prevention Institute (EBPI), University of Zurich (UZH), Zurich, Switzerland
| | - Kyra D Zens
- Epidemiology, Biostatistics and Prevention Institute (EBPI), University of Zurich (UZH), Zurich, Switzerland.,Institute for Experimental Immunology, University of Zurich (UZH), Zurich, Switzerland
| | - Tala Ballouz
- Epidemiology, Biostatistics and Prevention Institute (EBPI), University of Zurich (UZH), Zurich, Switzerland
| | - Nicole Caduff
- Epidemiology, Biostatistics and Prevention Institute (EBPI), University of Zurich (UZH), Zurich, Switzerland.,Institute for Experimental Immunology, University of Zurich (UZH), Zurich, Switzerland
| | - Daniel Llanas-Cornejo
- Epidemiology, Biostatistics and Prevention Institute (EBPI), University of Zurich (UZH), Zurich, Switzerland
| | - Hélène E Aschmann
- Epidemiology, Biostatistics and Prevention Institute (EBPI), University of Zurich (UZH), Zurich, Switzerland.,Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, USA
| | - Anja Domenghino
- Epidemiology, Biostatistics and Prevention Institute (EBPI), University of Zurich (UZH), Zurich, Switzerland.,Department of Visceral and Transplantation Surgery, University Hospital Zurich (USZ), University of Zurich (UZH), Zurich, Switzerland
| | - Céline Pellaton
- Service of Immunology and Allergy, Lausanne University Hospital (CHUV), University of Lausanne (UNIL), Lausanne, Switzerland
| | - Matthieu Perreau
- Service of Immunology and Allergy, Lausanne University Hospital (CHUV), University of Lausanne (UNIL), Lausanne, Switzerland
| | - Craig Fenwick
- Service of Immunology and Allergy, Lausanne University Hospital (CHUV), University of Lausanne (UNIL), Lausanne, Switzerland
| | - Giuseppe Pantaleo
- Service of Immunology and Allergy, Lausanne University Hospital (CHUV), University of Lausanne (UNIL), Lausanne, Switzerland
| | - Christian R Kahlert
- Division of Infectious Diseases and Hospital Epidemiology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland.,Division of Infectious Diseases and Hospital Epidemiology, Children's Hospital of Eastern Switzerland, St. Gallen, Switzerland
| | - Christian Münz
- Institute for Experimental Immunology, University of Zurich (UZH), Zurich, Switzerland
| | - Milo A Puhan
- Epidemiology, Biostatistics and Prevention Institute (EBPI), University of Zurich (UZH), Zurich, Switzerland.
| | - Jan S Fehr
- Epidemiology, Biostatistics and Prevention Institute (EBPI), University of Zurich (UZH), Zurich, Switzerland
| |
Collapse
|
185
|
Brehm J, Spaeth A, Dreßler L, Masetto T, Dannenberg R, Peter C, Grimmler M. SARS-CoV-2 antibody progression and neutralizing potential in mild symptomatic COVID-19 patients – a comparative long term post-infection study. Front Immunol 2022; 13:915338. [PMID: 36059441 PMCID: PMC9428854 DOI: 10.3389/fimmu.2022.915338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 07/26/2022] [Indexed: 01/08/2023] Open
Abstract
Background Since December 2019, SARS-CoV-2 has been keeping the world in suspense. Rapid tests, molecular diagnosis of acute infections, and vaccination campaigns with vaccines are building blocks of strategic pandemic control worldwide. For laboratory diagnostics, the quantification of the antibody titer of convalescents and vaccinated patients is thus increasingly coming to the fore. Methods Here we present an evaluation on the comparability of five serological tests on a cohort of 13 patients with mild COVID-19 disease. Also participants who were vaccinated after recovery were included in this study. All common immune methods (ELISA, CLIA, PETIA) and SARS-CoV-2 specific antigens (N-, S1- and RBD-) were specifically tracked and directly compared for up to 455 days. The titer of recovered participants was also set to the degree of symptoms during infection and the occurrence of Long-COVID. In addition, relative comparability of different serological tests, all standardized to WHO, was set in reference to the neutralizing potential of the corresponding participants. Findings The individual immune responses over 455 days after a mild SARS-CoV-2 infection remain stable, in contrast to vaccinated participants. All sero-tests reveal comparable performance and dynamics during the study and compared well to a surrogate neutralization test. Conclusion The information presented here will help clinicians in the daily laboratory work in the selection and evaluation of different serological tests offered. The data also will support in respect of a sero-test-based neutralization cutoff.
Collapse
Affiliation(s)
- Jessica Brehm
- MVZ Medizinische Labore Dessau Kassel GmbH, Dessau-Roßlau, Germany
| | - Alexander Spaeth
- MVZ Medizinische Labore Dessau Kassel GmbH, Dessau-Roßlau, Germany
| | - Lars Dreßler
- MVZ Medizinische Labore Dessau Kassel GmbH, Dessau-Roßlau, Germany
| | - Thomas Masetto
- Institute of Molecular Medicine I, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- DiaSys Diagnostic Systems GmbH, Holzheim, Germany
| | | | - Christoph Peter
- Institute of Molecular Medicine I, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Matthias Grimmler
- DiaSys Diagnostic Systems GmbH, Holzheim, Germany
- Hochschule Fresenius gGmbH, University of Applied Sciences, Idstein, Germany
- *Correspondence: Matthias Grimmler,
| |
Collapse
|
186
|
Arguni E, Dewi FST, Fachiroh J, Paramita DK, Lestari SK, Wiratama BS, Susilaningrum AR, Kharisma B, Meisyarah YH, Sari MP, Farahdilla ZA, Siswanto S, Sjaugi MF, Sasongko TH, Lazuardi L. Two-years antibody responses following SARS-CoV-2 infection in humans: A study protocol. PLoS One 2022; 17:e0272690. [PMID: 35972930 PMCID: PMC9380924 DOI: 10.1371/journal.pone.0272690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 07/25/2022] [Indexed: 01/08/2023] Open
Abstract
The long-term antibody response to the novel SARS-CoV-2 in infected patients and their residential neighborhood remains unknown in Indonesia. This information will provide insights into the antibody kinetics over a relatively long period as well as transmission risk factors in the community. We aim to prospectively observe and determine the kinetics of the anti-SARS-CoV-2 antibody for 2 years after infection in relation to disease severity and to determine the risk and protective factors of SARS CoV-2 infections in the community. A cohort of RT-PCR confirmed SARS-CoV-2 patients (case) will be prospectively followed for 2 years and will be compared to a control population. The control group comprises SARS-CoV-2 non-infected people who live within a one-kilometer radius from the corresponding case (location matching). This study will recruit at least 165 patients and 495 controls. Demographics, community variables, behavioral characteristics, and relevant clinical data will be collected. Serum samples taken at various time points will be tested for IgM anti-Spike protein of SARS-CoV-2 and IgG anti-Spike RBD of SARS-CoV-2 by using Chemiluminescent Microparticle Immunoassay (CMIA) method. The Kaplan-Meier method will be used to calculate cumulative seroconversion rates, and their association with disease severity will be estimated by logistic regression. The risk and protective factors associated with the SARS-CoV-2 infection will be determined using conditional (matched) logistic regression and presented as an odds ratio and 95% confidence interval.
