1
|
Fraser DD, Singh D, Cela E, Patel MA, Assaf M, Quintero M, Knauer M, Miller MR, Bellini M, Li A, Hahn P, Hrek M, Cruz-Aguado J. Neutralizing antibodies to SARS-CoV-2 variants of concern: a pediatric surveillance study. Sci Rep 2025; 15:11588. [PMID: 40185990 PMCID: PMC11971435 DOI: 10.1038/s41598-025-95956-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 03/25/2025] [Indexed: 04/07/2025] Open
Abstract
Knowledge regarding the pediatric immune response to SARS-CoV-2 infection and/or vaccination remains limited, particularly for the variants of concern (VOC). Our objective was to evaluate the neutralizing antibody response against SARS-CoV-2 VOC in the naturally infected and/or vaccinated pediatric population. Participants aged 5-12 years who presented to either an outpatient clinic or emergency room were eligible for participation in this study. Participants were divided into four groups based on infection and vaccination status. Plasma was tested using immunoassays targeting anti-SARS-CoV-2 IgG, spike protein, and nucleocapsid. A total of 619 participants met study inclusion. Natural infection was identified in 189/619 children (31%), 284/619 were vaccinated (46%) and 69/619 were both naturally infected and vaccinated (11%). Participants that were vaccinated had received one (n = 169/619; 27%) or two (n = 115/619; 19%) vaccine doses. The median time between the 1st and 2nd vaccine doses was 56 days, interquartile range 50-56. A general upward trend in antibody positivity was observed across all VOC as the study proceeded over a 5-month period. Omicron antibody responses were lower than those of other VOC, both in relation to the percentage of positive cases and over time. Neither asthma nor diabetes altered antibody responses, but antibody titres were reduced for a variety of VOC in those children receiving immunotherapy or with leukopenia. This study demonstrated decreased neutralizing antibody responses against the Omicron variant, regardless of past infection or vaccination status. These findings emphasize the need for continued neutralizing antibody surveillance.
Collapse
Affiliation(s)
- Douglas D Fraser
- Pediatrics, Western University, London, ON, Canada.
- Physiology and Pharmacology, Western University, London, ON, Canada.
- Clinical Neurological Sciences, Western University, London, ON, Canada.
- Children's Health Research Institute, London, ON, Canada.
| | - Devika Singh
- Pediatrics, Western University, London, ON, Canada
| | - Enis Cela
- Physiology and Pharmacology, Western University, London, ON, Canada
- Children's Health Research Institute, London, ON, Canada
| | - Maitray A Patel
- Epidemiology and Biostatistics, Western University, London, ON, Canada
| | - Maysaa Assaf
- Pediatrics, Western University, London, ON, Canada
| | | | - Michael Knauer
- Pathology and Laboratory Medicine, Western University, London, ON, Canada
| | - Michael R Miller
- Pediatrics, Western University, London, ON, Canada
- Children's Health Research Institute, London, ON, Canada
| | | | - Angela Li
- Diagnostics Biochem Canada Inc, London, ON, Canada
| | - Paul Hahn
- Diagnostics Biochem Canada Inc, London, ON, Canada
| | - Marta Hrek
- Diagnostics Biochem Canada Inc, London, ON, Canada
| | | |
Collapse
|
2
|
Adler AL, Waghmare A, Lacombe K, Dickerson JA, L. Greninger A, Briggs Hagen M, Pringle K, Fairlie T, Midgely CM, Englund JA. Seroprevalence of SARS-CoV-2 IgG antibodies in children seeking medical care in Seattle, WA June 2020 to December 2022. Microbiol Spectr 2025; 13:e0262524. [PMID: 40062892 PMCID: PMC11960482 DOI: 10.1128/spectrum.02625-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 02/18/2025] [Indexed: 04/03/2025] Open
Abstract
Seroprevalence studies play an important role in estimating the number of children infected with SARS-CoV-2. We report SARS-CoV-2 seroprevalence in children seeking medical care for any reason at a free-standing pediatric hospital in Seattle, WA over a 2.5-year period and four distinct pandemic waves. We randomly selected residual serum samples from children and young adults seeking medical care as inpatients and outpatients at Seattle Children's Hospital between June 2020 and December 2022 to test for the presence of anti-nucleocapsid (N) antibodies. Samples were categorized into four distinct pandemic waves based on Washington State epidemiology: Wave 1 (June 2020-October 2020), Wave 2 (November 2020-June 2021), Wave 3 (July 2021-November 2021), and Wave 4 (December 2021-December 2022). Patient characteristics and COVID-19 vaccine status were obtained, and zip codes were used to ascertain the Social Vulnerability Index (SVI). Multivariable Poisson regression models with robust variance estimates were used to examine the relationship between patient characteristics and anti-N-positivity for each wave. Among 8,040 samples from 7,102 patients included in the analyses, seroprevalence rose from 2.4% (95% CI, 2.0%-3.1%) in Wave 1 to 25.5% (95% CI 23.3%-27.8%) in Wave 4 (following the Omicron surge). High SVI, Hispanic ethnicity, or use of government insurance was associated with increased anti-N positivity in most waves. We observed a steady increase in anti-N seroprevalence followed by a sharp increase after the Omicron surge in early 2022. Our data demonstrate the burden of COVID-19 on specific groups with health disparities within our region throughout the pandemic.IMPORTANCEOur results highlight the importance of seropositivity studies as essential tools to provide information on the incidence and prevalence of SARS-CoV-2 seropositivity. Our results also reinforce other reports demonstrating the inequitable burden of COVID-19 on groups with health disparities and that this inequitable burden continued to persist throughout the pandemic, even in a region with high adherence to COVID-19 mitigation efforts. It also highlights SVI's value in identifying communities that must be part of pandemic research, and public health and vaccination strategies.
Collapse
Affiliation(s)
- Amanda L. Adler
- Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Alpana Waghmare
- Seattle Children’s Research Institute, Seattle, Washington, USA
- Department of Pediatrics, University of Washington, Seattle, Washington, USA
| | - Kirsten Lacombe
- Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Jane A. Dickerson
- Seattle Children’s Research Institute, Seattle, Washington, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Alexander L. Greninger
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Melissa Briggs Hagen
- Coronavirus and Other Respiratory Viruses Division, Centers for Disease Control and Prevention, Atlanta, USA
| | - Kimberly Pringle
- Coronavirus and Other Respiratory Viruses Division, Centers for Disease Control and Prevention, Atlanta, USA
| | - Tarayn Fairlie
- Coronavirus and Other Respiratory Viruses Division, Centers for Disease Control and Prevention, Atlanta, USA
| | - Claire M. Midgely
- Coronavirus and Other Respiratory Viruses Division, Centers for Disease Control and Prevention, Atlanta, USA
| | - Janet A. Englund
- Seattle Children’s Research Institute, Seattle, Washington, USA
- Department of Pediatrics, University of Washington, Seattle, Washington, USA
| |
Collapse
|
3
|
Florian DM, Bauer M, Popovitsch A, Fae I, Springer DN, Graninger M, Traugott M, Weseslindtner L, Aberle SW, Fischer G, Kundi M, Stiasny K, Zoufaly A, Landry SJ, Aberle JH. Enhanced and long-lasting SARS-CoV-2 immune memory in individuals with common cold coronavirus cross-reactive T cell immunity. Front Immunol 2025; 16:1501704. [PMID: 40191213 PMCID: PMC11968687 DOI: 10.3389/fimmu.2025.1501704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 03/03/2025] [Indexed: 04/09/2025] Open
Abstract
With the continuous emergence of novel SARS-CoV-2 variants, long-lasting and broadly reactive cellular and humoral immunity is critical for durable protection from COVID-19. We investigated SARS-CoV-2-specific T cell immunity in relation to antibodies, infection outcome and disease severity and assessed its durability in a longitudinal cohort over a three-year time course. We identified pre-existing T cells reactive to the seasonal coronavirus (CoV) OC43 that cross-react with the conserved SARS-CoV-2 spike S813-829 peptide. These cross-reactive T cells increased in frequency following SARS-CoV-2 infection or vaccination and correlated with enhanced spike-specific T cell responses and significantly reduced viral loads. Furthermore, our data revealed that CoV-cross-reactive T cells were maintained as part of the long-lasting memory response, contributing to increased T cell frequencies against omicron variants. These findings suggest a functional role of CoV-cross-reactive T cells that extends beyond the initial SARS-CoV-2 exposure, contributing to enhanced immunity against highly mutated SARS-CoV-2 variants.
Collapse
Affiliation(s)
- David M. Florian
- Center for Virology, Medical University of Vienna, Vienna, Austria
| | - Michael Bauer
- Center for Virology, Medical University of Vienna, Vienna, Austria
| | | | - Ingrid Fae
- Department of Transfusion Medicine and Cell Therapy, Medical University of Vienna, Vienna, Austria
| | | | | | | | | | | | - Gottfried Fischer
- Department of Transfusion Medicine and Cell Therapy, Medical University of Vienna, Vienna, Austria
| | - Michael Kundi
- Center for Public Health, Department for Environmental Health, Medical University of Vienna, Vienna, Austria
| | - Karin Stiasny
- Center for Virology, Medical University of Vienna, Vienna, Austria
| | - Alexander Zoufaly
- Department of Medicine IV, Klinik Favoriten, Vienna, Austria
- Faculty of Medicine, Sigmund Freud University, Vienna, Austria
| | - Samuel J. Landry
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, LA, United States
| | - Judith H. Aberle
- Center for Virology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
4
|
Geng C, Zhang M, Wang N, Li M, Cui L. SARS-CoV-2 compromises blastocyst quality by modifying the ovarian microenvironment. Signal Transduct Target Ther 2025; 10:68. [PMID: 39966380 PMCID: PMC11836307 DOI: 10.1038/s41392-025-02156-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 12/11/2024] [Accepted: 01/22/2025] [Indexed: 02/20/2025] Open
Affiliation(s)
- Chen Geng
- The Second Hospital of Shandong University, Institute of Women, Children and Reproductive Health, Shandong University, Jinan, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Shandong University, Jinan, Shandong, China
| | - Min Zhang
- The Second Hospital of Shandong University, Institute of Women, Children and Reproductive Health, Shandong University, Jinan, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Shandong University, Jinan, Shandong, China
| | - Ning Wang
- The Second Hospital of Shandong University, Institute of Women, Children and Reproductive Health, Shandong University, Jinan, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Shandong University, Jinan, Shandong, China
| | - Mei Li
- The Second Hospital of Shandong University, Institute of Women, Children and Reproductive Health, Shandong University, Jinan, China.
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Shandong University, Jinan, Shandong, China.
| | - Linlin Cui
- The Second Hospital of Shandong University, Institute of Women, Children and Reproductive Health, Shandong University, Jinan, China.
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Shandong University, Jinan, Shandong, China.
| |
Collapse
|
5
|
Trimbake D, Singh D, K. YG, Babar P, S. VD, Tripathy AS. Durability of Functional SARS-CoV-2-Specific Immunological Memory and T Cell Response up to 8-9 Months Postrecovery From COVID-19. J Immunol Res 2025; 2025:9743866. [PMID: 39963186 PMCID: PMC11832264 DOI: 10.1155/jimr/9743866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 12/19/2024] [Indexed: 02/20/2025] Open
Abstract
Research on long-term follow-up in individuals who have recovered from coronavirus disease-19 (COVID-19) would yield insights regarding their immunity status and identify those who need booster vaccinations. This study evaluated the longevity of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-specific cellular and humoral memory responses, as well as T cell effector functionalities, at 1-2 months (n = 40), 8-9 months (n = 40), and 12 months/1 year (n = 27) following recovery from SARS-CoV-2 infection. CTL response by enzyme-linked immunospot (ELISPOT); levels of cytokine by Bio-Plex, natural killer (NK), CD4+ helper, and CD8+ cytotoxic T cell functionalities using flow cytometry; anti-SARS-CoV-2 IgG by ELISA; and levels of neutralizing antibodies (NAbs) by surrogate virus NAb assay were assessed. The levels of SARS-CoV-2-specific IgG and NAb at 1-2 and 8-9 months postrecovery were hand in hand and appeared declining. SARS-CoV-2-specific B, memory B and plasma cells, and T cells sustained up to 8-9 months. Increased expression of CD107a/IFN-γ by NK cells and cytotoxic T cells at 8-9 months could be indicative of SARS-CoV-2-specific effector functions. Recovered individuals with positive and negative IgG antibody status displayed T cell response up to 1 year and 8-9 months, respectively, emphasizing the durabilty of effector immunity up to 8-9 months regardless of IgG antibody status. Overall, the recovered individuals exhibited robust immunological memory, sustained T cell response with effector functionality against SARS-CoV-2 that persists for at least 8-9 months.
Collapse
Affiliation(s)
- Diptee Trimbake
- Department of Dengue and Chikungunya, Indian Council of Medical Research-National Institute of Virology, 20-A, Dr Ambedkar Road, Pune 411001, India
| | - Dharmendra Singh
- Department of Dengue and Chikungunya, Indian Council of Medical Research-National Institute of Virology, 20-A, Dr Ambedkar Road, Pune 411001, India
| | - Yogesh Gurav K.
- Department of Dengue and Chikungunya, Indian Council of Medical Research-National Institute of Virology, 20-A, Dr Ambedkar Road, Pune 411001, India
| | - Prasad Babar
- Department of Dengue and Chikungunya, Indian Council of Medical Research-National Institute of Virology, 20-A, Dr Ambedkar Road, Pune 411001, India
| | - Varsha Dange S.
- Department of Medicine, Pimpri Chinchwad Municipal Corporation, Pimpri, Pune 411018, Maharashtra, India
| | - Anuradha S. Tripathy
- Department of Dengue and Chikungunya, Indian Council of Medical Research-National Institute of Virology, 20-A, Dr Ambedkar Road, Pune 411001, India
| |
Collapse
|
6
|
Cherif I, Kharroubi G, Darragi I, El Benna S, Gharbi A, Baccouche A, Souissi C, Bahri O, Ben Ahmed M, Bettaieb J. Dynamics of SARS-CoV-2 antibodies after natural infection: insights from a study on Pasteur Institute of Tunis employees. Libyan J Med 2024; 19:2348233. [PMID: 38693671 PMCID: PMC11067560 DOI: 10.1080/19932820.2024.2348233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 04/23/2024] [Indexed: 05/03/2024] Open
Abstract
This study aimed to assess the kinetics of antibodies against the SARS-CoV-2, following natural infection in a cohort of employees of the Institut Pasteur de Tunis (IPT) and to assess the risk of reinfection over a 12-months follow-up period. A prospective study was conducted among an open cohort of IPT employees with confirmed SARS-CoV-2 infection that were recruited between September 2020 and March 2021. Sera samples were taken at 1, 3, 6, 9 and 12 months after confirmation of COVID-19 infection and tested for SARS-CoV-2-specific immunoglobulin G (IgG) antibodies to the spike (S-RBD) protein (IgG anti-S-RBD) and for neutralizing antibodies. Participants who had an initial decline of IgG anti-S-RBD and neutralizing antibodies followed by a subsequent rise in antibody titers as well as those who tested positive for SARS-CoV-2 by RT-PCR after at least 60 days of follow up were considered as reinfected. In total, 137 individuals were included with a mean age of 44.7 ± 12.3 years and a sex-ratio (Male/Female) of 0.33. Nearly all participants (92.7%) were symptomatic, and 2.2% required hospitalization. Among the 70 participants with three or more prospective blood samples, 32.8% were reinfected among whom 11 (47.8%) reported COVID-19 like symptoms. Up to 12 months of follow up, 100% and 42.9% of participants had detectable IgG anti-S-RBD and neutralizing antibodies, respectively. This study showed that humoral immune response following COVID-19 infection may persist up to 12 months after infection despite the potential risk for reinfection that is mainly explained by the emergence of new variants.
Collapse
Affiliation(s)
- Ines Cherif
- Department of Medical Epidemiology, Institut Pasteur de Tunis, University of Tunis El Manar, Tunis, Tunisia
- Laboratory of Transmission, Control and Immunobiology of Infections (LR11IPT02), Institut Pasteur de Tunis, University of Tunis El Manar, Tunis, Tunisia
- Faculty of Medicine of Sousse, University of Sousse, Sousse, Tunisia
| | - Ghassen Kharroubi
- Department of Medical Epidemiology, Institut Pasteur de Tunis, University of Tunis El Manar, Tunis, Tunisia
- Laboratory of Transmission, Control and Immunobiology of Infections (LR11IPT02), Institut Pasteur de Tunis, University of Tunis El Manar, Tunis, Tunisia
| | - Imen Darragi
- Department of Medical Epidemiology, Institut Pasteur de Tunis, University of Tunis El Manar, Tunis, Tunisia
- Laboratory of Transmission, Control and Immunobiology of Infections (LR11IPT02), Institut Pasteur de Tunis, University of Tunis El Manar, Tunis, Tunisia
| | - Soumaya El Benna
- Laboratory of Microbiology-Biochemistry, Aziza Othmana Hospital, University of Tunis, Tunis, Tunisia
| | - Adel Gharbi
- Department of Medical Epidemiology, Institut Pasteur de Tunis, University of Tunis El Manar, Tunis, Tunisia
- Laboratory of Transmission, Control and Immunobiology of Infections (LR11IPT02), Institut Pasteur de Tunis, University of Tunis El Manar, Tunis, Tunisia
| | - Amor Baccouche
- Department of Medical Epidemiology, Institut Pasteur de Tunis, University of Tunis El Manar, Tunis, Tunisia
- Laboratory of Transmission, Control and Immunobiology of Infections (LR11IPT02), Institut Pasteur de Tunis, University of Tunis El Manar, Tunis, Tunisia
| | - Cyrine Souissi
- Laboratory of Transmission, Control and Immunobiology of Infections (LR11IPT02), Institut Pasteur de Tunis, University of Tunis El Manar, Tunis, Tunisia
- Laboratory of Clinical Immunology, Institut Pasteur de Tunis, University of Tunis El Manar, Tunis, Tunisia
| | - Olfa Bahri
- Laboratory of Microbiology-Biochemistry, Aziza Othmana Hospital, University of Tunis, Tunis, Tunisia
| | - Melika Ben Ahmed
- Laboratory of Transmission, Control and Immunobiology of Infections (LR11IPT02), Institut Pasteur de Tunis, University of Tunis El Manar, Tunis, Tunisia
- Laboratory of Clinical Immunology, Institut Pasteur de Tunis, University of Tunis El Manar, Tunis, Tunisia
| | - Jihene Bettaieb
- Department of Medical Epidemiology, Institut Pasteur de Tunis, University of Tunis El Manar, Tunis, Tunisia
- Laboratory of Transmission, Control and Immunobiology of Infections (LR11IPT02), Institut Pasteur de Tunis, University of Tunis El Manar, Tunis, Tunisia
| |
Collapse
|
7
|
Ali M, Longet S, Neale I, Rongkard P, Chowdhury FUH, Hill J, Brown A, Laidlaw S, Tipton T, Hoque A, Hassan N, Hackstein CP, Adele S, Akther HD, Abraham P, Paul S, Rahman MM, Alam MM, Parvin S, Mollah FH, Hoque MM, Moore SC, Biswas SK, Turtle L, de Silva TI, Ogbe A, Frater J, Barnes E, Tomic A, Carroll MW, Klenerman P, Kronsteiner B, Chowdhury FR, Dunachie SJ. Obesity differs from diabetes mellitus in antibody and T-cell responses post-COVID-19 recovery. Clin Exp Immunol 2024; 218:78-92. [PMID: 38642547 PMCID: PMC11404124 DOI: 10.1093/cei/uxae030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 01/05/2024] [Accepted: 04/18/2024] [Indexed: 04/22/2024] Open
Abstract
OBJECTIVE Obesity and type 2 diabetes (DM) are risk factors for severe coronavirus disease 2019 (COVID-19) outcomes, which disproportionately affect South Asian populations. This study aims to investigate the humoral and cellular immune responses to SARS-CoV-2 in adult COVID-19 survivors with overweight/obesity (Ov/Ob, BMI ≥ 23 kg/m2) and DM in Bangladesh. METHODS In this cross-sectional study, SARS-CoV-2-specific antibody and T-cell responses were investigated in 63 healthy and 75 PCR-confirmed COVID-19 recovered individuals in Bangladesh, during the pre-vaccination first wave of the COVID-19 pandemic in 2020. RESULTS In COVID-19 survivors, SARS-CoV-2 infection induced robust antibody and T-cell responses, which correlated with disease severity. After adjusting for age, sex, DM status, disease severity, and time since onset of symptoms, Ov/Ob was associated with decreased neutralizing antibody titers, and increased SARS-CoV-2 spike-specific IFN-γ response along with increased proliferation and IL-2 production by CD8 + T cells. In contrast, DM was not associated with SARS-CoV-2-specific antibody and T-cell responses after adjustment for obesity and other confounders. CONCLUSION Ov/Ob is associated with lower neutralizing antibody levels and higher T-cell responses to SARS-CoV-2 post-COVID-19 recovery, while antibody or T-cell responses remain unaltered in DM.