Collapse
Affiliation(s)
- Eggi Arguni
- Department of Child Health, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
- Health and Demographic Surveillance System Sleman, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Fatwa Sari Tetra Dewi
- Health and Demographic Surveillance System Sleman, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
- Department of Health Behavior, Environment, and Social Medicine, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Jajah Fachiroh
- Health and Demographic Surveillance System Sleman, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
- Department of Histology and Cell Biology, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Dewi Kartikawati Paramita
- Department of Histology and Cell Biology, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Septi Kurnia Lestari
- Health and Demographic Surveillance System Sleman, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Bayu Satria Wiratama
- Department Biostatistics, Epidemiology and Population Health, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Annisa Ryan Susilaningrum
- Health and Demographic Surveillance System Sleman, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Bara Kharisma
- Health and Demographic Surveillance System Sleman, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Yogi Hasna Meisyarah
- Health and Demographic Surveillance System Sleman, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Merlinda Permata Sari
- Health and Demographic Surveillance System Sleman, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Zakiya Ammalia Farahdilla
- Health and Demographic Surveillance System Sleman, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Siswanto Siswanto
- Universitas Gadjah Mada Academic Hospital, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Muhammad Farhan Sjaugi
- Perdana University Graduate School of Medicine and Perdana University Center for Research Excellence, Kuala Lumpur, Malaysia
| | - Teguh Haryo Sasongko
- Department of Physiology, School of Medicine and Institute for Research, Development, and Innovation, International Medical University Kuala Lumpur, Malaysia
| | - Lutfan Lazuardi
- Department of Health Policy and Management, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
| |
Collapse
|
187
|
Udaondo C, Cámara C, Miguel Berenguel L, Alcobendas Rueda R, Muñoz Gómez C, Millán Longo C, Díaz-Delgado B, Falces-Romero I, Díaz Almirón M, Ochando J, Méndez-Echevarría A, Remesal Camba A, Calvo C. Humoral and cellular immune response to mRNA SARS-CoV-2 BNT162b2 vaccine in adolescents with rheumatic diseases. Pediatr Rheumatol Online J 2022; 20:64. [PMID: 35964130 PMCID: PMC9375068 DOI: 10.1186/s12969-022-00724-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 07/18/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Data about safety and efficacy of the mRNA SARS-CoV-2 vaccine in adolescents with rheumatic diseases (RD) is scarce and whether these patients generate a sufficient immune response to the vaccine remains an outstanding question. OBJECTIVE To evaluate safety and humoral and cellular immunity of the BNT162b2 vaccine in adolescents 12 to 18 years with RD and immunosuppressive treatment compared with a healthy control group. METHODS Adolescents from 12 to 18 years with RD followed at Hospital La Paz in Madrid (n = 40) receiving the BNT162b2 mRNA vaccination were assessed 3 weeks after complete vaccination. Healthy adolescents served as controls (n = 24). Humoral response was measured by IgG antiSpike antibodies, and cellular response by the quantity of IFN-γ and IL-2 present in whole blood stimulated with SARS-CoV-2 Spike and M proteins. RESULTS There were no differences in spike-specific humoral or cellular response between groups (median IFN-γ response to S specific protein; 528.80 pg/ml in controls vs. 398.44 in RD patients, p 0.78, and median IL-2 response in controls: 635.68 pg/ml vs. 497.30 in RD patients, p 0.22. The most frequent diagnosis was juvenile idiopathic arthritis (26/40, 65%) followed by Lupus (6/40, 15%). 60% of cases (23/40) received TNF inhibitors and 35% (14/40) methotrexate. 40% of patients (26/64) had previous SARS-CoV-2 infection, 9 in the control group and 17 in the RD patients without differences. Of note, 70% of infections were asymptomatic. A higher IFN-γ production was found in COVID-19 recovered individuals than in naive subjects in both groups (controls: median 859 pg/ml in recovered patients vs. 450 in naïve p 0.017, and RD patients: 850 in recovered vs. 278 in naïve p 0.024). No serious adverse events or flares were reported following vaccination. CONCLUSIONS We conclude that standard of care treatment for adolescents with RD including TNF inhibitors and methotrexate did not affect the humoral and the cellular immunity to BNT162b2 mRNA vaccination compared to a healthy control group. The previous contact with SARS-CoV-2 was the most relevant factor in the immune response.
Collapse
Affiliation(s)
- Clara Udaondo
- Paediatric Rheumatology Unit, Hospital Infantil La Paz, Paseo de la Castellana 261, 28046, Madrid, Spain.
- La Paz Institute of Biomedical Research (IdiPAZ), 28046, Madrid, Spain.
- CIBERINFEC, Instituto de Salud Carlos III (ISCIII), Madrid, Spain.
| | - Carmen Cámara
- La Paz Institute of Biomedical Research (IdiPAZ), 28046, Madrid, Spain
- Department of Immunology, Hospital La Paz, 28046, Madrid, Spain
| | | | - Rosa Alcobendas Rueda
- Paediatric Rheumatology Unit, Hospital Infantil La Paz, Paseo de la Castellana 261, 28046, Madrid, Spain
| | - Celia Muñoz Gómez
- Paediatric Rheumatology Unit, Hospital Infantil La Paz, Paseo de la Castellana 261, 28046, Madrid, Spain
| | - Claudia Millán Longo
- Paediatric Rheumatology Unit, Hospital Infantil La Paz, Paseo de la Castellana 261, 28046, Madrid, Spain
| | - Blanca Díaz-Delgado
- Paediatric Rheumatology Unit, Hospital Infantil La Paz, Paseo de la Castellana 261, 28046, Madrid, Spain
| | - Iker Falces-Romero
- La Paz Institute of Biomedical Research (IdiPAZ), 28046, Madrid, Spain
- CIBERINFEC, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Microbiology and Parasitology Department, Hospital La Paz, 28046, Madrid, Spain
| | - Mariana Díaz Almirón
- La Paz Institute of Biomedical Research (IdiPAZ), 28046, Madrid, Spain
- Biostatistics, Hospital La Paz, 28046, Madrid, Spain
| | - Jordi Ochando
- National Microbiology Centre, Instituto de Salud Carlos III, 28220, Madrid, Spain
| | - Ana Méndez-Echevarría
- La Paz Institute of Biomedical Research (IdiPAZ), 28046, Madrid, Spain
- CIBERINFEC, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Paediatric and Infectious Diseases Department, Hospital La Paz, 28046, Madrid, Spain
- Paediatric Translational Network in Infectious Diseases (RITIP), Madrid, Spain
| | - Agustín Remesal Camba
- Paediatric Rheumatology Unit, Hospital Infantil La Paz, Paseo de la Castellana 261, 28046, Madrid, Spain
| | - Cristina Calvo
- La Paz Institute of Biomedical Research (IdiPAZ), 28046, Madrid, Spain
- CIBERINFEC, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Paediatric and Infectious Diseases Department, Hospital La Paz, 28046, Madrid, Spain
- Paediatric Translational Network in Infectious Diseases (RITIP), Madrid, Spain
| |
Collapse
|
188
|
Alasmari F, Mukahal M, Alqurashi AA, Huq M, Alabdrabalnabi F, AlJurayyan A, Alkahtani SM, Assari FS, Bashaweeh R, Salam R, Aldera S, Alkinani OM, Almutairi T, AlEnizi K, Tleyjeh I. Seroprevalence and longevity of SARS-CoV-2 nucleocapsid antigen-IgG among health care workers in a large COVID-19 public hospital in Saudi Arabia: A prospective cohort study. PLoS One 2022; 17:e0272818. [PMID: 35960736 PMCID: PMC9374211 DOI: 10.1371/journal.pone.0272818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 07/26/2022] [Indexed: 11/18/2022] Open
Abstract
Seroprevalence of SARS-CoV-2 IgG among health care workers (HCWs) is crucial to inform infection control programs. Conflicting reports have emerged on the longevity of SARS-CoV-2 IgG. Our objective is to describe the prevalence of SARS-CoV-2 IgG in HCWs and perform 8 months longitudinal follow-up (FU) to assess the duration of detectable IgG. In addition, we aim to explore the risk factors associated with positive SARS-CoV-2 IgG. The study was conducted at a large COVID-19 public hospital in Riyadh, Saudi Arabia. All HCWs were recruited by social media platform. The SARS-CoV-2 IgG assay against SARS-CoV-2 nucleocapsid antigen was used. Multivariable logistic regression was used to examine association between IgG seropositive status and clinical and epidemiological factors. A total of 2528 (33% of the 7737 eligible HCWs) participated in the survey and 2523 underwent baseline serological testing in June 2020. The largest occupation groups sampled were nurses [n = 1351(18%)], physicians [n = 456 (6%)], administrators [n = 277 (3.6%)], allied HCWs [n = 205(3%)], pharmacists [n = 95(1.2%)], respiratory therapists [n = 40(0.5%)], infection control staff [n = 21(0.27%], and others [n = 83 (1%)]. The total cohort median age was 36 (31-43) years and 66.3% were females. 273 were IgG seropositive at baseline with a seroprevalence of 10.8% 95% CI (9.6%-12.1%). 165/185 and 44/112 were persistently IgG positive, at 2-3 months and 6 months FU respectively. The median (25th- 75th percentile) IgG level at the 3 different time points was 5.86 (3.57-7.04), 3.91 (2.46-5.38), 2.52 (1.80-3.99) respectively. Respiratory therapists OR 2.38, (P = 0.035), and those with hypertension OR = 1.86, (P = 0.009) were more likely to be seropositive. A high proportion of seropositive staff had prior symptoms 214/273(78%), prior anosmia was associated with the presence of antibodies, with an odds ratio of 9.25 (P<0.001), as well as fever and cough. Being a non-smoker, non-Saudi, and previously diagnosed with COVID-19 infection by PCR were statistically significantly different by seroprevalence status. We found that the seroprevalence of IgG against SARS-CoV-2 nucleocapsid antigen was 10.8% in HCWs at the peak of the pandemic in Saudi Arabia. We also observed a decreasing temporal trend of IgG seropositivity over 8 months follow up period.