Collapse
Affiliation(s)
- Mohammad Ali
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
- Centre for Global Health Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
- Mahidol-Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand
- Directorate General of Health Services, Dhaka, Bangladesh
- Department of Biochemistry and Molecular Biology, Bangabandhu Sheikh Mujib Medical University, Dhaka, Bangladesh
| | - Stephanie Longet
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Isabel Neale
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
- Centre for Global Health Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
- Mahidol-Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand
| | - Patpong Rongkard
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
- Centre for Global Health Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
- Mahidol-Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand
| | | | - Jennifer Hill
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
- Centre for Global Health Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
- Mahidol-Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand
| | - Anthony Brown
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Stephen Laidlaw
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Tom Tipton
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Ashraful Hoque
- Department of Transfusion Medicine, Sheikh Hasina National Burn & Plastics Surgery Institute, Dhaka, Bangladesh
| | - Nazia Hassan
- Department of Internal Medicine, Bangabandhu Sheikh Mujib Medical University, Dhaka, Bangladesh
| | - Carl-Philipp Hackstein
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
- Translational Gastroenterology Unit, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Sandra Adele
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
- Centre for Global Health Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
- Mahidol-Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand
| | - Hossain Delowar Akther
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
- Translational Gastroenterology Unit, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Priyanka Abraham
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
- Centre for Global Health Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
- Mahidol-Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand
| | - Shrebash Paul
- Department of Internal Medicine, Bangabandhu Sheikh Mujib Medical University, Dhaka, Bangladesh
| | - Md Matiur Rahman
- Department of Biochemistry and Molecular Biology, Bangabandhu Sheikh Mujib Medical University, Dhaka, Bangladesh
| | - Md Masum Alam
- Department of Biochemistry and Molecular Biology, Bangabandhu Sheikh Mujib Medical University, Dhaka, Bangladesh
| | - Shamima Parvin
- Department of Biochemistry and Molecular Biology, Mugda Medical College, Dhaka, Bangladesh
| | - Forhadul Hoque Mollah
- Department of Biochemistry and Molecular Biology, Bangabandhu Sheikh Mujib Medical University, Dhaka, Bangladesh
| | - Md Mozammel Hoque
- Department of Biochemistry and Molecular Biology, Bangabandhu Sheikh Mujib Medical University, Dhaka, Bangladesh
| | - Shona C Moore
- Tropical and Infectious Disease Unit, Liverpool University Hospitals NHS Foundation Trust, Member of Liverpool Health Partners, Liverpool, UK
| | - Subrata K Biswas
- Department of Biochemistry and Molecular Biology, Bangabandhu Sheikh Mujib Medical University, Dhaka, Bangladesh
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, Connecticut, USA
| | - Lance Turtle
- Tropical and Infectious Disease Unit, Liverpool University Hospitals NHS Foundation Trust, Member of Liverpool Health Partners, Liverpool, UK
| | - Thushan I de Silva
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Ane Ogbe
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - John Frater
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Eleanor Barnes
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
- Centre for Global Health Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Adriana Tomic
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA
- Department of Microbiology, Boston University School of Medicine, Boston, MA, USA
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Miles W Carroll
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Paul Klenerman
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
- Centre for Global Health Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
- Translational Gastroenterology Unit, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Barbara Kronsteiner
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
- Centre for Global Health Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
- Mahidol-Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand
| | - Fazle Rabbi Chowdhury
- Mahidol-Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand
- Department of Internal Medicine, Bangabandhu Sheikh Mujib Medical University, Dhaka, Bangladesh
| | - Susanna J Dunachie
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
- Centre for Global Health Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
- Mahidol-Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand
- NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| |
Collapse
|
8
|
Tahsin A, Bhattacharjee P, Al Saba A, Yasmin T, Nabi AHMN. Genetic and epigenetic analyses of IFN-γ gene proximal promoter region underlying positive correlation between persistently high anti-SARS-CoV-2 IgG and IFN-γ among COVID-19 vaccinated Bangladeshi adults. Vaccine 2024; 42:126157. [PMID: 39079811 DOI: 10.1016/j.vaccine.2024.126157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 07/16/2024] [Accepted: 07/17/2024] [Indexed: 08/23/2024]
Abstract
IFN-γ is an immunological modulator influencing IgG isotype and concentration, which present a correlate of protection to evaluate vaccine efficacy. As transiently expressed, stable genetic and epigenetic signatures of the cytokine's expression may exist. This study investigates correlation between plasma IFN-γ and anti-SARS-CoV-2 IgG levels, seeking genetic polymorphisms and epigenetic variations within the IFN-γ gene proximal promoter. 200 COVID-19-vaccinated adults were classified into seropositive and seronegative groups based on plasma anti-SARS-CoV-2 IgG. Upon correlation analysis between anti-SARS-CoV-2 IgG and IFN-γ levels, IFN-γ gene proximal promoter region was subjected to nucleotide sequencing for two subsets: seronegative (21 < Days post-vaccination ≤180, n = 11) and seropositive (IgG > Q3 and Days post-vaccination >180, n = 24). Relative unmethylation of IFN-γ proximal promoter was assessed for the latter subset and its correlation with plasma IFN-γ and IgG levels was evaluated. A statistically significant positive correlation (r = 0.492, p = 0.018) was observed between IFN-γ and anti-SARS-CoV-2 IgG in the seropositive group with persistently high IgG titre (IgG > Q3, Days elapsed post-vaccination >180). A heterozygous 5'-UTR variant (rs776667149:C>T) identified in one seronegative individual revealed a potential impact on PKR-mediated translational attenuation of IFN-γ mRNA. No significant correlation was found between IFN-γ proximal promoter unmethylation and its plasma levels among HAR individuals with Days post-vaccination of either >180 (r = 0.14, p = 0.679) or < 180 (r = -0.062, p = 0.693). This study demonstrates an extent of humoral immunity against SARS-CoV-2 among COVID-19 vaccinated Bangladeshi population. This study suggests plasma IFN-γ may play a role in maintaining persistent anti-SARS-CoV-2 IgG levels, which warrants further investigation along with genetic and/or epigenetic basis to fully establish its protective nature in COVID-19 vaccination.
Collapse
Affiliation(s)
- Anika Tahsin
- Laboratory of Population Genetics, Department of Biochemistry and Molecular Biology, University of Dhaka, Bangladesh
| | - Piyash Bhattacharjee
- Laboratory of Population Genetics, Department of Biochemistry and Molecular Biology, University of Dhaka, Bangladesh
| | - Abdullah Al Saba
- Laboratory of Population Genetics, Department of Biochemistry and Molecular Biology, University of Dhaka, Bangladesh
| | - Tahirah Yasmin
- Laboratory of Population Genetics, Department of Biochemistry and Molecular Biology, University of Dhaka, Bangladesh
| | - A H M Nurun Nabi
- Laboratory of Population Genetics, Department of Biochemistry and Molecular Biology, University of Dhaka, Bangladesh.
| |
Collapse
|
9
|
Zhao K, So HC, Lin Z. scParser: sparse representation learning for scalable single-cell RNA sequencing data analysis. Genome Biol 2024; 25:223. [PMID: 39152499 PMCID: PMC11328435 DOI: 10.1186/s13059-024-03345-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 07/23/2024] [Indexed: 08/19/2024] Open
Abstract
The rapid rise in the availability and scale of scRNA-seq data needs scalable methods for integrative analysis. Though many methods for data integration have been developed, few focus on understanding the heterogeneous effects of biological conditions across different cell populations in integrative analysis. Our proposed scalable approach, scParser, models the heterogeneous effects from biological conditions, which unveils the key mechanisms by which gene expression contributes to phenotypes. Notably, the extended scParser pinpoints biological processes in cell subpopulations that contribute to disease pathogenesis. scParser achieves favorable performance in cell clustering compared to state-of-the-art methods and has a broad and diverse applicability.
Collapse
Affiliation(s)
- Kai Zhao
- Department of Statistics, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Hon-Cheong So
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China.
- KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research of Common Diseases, Kunming Institute of Zoology and The Chinese University of Hong Kong, Hong Kong SAR, China.
- Department of Psychiatry, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China.
- Margaret K.L. Cheung Research Centre for Management of Parkinsonism, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China.
- Brain and Mind Institute, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China.
- Hong Kong Branch of the Chinese Academy of Sciences Center for Excellence in Animal Evolution and Genetics, The Chinese University of Hong Kong, Hong Kong SAR, China.
| | - Zhixiang Lin
- Department of Statistics, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China.
| |
Collapse
|
10
|
Rothoeft T, Maier C, Talarico A, Hoffmann A, Schlegtendal A, Lange B, Petersmann A, Denz R, Timmesfeld N, Toepfner N, Vidal-Blanco E, Pfaender S, Lücke T, Brinkmann F. Natural and hybrid immunity after SARS-CoV-2 infection in children and adolescents. Infection 2024; 52:1449-1458. [PMID: 38499828 PMCID: PMC11288991 DOI: 10.1007/s15010-024-02225-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 02/24/2024] [Indexed: 03/20/2024]
Abstract
PURPOSE In contrast to adults, immune protection against SARS-CoV-2 in children and adolescents with natural or hybrid immunity is still poorly understood. The aim of this study was to analyze different immune compartments in different age groups and whether humoral immune reactions correlate with a cellular immune response. METHODS 72 children and adolescents with a preceding SARS-CoV-2 infection were recruited. 37 were vaccinated with an RNA vaccine (BNT162b2). Humoral immunity was analyzed 3-26 months (median 10 months) after infection by measuring Spike protein (S), nucleocapsid (NCP), and neutralizing antibodies (nAB). Cellular immunity was analyzed using a SARS-CoV-2-specific interferon-γ release assay (IGRA). RESULTS All children and adolescents had S antibodies; titers were higher in those with hybrid immunity (14,900 BAU/ml vs. 2118 BAU/ml). NCP antibodies were detectable in > 90%. Neutralizing antibodies (nAB) were more frequently detected (90%) with higher titers (1914 RLU) in adolescents with hybrid immunity than in children with natural immunity (62.5%, 476 RLU). Children with natural immunity were less likely to have reactive IGRAs (43.8%) than adolescents with hybrid immunity (85%). The amount of interferon-γ released by T cells was comparable in natural and hybrid immunity. CONCLUSION Spike antibodies are the most reliable markers to monitor an immune reaction against SARS-CoV-2. High antibody titers of spike antibodies and nAB correlated with cellular immunity, a phenomenon found only in adolescents with hybrid immunity. Hybrid immunity is associated with markedly higher antibody titers and a higher probability of a cellular immune response than a natural immunity.
Collapse
Affiliation(s)
- T Rothoeft
- University Hospital of Pediatrics and Adolescent Medicine, St. Josef-Hospital, Ruhr-University, Bochum, Germany.
| | - C Maier
- University Hospital of Pediatrics and Adolescent Medicine, St. Josef-Hospital, Ruhr-University, Bochum, Germany
| | - A Talarico
- University Hospital of Pediatrics and Adolescent Medicine, St. Josef-Hospital, Ruhr-University, Bochum, Germany
| | - A Hoffmann
- University Hospital of Pediatrics and Adolescent Medicine, St. Josef-Hospital, Ruhr-University, Bochum, Germany
| | - A Schlegtendal
- University Hospital of Pediatrics and Adolescent Medicine, St. Josef-Hospital, Ruhr-University, Bochum, Germany
| | - B Lange
- Department of Epidemiology, Helmholtz Centre for Infection Research, Brunswick, Germany
| | - A Petersmann
- University Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Oldenburg, Oldenburg, Germany
- University Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
| | - R Denz
- Department of Medical Informatics, Biometry and Epidemiology, Ruhr-University Bochum, Bochum, Germany
| | - N Timmesfeld
- Department of Medical Informatics, Biometry and Epidemiology, Ruhr-University Bochum, Bochum, Germany
| | - N Toepfner
- Department of Pediatrics, Faculty of Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - E Vidal-Blanco
- Department of Molecular and Medical Virology, Ruhr University Bochum, Bochum, Germany
| | - S Pfaender
- Department of Molecular and Medical Virology, Ruhr University Bochum, Bochum, Germany
| | - T Lücke
- University Hospital of Pediatrics and Adolescent Medicine, St. Josef-Hospital, Ruhr-University, Bochum, Germany
| | - F Brinkmann
- University Hospital of Pediatrics and Adolescent Medicine, St. Josef-Hospital, Ruhr-University, Bochum, Germany
- University Children's Hospital, Lübeck, Germany
- Airway Research Center North (ARCN), German Center for Lung Research (DZL), Lübeck, Germany
| |
Collapse
|
11
|
Brown PE, Fu SH, Newcombe L, Tang X, Nagelkerke N, Birnboim HC, Bansal A, Colwill K, Mailhot G, Delgado-Brand M, Tursun T, Qi F, Gingras AC, Slutsky AS, Pasic MD, Companion J, Bogoch II, Morawski E, Lam T, Reid A, Jha P. Hybrid immunity from severe acute respiratory syndrome coronavirus 2 infection and vaccination in Canadian adults: A cohort study. eLife 2024; 13:e89961. [PMID: 38916134 PMCID: PMC11281784 DOI: 10.7554/elife.89961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 06/20/2024] [Indexed: 06/26/2024] Open
Abstract
Background Few national-level studies have evaluated the impact of 'hybrid' immunity (vaccination coupled with recovery from infection) from the Omicron variants of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Methods From May 2020 to December 2022, we conducted serial assessments (each of ~4000-9000 adults) examining SARS-CoV-2 antibodies within a mostly representative Canadian cohort drawn from a national online polling platform. Adults, most of whom were vaccinated, reported viral test-confirmed infections and mailed self-collected dried blood spots (DBSs) to a central lab. Samples underwent highly sensitive and specific antibody assays to spike and nucleocapsid protein antigens, the latter triggered only by infection. We estimated cumulative SARS-CoV-2 incidence prior to the Omicron period and during the BA.1/1.1 and BA.2/5 waves. We assessed changes in antibody levels and in age-specific active immunity levels. Results Spike levels were higher in infected than in uninfected adults, regardless of vaccination doses. Among adults vaccinated at least thrice and infected more than 6 months earlier, spike levels fell notably and continuously for the 9-month post-vaccination. In contrast, among adults infected within 6 months, spike levels declined gradually. Declines were similar by sex, age group, and ethnicity. Recent vaccination attenuated declines in spike levels from older infections. In a convenience sample, spike antibody and cellular responses were correlated. Near the end of 2022, about 35% of adults above age 60 had their last vaccine dose more than 6 months ago, and about 25% remained uninfected. The cumulative incidence of SARS-CoV-2 infection rose from 13% (95% confidence interval 11-14%) before omicron to 78% (76-80%) by December 2022, equating to 25 million infected adults cumulatively. However, the coronavirus disease 2019 (COVID-19) weekly death rate during the BA.2/5 waves was less than half of that during the BA.1/1.1 wave, implying a protective role for hybrid immunity. Conclusions Strategies to maintain population-level hybrid immunity require up-to-date vaccination coverage, including among those recovering from infection. Population-based, self-collected DBSs are a practicable biological surveillance platform. Funding Funding was provided by the COVID-19 Immunity Task Force, Canadian Institutes of Health Research, Pfizer Global Medical Grants, and St. Michael's Hospital Foundation. PJ and ACG are funded by the Canada Research Chairs Program.
Collapse
Affiliation(s)
- Patrick E Brown
- Centre for Global Health Research, Unity Health Toronto and University of TorontoTorontoCanada
| | - Sze Hang Fu
- Centre for Global Health Research, Unity Health Toronto and University of TorontoTorontoCanada
| | - Leslie Newcombe
- Centre for Global Health Research, Unity Health Toronto and University of TorontoTorontoCanada
| | - Xuyang Tang
- Centre for Global Health Research, Unity Health Toronto and University of TorontoTorontoCanada
| | - Nico Nagelkerke
- Centre for Global Health Research, Unity Health Toronto and University of TorontoTorontoCanada
| | - H Chaim Birnboim
- Centre for Global Health Research, Unity Health Toronto and University of TorontoTorontoCanada
| | - Aiyush Bansal
- Centre for Global Health Research, Unity Health Toronto and University of TorontoTorontoCanada
| | - Karen Colwill
- Lunenfeld-Tanenbaum Research Institute, Sinai HealthTorontoCanada
| | | | | | - Tulunay Tursun
- Lunenfeld-Tanenbaum Research Institute, Sinai HealthTorontoCanada
| | - Freda Qi
- Lunenfeld-Tanenbaum Research Institute, Sinai HealthTorontoCanada
| | | | | | | | | | - Isaac I Bogoch
- Toronto General Hospital, University Hospital NetworkTorontoCanada
| | | | | | | | - Prabhat Jha
- Centre for Global Health Research, Unity Health Toronto and University of TorontoTorontoCanada
| | | |
Collapse
|
12
|
Bean DJ, Monroe J, Liang YM, Borberg E, Senussi Y, Swank Z, Chalise S, Walt D, Weinberg J, Sagar M. Heterotypic immunity from prior SARS-CoV-2 infection but not COVID-19 vaccination associates with lower endemic coronavirus incidence. Sci Transl Med 2024; 16:eado7588. [PMID: 38865483 PMCID: PMC11565543 DOI: 10.1126/scitranslmed.ado7588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 05/07/2024] [Indexed: 06/14/2024]
Abstract
Immune responses from prior severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and COVID-19 vaccination mitigate disease severity, but they do not fully prevent subsequent infections, especially from genetically divergent strains. We examined the incidence of and immune differences against human endemic coronaviruses (eCoVs) as a proxy for response against future genetically heterologous coronaviruses (CoVs). We assessed differences in symptomatic eCoV and non-CoV respiratory disease incidence among those with known prior SARS-CoV-2 infection or previous COVID-19 vaccination but no documented SARS-CoV-2 infection or neither exposure. Retrospective cohort analyses suggest that prior SARS-CoV-2 infection, but not previous COVID-19 vaccination alone, associates with a lower incidence of subsequent symptomatic eCoV infection. There was no difference in non-CoV incidence, implying that the observed difference was eCoV specific. In a second cohort where both cellular and humoral immunity were measured, those with prior SARS-CoV-2 spike protein exposure had lower eCoV-directed neutralizing antibodies, suggesting that neutralization is not responsible for the observed decreased eCoV disease. The three groups had similar cellular responses against the eCoV spike protein and nucleocapsid antigens. However, CD8+ T cell responses to the nonstructural eCoV proteins nsp12 and nsp13 were higher in individuals with previous SARS-CoV-2 infection as compared with the other groups. This association between prior SARS-CoV-2 infection and decreased incidence of eCoV disease may therefore be due to a boost in CD8+ T cell responses against eCoV nsp12 and nsp13, suggesting that incorporation of nonstructural viral antigens in a future pan-CoV vaccine may improve vaccine efficacy.
Collapse
Affiliation(s)
- David J. Bean
- Department of Virology, Immunology and Microbiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| | - Janet Monroe
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| | - Yan Mei Liang
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| | - Ella Borberg
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02215, USA
| | - Yasmeen Senussi
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02215, USA
| | - Zoe Swank
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02215, USA
| | - Sujata Chalise
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02215, USA
| | - David Walt
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02215, USA
| | - Janice Weinberg
- Department of Biostatistics, Boston University School of Public Health, Boston, MA 02118, USA
| | - Manish Sagar
- Department of Virology, Immunology and Microbiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| |
Collapse
|
13
|
Romero-Bogado L, Steiner M, Thuissard Vasallo IJ, Andreu Vázquez C, Navío T, Muñoz-Fernández S. [Impact of systemic therapies on SARS-CoV-2 antibody seroprevalence in patients with immune-mediated diseases]. Med Clin (Barc) 2024; 162:470-476. [PMID: 38418311 DOI: 10.1016/j.medcli.2023.12.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 12/02/2023] [Accepted: 12/05/2023] [Indexed: 03/01/2024]
Abstract
OBJECTIVE To determine the seroprevalence of SARS-CoV-2 in patients with immune-mediated inflammatory diseases (IMID) treated with biologic (bDMARDs) or synthetic targeted disease-modifying antirheumatic drugs (tsDMARDs). METHODS An observational, descriptive, prospective and cross-sectional study of analytical prevalence analysis was conducted in patients with IMID with bDMARDs or tsDMARDs. Seroprevalence was compared by measuring immunoglobulinG (IgG) against SARS-CoV-2 between October/2020 and May/2021. RESULTS A total of 550 IMID's patients were studied, all of them on treatment with bDMARDs or tsDMARDs. The seroprevalence of the total patient group was 16% (88/550). Patients receiving therapy with tumor necrosis factor alpha inhibitors (TNFi) had a higher seroprevalence compared to other biologic and synthetic targeted therapies (OR: 1.792 [95%CI: 1.088-2.951]; P=.021). The influence on seroprevalence of concomitant use with b/tsDMARDs of conventional synthetic DMARDs (csDMARDs) was also analyzed. A lower seroprevalence was demonstrated in the group of patients treated with TNFi and methotrexate together, compared with those on TNFi monotherapy, 10.1 vs 24.1% (OR: 0.355 [95%CI: 0.165-0.764]; P=.006). No significant differences were found with the other DMARDs. Regarding IMIDs, no differences in seroprevalence were identified between the different disease groups. CONCLUSION Patients on treatment with TNFα inhibitors have better humoral response compared to the other b/tsDMARDs. However, when associated with methotrexate the seroprevalence decreases significantly.
Collapse
Affiliation(s)
- Liz Romero-Bogado
- Sección de Reumatología, Hospital Universitario Infanta Sofía, Universidad Europea de Madrid, San Sebastián de los Reyes, Madrid, España.
| | - Martina Steiner
- Sección de Reumatología, Hospital Universitario Infanta Sofía, Universidad Europea de Madrid, San Sebastián de los Reyes, Madrid, España
| | | | - Cristina Andreu Vázquez
- Facultad de Ciencias Biomédicas y de la Salud, Universidad Europea de Madrid, Villaviciosa de Odón, Madrid, España
| | - Teresa Navío
- Sección de Reumatología, Hospital Universitario Infanta Leonor, Universidad Complutense de Madrid, Madrid, España
| | - Santiago Muñoz-Fernández
- Sección de Reumatología, Hospital Universitario Infanta Sofía, Universidad Europea de Madrid, San Sebastián de los Reyes, Madrid, España
| |
Collapse
|
14
|
Wang X, Zhang M, Wei K, Li C, Yang J, Jiang S, Zhao C, Zhao X, Qiao R, Cui Y, Chen Y, Li J, Cai G, Liu C, Yu J, Zhang W, Xie F, Wang P, Zhang Y. Longitudinal Analysis of Humoral and Cellular Immune Response up to 6 Months after SARS-CoV-2 BA.5/BF.7/XBB Breakthrough Infection and BA.5/BF.7-XBB Reinfection. Vaccines (Basel) 2024; 12:464. [PMID: 38793715 PMCID: PMC11125724 DOI: 10.3390/vaccines12050464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 04/19/2024] [Accepted: 04/23/2024] [Indexed: 05/26/2024] Open
Abstract
The rapid mutation of SARS-CoV-2 has led to multiple rounds of large-scale breakthrough infection and reinfection worldwide. However, the dynamic changes of humoral and cellular immunity responses to several subvariants after infection remain unclear. In our study, a 6-month longitudinal immune response evaluation was conducted on 118 sera and 50 PBMC samples from 49 healthy individuals who experienced BA.5/BF.7/XBB breakthrough infection or BA.5/BF.7-XBB reinfection. By studying antibody response, memory B cell, and IFN-γ secreting CD4+/CD8+ T cell response to several SARS-CoV-2 variants, we observed that each component of immune response exhibited distinct kinetics. Either BA.5/BF.7/XBB breakthrough infection or BA.5/BF.7-XBB reinfection induces relatively high level of binding and neutralizing antibody titers against Omicron subvariants at an early time point, which rapidly decreases over time. Most of the individuals at 6 months post-breakthrough infection completely lost their neutralizing activities against BQ.1.1, CH.1.1, BA.2.86, JN.1 and XBB subvariants. Individuals with BA.5/BF.7-XBB reinfection exhibit immune imprinting shifting and recall pre-existing BA.5/BF.7 neutralization antibodies. In the BA.5 breakthrough infection group, the frequency of BA.5 and XBB.1.16-RBD specific memory B cells, resting memory B cells, and intermediate memory B cells gradually increased over time. On the other hand, the frequency of IFN-γ secreting CD4+/CD8+ T cells induced by WT/BA.5/XBB.1.16 spike trimer remains stable over time. Overall, our research indicates that individuals with breakthrough infection have rapidly declining antibody levels but have a relatively stable cellular immunity that can provide some degree of protection from future exposure to new antigens.