Collapse
Affiliation(s)
- Faisal Alasmari
- Infection Control and Environmental Health Administration, King Fahad Medical City, Riyadh, Saudi Arabia
- Infectious Diseases Section, King Fahad Medical City, Riyadh, Saudi Arabia; College of Medicine, Al Faisal University, Riyadh, Saudi Arabia
| | - Mahmoud Mukahal
- Infection Control and Environmental Health Administration, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Alaa Ashraf Alqurashi
- Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Australia
| | - Molla Huq
- Immunology and Serology Laboratory, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Fatima Alabdrabalnabi
- Infection Control and Environmental Health Administration, King Fahad Medical City, Riyadh, Saudi Arabia
| | | | | | | | - Rahaf Bashaweeh
- Public Health College, Saudi Electronic University, Riyadh, Saudi Arabia
| | - Rana Salam
- Infectious Diseases Section, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Solaf Aldera
- Infection Control and Environmental Health Administration, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Ohud Mohammed Alkinani
- Pathology and Clinical Laboratory Administration, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Talal Almutairi
- Radiology Service Administration, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Kholoud AlEnizi
- Research Center, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Imad Tleyjeh
- Infectious Diseases Section, King Fahad Medical City, Riyadh, Saudi Arabia; College of Medicine, Al Faisal University, Riyadh, Saudi Arabia
| |
Collapse
|
189
|
SARS-CoV-2 antibody persistence after five and twelve months: A cohort study from South-Eastern Norway. PLoS One 2022; 17:e0264667. [PMID: 35947589 PMCID: PMC9365168 DOI: 10.1371/journal.pone.0264667] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 07/27/2022] [Indexed: 11/20/2022] Open
Abstract
Objectives To assess total antibody levels against Severe Acute Respiratory Syndrome CoronaVirus 2 (SARS CoV-2) spike protein up to 12 months after Coronavirus Disease (COVID-19) infection in non-vaccinated individuals and the possible predictors of antibody persistence. Methods This is the first part of a prospective multi-centre cohort study. Participants The study included SARS-CoV-2 real-time polymerase chain reaction (RT-PCR) positive and negative participants in South-Eastern Norway from February to December 2020. Possible predictors of SARS-CoV-2 total antibody persistence was assessed. The SARS-CoV-2 total antibody levels against spike protein were measured three to five months after PCR in 391 PCR-positive and 703 PCR-negative participants; 212 PCR-positive participants were included in follow-up measurements at 10 to 12 months. The participants completed a questionnaire including information about symptoms, comorbidities, allergies, body mass index (BMI), and hospitalisation. Primary outcome The SARS-CoV-2 total antibody levels against spike protein three to five and 10 to 12 months after PCR positive tests. Results SARS-CoV-2 total antibodies against spike protein were present in 366 (94%) non-vaccinated PCR-positive participants after three to five months, compared with nine (1%) PCR-negative participants. After 10 to 12 months, antibodies were present in 204 (96%) non-vaccinated PCR-positive participants. Of the PCR-positive participants, 369 (94%) were not hospitalised. The mean age of the PCR-positive participants was 48 years (SD 15, range 20–85) and 50% of them were male. BMI ≥ 25 kg/m2 was positively associated with decreased antibody levels (OR 2.34, 95% CI 1.06 to 5.42). Participants with higher age and self-reported initial fever with chills or sweating were less likely to have decreased antibody levels (age: OR 0.97, 95% CI 0.94 to 0.99; fever: OR 0.33, 95% CI 0.13 to 0.75). Conclusion Our results indicate that the level of SARS-CoV-2 total antibodies against spike protein persists for the vast majority of non-vaccinated PCR-positive persons at least 10 to 12 months after mild COVID-19.
Collapse
|
190
|
Oâ Connor MA, Hawman DW, Meade-White K, Leventhal S, Song W, Randall S, Archer J, Lewis TB, Brown B, Iwayama N, Ahrens C, Garrison W, Wangari S, Guerriero KA, Hanley P, Lovaglio J, Saturday G, Edlefsen PT, Khandhar A, Feldmann H, Fuller DH, Erasmus JH. A replicon RNA vaccine induces durable protective immunity from SARS-CoV-2 in nonhuman primates after neutralizing antibodies have waned. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022:2022.08.08.503239. [PMID: 35982677 PMCID: PMC9387133 DOI: 10.1101/2022.08.08.503239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
The global SARS-CoV-2 pandemic prompted rapid development of COVID-19 vaccines. Although several vaccines have received emergency approval through various public health agencies, the SARS-CoV-2 pandemic continues. Emergent variants of concern, waning immunity in the vaccinated, evidence that vaccines may not prevent transmission and inequity in vaccine distribution have driven continued development of vaccines against SARS-CoV-2 to address these public health needs. In this report, we evaluated a novel self-amplifying replicon RNA vaccine against SARS-CoV-2 in a pigtail macaque model of COVID-19 disease. We found that this vaccine elicited strong binding and neutralizing antibody responses. While binding antibody responses were sustained, neutralizing antibody waned to undetectable levels after six months but were rapidly recalled and conferred protection from disease when the animals were challenged 7 months after vaccination as evident by reduced viral replication and pathology in the lower respiratory tract, reduced viral shedding in the nasal cavity and lower concentrations of pro-inflammatory cytokines in the lung. Cumulatively, our data demonstrate in pigtail macaques that a self-amplifying replicon RNA vaccine can elicit durable and protective immunity to SARS-CoV-2 infection. Furthermore, these data provide evidence that this vaccine can provide durable protective efficacy and reduce viral shedding even after neutralizing antibody responses have waned to undetectable levels.