Collapse
Affiliation(s)
- Xun Wang
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai 200437, China; (X.W.); (C.L.); (C.Z.); (X.Z.); (R.Q.); (Y.C.); (Y.C.); (J.L.); (G.C.); (C.L.); (J.Y.)
| | - Meng Zhang
- Department of Infectious Diseases, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing 210023, China; (M.Z.); (J.Y.); (S.J.)
- Nanjing Research Center for Infectious Diseases of Integrated Traditional Chinese and Western Medicine, Nanjing 210001, China
| | - Kaifeng Wei
- College of Traditional Chinese Medicine·College of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China;
| | - Chen Li
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai 200437, China; (X.W.); (C.L.); (C.Z.); (X.Z.); (R.Q.); (Y.C.); (Y.C.); (J.L.); (G.C.); (C.L.); (J.Y.)
| | - Jinghui Yang
- Department of Infectious Diseases, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing 210023, China; (M.Z.); (J.Y.); (S.J.)
- Nanjing Research Center for Infectious Diseases of Integrated Traditional Chinese and Western Medicine, Nanjing 210001, China
| | - Shujun Jiang
- Department of Infectious Diseases, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing 210023, China; (M.Z.); (J.Y.); (S.J.)
- Nanjing Research Center for Infectious Diseases of Integrated Traditional Chinese and Western Medicine, Nanjing 210001, China
| | - Chaoyue Zhao
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai 200437, China; (X.W.); (C.L.); (C.Z.); (X.Z.); (R.Q.); (Y.C.); (Y.C.); (J.L.); (G.C.); (C.L.); (J.Y.)
| | - Xiaoyu Zhao
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai 200437, China; (X.W.); (C.L.); (C.Z.); (X.Z.); (R.Q.); (Y.C.); (Y.C.); (J.L.); (G.C.); (C.L.); (J.Y.)
| | - Rui Qiao
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai 200437, China; (X.W.); (C.L.); (C.Z.); (X.Z.); (R.Q.); (Y.C.); (Y.C.); (J.L.); (G.C.); (C.L.); (J.Y.)
| | - Yuchen Cui
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai 200437, China; (X.W.); (C.L.); (C.Z.); (X.Z.); (R.Q.); (Y.C.); (Y.C.); (J.L.); (G.C.); (C.L.); (J.Y.)
| | - Yanjia Chen
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai 200437, China; (X.W.); (C.L.); (C.Z.); (X.Z.); (R.Q.); (Y.C.); (Y.C.); (J.L.); (G.C.); (C.L.); (J.Y.)
| | - Jiayan Li
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai 200437, China; (X.W.); (C.L.); (C.Z.); (X.Z.); (R.Q.); (Y.C.); (Y.C.); (J.L.); (G.C.); (C.L.); (J.Y.)
| | - Guonan Cai
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai 200437, China; (X.W.); (C.L.); (C.Z.); (X.Z.); (R.Q.); (Y.C.); (Y.C.); (J.L.); (G.C.); (C.L.); (J.Y.)
| | - Changyi Liu
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai 200437, China; (X.W.); (C.L.); (C.Z.); (X.Z.); (R.Q.); (Y.C.); (Y.C.); (J.L.); (G.C.); (C.L.); (J.Y.)
| | - Jizhen Yu
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai 200437, China; (X.W.); (C.L.); (C.Z.); (X.Z.); (R.Q.); (Y.C.); (Y.C.); (J.L.); (G.C.); (C.L.); (J.Y.)
| | - Wenhong Zhang
- Department of Infectious Diseases, National Medical Center for Infectious Diseases and Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, Huashan Hospital, Fudan University, Shanghai 200437, China;
| | - Faren Xie
- Department of Infectious Diseases, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing 210023, China; (M.Z.); (J.Y.); (S.J.)
- Nanjing Research Center for Infectious Diseases of Integrated Traditional Chinese and Western Medicine, Nanjing 210001, China
| | - Pengfei Wang
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai 200437, China; (X.W.); (C.L.); (C.Z.); (X.Z.); (R.Q.); (Y.C.); (Y.C.); (J.L.); (G.C.); (C.L.); (J.Y.)
| | - Yanliang Zhang
- Department of Infectious Diseases, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing 210023, China; (M.Z.); (J.Y.); (S.J.)
- Nanjing Research Center for Infectious Diseases of Integrated Traditional Chinese and Western Medicine, Nanjing 210001, China
| |
Collapse
|
15
|
Zhao X, Pei W, Qi Y, Li Y, Kong X. Enhanced aerobic granular sludge with micro-electric field for sulfamethoxazole degradation: Efficiency, mechanism, and microbial community. CHEMOSPHERE 2024; 354:141741. [PMID: 38499071 DOI: 10.1016/j.chemosphere.2024.141741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 03/15/2024] [Accepted: 03/16/2024] [Indexed: 03/20/2024]
Abstract
In this study, an aerobic granular sludge electrochemical system (AGES) was established by applying the micro-electric field to an aerobic granular sludge (AGS) reactor for the degradation of sulfamethoxazole (SMZ). Under the stimulation of the micro-electric field, the granulation of sludge was improved and the degradation rate of SMZ was enhanced. The features of granular sludge were characterized by scanning electron microscopy and X-ray diffraction. The optimal degradation rate of SMZ (88%) was obtained at the voltage of 3 V and the effective electrode area of 800 mm2. The results of kinetics analyses revealed that the degradation of SMZ by AGES can be fitted with the second-order kinetic equation, showing a degradation rate constant (k) of 0.001 L mol-1·min-1. The degradation products of SMZ in the AGES system were detected by LC-MS and their possible degradation routes were elucidated. The micro-electric field in the AGES system played a selective role in microbes' enrichment and growth, changing the diversity of the microbial community. Pseudomonas, Tolumonas, and Acidovorax were the dominant bacteria in the AGES system, which is accountable for the abatement of SMZ and nutrients. This work provides a green means for improving AGS and paves the way for applying the AGS process to real-world wastewater treatment.
Collapse
Affiliation(s)
- Xia Zhao
- College of Petrochemical Engineering, Lanzhou University of Technology, Lanzhou 730050, PR China
| | - Weina Pei
- College of Petrochemical Engineering, Lanzhou University of Technology, Lanzhou 730050, PR China
| | - Yihan Qi
- College of Petrochemical Engineering, Lanzhou University of Technology, Lanzhou 730050, PR China.
| | - Yabin Li
- College of Petrochemical Engineering, Lanzhou University of Technology, Lanzhou 730050, PR China.
| | - Xiuqin Kong
- College of Petrochemical Engineering, Lanzhou University of Technology, Lanzhou 730050, PR China
| |
Collapse
|
16
|
Doan TA, Forward TS, Schafer JB, Lucas ED, Fleming I, Uecker-Martin A, Ayala E, Guthmiller JJ, Hesselberth JR, Morrison TE, Tamburini BAJ. Immunization-induced antigen archiving enhances local memory CD8+ T cell responses following an unrelated viral infection. NPJ Vaccines 2024; 9:66. [PMID: 38514656 PMCID: PMC10957963 DOI: 10.1038/s41541-024-00856-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 02/29/2024] [Indexed: 03/23/2024] Open
Abstract
Antigens from viruses or immunizations can persist or are archived in lymph node stromal cells such as lymphatic endothelial cells (LEC) and fibroblastic reticular cells (FRC). Here, we find that, during the time frame of antigen archiving, LEC apoptosis caused by a second, but unrelated, innate immune stimulus such as vaccina viral infection or CpG DNA administration resulted in cross-presentation of archived antigens and boosted memory CD8 + T cells specific to the archived antigen. In contrast to "bystander" activation associated with unrelated infections, the memory CD8 + T cells specific to the archived antigen from the immunization were significantly higher than memory CD8 + T cells of a different antigen specificity. Finally, the boosted memory CD8 + T cells resulted in increased protection against Listeria monocytogenes expressing the antigen from the immunization, but only for the duration that the antigen was archived. These findings outline an important mechanism by which lymph node stromal cell archived antigens, in addition to bystander activation, can augment memory CD8 + T cell responses during repeated inflammatory insults.
Collapse
Affiliation(s)
- Thu A Doan
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado School of Medicine, Aurora, CO, USA
- Immunology Graduate Program, University of Colorado School of Medicine, Aurora, CO, USA
| | - Tadg S Forward
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Johnathon B Schafer
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Erin D Lucas
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado School of Medicine, Aurora, CO, USA
- Immunology Graduate Program, University of Colorado School of Medicine, Aurora, CO, USA
| | - Ira Fleming
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado School of Medicine, Aurora, CO, USA
- Medical Scientist Training Program, University of Colorado School of Medicine, Aurora, CO, USA
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, CO, USA
| | - Aspen Uecker-Martin
- Medical Scientist Training Program, University of Colorado School of Medicine, Aurora, CO, USA
| | - Edgardo Ayala
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Jenna J Guthmiller
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Jay R Hesselberth
- Medical Scientist Training Program, University of Colorado School of Medicine, Aurora, CO, USA
| | - Thomas E Morrison
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Beth A Jirón Tamburini
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado School of Medicine, Aurora, CO, USA.
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, USA.
| |
Collapse
|
17
|
Inoue W, Kimura Y, Okamoto S, Nogimori T, Sakaguchi-Mikami A, Yamamoto T, Tsunetsugu-Yokota Y. SARS-CoV-2-Specific Immune Responses in Vaccination and Infection during the Pandemic in 2020-2022. Viruses 2024; 16:446. [PMID: 38543812 PMCID: PMC10974545 DOI: 10.3390/v16030446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 03/10/2024] [Accepted: 03/11/2024] [Indexed: 05/23/2024] Open
Abstract
To gain insight into how immunity develops against SARS-CoV-2 from 2020 to 2022, we analyzed the immune response of a small group of university staff and students who were either infected or vaccinated. We investigated the levels of receptor-binding domain (RBD)-specific and nucleocapsid (N)-specific IgG and IgA antibodies in serum and saliva samples taken early (around 10 days after infection or vaccination) and later (around 1 month later), as well as N-specific T-cell responses. One patient who had been infected in 2020 developed serum RBD and N-specific IgG antibodies, but declined eight months later, then mRNA vaccination in 2021 produced a higher level of anti-RBD IgG than natural infection. In the vaccination of naïve individuals, vaccines induced anti-RBD IgG, but it declined after six months. A third vaccination boosted the IgG level again, albeit to a lower level than after the second. In 2022, when the Omicron variant became dominant, familial transmission occurred among vaccinated people. In infected individuals, the levels of serum anti-RBD IgG antibodies increased later, while anti-N IgG peaked earlier. The N-specific activated T cells expressing IFN γ or CD107a were detected only early. Although SARS-CoV-2-specific salivary IgA was undetectable, two individuals showed a temporary peak in RBD- and N-specific IgA antibodies in their saliva on the second day after infection. Our study, despite having a small sample size, revealed that SARS-CoV-2 infection triggers the expected immune responses against acute viral infections. Moreover, our findings suggest that the temporary mucosal immune responses induced early during infection may provide better protection than the currently available intramuscular vaccines.
Collapse
Affiliation(s)
- Wakana Inoue
- Department of Medical Technology, School of Health Sciences and Graduate School of Medical Technology, Tokyo University of Technology, Tokyo 144-8535, Japan; (W.I.); (Y.K.); (S.O.); (A.S.-M.)
| | - Yuta Kimura
- Department of Medical Technology, School of Health Sciences and Graduate School of Medical Technology, Tokyo University of Technology, Tokyo 144-8535, Japan; (W.I.); (Y.K.); (S.O.); (A.S.-M.)
| | - Shion Okamoto
- Department of Medical Technology, School of Health Sciences and Graduate School of Medical Technology, Tokyo University of Technology, Tokyo 144-8535, Japan; (W.I.); (Y.K.); (S.O.); (A.S.-M.)
| | - Takuto Nogimori
- Laboratory of Precision Immunology, Center for Intractable Diseases and ImmunoGenomics, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka 567-0085, Japan; (T.N.); (T.Y.)
| | - Akane Sakaguchi-Mikami
- Department of Medical Technology, School of Health Sciences and Graduate School of Medical Technology, Tokyo University of Technology, Tokyo 144-8535, Japan; (W.I.); (Y.K.); (S.O.); (A.S.-M.)
| | - Takuya Yamamoto
- Laboratory of Precision Immunology, Center for Intractable Diseases and ImmunoGenomics, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka 567-0085, Japan; (T.N.); (T.Y.)
- Laboratory of Aging and Immune Regulation, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka 565-0871, Japan
- Department of Virology and Immunology, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | - Yasuko Tsunetsugu-Yokota
- Department of Medical Technology, School of Health Sciences and Graduate School of Medical Technology, Tokyo University of Technology, Tokyo 144-8535, Japan; (W.I.); (Y.K.); (S.O.); (A.S.-M.)
- Laboratory of Precision Immunology, Center for Intractable Diseases and ImmunoGenomics, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka 567-0085, Japan; (T.N.); (T.Y.)
- Research Institute, The World New Prosperity (WNP), Tokyo 169-0075, Japan
| |
Collapse
|
18
|
Malani A, Aiyar J, Sant A, Kamran N, Mohanan M, Taneja S, Woda B, Zhao W, Acharya A. Comparing population-level humoral and cellular immunity to SARS-Cov-2 in Bangalore, India. Sci Rep 2024; 14:5758. [PMID: 38459035 PMCID: PMC10923858 DOI: 10.1038/s41598-024-54922-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 02/18/2024] [Indexed: 03/10/2024] Open
Abstract
Two types of immunity, humoral and cellular, offer protection against COVID. Humoral protection, contributed by circulating neutralizing antibodies, can provide immediate protection but decays more quickly than cellular immunity and can lose effectiveness in the face of mutation and drift in the SARS-CoV-2 spike protein. Therefore, population-level seroprevalence surveys used to estimate population-level immunity may underestimate the degree to which a population is protected against COVID. In early 2021, before India began its vaccination campaign, we tested for humoral and cellular immunity to SARS-Cov-2 in representative samples of slum and non-slum populations in Bangalore, India. We found that 29.7% of samples (unweighted) had IgG antibodies to the spike protein and 15.5% had neutralizing antibodies, but at up to 46% showed evidence of cellular immunity. We also find that prevalence of cellular immunity is significantly higher in slums than in non-slums. These findings suggest (1) that a significantly larger proportion of the population in Bangalore, India, had cellular immunity to SARS-CoV-2 than had humoral immunity, as measured by serological surveys, and (2) that low socio-economic status communities display higher frequency of cellular immunity, likely because of greater exposure to infection due to population density.
Collapse
Affiliation(s)
| | | | - Andrea Sant
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, USA
| | | | - Manoj Mohanan
- Sanford School of Public Policy, Duke University, Durham, NC, USA
| | - Saloni Taneja
- University of Southern California, Los Angeles, CA, USA
| | - Bartek Woda
- University of Chicago, Chicago, IL, USA
- Amazon, Chicago, IL, USA
| | | | | |
Collapse
|
19
|
Feola S, Chiaro J, Fusciello M, Russo S, Kleino I, Ylösmäki L, Kekäläinen E, Hästbacka J, Pekkarinen PT, Ylösmäki E, Capone S, Folgori A, Raggioli A, Boni C, Tiezzi C, Vecchi A, Gelzo M, Kared H, Nardin A, Fehlings M, Barban V, Ahokas P, Viitala T, Castaldo G, Pastore L, Porter P, Pesonen S, Cerullo V. PeptiVAX: A new adaptable peptides-delivery platform for development of CTL-based, SARS-CoV-2 vaccines. Int J Biol Macromol 2024; 262:129926. [PMID: 38331062 DOI: 10.1016/j.ijbiomac.2024.129926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 01/30/2024] [Accepted: 01/31/2024] [Indexed: 02/10/2024]
Abstract
The emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) posed a threat to public health and the global economy, necessitating the development of various vaccination strategies. Mutations in the SPIKE protein gene, a crucial component of mRNA and adenovirus-based vaccines, raised concerns about vaccine efficacy, prompting the need for rapid vaccine updates. To address this, we leveraged PeptiCRAd, an oncolytic vaccine based on tumor antigen decorated oncolytic adenoviruses, creating a vaccine platform called PeptiVAX. First, we identified multiple CD8 T-cell epitopes from highly conserved regions across coronaviruses, expanding the range of T-cell responses to non-SPIKE proteins. We designed short segments containing the predicted epitopes presented by common HLA-Is in the global population. Testing the immunogenicity, we characterized T-cell responses to candidate peptides in peripheral blood mononuclear cells (PBMCs) from pre-pandemic healthy donors and ICU patients. As a proof of concept in mice, we selected a peptide with epitopes predicted to bind to murine MHC-I haplotypes. Our technology successfully elicited peptide-specific T-cell responses, unaffected by the use of unarmed adenoviral vectors or adeno-based vaccines encoding SPIKE. In conclusion, PeptiVAX represents a fast and adaptable SARS-CoV-2 vaccine delivery system that broadens T-cell responses beyond the SPIKE protein, offering potential benefits for vaccine effectiveness.
Collapse
Affiliation(s)
- Sara Feola
- Drug Research Program (DRP) ImmunoViroTherapy Lab (IVT), Division of Pharmaceutical Biosciences, Faculty of Pharmacy, Viikinkaari 5E, University of Helsinki, 00790 Helsinki, Finland; Helsinki Institute of Life Science (HiLIFE), Fabianinkatu 33, University of Helsinki, 00710 Helsinki, Finland; Translational Immunology Program (TRIMM), Faculty of Medicine Helsinki University, postal code Haartmaninkatu 8, University of Helsinki, 00290 Helsinki, Finland; Digital Precision Cancer Medicine Flagship (iCAN), University of Helsinki, FI-00014 Helsinki, Finland
| | - Jacopo Chiaro
- Drug Research Program (DRP) ImmunoViroTherapy Lab (IVT), Division of Pharmaceutical Biosciences, Faculty of Pharmacy, Viikinkaari 5E, University of Helsinki, 00790 Helsinki, Finland; Helsinki Institute of Life Science (HiLIFE), Fabianinkatu 33, University of Helsinki, 00710 Helsinki, Finland; Translational Immunology Program (TRIMM), Faculty of Medicine Helsinki University, postal code Haartmaninkatu 8, University of Helsinki, 00290 Helsinki, Finland; Digital Precision Cancer Medicine Flagship (iCAN), University of Helsinki, FI-00014 Helsinki, Finland
| | - Manlio Fusciello
- Drug Research Program (DRP) ImmunoViroTherapy Lab (IVT), Division of Pharmaceutical Biosciences, Faculty of Pharmacy, Viikinkaari 5E, University of Helsinki, 00790 Helsinki, Finland; Helsinki Institute of Life Science (HiLIFE), Fabianinkatu 33, University of Helsinki, 00710 Helsinki, Finland; Translational Immunology Program (TRIMM), Faculty of Medicine Helsinki University, postal code Haartmaninkatu 8, University of Helsinki, 00290 Helsinki, Finland; Digital Precision Cancer Medicine Flagship (iCAN), University of Helsinki, FI-00014 Helsinki, Finland
| | - Salvatore Russo
- Drug Research Program (DRP) ImmunoViroTherapy Lab (IVT), Division of Pharmaceutical Biosciences, Faculty of Pharmacy, Viikinkaari 5E, University of Helsinki, 00790 Helsinki, Finland; Helsinki Institute of Life Science (HiLIFE), Fabianinkatu 33, University of Helsinki, 00710 Helsinki, Finland; Translational Immunology Program (TRIMM), Faculty of Medicine Helsinki University, postal code Haartmaninkatu 8, University of Helsinki, 00290 Helsinki, Finland; Digital Precision Cancer Medicine Flagship (iCAN), University of Helsinki, FI-00014 Helsinki, Finland
| | - Iivari Kleino
- Turku Bioscience Centre, University of Turku and Åbo Akademi University Turku, Turku, Finland
| | | | - Eliisa Kekäläinen
- Translational Immunology Program (TRIMM), Faculty of Medicine Helsinki University, postal code Haartmaninkatu 8, University of Helsinki, 00290 Helsinki, Finland; HUSLAB Clinical Microbiology, HUS Diagnostic Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Johanna Hästbacka
- HUSLAB Clinical Microbiology, HUS Diagnostic Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Pirkka T Pekkarinen
- Translational Immunology Program (TRIMM), Faculty of Medicine Helsinki University, postal code Haartmaninkatu 8, University of Helsinki, 00290 Helsinki, Finland; Division of Intensive Care Medicine, Department of Anaesthesiology and Intensive Care, University of Helsinki and Helsinki University Hospital, Finland
| | - Erkko Ylösmäki
- Drug Research Program (DRP) ImmunoViroTherapy Lab (IVT), Division of Pharmaceutical Biosciences, Faculty of Pharmacy, Viikinkaari 5E, University of Helsinki, 00790 Helsinki, Finland; Helsinki Institute of Life Science (HiLIFE), Fabianinkatu 33, University of Helsinki, 00710 Helsinki, Finland; Translational Immunology Program (TRIMM), Faculty of Medicine Helsinki University, postal code Haartmaninkatu 8, University of Helsinki, 00290 Helsinki, Finland; Digital Precision Cancer Medicine Flagship (iCAN), University of Helsinki, FI-00014 Helsinki, Finland
| | | | | | | | - Carolina Boni
- Laboratory of Viral Immunopathology, Unit of Infectious Disease and Hepatology, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy
| | - Camilla Tiezzi
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Andrea Vecchi
- Laboratory of Viral Immunopathology, Unit of Infectious Disease and Hepatology, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy
| | - Monica Gelzo
- CEINGE-Biotecnologie Avanzate, Naples, Italy; Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, Naples, Italy
| | | | | | | | | | | | - Tapani Viitala
- Pharmaceutical Biophysics Research Group, Drug Research Program, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Giuseppe Castaldo
- CEINGE-Biotecnologie Avanzate, Naples, Italy; Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, Naples, Italy
| | - Lucio Pastore
- CEINGE-Biotecnologie Avanzate, Naples, Italy; Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, Naples, Italy; Department of Molecular Medicine and Medical Biotechnology, Naples University "Federico II", S. Pansini 5, Italy
| | - Paul Porter
- Valo Therapeutics Oy, Helsinki, Finland; School of Nursing, Curtin University, GPO Box U 1987, Perth, WA 6845, Australia
| | | | - Vincenzo Cerullo
- Drug Research Program (DRP) ImmunoViroTherapy Lab (IVT), Division of Pharmaceutical Biosciences, Faculty of Pharmacy, Viikinkaari 5E, University of Helsinki, 00790 Helsinki, Finland; Helsinki Institute of Life Science (HiLIFE), Fabianinkatu 33, University of Helsinki, 00710 Helsinki, Finland; Translational Immunology Program (TRIMM), Faculty of Medicine Helsinki University, postal code Haartmaninkatu 8, University of Helsinki, 00290 Helsinki, Finland; Digital Precision Cancer Medicine Flagship (iCAN), University of Helsinki, FI-00014 Helsinki, Finland; Institute for Molecular Medicine Finland, FIMM, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, FI-00014 Helsinki, Finland; Department of Molecular Medicine and Medical Biotechnology, Naples University "Federico II", S. Pansini 5, Italy.