Collapse
|
191
|
Kim Y, Bae JY, Kwon K, Chang HH, Lee WK, Park H, Kim J, Choi I, Park MS, Kim SW. Kinetics of neutralizing antibodies against SARS-CoV-2 infection according to sex, age, and disease severity. Sci Rep 2022; 12:13491. [PMID: 35931794 PMCID: PMC9356129 DOI: 10.1038/s41598-022-17605-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 07/28/2022] [Indexed: 11/09/2022] Open
Abstract
Knowledge of the factors affecting the difference in kinetics and longevity of the neutralizing antibody (nAb) response to SARS-CoV-2 is necessary to properly prioritize vaccination. In the present study, from March to December 2020, of the 143 patients who recovered from COVID-19, 87 underwent study visits scheduled every 3 months. Patient demographics and blood samples were collected followed by a plaque reduction neutralization test to analyze nAb titers. A linear mixed model was used to compare the effects of sex, age, and disease severity over time. Results demonstrated a gradual reduction in nAb titers over time with a significant decrease from 6 to 9 months post-COVID-19 infection (p < 0.001). In time-to-sex, age, and disease severity comparisons, reduction in nAb titers over time was unaffected by sex (p = 0.167), age (p = 0.188), or disease severity (p = 0.081). Additionally, the nAb titer was 1.46 times significantly higher in those aged ≥ 50 years than in those aged < 50 years (p = 0.036) irrespective of time Moreover, the nAb titer was 2.41 times higher in the moderate or above than that in the below moderate disease severity group (p < 0.001). However, no significant differences were observed in terms of sex (p = 0.300). Given the reduction in nAbs over time, maintaining protective neutralizing antibodies regardless of sex, age, or disease severity is needed.
Collapse
Affiliation(s)
- Yoonjung Kim
- Department of Internal Medicine, Kyungpook National University Hospital, School of Medicine, Kyungpook National University, 130, Dongdeok-ro, Jung-gu, Daegu, 41944, Republic of Korea
| | - Joon-Yong Bae
- Department of Microbiology, Institute for Viral Diseases, Biosafety center, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Kitae Kwon
- Department of Internal Medicine, Kyungpook National University Hospital, School of Medicine, Kyungpook National University, 130, Dongdeok-ro, Jung-gu, Daegu, 41944, Republic of Korea
| | - Hyun-Ha Chang
- Department of Internal Medicine, Kyungpook National University Hospital, School of Medicine, Kyungpook National University, 130, Dongdeok-ro, Jung-gu, Daegu, 41944, Republic of Korea
| | - Won Kee Lee
- Department of Medical Informatics, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Heedo Park
- Department of Microbiology, Institute for Viral Diseases, Biosafety center, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Jeonghun Kim
- Department of Microbiology, Institute for Viral Diseases, Biosafety center, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Isaac Choi
- Department of Microbiology, Institute for Viral Diseases, Biosafety center, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Man-Seong Park
- Department of Microbiology, Institute for Viral Diseases, Biosafety center, College of Medicine, Korea University, Seoul, Republic of Korea.
| | - Shin-Woo Kim
- Department of Internal Medicine, Kyungpook National University Hospital, School of Medicine, Kyungpook National University, 130, Dongdeok-ro, Jung-gu, Daegu, 41944, Republic of Korea.
| |
Collapse
|
192
|
Sharif N, Alzahrani KJ, Ahmed SN, Khan A, Banjer HJ, Alzahrani FM, Parvez AK, Dey SK. Genomic surveillance, evolution and global transmission of SARS-CoV-2 during 2019-2022. PLoS One 2022; 17:e0271074. [PMID: 35913920 PMCID: PMC9342790 DOI: 10.1371/journal.pone.0271074] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 06/23/2022] [Indexed: 12/19/2022] Open
Abstract
In spite of the availability of vaccine, the health burden associated with the COVID-19 pandemic continues to increase. An estimated 5 million people have died with SARS-CoV-2 infection. Analysis of evolution and genomic diversity can provide sufficient information to reduce the health burden of the pandemic. This study focused to conduct worldwide genomic surveillance. About 7.6 million genomic data were analyzed during 2019 to 2022. Multiple sequence alignment was conducted by using maximum likelihood method. Clade GK (52%) was the most predominant followed by GRY (12%), GRA (11%), GR (8%), GH (7%), G (6%), GV (3%), and O (1%), respectively. VOC Delta (66%) was the most prevalent variant followed by VOC Alpha (18%), VOC Omicron (13%), VOC Gamma (2%) and VOC Beta (1%), respectively. The frequency of point mutations including E484K, N501Y, N439K, and L452R at spike protein has increased 10%-92%. Evolutionary rate of the variants was 23.7 substitution per site per year. Substitution mutations E484K and N501Y had significant correlation with cases (r = .45, r = .23), fatalities (r = .15, r = .44) and growth rate R0 (r = .28, r = .54). This study will help to understand the genomic diversity, evolution and the impact of the variants on the outcome of the COVID-19 pandemic.
Collapse
Affiliation(s)
- Nadim Sharif
- Department of Microbiology, Jahangirnagar University, Savar, Dhaka, Bangladesh
| | - Khalid J. Alzahrani
- Department of Clinical Laboratories Sciences, College of Applied Medical Sciences, Taif University, Taif, Saudi Arabia
| | - Shamsun Nahar Ahmed
- Department of Microbiology, Jahangirnagar University, Savar, Dhaka, Bangladesh
| | - Afsana Khan
- Department of Statistics, Jahangirnagar University, Savar, Dhaka, Bangladesh
| | - Hamsa Jameel Banjer
- Department of Clinical Laboratories Sciences, College of Applied Medical Sciences, Taif University, Taif, Saudi Arabia
| | - Fuad M. Alzahrani
- Department of Clinical Laboratories Sciences, College of Applied Medical Sciences, Taif University, Taif, Saudi Arabia
| | | | - Shuvra Kanti Dey
- Department of Microbiology, Jahangirnagar University, Savar, Dhaka, Bangladesh
- * E-mail:
| |
Collapse
|
193
|
Gentile A, Castellano VE, Pacchiotti A, Weinberger N, Diana Menéndez S, Del Pino M, Carciofi G, Lamy P, Mistchenko AS. Long-term antibody response following SPUTNIK V primary vaccination in healthcare workers with and without history of SARS-CoV-2 infection: Prospective cohort study from a hospital in Argentina. Vaccine X 2022; 11:100187. [PMID: 35755140 PMCID: PMC9213035 DOI: 10.1016/j.jvacx.2022.100187] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 06/07/2022] [Accepted: 06/08/2022] [Indexed: 01/06/2023] Open
Abstract
After completing a two-dose Sputnik V schedule, all vaccinees developed antibodies. Prior infection was associated with higher antibody concentrations. Antibodies decreased significantly after 6 months in naïve subjects. In subjects without infection, 17% were negative at day 180. SARS-CoV2 infection after vaccination increased antibody concentrations.