| |
Collapse
|
20
|
Nesamari R, Omondi MA, Baguma R, Höft MA, Ngomti A, Nkayi AA, Besethi AS, Magugu SFJ, Mosala P, Walters A, Clark GM, Mennen M, Skelem S, Adriaanse M, Grifoni A, Sette A, Keeton RS, Ntusi NAB, Riou C, Burgers WA. Post-pandemic memory T cell response to SARS-CoV-2 is durable, broadly targeted, and cross-reactive to the hypermutated BA.2.86 variant. Cell Host Microbe 2024; 32:162-169.e3. [PMID: 38211583 PMCID: PMC10901529 DOI: 10.1016/j.chom.2023.12.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 11/28/2023] [Accepted: 12/11/2023] [Indexed: 01/13/2024]
Abstract
Ongoing severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) evolution has given rise to recombinant Omicron lineages that dominate globally (XBB.1), as well as the emergence of hypermutated variants (BA.2.86). In this context, durable and cross-reactive T cell immune memory is critical for continued protection against severe COVID-19. We examined T cell responses to SARS-CoV-2 approximately 1.5 years since Omicron first emerged. We describe sustained CD4+ and CD8+ spike-specific T cell memory responses in healthcare workers in South Africa (n = 39) who were vaccinated and experienced at least one SARS-CoV-2 infection. Spike-specific T cells are highly cross-reactive with all Omicron variants tested, including BA.2.86. Abundant nucleocapsid and membrane-specific T cells are detectable in most participants. The bulk of SARS-CoV-2-specific T cell responses have an early-differentiated phenotype, explaining their persistent nature. Overall, hybrid immunity leads to the accumulation of spike and non-spike T cells evident 3.5 years after the start of the pandemic, with preserved recognition of highly mutated SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Rofhiwa Nesamari
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa; Division of Medical Virology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Millicent A Omondi
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa; Division of Medical Virology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Richard Baguma
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa; Division of Medical Virology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Maxine A Höft
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa; Division of Medical Virology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Amkele Ngomti
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa; Division of Medical Virology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Anathi A Nkayi
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa; Division of Medical Virology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Asiphe S Besethi
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa; Division of Medical Virology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Siyabulela F J Magugu
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa; Division of Medical Virology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Paballo Mosala
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa; Division of Medical Virology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Avril Walters
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa; Division of Medical Virology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Gesina M Clark
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa; Division of Medical Virology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Mathilda Mennen
- Department of Medicine, University of Cape Town and Groote Schuur Hospital, Cape Town, South Africa; Cape Heart Institute, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa; South African Medical Research Council Extramural Unit on Intersection of Non-communicable Disease and Infectious Diseases, University of Cape Town, Cape Town, South Africa
| | - Sango Skelem
- Department of Medicine, University of Cape Town and Groote Schuur Hospital, Cape Town, South Africa; Cape Heart Institute, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa; South African Medical Research Council Extramural Unit on Intersection of Non-communicable Disease and Infectious Diseases, University of Cape Town, Cape Town, South Africa
| | - Marguerite Adriaanse
- Department of Medicine, University of Cape Town and Groote Schuur Hospital, Cape Town, South Africa; Cape Heart Institute, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa; South African Medical Research Council Extramural Unit on Intersection of Non-communicable Disease and Infectious Diseases, University of Cape Town, Cape Town, South Africa
| | - Alba Grifoni
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Alessandro Sette
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, USA; Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA, USA
| | - Roanne S Keeton
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa; Division of Medical Virology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Ntobeko A B Ntusi
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa; Department of Medicine, University of Cape Town and Groote Schuur Hospital, Cape Town, South Africa; Cape Heart Institute, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa; South African Medical Research Council Extramural Unit on Intersection of Non-communicable Disease and Infectious Diseases, University of Cape Town, Cape Town, South Africa; Wellcome Centre for Infectious Diseases Research in Africa, University of Cape Town, Cape Town, South Africa
| | - Catherine Riou
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa; Division of Medical Virology, Department of Pathology, University of Cape Town, Cape Town, South Africa; Wellcome Centre for Infectious Diseases Research in Africa, University of Cape Town, Cape Town, South Africa.
| | - Wendy A Burgers
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa; Division of Medical Virology, Department of Pathology, University of Cape Town, Cape Town, South Africa; Wellcome Centre for Infectious Diseases Research in Africa, University of Cape Town, Cape Town, South Africa.
| |
Collapse
|
21
|
Garay E, Whelan SPJ, DuBois RM, O’Rourke SM, Salgado-Escobar AE, Muñoz-Medina JE, Arias CF, López S. Immune response to SARS-CoV-2 variants after immunization with different vaccines in Mexico. Epidemiol Infect 2024; 152:e30. [PMID: 38312015 PMCID: PMC10894899 DOI: 10.1017/s0950268824000219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 01/11/2024] [Accepted: 01/24/2024] [Indexed: 02/06/2024] Open
Abstract
There is limited information on the antibody responses against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in subjects from developing countries with populations having a high incidence of co-morbidities. Here, we analysed the immunogenicity of homologous schemes using the ChAdOx1-S, Sputnik V, or BNT162b2 vaccines and the effect of a booster dose with ChAdOx1-S in middle-aged adults who were seropositive or seronegative to the SARS-CoV-2 spike protein before vaccination. The study was conducted post-vaccination with a follow-up of 4 months for antibody titre using enzyme-linked immunosorbent assay (ELISA) and pseudovirus (PV) neutralization assays (PNAs). All three vaccines elicited a superior IgG anti-receptor-binding domain (RBD) and neutralization response against the Alpha and Delta variants when administered to individuals with a previous infection by SARS-CoV-2. The booster dose spiked the neutralization activity among individuals with and without a prior SARS-CoV-2 infection. The ChAdOx1-S vaccine induced weaker antibody responses in infection-naive subjects. A follow-up of 4 months post-vaccination showed a drop in antibody titre, with about 20% of the infection-naive and 100% of SARS-CoV-2 pre-exposed participants with detectable neutralization capacity against Alpha pseudovirus (Alpha-PV) and Delta PV (Delta-PV). Our observations support the use of different vaccines in a country with high seroprevalence at the vaccination time.
Collapse
Affiliation(s)
- Erika Garay
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, Mexico
| | - Sean P. J. Whelan
- Department of Molecular Microbiology, Washington University in St. Louis, Saint Louis, United States
- Department of Microbiology, Harvard Medical School, Boston, United States
| | - Rebecca M. DuBois
- Department of Biomolecular Engineering, University of California, Santa Cruz, United States
| | - Sara M. O’Rourke
- Department of Biomolecular Engineering, University of California, Santa Cruz, United States
| | - Angel Eduardo Salgado-Escobar
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, Mexico
| | - José Esteban Muñoz-Medina
- Coordinación de Calidad de Insumos y Laboratorios Especializados, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Carlos F. Arias
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, Mexico
| | - Susana López
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, Mexico
| |
Collapse
|
22
|
Ferreras C, Hernández-Blanco C, Martín-Quirós A, Al-Akioui-Sanz K, Mora-Rillo M, Ibáñez F, Díaz-Almirón M, Cano-Ochando J, Lozano-Ojalvo D, Jiménez-González M, Goterris R, Sánchez-Zapardiel E, de Paz R, Guerra-García P, Queiruga-Parada J, Molina P, Briones ML, Ruz-Caracuel B, Borobia AM, Carcas AJ, Planelles D, Vicario JL, Moreno MÁ, Balas A, Llano M, Llorente A, Del Balzo Á, Cañada C, García MÁ, Calvin ME, Arenas I, Pérez de Diego R, Eguizábal C, Soria B, Solano C, Pérez-Martínez A. Results of phase 2 randomized multi-center study to evaluate the safety and efficacy of infusion of memory T cells as adoptive therapy in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pneumonia and/or lymphopenia (RELEASE NCT04578210). Cytotherapy 2024; 26:25-35. [PMID: 37897472 DOI: 10.1016/j.jcyt.2023.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 09/05/2023] [Accepted: 10/05/2023] [Indexed: 10/30/2023]
Abstract
BACKGROUND AIMS There are currently no effective anti-viral treatments for coronavirus disease 2019 (COVID-19)-hospitalized patients with hypoxemia. Lymphopenia is a biomarker of disease severity usually present in patients who are hospitalized. Approaches to increasing lymphocytes exerting an anti-viral effect must be considered to treat these patients. Following our phase 1 study, we performed a phase 2 randomized multicenter clinical trial in which we evaluated the efficacy of the infusion of allogeneic off-the-shelf CD45RA- memory T cells containing severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-specific T cells from convalescent donors plus the standard of care (SoC) versus just the SoC treatment. METHODS Eighty-four patients were enrolled in three Spanish centers. The patients were randomized into the infusion of 1 × 106/kg CD45RA- memory T cells or the SoC. We selected four unvaccinated donors based on the expression of interferon gamma SARS-CoV-2-specific response within the CD45RA- memory T cells and the most frequent human leukocyte antigen typing in the Spanish population. RESULTS We analyzed data from 81 patients. The primary outcome for recovery, defined as the proportion of participants in each group with normalization of fever, oxygen saturation sustained for at least 24 hours and lymphopenia recovery through day 14 or at discharge, was met for the experimental arm. We also observed faster lymphocyte recovery in the experimental group. We did not observe any treatment-related adverse events. CONCLUSIONS Adoptive cell therapy with off-the-shelf CD45RA- memory T cells containing SAR-CoV-2-specific T cells is safe, effective and accelerates lymphocyte recovery of patients with COVID-19 pneumonia and/or lymphopenia. TRIAL REGISTRATION NCT04578210.
Collapse
Affiliation(s)
- Cristina Ferreras
- Hospital La Paz Institute for Health Research, IdiPAZ, University Hospital La Paz, Madrid, Spain
| | - Clara Hernández-Blanco
- Internal Medicine Department, Hospital de Emergencias Enfermera Isabel Zendal, Madrid, Spain
| | | | - Karima Al-Akioui-Sanz
- Hospital La Paz Institute for Health Research, IdiPAZ, University Hospital La Paz, Madrid, Spain
| | - Marta Mora-Rillo
- Infectious Diseases Unit, Internal Medicine Department, University Hospital La Paz, Hospital La Paz Institute for Health Research, IdiPAZ, Consorcio Centro de Investigación Biomédica en Red CIBER-Infec, Madrid, Spain
| | - Fátima Ibáñez
- Internal Medicine Department, Hospital Puerta de Hierro, Madrid, Spain
| | | | - Jordi Cano-Ochando
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA; Centro Nacional de Microbiologia, Instituto de Salud Carlos III, 28220 Madrid, Spain; Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Daniel Lozano-Ojalvo
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - María Jiménez-González
- Infectious Diseases Unit, Internal Medicine Department, University Hospital La Paz, Hospital La Paz Institute for Health Research, IdiPAZ, Consorcio Centro de Investigación Biomédica en Red CIBER-Infec, Madrid, Spain; Clinical Trials Unit (UCICEC) at Hospital La Paz Institute for Health Research, IdiPAZ, University Hospital La Paz, Madrid, Spain
| | - Rosa Goterris
- Hematology Department, Hospital Clinico Universitario, Valencia, Spain
| | | | - Raquel de Paz
- Hematology Department, University Hospital La Paz, Madrid, Spain
| | - Pilar Guerra-García
- Hospital La Paz Institute for Health Research, IdiPAZ, University Hospital La Paz, Madrid, Spain; Pediatric Hemato-Oncology Department, University Hospital La Paz, Madrid, Spain
| | | | - Pablo Molina
- Clinical Pharmacology Department, University Hospital La Paz, Madrid, Spain
| | | | - Beatriz Ruz-Caracuel
- Hospital La Paz Institute for Health Research, IdiPAZ, University Hospital La Paz, Madrid, Spain
| | - Alberto M Borobia
- Hospital La Paz Institute for Health Research, IdiPAZ, University Hospital La Paz, Madrid, Spain; Clinical Trials Unit (UCICEC) at Hospital La Paz Institute for Health Research, IdiPAZ, University Hospital La Paz, Madrid, Spain; Clinical Pharmacology Department, University Hospital La Paz, Madrid, Spain
| | - Antonio J Carcas
- Clinical Trials Unit (UCICEC) at Hospital La Paz Institute for Health Research, IdiPAZ, University Hospital La Paz, Madrid, Spain; Clinical Pharmacology Department, University Hospital La Paz, Madrid, Spain; Faculty of Medicine Universidad Autónoma de Madrid, Madrid, Spain
| | - Dolores Planelles
- Department of Histocompatibility, Centro de Transfusión de la Comunidad Valenciana, Valencia, Spain
| | - José Luis Vicario
- Histocompatibility, Centro de Transfusión de la Comunidad de Madrid, Madrid, Spain
| | - Miguel Ángel Moreno
- Histocompatibility, Centro de Transfusión de la Comunidad de Madrid, Madrid, Spain
| | - Antonio Balas
- Histocompatibility, Centro de Transfusión de la Comunidad de Madrid, Madrid, Spain
| | - Marta Llano
- Infectious Diseases Department, Hospital de Emergencias Enfermera Isabel Zendal, Madrid, Spain
| | - Andrea Llorente
- Infectious Diseases Department, Hospital de Emergencias Enfermera Isabel Zendal, Madrid, Spain
| | - Álvaro Del Balzo
- Emergency Unit, Internal Medicine Department, University Hospital La Paz, Madrid, Spain
| | - Carlos Cañada
- Emergency Unit, Internal Medicine Department, University Hospital La Paz, Madrid, Spain
| | - Miguel Ángel García
- Emergency Unit, Internal Medicine Department, University Hospital La Paz, Madrid, Spain
| | - María Elena Calvin
- Emergency Unit, Internal Medicine Department, University Hospital La Paz, Madrid, Spain
| | - Isabel Arenas
- Emergency Unit, Internal Medicine Department, University Hospital La Paz, Madrid, Spain
| | - Rebeca Pérez de Diego
- Laboratory of Immunogenetics of Human Diseases, IdiPAZ Institute for Health Research, La Paz University Hospital, Madrid, Spain; Innate Immunity Group, IdiPAZ Institute for Health Research, La Paz University Hospital, Madrid, Spain; Interdepartmental Group of Immunodeficiencies, Madrid, Spain
| | - Cristina Eguizábal
- Research Unit, Basque Centre for Blood Transfusion and Human Tissues, Osakidetza, Bizkaia, Spain; Biocruces Bizkaia Health Research Institute, Bizkaia, Spain
| | - Bernat Soria
- Health Research Institute-ISABIAL, Alicante University Hospital and Institute of Bioengineering, Miguel Hernández University, Alicante, Spain; University Pablo de Olavide, Sevilla, Spain
| | - Carlos Solano
- Hematology Department, Hospital Clinico Universitario, Valencia, Spain; Department of Medicine, University of Valencia, Valencia, Spain
| | - Antonio Pérez-Martínez
- Hospital La Paz Institute for Health Research, IdiPAZ, University Hospital La Paz, Madrid, Spain; Pediatric Hemato-Oncology Department, University Hospital La Paz, Madrid, Spain; Faculty of Medicine Universidad Autónoma de Madrid, Madrid, Spain.
| |
Collapse
|
23
|
Li L, Xie Z, Li Y, Luo M, Zhang L, Feng C, Tang G, Huang H, Hou R, Xu Y, Jia S, Shi J, Fan Q, Gan Q, Yu N, Hu F, Li Y, Lan Y, Tang X, Li F, Deng X. Immune response and severity of Omicron BA.5 reinfection among individuals previously infected with different SARS-CoV-2 variants. Front Cell Infect Microbiol 2023; 13:1277880. [PMID: 38188634 PMCID: PMC10766752 DOI: 10.3389/fcimb.2023.1277880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 11/28/2023] [Indexed: 01/09/2024] Open
Abstract
Introduction COVID-19 continues to spread worldwide, with an increasing number of individuals experiencing reinfection after recovering from their primary infection. However, the nature and progression of this infection remain poorly understood. We aimed to investigate the immune response, severity and outcomes of Omicron BA.5 reinfection among individuals previously infected with different SARS-CoV-2 variants. Methods We enrolled 432 COVID-19 cases who had experienced prior infection with the ancestral SARS-CoV-2 virus, Delta variant or Omicron BA.2 variant between January 2020 and May 2022 in Guangzhou, China. All cases underwent follow-up from March to April, 2023 through telephone questionnaires and clinical visits. Nasal lavage fluid and peripheral blood were collected to assess anti-RBD IgA, anti-RBD IgG and virus-specific IFN-γ secreting T cells. Results Our study shows that 73.1%, 56.7% and 12.5% of individuals with a prior infection of the ancestral virus, Delta or Omicron BA.2 variant experienced reinfection with the BA.5 variant, respectively. Fever, cough and sore throat were the most common symptoms of BA.5 reinfection, with most improving within one week and none progressing to a critical condition. Compared with individuals without reinfection, reinfected patients with a prior Delta infection exhibited elevated levels of nasal anti-RBD IgA, serum anti-RBD IgG and IFN-γ secreting T cells, whereas there was no noticeable change in reinfected individuals with a prior BA.2 infection. Conclusion These results suggest that BA.5 reinfection is common but severe outcomes are relatively rare. Reinfection with a novel SARS-CoV-2 variant different from the prior infection may induce a more robust immune protection, which should be taken into account during vaccine development.
Collapse
Affiliation(s)
- Lu Li
- Institute of Infectious Diseases, Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou, China
| | - Zhiwei Xie
- Institute of Infectious Diseases, Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou, China
| | - Youxia Li
- Department of Critical Care Medicine, Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou, China
| | - Minhan Luo
- Institute of Infectious Diseases, Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou, China
| | - Lieguang Zhang
- Department of Radiology, Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou, China
| | - Chengqian Feng
- Institute of Infectious Diseases, Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou, China
| | - Guofang Tang
- Institute of Infectious Diseases, Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou, China
| | - Huang Huang
- Department of Critical Care Medicine, Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou, China
| | - Ruitian Hou
- Institute of Infectious Diseases, Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yujuan Xu
- Institute of Infectious Diseases, Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou, China
| | - Shijie Jia
- Department of Traditional Chinese Medicine, Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jingrong Shi
- Institute of Infectious Diseases, Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou, China
| | - Qinghong Fan
- Institute of Infectious Diseases, Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou, China
| | - Qingxin Gan
- Department of Radiology, Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou, China
| | - Na Yu
- Institute of Infectious Diseases, Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou, China
| | - Fengyu Hu
- Institute of Infectious Diseases, Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou, China
- Guangzhou Laboratory, Bio-Island, Guangzhou, China
| | - Yueping Li
- Department of Infectious Critical Care Medicine, Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yun Lan
- Institute of Infectious Diseases, Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou, China
| | - Xiaoping Tang
- Institute of Infectious Diseases, Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou, China
- Guangzhou Laboratory, Bio-Island, Guangzhou, China
| | - Feng Li
- Institute of Infectious Diseases, Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou, China
- Guangzhou Laboratory, Bio-Island, Guangzhou, China
| | - Xilong Deng
- Department of Critical Care Medicine, Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
24
|
Zaidi AK, Bajpai S, Dehgani-Mobaraki P. B cell responses to SARS-CoV-2. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2023; 202:155-181. [PMID: 38237985 DOI: 10.1016/bs.pmbts.2023.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2024]
Abstract
This chapter provides an overview of B cell responses in COVID-19, highlighting the structure of SARS-CoV-2 and its impact on B cell immunity. It explores the production and maturation of SARS-CoV-2-specific B cells, with a focus on the two distinct phases of the humoral immune response: the extrafollicular (EF) phase and the germinal center (GC) phase. Furthermore, the interplay between B cells, follicular T helper cells, CD4+ T cells, and plasma cells is discussed, emphasizing their collaborative role in mounting an effective humoral immune response against SARS-CoV-2. The concept of immunological memory is explored, highlighting the roles of plasma cells and B memory cells in providing long-term protection. The chapter delves into the antibody response during SARS-CoV-2 infection, categorizing the types of antibodies generated. This includes a detailed analysis of neutralizing antibodies, such as those directed against the receptor-binding domain (RBD) and the N-terminal domain (NTD), as well as non-neutralizing antibodies. The role of mucosal antibodies, cross-reactive antibodies, and auto-reactive antibodies is also discussed. Factors influencing the dynamics of anti-SARS-CoV-2 antibodies are examined, including the duration and strength of the humoral response. Additionally, the chapter highlights the impact of the Omicron variant on humoral immune responses and its implications for vaccine efficacy and antibody-mediated protection.