Background In December 2020, Sputnik V was incorporated to the National COVID-19 Immunization Plan in Argentina. Studies had shown 98% of antibody response rate. To date, data on immunogenicity and antibody persistence in Argentina are scarce. The objective was to assess humoral immune response after two doses of Sputnik V in Health Care Workers (HCWs) at the Ricardo Gutierrez Children’s Hospital (RGCH). Methods A prospective, cohort study in HCWs immunized with two doses of Sputnik V between February and March 2021. The following variables were assessed: age, gender, risk factors for severe COVID-19 or mortality, immunosuppressive therapy and history of SARS-CoV-2. Blood samples were drawn on the day of the first dose, 28 days and 180 days after the second. Anti-Spike IgG was measured using an ELISA assay. Differences in immune response were evaluated according to study variables. Comparison analyses between groups with or without history of infection were performed, with T-test and ANOVA or Mann-Whitney tests. For each subject, we compared baseline values with 28 days and 180 days after the second vaccine. STATA version 14 and R Sofware were used for data analyses. Results We included 528 individuals, mean age 41.5 years, 82.9% female, 14.4% (76/528) reported previous SARS-CoV-2 infection. All subjects developed antibodies post-vaccination. At day 28, concentrations were significantly higher in previously infected than naïve subjects (p < 0.001) with no differences according to age, gender and comorbidities. At day 180, 17% (95% CI 13.17–21.53) of naïve subjects were negative. Antibody concentrations decreased significantly in all subjects except in those who reported SARS-CoV-2 infection after vaccination (n = 31). This last group had significantly higher antibody concentrations. Conclusion This study assessed immune response to a new COVID-19 vaccine in real life in a cohort of subjects. Antibody concentrations varied according to history of SARS-COV-2 infection and decreased over time.
Collapse
Affiliation(s)
- A Gentile
- Epidemiology, Ricardo Gutierrez Children's Hospital, Gallo 1330, Buenos Aires City, Argentina
| | - V E Castellano
- Epidemiology, Ricardo Gutierrez Children's Hospital, Gallo 1330, Buenos Aires City, Argentina
| | - A Pacchiotti
- Epidemiology, Ricardo Gutierrez Children's Hospital, Gallo 1330, Buenos Aires City, Argentina
| | - N Weinberger
- Department of Virology, Ricardo Gutierrez Children's Hospital, Gallo 1330, Buenos Aires City, Argentina
| | - S Diana Menéndez
- Epidemiology, Ricardo Gutierrez Children's Hospital, Gallo 1330, Buenos Aires City, Argentina
| | - M Del Pino
- Epidemiology, Ricardo Gutierrez Children's Hospital, Gallo 1330, Buenos Aires City, Argentina
| | - G Carciofi
- Department of Virology, Ricardo Gutierrez Children's Hospital, Gallo 1330, Buenos Aires City, Argentina
| | - P Lamy
- Epidemiology, Ricardo Gutierrez Children's Hospital, Gallo 1330, Buenos Aires City, Argentina
| | - A S Mistchenko
- Commission of Scientific Investigations of the Province of Buenos Aires, Calle 526, La Plata, Buenos Aires Province, Argentina
| |
Collapse
|
194
|
Mastroianni F, Guida P, Bellanova G, Valentina De Nicolò E, Righetti G, Formoso M, Celani F. SARS-CoV-2 antibody response after BNT162b2 mRNA vaccine in healthcare workers: Nine-month of follow-up. Vaccine X 2022; 11:100175. [PMID: 35692461 PMCID: PMC9170276 DOI: 10.1016/j.jvacx.2022.100175] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 05/10/2022] [Accepted: 06/02/2022] [Indexed: 11/25/2022] Open
Abstract
We collected sequential serum samples (0, 4, 12 weeks, 9 months) for the determination of S-RDB IgG levels from 103 vaccinated healthy subjects (age 45 ± 13 years; 60 women), in order to evaluate neutralizing antibody response against SARS-CoV-2 in healthy healthcare workers (HCWs) after the administration of two doses of BNT162b2 SARS-CoV-2 mRNA vaccine. Every subject received two doses of mRNA vaccine BNT162b2 (Pfizer-BioNTech), 21 days apart (January-February 2021). Furthermore, antibody titer of 14 subjects who were hospitalized for symptomatic COVID-19 was evaluated. Antibody response was (median, interquartile range) 35 U/mL (10-104) at baseline, 1960 (1241-3221) at 4 weeks, 791 (388-1179) at 12 weeks and 524 (273-931) at 6 months. Antibody response was inversely correlated with age at all timepoints (p < 0.001) while gender and Body Mass Index had no significant effect. At multivariate analysis, post-baseline values were significantly higher than baseline (p < 0.001) with a reduction at 12 weeks and 9 months (p < 0.001). Antibody response of hospitalized subjects who did not receive vaccination, symptomatic for COVID 19 infection, was 103 (25-557) U/mL, significantly higher than baseline (p = 0.007) of study population but lower than all post-baseline determinations (p < 0.001). Younger subjects showed a stronger response and a lower decrease of antibody titers compared to the classes of older subjects. SARS-CoV2 infection was excluded by performing 1017 nasopharyngeal RT-PCR swabs on the study cohort. The second dose of mRNA vaccine resulted in an antibody response effective in preventing infection in a population of healthcare professionals. The antibody level was stable through week 12, showing a reduction in the following six months.
Collapse
Affiliation(s)
- Franco Mastroianni
- Regional General Hospital “F. Miulli”, Acquaviva delle Fonti, Bari, Italy
| | - Pietro Guida
- Regional General Hospital “F. Miulli”, Acquaviva delle Fonti, Bari, Italy
| | - Grazia Bellanova
- Regional General Hospital “F. Miulli”, Acquaviva delle Fonti, Bari, Italy
| | | | - Giulia Righetti
- Regional General Hospital “F. Miulli”, Acquaviva delle Fonti, Bari, Italy
| | - Maurizio Formoso
- Regional General Hospital “F. Miulli”, Acquaviva delle Fonti, Bari, Italy
| | - Fabrizio Celani
- Regional General Hospital “F. Miulli”, Acquaviva delle Fonti, Bari, Italy
| |
Collapse
|
195
|
Hanssen DAT, Penders J, Heijgele K, de Leede S, Mulder M, Bank LEA, Slaats MHC, Savelkoul PHM, van Loo IHM. Antibodies against SARS-CoV-2 after natural infection in healthcare workers and clinical characteristics as putative antibody production prediction. JOURNAL OF CLINICAL VIROLOGY PLUS 2022; 2:100089. [PMID: 35755211 PMCID: PMC9213037 DOI: 10.1016/j.jcvp.2022.100089] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 05/21/2022] [Accepted: 06/06/2022] [Indexed: 12/03/2022] Open
Abstract
Introduction There is a need for detailed data on early antibody responses against SARS-CoV-2 as this may contribute to the prediction of the clinical course of COVID-19 and the optimization of convalescent plasma treatment. This study aims to gain insight into developing antibodies to SARS-CoV-2 in health care workers (HCWs) infected in the first wave of the SARS-CoV-2 pandemic in the Netherlands. Materials and methods In this retrospective analysis, sera from PCR-confirmed COVID-19 positive HCWs are included at the time of the initial PCR (T = 0, n = 95) and at least 21 days after the initial serum (T ≥ 21, n = 133). This study assesses correlations between qualitative total Ig, IgM, IgA, IgG, and quantitative anti-S-RBD antibody responses and participant characteristics. Results Higher Ct values were associated with higher antibody positivity rates for total Ig (OR 1.261 (95% CI 1.095–1.452)), IgM (OR 1.373 (95% CI 1.125–1.675)), and IgA (OR 1.222 (95% CI 1.013–1.475)). Gender was predictive of IgM and IgA antibody positivity rates at T = 0 (OR 0.018 (95% CI 0.001–0.268)) and (OR 0.070 (95% CI 0.008–0.646)). At T ≥ 21, a substantial proportion of HCWs developed IgM (103/133; 77.4%) and total Ig (128/133; 96.2%) antibodies. IgA and IgG seroconversions were observed in only 51.1% (67/131) and 55.7% (73/131) of HCWs. Anti-S-RBD responses were higher when the interval between onset of symptoms and sampling was longer. Conclusion The findings of this study give insight into early antibody responses and may have implications for the selection of convalescent plasma donors and the further development of monoclonal antibody treatment.
Collapse
Affiliation(s)
- D A T Hanssen
- Department of Medical Microbiology, infectious diseases & Infection prevention, Maastricht University Medical Center, the Netherlands
- Care and Primary Health Research Institute (CAPHRI), Maastricht University, the Netherlands
| | - J Penders
- Department of Medical Microbiology, infectious diseases & Infection prevention, Maastricht University Medical Center, the Netherlands
- Care and Primary Health Research Institute (CAPHRI), Maastricht University, the Netherlands
- NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, the Netherlands
| | - K Heijgele
- Department of Medical Microbiology, infectious diseases & Infection prevention, Maastricht University Medical Center, the Netherlands
| | - S de Leede
- Department of Medical Microbiology, infectious diseases & Infection prevention, Maastricht University Medical Center, the Netherlands
| | - M Mulder
- Department of Medical Microbiology, infectious diseases & Infection prevention, Maastricht University Medical Center, the Netherlands
| | - L E A Bank
- Department of Medical Microbiology, infectious diseases & Infection prevention, Maastricht University Medical Center, the Netherlands
| | - M H C Slaats
- Department of Medical Microbiology, infectious diseases & Infection prevention, Maastricht University Medical Center, the Netherlands
| | - P H M Savelkoul
- Department of Medical Microbiology, infectious diseases & Infection prevention, Maastricht University Medical Center, the Netherlands
- Care and Primary Health Research Institute (CAPHRI), Maastricht University, the Netherlands
| | - I H M van Loo
- Department of Medical Microbiology, infectious diseases & Infection prevention, Maastricht University Medical Center, the Netherlands
- Care and Primary Health Research Institute (CAPHRI), Maastricht University, the Netherlands
| |
Collapse
|
196
|
Khairullin B, Zakarya K, Orynbayev M, Abduraimov Y, Kassenov M, Sarsenbayeva G, Sultankulova K, Chervyakova O, Myrzakhmetova B, Nakhanov A, Nurpeisova A, Zhugunissov K, Assanzhanova N, Nurabayev S, Kerimbayev A, Yershebulov Z, Burashev Y, Kulmagambetov I, Davlyatshin T, Sergeeva M, Buzitskaya Z, Stukova M, Kutumbetov L. Efficacy and safety of an inactivated whole-virion vaccine against COVID-19, QazCovid-in®, in healthy adults: A multicentre, randomised, single-blind, placebo-controlled phase 3 clinical trial with a 6-month follow-up. EClinicalMedicine 2022; 50:101526. [PMID: 35770251 PMCID: PMC9233449 DOI: 10.1016/j.eclinm.2022.101526] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 05/16/2022] [Accepted: 06/01/2022] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Vaccination remains the primary measure to prevent the spread of the SARS-CoV-2 virus, further necessitating the use of effective licensed vaccines. METHODS From Dec 25, 2020, to July 11, 2021, we conducted a multicenter, randomised, single-blind, placebo-controlled phase 3 efficacy trial of the QazCovid-in® vaccine with a 180-day follow-up period in three clinical centres in Kazakhstan. A total of 3000 eligible participants aged 18 years or older were randomly assigned (4:1) to receive two doses of the vaccine (5 μg each, 21 days apart) or placebo administered intramuscularly. QazCovid-in® is a whole-virion formaldehyde-inactivated anti-COVID-19 vaccine, adjuvanted with aluminium hydroxide. The primary endpoint was the incidence of symptomatic cases of the SARS-CoV-2 infection confirmed by RT-PCR starting from day 14 after the first immunisation. The trial was registered with ClinicalTrials.gov NCT04691908. FINDINGS The QazCovid-in® vaccine was safe over the 6-month monitoring period after two intramuscular immunisations inducing only local short-lived adverse events. The concomitant diseases of participants did not affect the vaccine safety. Out of 2400 vaccinated participants, 31 were diagnosed with COVID-19; 43 COVID-19 cases were recorded in 600 placebo participants with onset of 14 days after the first dose within the 180-day observation period. Only one severe COVID-19 case was identified in a vaccine recipient with a comorbid chronic heart failure. The protective efficacy of the QazCovid-in® vaccine reached 82·0% (95% CI 71.1-88.5) within the 180-day observation period. INTERPRETATION Two immunisations with the inactivated QazCovid-in® vaccine achieved 82·0% (95% CI 71.1-88.5) protective efficacy against COVID-19 within a 180-day follow-up period. FUNDING The work was funded by the Science Committee of the Ministry of Education and Science of Kazakhstan within the framework of the Scientific and Technical Program "Development of a vaccine against coronavirus infection COVID-19". State registration number 0.0927.
Collapse
Affiliation(s)
- Berik Khairullin
- Research Institute of Biological Safety Problems of the Science Committee of the Ministry of Education and Science of the Republic of Kazakhstan, Gvardeysk, Kazakhstan
- Corresponding author at: Research Institute of Biological Safety Problems of the Science Committee of the Ministry of Education and Science of the Republic of Kazakhstan, Gvardeysk, 080409, Kazakhstan.
| | - Kunsulu Zakarya
- Research Institute of Biological Safety Problems of the Science Committee of the Ministry of Education and Science of the Republic of Kazakhstan, Gvardeysk, Kazakhstan
| | - Mukhit Orynbayev
- Research Institute of Biological Safety Problems of the Science Committee of the Ministry of Education and Science of the Republic of Kazakhstan, Gvardeysk, Kazakhstan
| | - Yergali Abduraimov
- Research Institute of Biological Safety Problems of the Science Committee of the Ministry of Education and Science of the Republic of Kazakhstan, Gvardeysk, Kazakhstan
| | - Markhabat Kassenov
- Research Institute of Biological Safety Problems of the Science Committee of the Ministry of Education and Science of the Republic of Kazakhstan, Gvardeysk, Kazakhstan
| | - Gulbanu Sarsenbayeva
- Research Institute of Biological Safety Problems of the Science Committee of the Ministry of Education and Science of the Republic of Kazakhstan, Gvardeysk, Kazakhstan
| | - Kulyaisan Sultankulova
- Research Institute of Biological Safety Problems of the Science Committee of the Ministry of Education and Science of the Republic of Kazakhstan, Gvardeysk, Kazakhstan
| | - Olga Chervyakova
- Research Institute of Biological Safety Problems of the Science Committee of the Ministry of Education and Science of the Republic of Kazakhstan, Gvardeysk, Kazakhstan
| | - Balzhan Myrzakhmetova
- Research Institute of Biological Safety Problems of the Science Committee of the Ministry of Education and Science of the Republic of Kazakhstan, Gvardeysk, Kazakhstan
| | - Aziz Nakhanov
- Research Institute of Biological Safety Problems of the Science Committee of the Ministry of Education and Science of the Republic of Kazakhstan, Gvardeysk, Kazakhstan
| | - Ainur Nurpeisova
- Research Institute of Biological Safety Problems of the Science Committee of the Ministry of Education and Science of the Republic of Kazakhstan, Gvardeysk, Kazakhstan
| | - Kuandyk Zhugunissov
- Research Institute of Biological Safety Problems of the Science Committee of the Ministry of Education and Science of the Republic of Kazakhstan, Gvardeysk, Kazakhstan
| | - Nurika Assanzhanova
- Research Institute of Biological Safety Problems of the Science Committee of the Ministry of Education and Science of the Republic of Kazakhstan, Gvardeysk, Kazakhstan
| | - Sergazy Nurabayev
- Research Institute of Biological Safety Problems of the Science Committee of the Ministry of Education and Science of the Republic of Kazakhstan, Gvardeysk, Kazakhstan
| | - Aslan Kerimbayev
- Research Institute of Biological Safety Problems of the Science Committee of the Ministry of Education and Science of the Republic of Kazakhstan, Gvardeysk, Kazakhstan
| | - Zakir Yershebulov
- Research Institute of Biological Safety Problems of the Science Committee of the Ministry of Education and Science of the Republic of Kazakhstan, Gvardeysk, Kazakhstan