Collapse
Affiliation(s)
| | - Sanchit Bajpai
- Consultant ENT & Head and Neck Surgeon at TSM Medical College and Multispeciality Hospital, Lucknow, India.
| | - Puya Dehgani-Mobaraki
- Founder and President, Associazione Naso Sano, Ringgold Institution ID 567754, San Mariano, Italy
| |
Collapse
|
25
|
Altorki TA, Abdulal RH, Suliman BA, Aljeraisi TM, Alsharef A, Abdulaal WH, Alfaleh MA, Algaissi AA, Alhabbab RY, Ozbak H, Eid HM, Almutawif YA, Li X, Al-Rabia MW, Zhang Q, Mahmoud AB, Mahallawi WH, Hashem AM. Robust memory humoral immune response to SARS-CoV-2 in the tonsils of adults and children. Front Immunol 2023; 14:1291534. [PMID: 38149243 PMCID: PMC10750384 DOI: 10.3389/fimmu.2023.1291534] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Accepted: 11/17/2023] [Indexed: 12/28/2023] Open
Abstract
Background Adaptive humoral immunity against SARS-CoV-2 has mainly been evaluated in peripheral blood. Human secondary lymphoid tissues (such as tonsils) contain large numbers of plasma cells that secrete immunoglobulins at mucosal sites. Yet, the role of mucosal memory immunity induced by vaccines or natural infection against SARS-CoV-2 and its variants is not fully understood. Methods Tonsillar mononuclear cells (TMNCs) from adults (n=10) and children (n=11) were isolated and stimulated using positive SARS-CoV-2 nasal swabs. We used endpoint enzyme-linked immunosorbent assays (ELISAs) for the measurement of anti-S1, -RBD, and -N IgG antibody levels and a pseudovirus microneutralization assay to assess neutralizing antibodies (nAbs) in paired serum and supernatants from stimulated TMNCs. Results Strong systemic humoral response in previously SARS-CoV-2 infected and vaccinated adults and children was observed in accordance with the reported history of the participants. Interestingly, we found a significant increase in anti-RBD IgG (305 and 834 folds) and anti-S1 IgG (475 and 443 folds) in the stimulated TMNCs from adults and children, respectively, compared to unstimulated cells. Consistently, the stimulated TMNCs secreted higher levels of nAbs against the ancestral Wuhan strain and the Omicron BA.1 variant compared to unstimulated cells by several folds. This increase was seen in all participants including children with no known history of infection, suggesting that these participants might have been previously exposed to SARS-CoV-2 and that not all asymptomatic cases necessarily could be detected by serum antibodies. Furthermore, nAb levels against both strains were significantly correlated in adults (r=0.8788; p = 0.0008) and children (r = 0.7521; p = 0.0076), and they strongly correlated with S1 and RBD-specific IgG antibodies. Conclusion Our results provide evidence for persistent mucosal humoral memory in tonsils from previously infected and/or vaccinated adults and children against recent and old variants upon re-exposure. They also highlight the importance of targeting mucosal sites with vaccines to help control infection at the primary sites and prevent potential breakthrough infections.
Collapse
Affiliation(s)
- Tarfa A. Altorki
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Rwaa H. Abdulal
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Bandar A. Suliman
- Medical Laboratory Technology Department, College of Applied Medical Sciences, Taibah University, Madinah, Saudi Arabia
| | - Talal M. Aljeraisi
- Otorhinolaryngology, Head and Neck Surgery Department, Faculty of Medicine, Taibah University, Madinah, Saudi Arabia
| | - Asem Alsharef
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Wesam H. Abdulaal
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mohamed A. Alfaleh
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Abdullah A. Algaissi
- Department of Medical Laboratories Technology, College of Applied Medical Sciences, Jazan University, Jazan, Saudi Arabia
| | - Rowa Y. Alhabbab
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Hani Ozbak
- Medical Laboratory Technology Department, College of Applied Medical Sciences, Taibah University, Madinah, Saudi Arabia
| | - Hamza Mohammed Eid
- Medical Laboratory Technology Department, College of Applied Medical Sciences, Taibah University, Madinah, Saudi Arabia
| | - Yahya Ahmad Almutawif
- Medical Laboratory Technology Department, College of Applied Medical Sciences, Taibah University, Madinah, Saudi Arabia
| | - Xuguang Li
- Centre for Oncology and Regulatory Research, Biologic and Radiopharmaceutical Drugs Directorate, Health Products and Food Branch, Health Canada and World Health Organization Collaborating Center for Standardization and Evaluation of Biologicals, Ottawa, ON, Canada
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Mohammed W. Al-Rabia
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Qibo Zhang
- Academic and Research Departments, Section of Immunology, School of Biosciences and Medicine University of Surrey, Surrey, United Kingdom
| | - Ahmed Bakur Mahmoud
- Medical Laboratory Technology Department, College of Applied Medical Sciences, Taibah University, Madinah, Saudi Arabia
- Health and Life Research Center, Taibah University, Madinah, Saudi Arabia
| | - Waleed H. Mahallawi
- Medical Laboratory Technology Department, College of Applied Medical Sciences, Taibah University, Madinah, Saudi Arabia
| | - Anwar M. Hashem
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
26
|
Jafari M, Asli S, Moghbeli F, Fesharaki MG, Hajiahmadi N, Mojtahedzadeh F, Amel Jamehdar S, Bamdad T. Persistence of SARS-CoV-2-antibodies against N, S and RBD after natural infection. IRANIAN JOURNAL OF MICROBIOLOGY 2023; 15:803-810. [PMID: 38156306 PMCID: PMC10751617 DOI: 10.18502/ijm.v15i6.14161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/30/2023]
Abstract
Background and Objectives Coronavirus disease 2019 (COVID-19) pandemic has affected most countries in the world. Monitoring the humoral immune responses during the natural course of Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) infection and the duration of them provide useful information for the development of vaccination strategies against this virus and its emerging variants. The importance of the antibody response especially neutralizing antibodies in long-term immunity to SARS-CoV-2 is significant. Materials and Methods The present study is a cross-sectional study of sero-epidemiological type that has been proposed to compare the persistence of Immunoglobulin G (IgG) against N (nucleocapsid), S (spike) and RBD (receptor-binding domain) proteins in the community after the time of primary disease. A total of 652 serum samples were collected from hospital staff working in COVID wards, as well as a number of community members with different occupations, among those with positive antibody titers, 86 participated in the resampling test before vaccination. Results There was no association between antibody titer and disease severity (p>0.05). A significant decrease in Ab levels was observed in the paired second samples. The highest rate of decrease was related to anti-N, then anti-RBD and anti-S IgG levels, respectively. There is a significant relationship between the initial antibody titer and its reduction over time (p-value <0.05). Conclusion Our data revealed that humoral immunity following natural infection of SARS-CoV-2 is detectable for at least 4 months, regardless of disease severity. The most decrease in antibody titer over time was related to anti-N IgG levels.
Collapse
Affiliation(s)
- Mitra Jafari
- Department of Virology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Samira Asli
- Clinical Research Development, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fateme Moghbeli
- Department of Health Information Technology, Varastegan Institute for Medical Sciences, Mashhad, Iran
| | | | - Nazila Hajiahmadi
- Department of Virology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Faezeh Mojtahedzadeh
- Department of Virology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Saeid Amel Jamehdar
- Clinical Research Development, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Antimicrobial Resistance Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Taravat Bamdad
- Department of Virology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
27
|
Xu M, O’Brien MP, Hooper AT, Forleo-Neto E, Isa F, Hou P, Chan KC, Cohen MS, Marovich MA, Hamilton JD, Hirshberg B, Herman GA, Musser BJ. Nasopharyngeal Viral Load Is the Major Driver of Incident Antibody Immune Response to SARS-CoV-2 Infection. Open Forum Infect Dis 2023; 10:ofad598. [PMID: 38111750 PMCID: PMC10727195 DOI: 10.1093/ofid/ofad598] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 11/29/2023] [Indexed: 12/20/2023] Open
Abstract
Background Virologic determinants of seroconversion to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection were defined in a post hoc analysis of prospectively studied vaccine- and infection-naïve individuals at high risk for coronavirus disease 2019 (COVID-19). Methods This phase 3 COVID-19 prevention trial (NCT04452318) with casirivimab and imdevimab was conducted in July 2020-February 2021, before widespread vaccine availability. Placebo-treated participants who were uninfected (SARS-CoV-2 quantitative reverse transcription polymerase chain reaction [RT-qPCR] negative) and seronegative were assessed weekly for 28 days (efficacy assessment period [EAP]) for COVID-19 symptoms and SARS-CoV-2 infection by RT-qPCR of nasopharyngeal swab samples and for serostatus by antinucleocapsid immunoglobulin (Ig) G. Regression-based modeling, including causal mediation analysis, estimated the effects of viral load on seroconversion. Results Of 157/1069 (14.7%) uninfected and seronegative (for antispike IgG, antispike IgA, and antinucleocapsid IgG) participants who became infected during the EAP, 105 (65%) seroconverted. The mean (SD) maximum viral load of seroconverters was 7.23 (1.68) log10 copies/mL vs 4.8 (2.2) log10 copies/mL in those who remained seronegative; viral loads of ∼6.0 log10 copies/mL better predicted seroconversion. The mean of the maximum viral load was 7.11 log10 copies/mL in symptomatic participants vs 5.58 log10 copies/mL in asymptomatic participants. The mean duration of detectable viral load was longer in seroconverted vs seronegative participants: 3.24 vs 1.63 weeks. Conclusions Maximum SARS-CoV-2 viral load is a major driver of seroconversion and symptomatic COVID-19, with high viral loads (∼6.0 log10 copies/mL) better predicting seroconversion. Serology underestimates infection rates, incidence, and prevalence of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Meng Xu
- Regeneron Pharmaceuticals, Inc., Tarrytown, NewYork, USA
| | | | | | | | - Flonza Isa
- Regeneron Pharmaceuticals, Inc., Tarrytown, NewYork, USA
| | - Peijie Hou
- Regeneron Pharmaceuticals, Inc., Tarrytown, NewYork, USA
| | - Kuo-Chen Chan
- Regeneron Pharmaceuticals, Inc., Tarrytown, NewYork, USA
| | - Myron S Cohen
- University of North Carolina Chapel Hill School of Medicine, Institute for Global Health and Infectious Diseases, Chapel Hill, North Carolina, USA
| | - Mary A Marovich
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| | | | - Boaz Hirshberg
- Regeneron Pharmaceuticals, Inc., Tarrytown, NewYork, USA
| | - Gary A Herman
- Regeneron Pharmaceuticals, Inc., Tarrytown, NewYork, USA
| | - Bret J Musser
- Regeneron Pharmaceuticals, Inc., Tarrytown, NewYork, USA
| |
Collapse
|
28
|
Pitiriga VC, Papamentzelopoulou M, Konstantinakou KE, Vasileiou IV, Sakellariou KS, Spyrou NI, Tsakris A. Persistence of T-Cell Immunity Responses against SARS-CoV-2 for over 12 Months Post COVID-19 Infection in Unvaccinated Individuals with No Detectable IgG Antibodies. Vaccines (Basel) 2023; 11:1764. [PMID: 38140169 PMCID: PMC10747023 DOI: 10.3390/vaccines11121764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 11/09/2023] [Accepted: 11/24/2023] [Indexed: 12/24/2023] Open
Abstract
BACKGROUND Immune response to SARS-CoV-2 is crucial for preventing reinfection or reducing disease severity. T-cells' long-term protection, elicited either by COVID-19 vaccines or natural infection, has been extensively studied thus far; however, it is still attracting considerable scientific interest. The aim of the present epidemiological study was to define the levels of T-cellular immunity response in a specific group of unvaccinated individuals from the general population with a prior confirmed COVID-19 infection and no measurable levels of IgG antibodies. METHODS We performed a retrospective descriptive analysis of data collected from the medical records of consecutive unvaccinated individuals recovered from COVID-19, who had proceeded to a large private medical center in the Attica region from September 2021 to September 2022 in order to be examined on their own initiative for SARS-CoV-2 T-cell immunity response. The analysis of T-cell responses was divided into three time periods post infection: Group A: up to 6 months; Group B: 6-12 months; Group C: >12 months. The SARS-CoV-2 T-cell response was estimated against spike (S) and nucleocapsid (N) structural proteins by performing the T-SPOT. COVID test methodology. SARS-CoV-2 IgG antibody levels were measured by the SARS-CoV-2 IgG II Quant assay (Abbott Diagnostics). RESULTS A total of 182 subjects were retrospectively included in the study, 85 females (46.7%) and 97 (53.3%) males, ranging from 19 to 91 years old (mean 50.84 ± 17.2 years). Among them, 59 (32.4%) had been infected within the previous 6 months from the examination date (Group A), 69 (37.9%) had been infected within a time period > 6 months and <1 year (Group B) and 54 (29.7%) had been infected within a time period longer than 1 year from the examination date (Group C). Among the three groups, a positive T-cell reaction against the S antigen was reported in 47/58 (81%) of Group A, 61/69 (88.4%) of Group B and 40/54 (74.1%) of Group C (chi square, p = 0.27). T-cell reaction against the N antigen was present in 45/58 (77.6%) of Group A, 61/69 (88.4%) of Group B and 36/54 (66.7%) of Group C (chi square, p = 0.02). The median Spot-Forming Cells (SFC) count for the S antigen was 18 (range from 0-160) in Group A, 19 (range from 0-130) in Group B and 17 (range from 0-160) in Group C (Kruskal-Wallis test, p = 0.11; pairwise comparisons: groups A-B, p = 0.95; groups A-C, p = 0.89; groups B-C, p = 0.11). The median SFCs count for the N antigen was 14.5 (ranging from 0 to 116) for Group A, 24 (ranging from 0-168) in Group B and 16 (ranging from 0-112) for Group C (Kruskal-Wallis test, p = 0.01; pairwise comparisons: groups A-B, p = 0.02; groups A-C, p = 0.97; groups B-C, p = 0.03). CONCLUSIONS Our data suggest that protective adaptive T-cellular immunity following natural infection by SARS-CoV-2 may persist for over 12 months, despite the undetectable humoral element.
Collapse
Affiliation(s)
- Vassiliki C. Pitiriga
- Department of Microbiology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Street, 11527 Athens, Greece;
| | - Myrto Papamentzelopoulou
- Molecular Biology Unit, 1st Department of Obstetrics and Gynecology, National and Kapodistrian University of Athens, 11528 Athens, Greece;
| | - Kanella E. Konstantinakou
- Bioiatriki Healthcare Group, Kifisias 132 and Papada Street, 11526 Athens, Greece; (K.E.K.); (I.V.V.); (K.S.S.); (N.I.S.)
| | - Irene V. Vasileiou
- Bioiatriki Healthcare Group, Kifisias 132 and Papada Street, 11526 Athens, Greece; (K.E.K.); (I.V.V.); (K.S.S.); (N.I.S.)
| | - Konstantina S. Sakellariou
- Bioiatriki Healthcare Group, Kifisias 132 and Papada Street, 11526 Athens, Greece; (K.E.K.); (I.V.V.); (K.S.S.); (N.I.S.)
| | - Natalia I. Spyrou
- Bioiatriki Healthcare Group, Kifisias 132 and Papada Street, 11526 Athens, Greece; (K.E.K.); (I.V.V.); (K.S.S.); (N.I.S.)
| | - Athanasios Tsakris
- Department of Microbiology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Street, 11527 Athens, Greece;
| |
Collapse
|
29
|
Pathakumari B, Marty PK, Shah M, Van Keulen VP, Erskine CL, Block MS, Arias-Sanchez P, Escalante P, Peikert T. Convalescent Adaptive Immunity Is Highly Heterogenous after SARS-CoV-2 Infection. J Clin Med 2023; 12:7136. [PMID: 38002748 PMCID: PMC10672050 DOI: 10.3390/jcm12227136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 11/03/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023] Open
Abstract
The optimal detection strategies for effective convalescent immunity after SARS-CoV-2 infection and vaccination remain unclear. The objective of this study was to characterize convalescent immunity targeting the SARS-CoV-2 spike protein using a multiparametric approach. At the beginning of the pandemic, we recruited 30 unvaccinated convalescent donors who had previously been infected with COVID-19 and 7 unexposed asymptomatic controls. Peripheral blood mononuclear cells (PBMCs) were obtained from leukapheresis cones. The humoral immune response was assessed by measuring serum anti-SARS-CoV-2 spike S1 subunit IgG via semiquantitative ELISA, and T-cell immunity against S1 and S2 subunits were studied via IFN-γ enzyme-linked immunosorbent spot (ELISpot) and flow cytometric (FC) activation-induced marker (AIM) assays and the assessment of cytotoxic CD8+ T-cell function (in the subset of HLA-A2-positive patients). No single immunoassay was sufficient in identifying anti-spike convalescent immunity among all patients. There was no consistent correlation between adaptive humoral and cellular anti-spike responses. Our data indicate that the magnitude of anti-spike convalescent humoral and cellular immunity is highly heterogeneous and highlights the need for using multiple assays to comprehensively measure SARS-CoV-2 convalescent immunity. These observations might have implications for COVID-19 surveillance, and the determination of optimal vaccination strategies for emerging variants. Further studies are needed to determine the optimal assessment of adaptive humoral and cellular immunity following SARS-CoV-2 infection, especially in the context of emerging variants and unclear vaccination schedules.
Collapse
Affiliation(s)
- Balaji Pathakumari
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA; (B.P.); (P.K.M.); (M.S.); (V.P.V.K.); (P.A.-S.); (P.E.)
| | - Paige K. Marty
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA; (B.P.); (P.K.M.); (M.S.); (V.P.V.K.); (P.A.-S.); (P.E.)
| | - Maleeha Shah
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA; (B.P.); (P.K.M.); (M.S.); (V.P.V.K.); (P.A.-S.); (P.E.)
| | - Virginia P. Van Keulen
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA; (B.P.); (P.K.M.); (M.S.); (V.P.V.K.); (P.A.-S.); (P.E.)
- Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA; (C.L.E.); (M.S.B.)
| | - Courtney L. Erskine
- Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA; (C.L.E.); (M.S.B.)
| | - Matthew S. Block
- Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA; (C.L.E.); (M.S.B.)
- Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA
| | - Pedro Arias-Sanchez
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA; (B.P.); (P.K.M.); (M.S.); (V.P.V.K.); (P.A.-S.); (P.E.)
| | - Patricio Escalante
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA; (B.P.); (P.K.M.); (M.S.); (V.P.V.K.); (P.A.-S.); (P.E.)
| | - Tobias Peikert
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA; (B.P.); (P.K.M.); (M.S.); (V.P.V.K.); (P.A.-S.); (P.E.)
- Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA; (C.L.E.); (M.S.B.)
| |
Collapse
|
30
|
Guan X, Huang Q, Dong M, Li M, Xie H, Wei X, Kang L, Wang X, Li A, Wang Q, Huang F, Wang Q. SARS-CoV-2-specific antibody and T-cell immunity in convalescents after infection wave in Beijing in late 2022. J Infect 2023; 87:413-419. [PMID: 37652314 DOI: 10.1016/j.jinf.2023.08.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 08/24/2023] [Accepted: 08/27/2023] [Indexed: 09/02/2023]
Abstract
OBJECTIVES To evaluate SARS-CoV-2-specific antibody and T-cell responses in convalescents 5 months after infection wave in Beijing from December 2022 to January 2023 to prevent reinfection and severe disease. METHODS Convalescents and uninfected individuals vaccinated with different doses were enrolled to assess the IFNγ T-cell responses against SARS-CoV-2 prototype strain, BF.7, BQ.1, and XBB. Neutralizing antibodies against prototype strain, BF.7, BA.5, and XBB and immunoglobulin G antibody were further analyzed. RESULTS In convalescents, the IFNγ T-cell response was significantly higher than that of uninfected individuals (all P < 0.001), and the T-cell response against XBB had no significant difference from that of SARS-CoV-2 prototype strain and BF.7 and BQ.1 (all P > 0.05). The seropositive rates of IgG antibodies were 100% (303/303) with a median concentration of 90.52 (95% CI, 82.52-99.37). The neutralizing antibodies titers of convalescents against BF.7 and BA.5 were higher than that against the prototype strain (both P < 0.001), while XBB.1.5 was lower (P < 0.001). T-cell response, IgG and neutralizing antibodies had no significant difference in convalescents vaccinated with different doses (all P > 0.05). CONCLUSIONS The immunities may have some protective effect against possible future outbreaks and severe diseases of COVID-19.
Collapse
Affiliation(s)
- Xuejiao Guan
- Beijing Center for Disease Prevention and Control, Beijing Academy for Preventive Medicine, Beijing Institute of Tuberculosis Control Research and Prevention, Beijing 100013, People's Republic of China
| | - Qi Huang
- Beijing Center for Disease Prevention and Control, Beijing Academy for Preventive Medicine, Beijing Institute of Tuberculosis Control Research and Prevention, Beijing 100013, People's Republic of China; School of Public Health, Capital Medical University, Beijing 100069, People's Republic of China
| | - Mei Dong
- Beijing Center for Disease Prevention and Control, Beijing Academy for Preventive Medicine, Beijing Institute of Tuberculosis Control Research and Prevention, Beijing 100013, People's Republic of China
| | - Maozhong Li
- Beijing Center for Disease Prevention and Control, Beijing Academy for Preventive Medicine, Beijing Institute of Tuberculosis Control Research and Prevention, Beijing 100013, People's Republic of China
| | - Hui Xie
- Beijing Center for Disease Prevention and Control, Beijing Academy for Preventive Medicine, Beijing Institute of Tuberculosis Control Research and Prevention, Beijing 100013, People's Republic of China
| | - Xiaofeng Wei
- Beijing Center for Disease Prevention and Control, Beijing Academy for Preventive Medicine, Beijing Institute of Tuberculosis Control Research and Prevention, Beijing 100013, People's Republic of China; School of Public Health, Capital Medical University, Beijing 100069, People's Republic of China
| | - Lu Kang
- Beijing Center for Disease Prevention and Control, Beijing Academy for Preventive Medicine, Beijing Institute of Tuberculosis Control Research and Prevention, Beijing 100013, People's Republic of China
| | - Xue Wang
- Beijing Center for Disease Prevention and Control, Beijing Academy for Preventive Medicine, Beijing Institute of Tuberculosis Control Research and Prevention, Beijing 100013, People's Republic of China
| | - Aihua Li
- Beijing Center for Disease Prevention and Control, Beijing Academy for Preventive Medicine, Beijing Institute of Tuberculosis Control Research and Prevention, Beijing 100013, People's Republic of China
| | - Qing Wang
- Beijing Center for Disease Prevention and Control, Beijing Academy for Preventive Medicine, Beijing Institute of Tuberculosis Control Research and Prevention, Beijing 100013, People's Republic of China; School of Public Health, Capital Medical University, Beijing 100069, People's Republic of China
| | - Fang Huang
- Beijing Center for Disease Prevention and Control, Beijing Academy for Preventive Medicine, Beijing Institute of Tuberculosis Control Research and Prevention, Beijing 100013, People's Republic of China; Beijing Research Center for Respiratory Infectious Diseases, People's Republic of China.
| | - Quanyi Wang
- Beijing Center for Disease Prevention and Control, Beijing Academy for Preventive Medicine, Beijing Institute of Tuberculosis Control Research and Prevention, Beijing 100013, People's Republic of China; Beijing Research Center for Respiratory Infectious Diseases, People's Republic of China.
| |
Collapse
|
31
|
Bean DJ, Monroe J, Liang YM, Borberg E, Senussi Y, Swank Z, Chalise S, Walt D, Weinberg J, Sagar M. Heterotypic responses against nsp12/nsp13 from prior SARS-CoV-2 infection associates with lower subsequent endemic coronavirus incidence. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.23.563621. [PMID: 37961343 PMCID: PMC10634759 DOI: 10.1101/2023.10.23.563621] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Immune responses from prior SARS-CoV-2 infection and COVID-19 vaccination do not prevent re-infections and may not protect against future novel coronaviruses (CoVs). We examined the incidence of and immune differences against human endemic CoVs (eCoV) as a proxy for response against future emerging CoVs. Assessment was among those with known SARS-CoV-2 infection, COVID-19 vaccination but no documented SARS-CoV-2 infection, or neither exposure. Retrospective cohort analyses suggest that prior SARS-CoV-2 infection, but not COVID-19 vaccination alone, protects against subsequent symptomatic eCoV infection. CD8+ T cell responses to the non-structural eCoV proteins, nsp12 and nsp13, were significantly higher in individuals with previous SARS-CoV-2 infection as compared to the other groups. The three groups had similar cellular responses against the eCoV spike and nucleocapsid, and those with prior spike exposure had lower eCoV-directed neutralizing antibodies. Incorporation of non-structural viral antigens in a future pan-CoV vaccine may improve protection against future heterologous CoV infections.