| | - Yerbol Burashev
- Research Institute of Biological Safety Problems of the Science Committee of the Ministry of Education and Science of the Republic of Kazakhstan, Gvardeysk, Kazakhstan
| | | | | | - Maria Sergeeva
- Smorodintsev Research Institute of Influenza, Saint Petersburg, Russia
| | - Zhanna Buzitskaya
- Smorodintsev Research Institute of Influenza, Saint Petersburg, Russia
| | - Marina Stukova
- Smorodintsev Research Institute of Influenza, Saint Petersburg, Russia
| | - Lespek Kutumbetov
- Research Institute of Biological Safety Problems of the Science Committee of the Ministry of Education and Science of the Republic of Kazakhstan, Gvardeysk, Kazakhstan
| |
Collapse
|
197
|
Ochoa-Azze R, Chang-Monteagudo A, Climent-Ruiz Y, Macías-Abraham C, Valenzuela-Silva C, de Los Ángeles García-García M, Jerez-Barceló Y, Triana-Marrero Y, Ruiz-Villegas L, Dairon Rodríguez-Prieto L, Guerra-Chaviano PP, Sánchez-Ramírez B, Hernández-García T, Orosa-Vázquez I, Díaz-Hernández M, Chiodo F, Calcagno A, Ghisetti V, Rodríguez-Acosta M, Noa-Romero E, Enríquez-Puertas J, Ortega-León D, Valdivia-Álvarez I, Delahanty-Fernández A, Palenzuela-Díaz A, Rodríguez-Noda L, González-Mugica R, Valdés-Balbín Y, García-Rivera D, Verez-Bencomo V. Safety and immunogenicity of the FINLAY-FR-1A vaccine in COVID-19 convalescent participants: an open-label phase 2a and double-blind, randomised, placebo-controlled, phase 2b, seamless, clinical trial. THE LANCET. RESPIRATORY MEDICINE 2022; 10:785-795. [PMID: 35691295 PMCID: PMC9183216 DOI: 10.1016/s2213-2600(22)00100-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 03/15/2022] [Accepted: 03/15/2022] [Indexed: 12/26/2022]
Abstract
BACKGROUND A phase 1, clinical trial to evaluate FINLAY-FR-1A vaccine in COVID-19 convalescent individuals was completed. Here, we report results of the phase 2, clinical trial. METHODS We studied 450 convalescent participants with a history of asymptomatic, mild, or moderate COVID-19 at the National Institute of Hematology and Immunology and the National Centre for Sexual Education in Havana, Cuba. The study included adults aged 19-78 years who had recovered from COVID-19 and had had a negative PCR test at least 2 months before the initiation of the study. Phase 2 was done sequentially in two stages. The first stage to assess safety comprised an open, non-controlled phase 2a study in participants aged 60-78 years who received a single dose of the FINLAY-FR-1A vaccine (50 μg of recombinant dimeric receptor binding domain [RBD]). The second stage comprised the placebo-controlled, double-blind, phase 2b trial in participants aged 19-78 years, where participants were randomly assigned (4:1) into two groups: an experimental group vaccinated with a single dose of the FINLAY-FR-1A vaccine, and a control (placebo) group injected with vaccine excipient. The primary outcomes were safety, evaluated 28 days after vaccination by the occurrence of serious adverse events in all participants, and successful immune response, assessed by neutralising antibody ELISA, and defined as half-maximal surrogate virus neutralisation titres of 250 or more. Secondary endpoints included vaccine immunogenicity assessed by ELISA anti-RBD and live-virus neutralisation test. All randomly assigned participants were included in the safety analysis (safety population), and immunogenicity was evaluated in participants without study interruptions (per-protocol population). The trial is registered with the Cuban Public Registry of Clinical Trials, RPCEC00000366-En and WHO-ICTRP and is complete. FINDINGS From April 9, 2021, to April 17, 2021, 663 COVID-19 convalescent participants were enrolled in the study; 213 participants did not meet the selection criteria and 450 volunteers were recruited. 20 participants aged 60-78 years were included in the open, single-group, phase 2a study and 430 participants were randomly assigned to the experimental (n=344) or control groups (n=86) in the phase 2b study of participants aged 19-78 years. 19 (95%) of 20 phase 2a volunteers achieved a successful immune response after vaccination. No vaccine-associated serious adverse events were reported in the whole study population. Minor adverse events were found, the most common being pain at the injection site (105 [29%] of 364 in the intervention group; 13 [15%] of 86 in the placebo group). A successful immune response was found in 289 (81%) of 358 participants 28 days after vaccination. The vaccine elicited a greater than 31-times increase in anti-RBD-IgG antibodies compared with prevaccination rates, and the seroconversion rate was 302 (84%) of 358 on day 28 after vaccination; the geometric mean titres of live-virus neutralisation test increased from 15·4 (95% CI 10·3-23·2) to 400·3 (272·4-588·1) and high response was found against alpha, beta, and delta variants of concern. INTERPRETATION A single dose of the FINLAY-FR-1A vaccine against SARS-CoV-2 strengthened the pre-existing natural immunity, with excellent safety profile. FUNDING Cuba's Ministry of Science, Technology, and Environment.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Fabrizio Chiodo
- Finlay Vaccine Institute, Atabey, Playa, Havana, Cuba; Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit Amsterdam, Netherlands and Institute of Biomolecular Chemistry, National Research Council, Pozzuoli, Napoli, Italy
| | - Andrea Calcagno
- Unit of Infectious Diseases, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Valeria Ghisetti
- Laboratory of Microbiology and Virology, Amedeo di Savoia Hospital, ASL, Turin, Italy
| | | | - Enrique Noa-Romero
- Research Centre of Civil Defence, San José de las Lajas, Mayabeque, Cuba
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
198
|
Chitwood MH, Russi M, Gunasekera K, Havumaki J, Klaassen F, Pitzer VE, Salomon JA, Swartwood NA, Warren JL, Weinberger DM, Cohen T, Menzies NA. Reconstructing the course of the COVID-19 epidemic over 2020 for US states and counties: Results of a Bayesian evidence synthesis model. PLoS Comput Biol 2022; 18:e1010465. [PMID: 36040963 PMCID: PMC9467347 DOI: 10.1371/journal.pcbi.1010465] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 09/12/2022] [Accepted: 08/03/2022] [Indexed: 12/11/2022] Open
Abstract
Reported COVID-19 cases and deaths provide a delayed and incomplete picture of SARS-CoV-2 infections in the United States (US). Accurate estimates of both the timing and magnitude of infections are needed to characterize viral transmission dynamics and better understand COVID-19 disease burden. We estimated time trends in SARS-CoV-2 transmission and other COVID-19 outcomes for every county in the US, from the first reported COVID-19 case in January 13, 2020 through January 1, 2021. To do so we employed a Bayesian modeling approach that explicitly accounts for reporting delays and variation in case ascertainment, and generates daily estimates of incident SARS-CoV-2 infections on the basis of reported COVID-19 cases and deaths. The model is freely available as the covidestim R package. Nationally, we estimated there had been 49 million symptomatic COVID-19 cases and 404,214 COVID-19 deaths by the end of 2020, and that 28% of the US population had been infected. There was county-level variability in the timing and magnitude of incidence, with local epidemiological trends differing substantially from state or regional averages, leading to large differences in the estimated proportion of the population infected by the end of 2020. Our estimates of true COVID-19 related deaths are consistent with independent estimates of excess mortality, and our estimated trends in cumulative incidence of SARS-CoV-2 infection are consistent with trends in seroprevalence estimates from available antibody testing studies. Reconstructing the underlying incidence of SARS-CoV-2 infections across US counties allows for a more granular understanding of disease trends and the potential impact of epidemiological drivers.
Collapse
Affiliation(s)
- Melanie H Chitwood
- Department of Epidemiology of Microbial Diseases and Public Health Modeling Unit, Yale School of Public Health, Yale University, New Haven, Connecticut United States of America
| | - Marcus Russi
- Department of Epidemiology of Microbial Diseases and Public Health Modeling Unit, Yale School of Public Health, Yale University, New Haven, Connecticut United States of America
| | - Kenneth Gunasekera
- Department of Epidemiology of Microbial Diseases and Public Health Modeling Unit, Yale School of Public Health, Yale University, New Haven, Connecticut United States of America
| | - Joshua Havumaki
- Department of Epidemiology of Microbial Diseases and Public Health Modeling Unit, Yale School of Public Health, Yale University, New Haven, Connecticut United States of America
| | - Fayette Klaassen
- Department of Global Health and Population, Harvard T.H. Chan School of Public Health, Harvard University, Boston, Massachusetts United States of America
| | - Virginia E Pitzer
- Department of Epidemiology of Microbial Diseases and Public Health Modeling Unit, Yale School of Public Health, Yale University, New Haven, Connecticut United States of America
| | - Joshua A Salomon
- Department of Health Policy, Stanford University, Stanford, California United States of America
| | - Nicole A Swartwood
- Department of Global Health and Population, Harvard T.H. Chan School of Public Health, Harvard University, Boston, Massachusetts United States of America
| | - Joshua L Warren
- Department of Biostatistics and Public Health Modeling Unit, Yale School of Public Health, Yale University, New Haven, Connecticut United States of America
| | - Daniel M Weinberger
- Department of Epidemiology of Microbial Diseases and Public Health Modeling Unit, Yale School of Public Health, Yale University, New Haven, Connecticut United States of America
| | - Ted Cohen
- Department of Epidemiology of Microbial Diseases and Public Health Modeling Unit, Yale School of Public Health, Yale University, New Haven, Connecticut United States of America
| | - Nicolas A Menzies
- Department of Global Health and Population, Harvard T.H. Chan School of Public Health, Harvard University, Boston, Massachusetts United States of America
| |
Collapse
|
199
|
Korodi M, Horváth I, Rákosi K, Jenei Z, Hudák G, Kákes M, Dallos-Fejér K, Simai E, Páll O, Staver N, Briciu V, Lupșe M, Flonta M, Almaș A, Birlutiu V, Daniela Lupu C, Magdalena Ghibu A, Pianoschi D, Terza LM, Fejer SN. Longitudinal determination of BNT162b2 vaccine induced strongly binding SARS-CoV-2 IgG antibodies in a cohort of Romanian healthcare workers. Vaccine 2022; 40:5445-5451. [PMID: 35931634 PMCID: PMC9339977 DOI: 10.1016/j.vaccine.2022.07.040] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 06/22/2022] [Accepted: 07/26/2022] [Indexed: 12/09/2022]
Abstract
Mass vaccination against the disease caused by the novel coronavirus (COVID-19) was a crucial step in slowing the spread of SARS-CoV-2 in 2021. Even in the face of new variants, it still remains extremely important for reducing hospitalizations and COVID-19 deaths. In order to better understand the short- and long-term dynamics of humoral immune response, we present a longitudinal analysis of post-vaccination IgG levels in a cohort of 166 Romanian healthcare workers vaccinated with BNT162b2 with weekly follow-up until 35 days past the first dose and monthly follow-up up to 6 months post-vaccination. A subset of the patients continued with follow-up after 6 months and either received a booster dose or got infected during the Delta wave in Romania. Tests were carried out on 1694 samples using a CE-marked IgG ELISA assay developed in-house, containing S1 and N antigens of the wild type virus. Participants infected with SARS-CoV-2 before vaccination mount a quick immune response, reaching peak IgG levels two weeks after the first dose, while IgG levels of previously uninfected participants mount gradually, increasing abruptly after the second dose. Overall higher IgG levels are maintained for the previously infected group throughout the six month primary observation period (e.g. 36–65 days after the first dose, the median value in the previously infected group is 5.29 AU/ml, versus 3.58 AU/ml in the infection naïve group, p less than 0.001). The decrease of IgG levels is gradual, with lower median values in the infection naïve cohort even 7–8 months after vaccination, compared to the previously infected cohort (0.7 AU/ml versus 1.29 AU/ml, p = 0.006). Administration of a booster dose yielded higher median IgG antibody levels than post second dose in the infection naïve group and comparable levels in the previously infected group.
Collapse
|
200
|
Mehew J, Johnson R, Roberts D, Griffiths A, Harvala H. Convalescent plasma for COVID-19: Donor demographic factors associated high neutralising antibody titres. Transfus Med 2022; 32:327-337. [PMID: 35434868 PMCID: PMC9115414 DOI: 10.1111/tme.12868] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 01/27/2022] [Accepted: 04/03/2022] [Indexed: 01/08/2023]
Abstract
BACKGROUND Convalescent plasma containing high levels of SARS-CoV-2 antibodies has been studied as a possible treatment for COVID-19. Better understanding of predictors of high antibody levels is needed for improving supply of high-quality therapeutic plasma. AIMS We have evaluated demographic and clinical factors associated with the probability of a convalescent plasma donor having high SARS-CoV-2 IgG antibody levels. METHODS A total of 29,585 convalescent plasma donors employed during the first and second waves of the COVID-19 pandemic in England were included in this study. All had been tested for SARS-CoV-2 IgG antibodies by EUROimmun ELISA. A multivariable logistic regression model was used to quantify the association of the demographic and clinical factors with high (EUROimmun S/Co>6.0) SARS-CoV-2 IgG antibody level. RESULTS Most of the donors were male (23,024; 78%), with white ethnic background (24,598;83%) and had not been tested for SARS-CoV-2 (15,266; 52%).Overall, less than 20% of convalescent plasma donors with confirmed or suspected SARS-CoV-2 infection harboured high SARS-CoV-2 antibody levels (n = 4,978). We found that older male donors who had been hospitalised with COVID-19 were most likely to harbour high levels of antibodies. White donors were less likely to have high SARS-CoV-2 antibody levels than donors with Asian orblack ethnic backgrounds residing in affluent areas likely reflecting ethnic inequality previously associated with SARS-CoV-2 infection. DISCUSSION In a time of great uncertainty, and predicted new waves associated with newly emerging SARS-CoV-2 variants, these results will help us to target future convalescent plasma collections.
Collapse
Affiliation(s)
- Jennifer Mehew
- Statistics and Clinical Studies, NHS Blood and TransplantBristolUK
| | - Rachel Johnson
- Statistics and Clinical Studies, NHS Blood and TransplantBristolUK
| | - David Roberts
- Clinical, Research and Development, NHS Blood and TransplantOxfordUK
- Radcliffe Department of Medicine and BRC Haematology ThemeUniversity of Oxford, John Radcliffe HospitalOxfordUK
| | - Alex Griffiths
- Statistics and Clinical Studies, NHS Blood and TransplantBristolUK
| | - Heli Harvala
- Microbiology ServicesNHS Blood and TransplantLondonUK
| |
Collapse
|