Collapse
Affiliation(s)
- David J. Bean
- Department of Virology, Immunology and Microbiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA
| | - Janet Monroe
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA
| | - Yan Mei Liang
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA
| | - Ella Borberg
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA
| | - Yasmeen Senussi
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA
| | - Zoe Swank
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA
| | - Sujata Chalise
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA
| | - David Walt
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA
| | - Janice Weinberg
- Department of Biostatistics, Boston University School of Public Health, Boston, MA
| | - Manish Sagar
- Department of Virology, Immunology and Microbiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA
| |
Collapse
|
32
|
Tamburini B, Doan T, Forward T, Lucas E, Fleming I, Uecker-Martin A, Hesselberth J, Morrison T. Vaccine-induced antigen archiving enhances local memory CD8+ T cell responses following an unrelated viral infection. RESEARCH SQUARE 2023:rs.3.rs-3307809. [PMID: 37841845 PMCID: PMC10571600 DOI: 10.21203/rs.3.rs-3307809/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/17/2023]
Abstract
Viral and vaccine antigens persist or are archived in lymph node stromal cells (LNSC) such as lymphatic endothelial cells (LEC) and fibroblastic reticular cells (FRC). Here, we find that, during the time frame of antigen archiving, LEC apoptosis caused by a second, but unrelated, innate immune stimulus such as vaccina viral infection or CpG DNA administration boosted memory CD8+ T cells specific to the archived antigen. In contrast to "bystander" activation associated with unrelated infections, the memory CD8+ T cells specific to the vaccine archived antigen were significantly higher than memory CD8+ T cells of a different antigen specificity. Finally, the boosted memory CD8+ T cells resulted in increased protection against Listeria monocytogenes expressing the vaccine antigen, but only for the duration that the vaccine antigen was archived. These findings outline a novel mechanism by which LNSC archived antigens, in addition to bystander activation, can augment memory CD8+ T cell responses during repeated inflammatory insults.
Collapse
Affiliation(s)
| | - Thu Doan
- University of Colorado Anschutz Medical Campus
| | | | - Erin Lucas
- University of Colorado Anschutz Medical Campus
| | - Ira Fleming
- University of Colorado Anschutz Medical Campus
| | | | | | | |
Collapse
|
33
|
Marty PK, Pathakumari B, Shah M, Keulen VP, Erskine CL, Block MS, Arias-Sanchez P, Escalante P, Peikert T. Convalescent Adaptive Immunity is Highly Heterogenous after SARS-CoV-2 Infection. RESEARCH SQUARE 2023:rs.3.rs-3222112. [PMID: 37674707 PMCID: PMC10479471 DOI: 10.21203/rs.3.rs-3222112/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/08/2023]
Abstract
Optimal detection strategies for effective convalescent immunity after SARS-CoV-2 infection and vaccination remain unclear. The objective of this study was to characterize convalescent immunity targeting the SARS-CoV-2 spike protein using a multiparametric approach. At the beginning of the pandemic, between April 23, 2020, to May 11, 2020, we recruited 30 COVID-19 unvaccinated convalescent donors and 7 unexposed asymptomatic donors. Peripheral blood mononuclear cells (PBMCs) were obtained from leukapheresis cones. The humoral immune response was assessed by measuring serum anti-SARS-CoV-2 spike S1 subunit IgG semiquantitative ELISA and T cell immunity against S1 and S2 subunits were studied by IFN-γ Enzyme-Linked Immune absorbent Spot (ELISpot), flow cytometric (FC) activation-induced marker (AIM) assays and the assessment of cytotoxic CD8+ T-cell function (in the subset of HLA-A2 positive patients). No single immunoassay was sufficient in identifying anti-spike convalescent immunity among all patients. There was no consistent correlation between adaptive humoral and cellular anti-spike responses. Our data indicate that the magnitude of anti-spike convalescent humoral and cellular immunity is highly heterogeneous and highlights the need for using multiple assays to comprehensively measure SARS-CoV-2 convalescent immunity. These observations might have implications for COVID-19 surveillance, and optimal vaccination strategies for emerging variants. Further studies are needed to determine the optimal assessment of adaptive humoral and cellular immunity following SARSCoV-2 infection, especially in the context of emerging variants and unclear vaccination schedules.
Collapse
|
34
|
Antinori A, Bausch-Jurken M. The Burden of COVID-19 in the Immunocompromised Patient: Implications for Vaccination and Needs for the Future. J Infect Dis 2023; 228:S4-S12. [PMID: 37539764 PMCID: PMC10401620 DOI: 10.1093/infdis/jiad181] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/05/2023] Open
Abstract
Approximately 3% of US adults are immunocompromised and less capable of fighting infections such as SARS-CoV-2 (the causative agent of COVID-19). Individuals may be immunocompromised for reasons related to an underlying medical condition or to immunomodulatory therapies that alter the immune response. In general, vaccination with mRNA-based vaccines is effective at reducing COVID-19-associated hospitalization and death among immunocompromised populations, particularly after 3 or more doses. However, the immunocompromised population is heterogeneous, with COVID-19 vaccine-elicited immune responses and risk for severe COVID-19 existing on a continuum. Therefore, understanding the impact of vaccination and the complexity of immune responses across heterogeneous immunocompromised individuals is essential for guiding effective vaccination regimens including additional (booster) doses. In this article, we provide an overview of the immunocompromised population and the burden of disease attributable to COVID-19, while discussing key opportunities and challenges of vaccinating immunocompromised individuals.
Collapse
Affiliation(s)
- Andrea Antinori
- Correspondence: Andrea Antinori, MD, National Institute for Infectious Diseases Lazzaro Spallanzani, IRCCS, Via Portuense, 292, 00149 Roma RM, Italy (); Mary Bausch-Jurken, PhD, Moderna, Inc., 200 Technology Square, Cambridge, MA 02139, USA ()
| | - Mary Bausch-Jurken
- Correspondence: Andrea Antinori, MD, National Institute for Infectious Diseases Lazzaro Spallanzani, IRCCS, Via Portuense, 292, 00149 Roma RM, Italy (); Mary Bausch-Jurken, PhD, Moderna, Inc., 200 Technology Square, Cambridge, MA 02139, USA ()
| |
Collapse
|
35
|
Kowatsch MM, Lajoie J, Mwangi L, Omollo K, Oyugi J, Hollett N, Kimani J, Fowke KR. Hydroxychloroquine reduces T cells activation recall antigen responses. PLoS One 2023; 18:e0287738. [PMID: 37531383 PMCID: PMC10395872 DOI: 10.1371/journal.pone.0287738] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 06/04/2023] [Indexed: 08/04/2023] Open
Abstract
BACKGROUND In the context of the current COVID-19 pandemic, there is still limited information about how people suffering from autoimmune diseases respond to the different COVID vaccines. The fact that they are taking an immunosuppressant or other drugs that aim to decrease the immune system activities, such as hydroxychloroquine (HCQ), could also impact their ability to respond to a COVID vaccine and vaccines in general. METHODS Heathy donors were given 200mg of HCQ daily for 6-weeks to assess HCQs impact on the systemic T cells and humoral immune response. Peripheral blood mononuclear cells (PBMC) and plasma were obtained at baseline and 6-weeks after starting daily HCQ. Flow cytometry assays were designed to determine changes in T cell activation and T cell responses. Bead array multiplex were used to analyse antibodies and cytokine levels before and after HCQ intake. RESULTS As anticipated, HCQ treatment decreased ex vivo T cell activation. We observed a decrease in CD4+CD161- expressing CCR5 (p = 0.015) and CD69 (p = 0.004) as well as in CD8+CCR5+ (p = 0.003), CD8+CD161+CCR5+ (p = 0.002) and CD8+CD161+CD95+ (p = 0.004). Additionally, HCQ decreased the proportion of Th17 expressing CD29 (p = 0.019), a subset associated with persistent inflammation. The proportion of T regulatory cells expressing the inhibitory molecule TIGIT was also reduced by HCQ (p = 0.003). As well, T cells from people on HCQ were less responsive to activation and cytokine production following stimulation with recall antigens and memory T cells were less likely to produce both IFNγ and TNFα following stimulation. CONCLUSION This study shows HCQ is associated with lower T cell activation and decreased T cell cytokine production. While this study was not performed with the intent of looking at COVID vaccine response, it does provide important information about the changes in immune response that may occur in patient taking HCQ as a treatment for their autoimmune disease.
Collapse
Affiliation(s)
- Monika M Kowatsch
- Laboratory of Viral Immunology, Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Julie Lajoie
- Laboratory of Viral Immunology, Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada
- Department of Medical Microbiology, University of Nairobi, Nairobi, Kenya
| | - Lucy Mwangi
- Department of Medical Microbiology, University of Nairobi, Nairobi, Kenya
| | - Kenneth Omollo
- Department of Medical Microbiology, University of Nairobi, Nairobi, Kenya
| | - Julius Oyugi
- Laboratory of Viral Immunology, Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada
- Partners for Health and Development in Africa, Nairobi, Kenya
- University of Nairobi Institute for Tropical and Infectious Diseases, University of Nairobi, Nairobi, Kenya
| | - Natasha Hollett
- Laboratory of Viral Immunology, Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Joshua Kimani
- Laboratory of Viral Immunology, Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada
- Partners for Health and Development in Africa, Nairobi, Kenya
- University of Nairobi Institute for Tropical and Infectious Diseases, University of Nairobi, Nairobi, Kenya
| | - Keith R Fowke
- Laboratory of Viral Immunology, Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada
- Department of Medical Microbiology, University of Nairobi, Nairobi, Kenya
- Partners for Health and Development in Africa, Nairobi, Kenya
- Department of Community Health Science, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
36
|
Li Q, Chen L, Li F, He A. Long-term evaluation of the seroprevalence of SARS-CoV-2 IgG and IgM antibodies in recovered patients: a meta-analysis. BMC Infect Dis 2023; 23:444. [PMID: 37393304 DOI: 10.1186/s12879-023-08425-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 06/24/2023] [Indexed: 07/03/2023] Open
Abstract
Estimating severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) -specific immunoglobulin G (IgG) immunoglobulin M (IgM) antibodies are increasingly important for tracking the spread of infection and defining herd immunity barrier and individual immunization levels in the ongoing coronavirus disease 2019 (COVID-19) pandemic. Therefore, we conducted the present systematic review and meta-analysis to evaluate the seroprevalence of SARS-CoV-2 IgM and IgG antibodies of recovered COVID-19 patients in long-term follow-up studies. A systematic search of the MEDLINE, Embase, COVID-19 Primer, PubMed, CNKI, and the Public Health England library databases was conducted. Twenty-fourth eligible studies were included. Meta-analysis showed that 27% (95%CI: 0.04-0.49) and 66% (95%CI:0.47-0.85) were seropositive for SARS-CoV-2 IgM and IgG, respectively, while in long-term 12 months following up studies, the seroprevalences of IgM antibody (17%) decreased and IgG antibody (75%) was higher than 6 months follow-up patients. However, due to the limited number of relevant studies, the high level of heterogeneity, and the large gap in studies conducted, the findings of our study may not accurately reflect the true seroprevalence status of SARS-CoV-2 infection. Nevertheless, sequential vaccination or booster immunization is considered to be a necessary long-term strategy to sustain the fight against the pandemic.
Collapse
Affiliation(s)
- Qiu Li
- Laboratory Medicine Center, Chenzhou First People's Hospital, Chenzhou, 423000, P.R. China
| | - Lu Chen
- Baoshan Community Hospital, Chenzhou, 424400, P.R. China
| | - Fen Li
- Laboratory Medicine Center, Chenzhou First People's Hospital, Chenzhou, 423000, P.R. China
| | - An He
- Laboratory Medicine Center, Chenzhou First People's Hospital, Chenzhou, 423000, P.R. China.
| |
Collapse
|
37
|
Gonzales MLAM, Dans LF, Tan-Lim CSC, Uy E, Cutiongco-dela Paz E, Sulit MVV, Alejandria MM, Lansang MAD, Dans AL, Dator MA, Cordero CP, Pardilla GF. Durability and extent of protection of SARS-CoV-2 antibodies among patients with COVID-19 in Metro Manila, Philippines. Front Immunol 2023; 14:1190093. [PMID: 37457735 PMCID: PMC10338854 DOI: 10.3389/fimmu.2023.1190093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 06/09/2023] [Indexed: 07/18/2023] Open
Abstract
Introduction Information on the magnitude and durability of humoral immunity against COVID-19 among specific populations can guide policies on vaccination, return from isolation and physical distancing measures. The study determined the durability of SARS-CoV-2 antibodies after an initial infection among Filipinos in Metro Manila, Philippines, and the extent of protection SARS-CoV-2 antibodies confer against reinfection. Methods We conducted a cohort study to monitor the antibody levels of patients diagnosed with COVID-19. Receptor-binding domain (RBD)-specific antibodies were measured at Days 21, 90, 180, 270 and 360. Antibody levels were reported as geometric mean titers (GMT) with geometric standard deviation (GSD). Differences in GMT were tested using Friedman test and Kruskal Wallis test, with Bonferroni multiple comparisons procedure. Adjusted hazard ratios on the development of probable reinfection were estimated using Cox proportional models. Results There were 307 study participants included in the study, with 13 dropouts. Study participants received SARS-CoV-2 vaccines at varying times, with 278 participants (90.5%) fully vaccinated by the end of study. The GMT of the study cohort increased over time, from 19.7 U/mL (GSD 11) at Day 21; to 284.5 U/mL (GSD 9.6) at Day 90; 1,061 U/mL (GSD 5.3) at Day 180; 2,003 U/mL (GSD 6.7) at Day 270; and 8,403 U/mL (GSD 3.1) at Day 360. The increase was statistically significant from Day 21 to Day 90 (p<0.0001), Day 90 to Day 180 (p=0.0005), and Day 270 to Day 360 (p<0.0001). Participants with more severe initial infection demonstrated significantly higher antibody levels compared to those with milder infection at Day 21. Sixty-four patients had probable COVID-19 reinfection (incidence of 20.8%, 95% CI 16.4, 25.8%). The GMT of these 64 patients was 411.8 U/mL (GSD 6.9) prior to the occurrence of the probable reinfection. Majority (87.5%) were fully vaccinated. Antibody titers significantly affected the risk of developing reinfection, with adjusted hazard ratio of 0.994, 95% CI 0.992-0.996, p<0.001. Conclusion Antibody levels against SARS-CoV-2 increased over a one-year follow-up. Higher antibody levels were observed among those with more severe initial infection and those vaccinated. Higher antibody levels are associated with a lower risk of probable reinfection.
Collapse
Affiliation(s)
| | - Leonila F. Dans
- Department of Pediatrics, College of Medicine, University of the Philippines Manila, Manila, Philippines
- Department of Clinical Epidemiology, College of Medicine, University of the Philippines Manila, Manila, Philippines
| | - Carol Stephanie C. Tan-Lim
- Department of Clinical Epidemiology, College of Medicine, University of the Philippines Manila, Manila, Philippines
| | - Elenore Uy
- Asia-Pacific Centre for Evidence-Based Healthcare, Manila, Philippines
| | - Eva Cutiongco-dela Paz
- Institute of Human Genetics, National Institutes of Health, University of the Philippines Manila, Manila, Philippines
| | - Maria Vanessa V. Sulit
- Institute of Clinical Epidemiology, National Institutes of Health, University of the Philippines Manila, Manila, Philippines
| | - Marissa M. Alejandria
- Department of Clinical Epidemiology, College of Medicine, University of the Philippines Manila, Manila, Philippines
| | - Mary Ann D. Lansang
- Department of Clinical Epidemiology, College of Medicine, University of the Philippines Manila, Manila, Philippines
| | - Antonio L. Dans
- College of Medicine, University of the Philippines Manila, Manila, Philippines
| | - Melissa A. Dator
- Department of Pediatrics, College of Medicine, University of the Philippines Manila, Manila, Philippines
| | - Cynthia P. Cordero
- Department of Clinical Epidemiology, College of Medicine, University of the Philippines Manila, Manila, Philippines
| | - Gina F. Pardilla
- Manila Health Department Delpan Evacuation Center Quarantine Facility, Manila, Philippines
| |
Collapse
|
38
|
Seo YB, Ko A, Shin D, Kim J, Suh YS, Na J, Ryu JI, Lee S, Oh MJ, Sung YC. Potentiating the Cross-Reactive IFN-γ T Cell and Polyfunctional T Cell Responses by Heterologous GX-19N DNA Booster in Mice Primed with Either a COVID-19 mRNA Vaccine or Inactivated Vaccine. Int J Mol Sci 2023; 24:ijms24119753. [PMID: 37298704 DOI: 10.3390/ijms24119753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 05/31/2023] [Accepted: 05/31/2023] [Indexed: 06/12/2023] Open
Abstract
Waning vaccine-induced immunity, coupled with the emergence of SARS-CoV-2 variants, has inspired the widespread implementation of COVID-19 booster vaccinations. Here, we evaluated the potential of the GX-19N DNA vaccine as a heterologous booster to enhance the protective immune response to SARS-CoV-2 in mice primed with either an inactivated virus particle (VP) or an mRNA vaccine. We found that in the VP-primed condition, GX-19N enhanced the response of both vaccine-specific antibodies and cross-reactive T Cells to the SARS-CoV-2 variant of concern (VOC), compared to the homologous VP vaccine prime-boost. Under the mRNA-primed condition, GX-19N induced higher vaccine-induced T Cell responses but lower antibody responses than the homologous mRNA vaccine prime-boost. Furthermore, the heterologous GX-19N boost induced higher S-specific polyfunctional CD4+ and CD8+ T cell responses than the homologous VP or mRNA prime-boost vaccinations. Our results provide new insights into booster vaccination strategies for the management of novel COVID-19 variants.
Collapse
Affiliation(s)
- Yong Bok Seo
- Research Institute, SL VaxiGen Inc., Korea Bio Park, Seongnam 13488, Republic of Korea
| | - Ara Ko
- Research Institute, SL VaxiGen Inc., Korea Bio Park, Seongnam 13488, Republic of Korea
| | - Duckhyang Shin
- Research Institute, Genexine Inc., Korea Bio Park, Seongnam 13488, Republic of Korea
| | - Junyoung Kim
- Research Institute, SL VaxiGen Inc., Korea Bio Park, Seongnam 13488, Republic of Korea
| | - You Suk Suh
- Research Institute, Genexine Inc., Korea Bio Park, Seongnam 13488, Republic of Korea
| | - Juyoung Na
- Research Institute, Genexine Inc., Korea Bio Park, Seongnam 13488, Republic of Korea
| | - Ji In Ryu
- Research Institute, SL VaxiGen Inc., Korea Bio Park, Seongnam 13488, Republic of Korea
| | - Suyeon Lee
- Research Institute, SL VaxiGen Inc., Korea Bio Park, Seongnam 13488, Republic of Korea
| | - Min Ji Oh
- Research Institute, SL VaxiGen Inc., Korea Bio Park, Seongnam 13488, Republic of Korea
| | - Young Chul Sung
- Research Institute, Genexine Inc., Korea Bio Park, Seongnam 13488, Republic of Korea
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang 37673, Republic of Korea
| |
Collapse
|
39
|
Huang Q, Jia M, Sun Y, Jiang B, Cui D, Feng L, Yang W. One-Year Temporal Changes in Long COVID Prevalence and Characteristics: A Systematic Review and Meta-Analysis. VALUE IN HEALTH : THE JOURNAL OF THE INTERNATIONAL SOCIETY FOR PHARMACOECONOMICS AND OUTCOMES RESEARCH 2023; 26:934-942. [PMID: 36436792 DOI: 10.1016/j.jval.2022.11.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 10/24/2022] [Accepted: 11/16/2022] [Indexed: 06/04/2023]
Abstract
OBJECTIVES This study aimed to explore the 1-year temporal change in prevalence, variety, and potential risk factors of long COVID symptoms and to further predict the prognostic trends of long COVID. METHODS We searched electronic databases for related studies published from January 2020 to February 2022 and conducted 1-group meta-analysis and locally weighted regression to explore the monthly temporal change in the prevalence of each long COVID symptom in 1-year follow-up period. RESULTS A total of 137 studies were included in meta-analysis, including 134 093 participants. The temporal change of any long COVID symptom showed a steep decrease initially (from 92% at acute phase to 55% at 1-month follow-up), followed by stabilization at approximately 50% during 1-year follow-up. Six months or more after the acute phase, the odds ratio of population characteristic-related factors increased, such as female (from 1.62 to 1.82), whereas the odds ratio value of acute phase-related factors (severe or critical cases and hospitalization) decreased. As for specific symptoms, approximately two-thirds of the symptoms did not significantly reduce during the 1-year follow-up, and the neuropsychiatric symptoms showed a higher long-term prevalence (approximately 25%) and longer persistence than physical symptoms. CONCLUSIONS The temporal changes in the prevalence and characteristics speculate that long COVID may persist longer than expected. In particular, we should pay more attention to neuropsychiatric symptoms and other symptoms for which there is no significant downward trend in prevalence. The influence of acute phase-related factors for long COVID gradually decreases over time, whereas the influence of population characteristic-related factors gradually increases.
Collapse
Affiliation(s)
- Qiangru Huang
- School of Population Medicine and Public Health, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Mengmeng Jia
- School of Population Medicine and Public Health, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yanxia Sun
- School of Population Medicine and Public Health, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Binshan Jiang
- School of Population Medicine and Public Health, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Dan Cui
- Department of Pulmonary and Critical Care Medicine, The 2nd Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, China; Department of Pulmonary and Critical Care Medicine, National Center for Respiratory Medicine, Center of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, China-Japan Friendship Hospital, Beijing, China
| | - Luzhao Feng
- School of Population Medicine and Public Health, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Weizhong Yang
- School of Population Medicine and Public Health, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
40
|
Doucette EJ, Gray J, Fonseca K, Charlton C, Kanji JN, Tipples G, Kuhn S, Dunn J, Sayers P, Symonds N, Wu G, Freedman SB, Kellner JD. A longitudinal seroepidemiology study to evaluate antibody response to SARS-CoV-2 virus infection and vaccination in children in Calgary, Canada from July 2020 to April 2022: Alberta COVID-19 Childhood Cohort (AB3C) Study. PLoS One 2023; 18:e0284046. [PMID: 37023007 PMCID: PMC10079115 DOI: 10.1371/journal.pone.0284046] [Citation(s) in RCA: 8] [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: 11/08/2022] [Accepted: 03/22/2023] [Indexed: 04/07/2023] Open
Abstract
BACKGROUND Measurement of SARS-CoV-2 antibody seropositivity is important to accurately understand exposure to infection and/or vaccination in specific populations. This study aimed to estimate the serologic response to SARS-CoV-2 virus infection and vaccination in children in Calgary, Alberta over a two-year period. METHODS Children with or without prior SARS-CoV-2 infections, were enrolled in Calgary, Canada in 2020. Venous blood was sampled 4 times from July 2020 to April 2022 for SARS-CoV-2 nucleocapsid and spike antibodies. Demographic and clinical information was obtained including SARS-CoV-2 testing results and vaccination records. RESULTS 1035 children were enrolled and 88.9% completed all 4 visits; median age 9 years (IQR: 5,13); 519 (50.1%) female; and 815 (78.7%) Caucasian. Before enrolment, 118 (11.4%) had confirmed or probable SARS-CoV-2. By April 2022, 39.5% of previously uninfected participants had a SARS-CoV-2 infection. Nucleocapsid antibody seropositivity declined to 16.4% of all infected children after more than 200 days post diagnosis. Spike antibodies remained elevated in 93.6% of unvaccinated infected children after more than 200 days post diagnosis. By April 2022, 408 (95.6%) children 12 years and older had received 2 or more vaccine doses, and 241 (61.6%) 5 to 11 year-old children had received 2 vaccine doses. At that time, all 685 vaccinated children had spike antibodies, compared with 94/176 (53.4%) of unvaccinated children. CONCLUSIONS In our population, after the first peak of Omicron variant infections and introduction of COVID-19 vaccines for children, all vaccinated children, but just over one-half of unvaccinated children, had SARS-CoV-2 spike antibodies indicating infection and/or vaccination, highlighting the benefit of vaccination. It is not yet known whether a high proportion of seropositivity at the present time predicts sustained population-level protection against future SARS-CoV-2 transmission, infection or severe COVID-19 outcomes in children.
Collapse
Affiliation(s)
- Emily J. Doucette
- Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Joslyn Gray
- Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Kevin Fonseca
- Department of Microbiology, Immunology & Infectious Diseases, University of Calgary, Calgary, Alberta, Canada
- Public Health Laboratory, Alberta Precision Laboratories, Alberta, Canada
| | - Carmen Charlton
- Public Health Laboratory, Alberta Precision Laboratories, Alberta, Canada
- Department of Pathology and Laboratory Medicine, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Alberta, Canada
| | - Jamil N. Kanji
- Public Health Laboratory, Alberta Precision Laboratories, Alberta, Canada
- Department of Pathology and Laboratory Medicine, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Laboratory Medicine and Pathology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
- Division of Infectious Diseases, Department of Medicine, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Graham Tipples
- Public Health Laboratory, Alberta Precision Laboratories, Alberta, Canada
- Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Alberta, Canada
- Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Susan Kuhn
- Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Jessica Dunn
- Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Payton Sayers
- Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Nicola Symonds
- School of Medicine, Queen’s University, Kingston, Ontario, Canada
| | - Guosong Wu
- Department of Community Health Sciences, University of Calgary, Calgary, Alberta, Canada
| | - Stephen B. Freedman
- Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Emergency Medicine, University of Calgary, Calgary, Alberta, Canada
| | - James D. Kellner
- Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
41
|
Mohammadzadeh Hosseini Moghri SAH, Ranjbar M, Hassannia H, Khakdan F. Comparison and monitoring of antibody response in convalescent and healthy vaccinated individuals against RBD and PCS of SARS-CoV-2 spike protein. J Biomol Struct Dyn 2023; 41:14224-14231. [PMID: 36961201 DOI: 10.1080/07391102.2023.2193981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 02/06/2023] [Indexed: 03/25/2023]
Abstract
The prevalence of SARS-CoV-2 as a global health threat has called for population-wide vaccination to curb COVID-19. Hence, the World Health Organization (WHO) has approved several platforms of SARS-CoV-2 vaccines for emergency use. Therefore, a more comprehensive study on the immune response induced by vaccines in diverse individuals is still required. Here, we expressed a local variant of SARS-CoV-2 receptor-binding domain (RBD) and protease cleavage site (PCS), playing a vital role in binding and fusion in Rosetta (DE3). We then characterized it through SDS-PAGE analysis and western blotting. Moreover, we compared and monitored ChAdOx1 nCoV-19 vaccination-induced antibody response in convalescent and healthy vaccinated individuals after the first and second vaccine doses through serologic assay against RBD and PCS, which have not yet been compared. We investigated a cohort of 100 sera samples; based on our parameters, 25 serum samples were selected as convalescent samples and 25 serum samples as healthy samples for comparison. These findings demonstrate that most of the convalescent sera show more reactivity with PCS (80%) than with RBD (56%). Interestingly, IgG antibody response against PCS was more significant in both pre- and post-vaccination in convalescent individuals than in healthy individuals. Indeed, anti-RBD antibody titers were most significant in pre-vaccination and post-first vaccination in convalescent individuals than in healthy individuals and not in pre-vaccination and post-second vaccination. Besides monitoring IgG antibody response against COVID-19, these findings could shed light on the progress, assessment, and efficacy evaluation of SARS-CoV-2 vaccines.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
| | - Mojtaba Ranjbar
- Department of Microbial Biotechnology, Faculty of Biotechnology, Amol University of Special Modern Technologies, Amol, Iran
| | - Hadi Hassannia
- Immunogenetic Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | | |
Collapse
|
42
|
Chauvin C, Levillayer L, Roumier M, Nielly H, Roth C, Karnam A, Bonam SR, Bourgarit A, Dubost C, Bousquet A, Le Burel S, Mestiri R, Sene D, Galland J, Vasse M, Groh M, Le Marchand M, Vassord-Dang C, Gautier JF, Pham-Thi N, Verny C, Pitard B, Planchais C, Mouquet H, Paul R, Simon-Loriere E, Bayry J, Gilardin L, Sakuntabhai A. Tocilizumab-treated convalescent COVID-19 patients retain the cross-neutralization potential against SARS-CoV-2 variants. iScience 2023; 26:106124. [PMID: 36776936 PMCID: PMC9894676 DOI: 10.1016/j.isci.2023.106124] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 10/10/2022] [Accepted: 01/30/2023] [Indexed: 02/05/2023] Open
Abstract
Although tocilizumab treatment in severe and critical coronavirus disease 2019 (COVID-19) patients has proven its efficacy at the clinical level, there is little evidence supporting the effect of short-term use of interleukin-6 receptor blocking therapy on the B cell sub-populations and the cross-neutralization of SARS-CoV-2 variants in convalescent COVID-19 patients. We performed immunological profiling of 69 tocilizumab-treated and non-treated convalescent COVID-19 patients in total. We observed that SARS-CoV-2-specific IgG1 titers depended on disease severity but not on tocilizumab treatment. The plasma of both treated and non-treated patients infected with the ancestral variant exhibit strong neutralizing activity against the ancestral virus and the Alpha, Beta, and Delta variants of SARS-CoV-2, whereas the Gamma and Omicron viruses were less sensitive to seroneutralization. Overall, we observed that, despite the clinical benefits of short-term tocilizumab therapy in modifying the cytokine storm associated with COVID-19 infections, there were no modifications in the robustness of B cell and IgG responses to Spike antigens.
Collapse
Affiliation(s)
- Camille Chauvin
- Institut Pasteur, Université de Paris, Functional Genetics of Infectious Diseases Unit, Department of Global Health, 75015 Paris, France.,Centre National de la Recherche Scientifique (CNRS), UMR2000, Paris Cedex 15, France
| | - Laurine Levillayer
- Institut Pasteur, Université de Paris, Functional Genetics of Infectious Diseases Unit, Department of Global Health, 75015 Paris, France.,Centre National de la Recherche Scientifique (CNRS), UMR2000, Paris Cedex 15, France
| | - Mathilde Roumier
- Service de Médecine Interne, Hôpital Foch, 92151 Suresnes, France
| | - Hubert Nielly
- Service de Médecine Interne, Hôpital d'Instruction des Armées Bégin, 94160 Saint Mandé, France
| | - Claude Roth
- Institut Pasteur, Université de Paris, Functional Genetics of Infectious Diseases Unit, Department of Global Health, 75015 Paris, France.,Centre National de la Recherche Scientifique (CNRS), UMR2000, Paris Cedex 15, France
| | - Anupama Karnam
- Institut National de la Santé et de la Recherche Médicale, Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, Paris, 75006, France
| | - Srinivasa Reddy Bonam
- Institut National de la Santé et de la Recherche Médicale, Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, Paris, 75006, France
| | - Anne Bourgarit
- Hôpital Jean Verdier, HUPSSD, AP-HP, 93140 Bondy, France.,Sorbonne Paris-Nord University (Paris 13), 93000 Bobigny, France.,Inserm, UMR 1135 CIMI, 75013 Paris, France
| | - Clément Dubost
- Service de réanimation, Hôpital militaire Bégin, 94120 Saint Mandé, France.,Université Paris-Saclay, ENS Paris-Saclay, CNRS, Centre Borelli, 91190, Gif-sur-Yvette, France
| | - Aurore Bousquet
- Département des laboratoires, Hôpital militaire Bégin, 94120 Saint Mandé, France
| | - Sébastien Le Burel
- Service de Médecine Interne, Hôpital d'Instruction des Armées Bégin, 94160 Saint Mandé, France
| | - Raphaële Mestiri
- Service de Médecine Interne, Hôpital d'Instruction des Armées Bégin, 94160 Saint Mandé, France
| | - Damien Sene
- Département de médecine interne, Hôpital Lariboisière, Université de Paris (Diderot), AP-HP, 75010 Paris, France
| | - Joris Galland
- Département de médecine interne, Hôpital Lariboisière, Université de Paris (Diderot), AP-HP, 75010 Paris, France
| | - Marc Vasse
- Laboratoire de Biologie Médicale, Hôpital Foch, 92151 Suresnes, France.,UMRS-1176, Le Kremlin Bicêtre, France
| | - Matthieu Groh
- Service de Médecine Interne, Hôpital Foch, 92151 Suresnes, France
| | - Mathilde Le Marchand
- Department of Clinical Research and Innovation, Foch Hospital, 40 rue Worth, 92150 Suresnes, France
| | - Camille Vassord-Dang
- Department of Clinical Research and Innovation, Foch Hospital, 40 rue Worth, 92150 Suresnes, France
| | - Jean-François Gautier
- Departement of Diabetes and Endocrinology, Hôpital Lariboisière, APHP, and INSERM U1138 Paris, France.,Université de Paris, 75006 Paris, France
| | - Nhan Pham-Thi
- Unité de Neurophysiologie du Stress, Département des Neurosciences, Institut de Recherche Biomédicale des Armées (IRBA), BP 73 91223 Brétigny sur Orge Cedex, France
| | - Christiane Verny
- Unité de Neurophysiologie du Stress, Département des Neurosciences, Institut de Recherche Biomédicale des Armées (IRBA), BP 73 91223 Brétigny sur Orge Cedex, France
| | - Bruno Pitard
- Nantes Université, INSERM, CNRS, Immunology and New Concepts in ImmunoTherapy, INCIT,UMR 1302, F-44000 Nantes, France
| | - Cyril Planchais
- Institut Pasteur, Université de Paris, Humoral Immunology Unit, Department of Immunology, 75015 Paris, France
| | - Hugo Mouquet
- Institut Pasteur, Université de Paris, Humoral Immunology Unit, Department of Immunology, 75015 Paris, France
| | - Richard Paul
- Institut Pasteur, Université de Paris, Functional Genetics of Infectious Diseases Unit, Department of Global Health, 75015 Paris, France.,Centre National de la Recherche Scientifique (CNRS), UMR2000, Paris Cedex 15, France
| | - Etienne Simon-Loriere
- Institut Pasteur, Université de Paris, G5 Evolutionary Genomics of RNA viruses, 75015 Paris, France
| | - Jagadeesh Bayry
- Institut National de la Santé et de la Recherche Médicale, Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, Paris, 75006, France.,Department of Biological Sciences & Engineering, Indian Institute of Technology Palakkad, Palakkad 678623, India
| | - Laurent Gilardin
- Institut National de la Santé et de la Recherche Médicale, Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, Paris, 75006, France.,Sorbonne Paris-Nord University (Paris 13), 93000 Bobigny, France.,Service de médecine interne, Hôpital Jean Verdier, HUPSSD, AP-HP, 93140 Bondy, France
| | - Anavaj Sakuntabhai
- Institut Pasteur, Université de Paris, Functional Genetics of Infectious Diseases Unit, Department of Global Health, 75015 Paris, France.,Centre National de la Recherche Scientifique (CNRS), UMR2000, Paris Cedex 15, France.,International Vaccine Design Center (vDesC), The Institute of Medical Science, The University of Tokyo (IMSUT), Tokyo, Japan
| |
Collapse
|
43
|
Zhao Z, Kumanovics A, Love T, Melanson SEF, Meng QH, Wu AHB, Wiencek J, Apple FS, Ondracek CR, Koch DD, Christenson RH, Zhang YV. T Cell Responses Correlate with Self-Reported Disease Severity and Neutralizing Antibody Responses Predict Protection against SARS-CoV-2 Breakthrough Infection. Viruses 2023; 15:709. [PMID: 36992418 PMCID: PMC10058409 DOI: 10.3390/v15030709] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 03/06/2023] [Accepted: 03/07/2023] [Indexed: 03/31/2023] Open
Abstract
OBJECTIVES The objective of this prospective study was to investigate the role of adaptive immunity in response to SARS-CoV-2 vaccines. DESIGN AND METHODS A cohort of 677 vaccinated individuals participated in a comprehensive survey of their vaccination status and associated side effects, and donated blood to evaluate their adaptive immune responses by neutralizing antibody (NAb) and T cell responses. The cohort then completed a follow-up survey to investigate the occurrence of breakthrough infections. RESULTS NAb levels were the highest in participants vaccinated with Moderna, followed by Pfizer and Johnson & Johnson. NAb levels decreased with time after vaccination with Pfizer and Johnson & Johnson. T cell responses showed no significant difference among the different vaccines and remained stable up to 10 months after the study period for all vaccine types. In multivariate analyses, NAb responses (<95 U/mL) predicted breakthrough infection, whereas previous infection, the type of vaccine, and T cell responses did not. T cell responses to viral epitopes (<0.120 IU/mL) showed a significant association with the self-reported severity of COVID-19 disease. CONCLUSION This study provides evidence that NAb responses to SARS-CoV-2 vaccination correlate with protection against infection, whereas the T cell memory responses may contribute to protection against severe disease but not against infection.
Collapse
Affiliation(s)
- Zhen Zhao
- Department of Laboratory Medicine and Pathology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Attila Kumanovics
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA
| | - Tanzy Love
- Department of Biostatistics and Computational Biology, University of Rochester, Rochester, NY 14642, USA
| | - Stacy E. F. Melanson
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Qing H. Meng
- Department of Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Alan H. B. Wu
- Department of Laboratory Medicine, University of California, San Francisco, CA 94143, USA
| | - Joesph Wiencek
- Department of Pathology, Microbiology and Immunology, Vanderbilt School of Medicine, Nashville, TN 37240, USA
| | - Fred S. Apple
- Department of Laboratory Medicine and Pathology, Hennepin Healthcare/Hennepin County Medical Center, Minneapolis, MN 55415, USA
- Hennepin Healthcare Research Institute, Minneapolis, MN 55404, USA
| | | | - David D. Koch
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA 30303, USA
| | - Robert H. Christenson
- Department of Pathology, University of Maryland School of Medicine, 685 W. Baltimore Street, Baltimore, MD 21201, USA
| | - Yan Victoria Zhang
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, 601 Elmwood Avenue, Box 626, Rochester, NY 14642, USA
| |
Collapse
|
44
|
Al-Akioui-Sanz K, Pascual-Miguel B, Díaz-Almirón M, Mestre-Durán C, Navarro-Zapata A, Clares-Villa L, Martín-Cortázar C, Vicario JL, Moreno MÁ, Balas A, De Paz R, Minguillón J, Pérez-Martínez A, Ferreras C. Donor selection for adoptive cell therapy with CD45RA - memory T cells for patients with coronavirus disease 2019, and dexamethasone and interleukin-15 effects on the phenotype, proliferation and interferon gamma release. Cytotherapy 2023; 25:330-340. [PMID: 36585293 PMCID: PMC9742221 DOI: 10.1016/j.jcyt.2022.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 12/02/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022]
Abstract
BACKGROUND AIMS We have previously demonstrated the safety and feasibility of adoptive cell therapy with CD45RA- memory T cells containing severe acute respiratory syndrome coronavirus 2-specific T cells for patients with coronavirus disease 2019 from an unvaccinated donor who was chosen based on human leukocyte antigen compatibility and cellular response. In this study, we examined the durability of cellular and humoral immunity within CD45RA- memory T cells and the effect of dexamethasone, the current standard of care treatment, and interleukin-15, a cytokine critically involved in T-cell maintenance and survival. METHODS We performed a longitudinal analysis from previously severe acute respiratory syndrome coronavirus 2-infected and infection-naïve individuals covering 21 months from infection and 10 months after full vaccination with the BNT162b2 Pfizer/BioNTech vaccine. RESULTS We observed that cellular responses are maintained over time. Humoral responses increased after vaccination but were gradually lost. In addition, dexamethasone did not alter cell functionality or proliferation of CD45RA- T cells, and interleukin-15 increased the memory T-cell activation state, regulatory T cell expression, and interferon gamma release. CONCLUSIONS Our results suggest that the best donors for adoptive cell therapy would be recovered individuals and 2 months after vaccination, although further studies with larger cohorts would be needed to confirm this finding. Dexamethasone did not affect the characteristics of the memory T cells at a concentration used in the clinical practice and IL-15 showed a positive effect on SARS-CoV-2-specific CD45RA- T cells.
Collapse
Affiliation(s)
- Karima Al-Akioui-Sanz
- IdiPAZ, Hospital La Paz Institute for Health Research, La Paz University Hospital, Madrid, Spain
| | - Bárbara Pascual-Miguel
- IdiPAZ, Hospital La Paz Institute for Health Research, La Paz University Hospital, Madrid, Spain
| | | | - Carmen Mestre-Durán
- IdiPAZ, Hospital La Paz Institute for Health Research, La Paz University Hospital, Madrid, Spain
| | - Alfonso Navarro-Zapata
- IdiPAZ, Hospital La Paz Institute for Health Research, La Paz University Hospital, Madrid, Spain
| | - Laura Clares-Villa
- IdiPAZ, Hospital La Paz Institute for Health Research, La Paz University Hospital, Madrid, Spain
| | - Carla Martín-Cortázar
- IdiPAZ, Hospital La Paz Institute for Health Research, La Paz University Hospital, Madrid, Spain
| | - José Luis Vicario
- Histocompatibility Unit, Transfusion Center of Madrid, Madrid, Spain
| | | | - Antonio Balas
- Histocompatibility Unit, Transfusion Center of Madrid, Madrid, Spain
| | - Raquel De Paz
- Cell Therapy Unit, Hematology Department, La Paz University Hospital, Madrid, Spain
| | - Jordi Minguillón
- IdiPAZ, Hospital La Paz Institute for Health Research, La Paz University Hospital, Madrid, Spain
| | - Antonio Pérez-Martínez
- IdiPAZ, Hospital La Paz Institute for Health Research, La Paz University Hospital, Madrid, Spain; Pediatric Hemato-oncology Department, La Paz University Hospital, Madrid, Spain; Faculty of Medicine Autonomous, University of Madrid, Madrid, Spain
| | - Cristina Ferreras
- IdiPAZ, Hospital La Paz Institute for Health Research, La Paz University Hospital, Madrid, Spain.
| |
Collapse
|
45
|
Poolchanuan P, Matsee W, Sengyee S, Siripoon T, Dulsuk A, Phunpang R, Pisutsan P, Piyaphanee W, Luvira V, Chantratita N. Dynamics of Different Classes and Subclasses of Antibody Responses to Severe Acute Respiratory Syndrome Coronavirus 2 Variants after Coronavirus Disease 2019 and CoronaVac Vaccination in Thailand. mSphere 2023; 8:e0046522. [PMID: 36688637 PMCID: PMC9942573 DOI: 10.1128/msphere.00465-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 12/20/2022] [Indexed: 01/24/2023] Open
Abstract
The humoral immune response plays a key role in protecting the population from SARS-CoV-2 transmission. Patients who recovered from COVID-19 as well as fully vaccinated individuals have elevated levels of antibodies. The dynamic levels of the classes and subclasses of antibody responses to new variants that occur in different populations remain unclear. We prospectively recruited 60 participants, including COVID-19 patients and CoronaVac-vaccinated individuals, in Thailand from May to August 2021. Plasma samples were collected on day 0, day 14, and day 28 to determine the dynamic levels of the classes and subclasses of plasma antibodies against the receptor-binding domain (RBD) in the spike protein (S) of four SARS-CoV-2 strains (Wuhan, Alpha, Delta, and Omicron) via enzyme-linked immunosorbent assay. Our results indicated that the patients with SARS-CoV-2 infections had broader class and subclass profiles as well as higher levels of anti-S RBD antibodies to the Wuhan, Alpha, and Delta strains than did the CoronaVac-vaccinated individuals. The median antibody levels increased and subsequently declined in a month in the COVID-19 patients and in the vaccinated group. Correlations of the classes and subclasses of antibodies were observed in the COVID-19 patients but not in the vaccinated individuals. The levels of all of the anti-S RBD antibodies against the Omicron variant were low in the patients and in the vaccinated individuals. Our study revealed distinct antibody profiles between the two cohorts, suggesting different pathways of immune activation. This could have an impact on protection from infections by new variants of concern (VOC). IMPORTANCE The antibody responses to new SARS-CoV-2 variants that occur in different populations remain unclear. In this study, we recruited 60 participants, including COVID-19 patients and CoronaVac-vaccinated individuals, in Thailand and determined the dynamic levels of the IgG, IgA, IgM, and IgG subclasses of antibodies against the spike protein (S) of four SARS-CoV-2 strains. Our results showed that the patients with SARS-CoV-2 infections had broader profiles and higher levels of antibodies to the Wuhan, Alpha, and Delta strains than did the CoronaVac-vaccinated individuals. The antibody levels of both groups increased and subsequently decreased within 1 month. Higher and functional correlations of these antibodies were observed in the COVID-19 patients. The levels of all anti-S RBD antibodies against the Omicron variant were low in patients and vaccinated individuals. Our study revealed distinct antibody responses between the two groups, suggesting different pathways of immune response, which may have an impact on protection from infections by new SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Prapassorn Poolchanuan
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Wasin Matsee
- Thai Travel Clinic, Hospital for Tropical Diseases, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Sineenart Sengyee
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Tanaya Siripoon
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Adul Dulsuk
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Rungnapa Phunpang
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Phimphan Pisutsan
- Thai Travel Clinic, Hospital for Tropical Diseases, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Watcharapong Piyaphanee
- Thai Travel Clinic, Hospital for Tropical Diseases, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Viravarn Luvira
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Narisara Chantratita
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| |
Collapse
|
46
|
Monti S, Fornara C, Delvino P, Bartoletti A, Bergami F, Comolli G, Sammartino JC, Biglia A, Bozzalla Cassione E, Cassaniti I, Baldanti F, Lilleri D, Montecucco C. Immunosuppressive treatments selectively affect the humoral and cellular response to SARS-CoV-2 in vaccinated patients with vasculitis. Rheumatology (Oxford) 2023; 62:726-734. [PMID: 35736379 PMCID: PMC9278207 DOI: 10.1093/rheumatology/keac365] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 06/08/2022] [Accepted: 06/08/2022] [Indexed: 02/04/2023] Open
Abstract
OBJECTIVES To analyse humoral and cellular immune response to mRNA COVID-19 vaccines in patients with GCA. METHODS Consecutive patients with a diagnosis of GCA receiving two doses of BNT162b2 vaccine were assessed at baseline and 3 weeks from the second vaccine dose. Healthy subjects (n = 51) were included as controls (HC). Humoral response was assessed with Spike-specific IgG antibody response (S-IgG) and neutralizing antibodies (NtAb). Specific T cell response was assessed by enzyme linked immunosorbent spot (ELISpot). RESULTS Of 56 included patients with GCA, 44 were eligible after exclusion of previous evidence of COVID-19 and incomplete follow-up. A significant proportion of patients with GCA (91%) demonstrated antibody (S-IgG) response, but this was significantly lower than HCs (100%); P < 0.0001. Neutralizing activity was not detected in 16% of patients with GCA. Antibody titres (S-IgG and NtAb) were significantly lower compared with HCs. Humoral response (S-IgG and NtAb) was significantly hampered by treatment with MTX. Cellular response was lacking in 30% of patients with GCA (vs 0% in HCs; P < 0.0001). Cellular response was significantly influenced by the levels of baseline peripheral T-lymphocytes and by glucocorticoid treatment. Treatment with tocilizumab did not affect any level of the immune response elicited by vaccination. CONCLUSIONS Although patients with GCA apparently achieve a robust antibody seroconversion, there is a significant impairment of the neutralizing activity. MTX significantly reduced all levels of the humoral response. Up to one-third of patients do not develop a cellular immune protection in response to COVID-19 vaccination.
Collapse
Affiliation(s)
- Sara Monti
- Department of Rheumatology, Fondazione IRCCS Policlinico San Matteo Pavia, University of Pavia
| | - Chiara Fornara
- Molecular Virology Unit, Department of Microbiology and Virology
| | - Paolo Delvino
- Department of Rheumatology, Fondazione IRCCS Policlinico San Matteo Pavia, University of Pavia
| | - Alice Bartoletti
- Department of Rheumatology, Fondazione IRCCS Policlinico San Matteo Pavia, University of Pavia
| | - Federica Bergami
- Molecular Virology Unit, Department of Microbiology and Virology
| | - Giuditta Comolli
- Molecular Virology Unit, Department of Microbiology and Virology.,Experimental Research Laboratories, Biotechnology Area, Fondazione IRCCS Policlinico San Matteo
| | | | - Alessandro Biglia
- Department of Rheumatology, Fondazione IRCCS Policlinico San Matteo Pavia, University of Pavia
| | | | - Irene Cassaniti
- Molecular Virology Unit, Department of Microbiology and Virology
| | - Fausto Baldanti
- Molecular Virology Unit, Department of Microbiology and Virology.,Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy
| | - Daniele Lilleri
- Molecular Virology Unit, Department of Microbiology and Virology
| | | |
Collapse
|
47
|
Zhang B, Upadhyay R, Hao Y, Samanovic MI, Herati RS, Blair J, Axelrad J, Mulligan MJ, Littman DR, Satija R. Multimodal characterization of antigen-specific CD8 + T cells across SARS-CoV-2 vaccination and infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.24.525203. [PMID: 36747786 PMCID: PMC9900816 DOI: 10.1101/2023.01.24.525203] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The human immune response to SARS-CoV-2 antigen after infection or vaccination is defined by the durable production of antibodies and T cells. Population-based monitoring typically focuses on antibody titer, but there is a need for improved characterization and quantification of T cell responses. Here, we utilize multimodal sequencing technologies to perform a longitudinal analysis of circulating human leukocytes collected before and after BNT162b2 immunization. Our data reveal distinct subpopulations of CD8 + T cells which reliably appear 28 days after prime vaccination (7 days post boost). Using a suite of cross-modality integration tools, we define their transcriptome, accessible chromatin landscape, and immunophenotype, and identify unique biomarkers within each modality. By leveraging DNA-oligo-tagged peptide-MHC multimers and T cell receptor sequencing, we demonstrate that this vaccine-induced population is SARS-CoV-2 antigen-specific and capable of rapid clonal expansion. Moreover, we also identify these CD8 + populations in scRNA-seq datasets from COVID-19 patients and find that their relative frequency and differentiation outcomes are predictive of subsequent clinical outcomes. Our work contributes to our understanding of T cell immunity, and highlights the potential for integrative and multimodal analysis to characterize rare cell populations.
Collapse
Affiliation(s)
- Bingjie Zhang
- New York Genome Center, New York, NY, USA
- Center for Genomics and Systems Biology, New York University, New York, NY, USA
- Department of Cell Biology and Regenerative Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Rabi Upadhyay
- Department of Cell Biology and Regenerative Medicine, New York University Grossman School of Medicine, New York, NY, USA
- Perlmutter Cancer Center, New York University Langone Health, New York, NY, USA
- Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Yuhan Hao
- New York Genome Center, New York, NY, USA
- Center for Genomics and Systems Biology, New York University, New York, NY, USA
| | - Marie I. Samanovic
- Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
- New York University Langone Vaccine Center, New York, NY, USA
| | - Ramin S. Herati
- Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
- New York University Langone Vaccine Center, New York, NY, USA
| | - John Blair
- New York Genome Center, New York, NY, USA
- Center for Genomics and Systems Biology, New York University, New York, NY, USA
| | - Jordan Axelrad
- Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Mark J. Mulligan
- Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
- New York University Langone Vaccine Center, New York, NY, USA
| | - Dan R. Littman
- Department of Cell Biology and Regenerative Medicine, New York University Grossman School of Medicine, New York, NY, USA
- Perlmutter Cancer Center, New York University Langone Health, New York, NY, USA
- Howard Hughes Medical Institute, New York, NY, USA
| | - Rahul Satija
- New York Genome Center, New York, NY, USA
- Center for Genomics and Systems Biology, New York University, New York, NY, USA
| |
Collapse
|
48
|
Carvalho Á, Henriques AR, Queirós P, Rodrigues J, Mendonça N, Rodrigues AM, Canhão H, de Sousa G, Antunes F, Guimarães M. Persistence of IgG COVID-19 antibodies: A longitudinal analysis. Front Public Health 2023; 10:1069898. [PMID: 36703818 PMCID: PMC9872107 DOI: 10.3389/fpubh.2022.1069898] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 12/19/2022] [Indexed: 01/12/2023] Open
Abstract
Background and aim The kinetics of antibody production in response to coronavirus disease 2019 (COVID-19) infection is not well-defined yet. This study aimed to evaluate the antibody responses to SARS-CoV-2 and its dynamics during 9-months in a cohort of patients infected during the first phase of the pandemic. As a secondary aim, it was intended to evaluate the factors associated with different concentrations of IgG antibodies. Methods A prospective cohort study was conducted from June 2020 to January 2021. This study recruited a convenience sample of adult individuals who where recently diagnosed with COVID-19 and were living in mainland Portugal. A total of 1,695 blood samples were collected from 585 recovered COVID-19 patients up to 9 months after SARS-CoV-2 acute infection. A blood sample was collected at baseline and three, 6 and 9 months after SARS-CoV-2 acute infection to assess the concentration of IgG antibody against SARS-CoV-2. Results The positivity rate of IgG reached 77.7% in the first 3 months after symptom onset. The IgG persists at all subsequent follow-up time-points, which was 87.7 and 89.2% in the 6th and 9th months after symptom onset, respectively. Three distinct kinetics of antibody response were found within the 9 months after infection. Kinetic 1 (K1) was characterized by a constant low IgG antibody concentration kinetic (group size: 65.2%); kinetic 2 (K2), composed by constant moderate IgG kinetic (group size: 27.5%) and kinetic 3 (K3) characterized by higher IgG kinetic (group size: 7.3%). People with ≥56 years old (OR: 3.33; CI 95%: [1.64; 6.67]; p-value: 0.001) and symptomatic COVID-19 (OR: 2.08; CI 95%: [1.08; 4.00]; p-value: 0.031) had higher odds of a "Moderate IgG kinetic." No significant association were found regarding the "Higher IgG kinetic." Conclusion Our results demonstrate a lasting anti-spike (anti-S) IgG antibody response at least 9 months after infection in the majority of patients with COVID-19. Younger participants with asymptomatic disease have lower IgG antibody positivity and possibly more susceptible to reinfection. This information contributes to expanding knowledge of SARS-CoV-2 immune response and has direct implications in the adoption of preventive strategies and public health policies.
Collapse
Affiliation(s)
| | - Ana Rita Henriques
- CHRC, NOVA Medical School, Universidade NOVA de Lisboa, Lisboa, Portugal,*Correspondence: Ana Rita Henriques ✉
| | | | | | - Nuno Mendonça
- CHRC, NOVA Medical School, Universidade NOVA de Lisboa, Lisboa, Portugal
| | | | - Helena Canhão
- CHRC, NOVA Medical School, Universidade NOVA de Lisboa, Lisboa, Portugal
| | - Germano de Sousa
- Germano de Sousa Group- Centro de Medicina Laboratorial, Pólo Tecnológico de Lisboa, Lisboa, Portugal
| | - Francisco Antunes
- Instituto de Saúde Ambiental, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal,Laboratório Associado TERRA, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | | |
Collapse
|
49
|
Yan LN, Li D, Wang ZD, Jiang ZZ, Xiao X, Yu XJ. Neutralizing antibodies and T-cell responses to inactivated SARS-CoV-2 vaccine in COVID-19 convalescents one and a half years after infection. Virus Res 2023; 323:198977. [PMID: 36283534 PMCID: PMC9595495 DOI: 10.1016/j.virusres.2022.198977] [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: 06/08/2022] [Revised: 10/16/2022] [Accepted: 10/21/2022] [Indexed: 11/09/2022]
Abstract
Vaccines have been considered the most promising solution for ending the coronavirus disease 2019 (COVID-19) pandemic. Information regarding neutralizing antibodies (NAbs) and T-cell immune response in inactivated SARS-CoV-2 vaccine-immunized COVID-19 convalescent patients were either only available for a short time after illness recovered or not available at all (T-cell immunity). We evaluated SARS-CoV-2 NAbs and cellular immune responses to the SARS-CoV-2 inactivated vaccine in convalescent patients who recovered from infection for about one and a half years. We found that compared to before vaccination, SARS-CoV-2 NAbs and specific T-cell responses were significantly boosted by the inactivated vaccine in convalescent patients, which confirmed the pre-existing adaptive immunity in SARS-CoV-2 infected people. We observed that NAbs and IFN-γ-secreting T-cell response elicited by a single vaccine dose in subjects with prior COVID-19 infection were higher than after two doses of vaccine in SARS-CoV-2 naïve subjects. Both humoral and cellular immune responses elicited by one and two doses of inactivated vaccine were comparable in COVID-19-recovered persons. In conclusion, inactivated COVID-19 vaccine induced robust NAbs and T-cell responses to SARS-CoV-2 in COVID-19 convalescent patients and immune responses after one dose were equal to that after receiving two doses, which highlighted that robust humoral and cellular immune response can be reactivated by the inactivated vaccine in SARS-CoV-2 convalescent patients.
Collapse
Affiliation(s)
- Li-Na Yan
- State Key Laboratory of Virology, School of Public Health, Wuhan University, Wuhan 430070, PR China
| | - Dan Li
- State Key Laboratory of Virology, School of Public Health, Wuhan University, Wuhan 430070, PR China
| | - Zhen-Dong Wang
- School of Public Health, Xi'an Medical University, Xi'an 710021, PR China
| | - Ze-Zheng Jiang
- State Key Laboratory of Virology, School of Public Health, Wuhan University, Wuhan 430070, PR China
| | - Xiao Xiao
- Institute of Epidemic Research, Hubei University of Chinese Medicine, Wuhan 430065, PR China.
| | - Xue-Jie Yu
- State Key Laboratory of Virology, School of Public Health, Wuhan University, Wuhan 430070, PR China.
| |
Collapse
|
50
|
Egri N, Calderón H, Martinez R, Vazquez M, Gómez-Caverzaschi V, Pascal M, Araújo O, Juan M, González-Navarro EA, Hernández-Rodríguez J. Cellular and humoral responses after second and third SARS-CoV-2 vaccinations in patients with autoimmune diseases treated with rituximab: specific T cell immunity remains longer and plays a protective role against SARS-CoV-2 reinfections. Front Immunol 2023; 14:1146841. [PMID: 37180097 PMCID: PMC10174323 DOI: 10.3389/fimmu.2023.1146841] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 04/07/2023] [Indexed: 05/15/2023] Open
Abstract
Background Humoral and cellular immune responses are known to be crucial for patients to recover from COVID-19 and to protect them against SARS-CoV-2 reinfection once infected or vaccinated. Objectives This study aimed to investigate humoral and T cell responses to SARS-CoV-2 vaccination in patients with autoimmune diseases after the second and third vaccine doses while on rituximab and their potential protective role against reinfection. Methods Ten COVID-19-naïve patients were included. Three time points were used for monitoring cellular and humoral responses: pre-vaccine to exclude virus exposure (time point 1) and post-second and post-third vaccine (time points 2 and 3). Specific IgG antibodies were monitored by Luminex and T cells against SARS-CoV-2 spike-protein by ELISpot and CoVITEST. All episodes of symptomatic COVID-19 were recorded. Results Nine patients with antineutrophil cytoplasmic antibody (ANCA)-associated vasculitis and one with an undifferentiated autoimmune disease were included. Nine patients received mRNA vaccines. The last rituximab infusion was administered for a mean (SD) of 15 (10) weeks before the first vaccine and six patients were CD19-B cell-depleted. After a mean (SD) of 19 (10) and 16 (2) days from the second and third vaccine dose, IgG anti-SARS-CoV-2 antibodies were detected in six (60%) and eight (80%) patients, respectively. All patients developed specific T cell responses by ELISpot and CoVITEST in time points 2 and 3. Previous B cell depletion correlated with anti-SARS-CoV-2 IgG levels. Nine (90%) patients developed mild COVID-19 after a median of 7 months of the third dose. Conclusion Rituximab in patients with autoimmune diseases reduces humoral responses but does not avoid the development of T cell responses to SARS-CoV-2 vaccination, which remain present after a booster dose. A steady cellular immunity appears to be protective against subsequent reinfections.
Collapse
Affiliation(s)
- Natalia Egri
- Department of Immunology, Centre de Diagnòstic Biomèdic, Hospital Clínic of Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Center of the European Reference Network (ERN) for Rare Immunodeficiency, Autoinflammatory and Autoimmune Diseases (RITA) and ERN on Connective Tissue and Musculoskeletal Diseases (ReCONNET); Spanish Center of the Centros, Servicios y Unidades de Referencia (CSUR) and Catalan Center of the Xarxa d’Unitats d’Expertesa Clínica (XUEC) for Autoinflammatory Diseases, Autoimmune Diseases and Primary Immunodeficiencies, Barcelona, Spain
| | - Hugo Calderón
- Department of Immunology, Centre de Diagnòstic Biomèdic, Hospital Clínic of Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Center of the European Reference Network (ERN) for Rare Immunodeficiency, Autoinflammatory and Autoimmune Diseases (RITA) and ERN on Connective Tissue and Musculoskeletal Diseases (ReCONNET); Spanish Center of the Centros, Servicios y Unidades de Referencia (CSUR) and Catalan Center of the Xarxa d’Unitats d’Expertesa Clínica (XUEC) for Autoinflammatory Diseases, Autoimmune Diseases and Primary Immunodeficiencies, Barcelona, Spain
| | - Robert Martinez
- Department of Immunology, Centre de Diagnòstic Biomèdic, Hospital Clínic of Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Center of the European Reference Network (ERN) for Rare Immunodeficiency, Autoinflammatory and Autoimmune Diseases (RITA) and ERN on Connective Tissue and Musculoskeletal Diseases (ReCONNET); Spanish Center of the Centros, Servicios y Unidades de Referencia (CSUR) and Catalan Center of the Xarxa d’Unitats d’Expertesa Clínica (XUEC) for Autoinflammatory Diseases, Autoimmune Diseases and Primary Immunodeficiencies, Barcelona, Spain
| | - Mario Vazquez
- Department of Immunology, Centre de Diagnòstic Biomèdic, Hospital Clínic of Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Center of the European Reference Network (ERN) for Rare Immunodeficiency, Autoinflammatory and Autoimmune Diseases (RITA) and ERN on Connective Tissue and Musculoskeletal Diseases (ReCONNET); Spanish Center of the Centros, Servicios y Unidades de Referencia (CSUR) and Catalan Center of the Xarxa d’Unitats d’Expertesa Clínica (XUEC) for Autoinflammatory Diseases, Autoimmune Diseases and Primary Immunodeficiencies, Barcelona, Spain
| | - Verónica Gómez-Caverzaschi
- Clinical Unit of Autoinflammatory Diseases and Vasculitis Research Unit, Department of Autoimmune Diseases, Hospital Clínic of Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Center of the European Reference Network (ERN) for Rare Immunodeficiency, Autoinflammatory and Autoimmune Diseases (RITA) and ERN on Connective Tissue and Musculoskeletal Diseases (ReCONNET), Spanish Center of the Centros, Servicios y Unidades de Referencia (CSUR) and Catalan Center of the Xarxa d’Unitats d’Expertesa Clínica (XUEC) for Autoinflammatory Diseases, Autoimmune Diseases and Primary Immunodeficiencies, Barcelona, Spain
| | - Mariona Pascal
- Department of Immunology, Centre de Diagnòstic Biomèdic, Hospital Clínic of Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Center of the European Reference Network (ERN) for Rare Immunodeficiency, Autoinflammatory and Autoimmune Diseases (RITA) and ERN on Connective Tissue and Musculoskeletal Diseases (ReCONNET); Spanish Center of the Centros, Servicios y Unidades de Referencia (CSUR) and Catalan Center of the Xarxa d’Unitats d’Expertesa Clínica (XUEC) for Autoinflammatory Diseases, Autoimmune Diseases and Primary Immunodeficiencies, Barcelona, Spain
| | - Olga Araújo
- Clinical Unit of Autoinflammatory Diseases and Vasculitis Research Unit, Department of Autoimmune Diseases, Hospital Clínic of Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Center of the European Reference Network (ERN) for Rare Immunodeficiency, Autoinflammatory and Autoimmune Diseases (RITA) and ERN on Connective Tissue and Musculoskeletal Diseases (ReCONNET), Spanish Center of the Centros, Servicios y Unidades de Referencia (CSUR) and Catalan Center of the Xarxa d’Unitats d’Expertesa Clínica (XUEC) for Autoinflammatory Diseases, Autoimmune Diseases and Primary Immunodeficiencies, Barcelona, Spain
| | - Manel Juan
- Department of Immunology, Centre de Diagnòstic Biomèdic, Hospital Clínic of Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Center of the European Reference Network (ERN) for Rare Immunodeficiency, Autoinflammatory and Autoimmune Diseases (RITA) and ERN on Connective Tissue and Musculoskeletal Diseases (ReCONNET); Spanish Center of the Centros, Servicios y Unidades de Referencia (CSUR) and Catalan Center of the Xarxa d’Unitats d’Expertesa Clínica (XUEC) for Autoinflammatory Diseases, Autoimmune Diseases and Primary Immunodeficiencies, Barcelona, Spain
| | - Europa Azucena González-Navarro
- Department of Immunology, Centre de Diagnòstic Biomèdic, Hospital Clínic of Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Center of the European Reference Network (ERN) for Rare Immunodeficiency, Autoinflammatory and Autoimmune Diseases (RITA) and ERN on Connective Tissue and Musculoskeletal Diseases (ReCONNET); Spanish Center of the Centros, Servicios y Unidades de Referencia (CSUR) and Catalan Center of the Xarxa d’Unitats d’Expertesa Clínica (XUEC) for Autoinflammatory Diseases, Autoimmune Diseases and Primary Immunodeficiencies, Barcelona, Spain
| | - José Hernández-Rodríguez
- Clinical Unit of Autoinflammatory Diseases and Vasculitis Research Unit, Department of Autoimmune Diseases, Hospital Clínic of Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Center of the European Reference Network (ERN) for Rare Immunodeficiency, Autoinflammatory and Autoimmune Diseases (RITA) and ERN on Connective Tissue and Musculoskeletal Diseases (ReCONNET), Spanish Center of the Centros, Servicios y Unidades de Referencia (CSUR) and Catalan Center of the Xarxa d’Unitats d’Expertesa Clínica (XUEC) for Autoinflammatory Diseases, Autoimmune Diseases and Primary Immunodeficiencies, Barcelona, Spain
- *Correspondence: José Hernández-Rodríguez,
| |
Collapse
|