1
|
Kwesi-Maliepaard EM, Alhassan Y, Quaye EK, Kotey VM, Mohammed AM, Agyemang S, Sromani AK, Darko S, Buadii E, Tackie R, Akligoh H, Ibrahim B, Hutchful D, Paemka L, Amoako E, Ngoi JM, Manu A, HERITAGE study team, Greenwood D, Carr EJ, Wu MY, Bauer DLV, Wall EC, Crick Legacy Consortium, Dey D, Quao AR, Ayisi A, Amponsa-Achiano K, Bekoe FA, Awandare G, Quashie PK, Bediako Y. Adults in Ghana generate higher and more durable neutralising antibody titres following primary course COVID-19 vaccination than matched UK adults: The HERITAGE Study. BMC Med 2025; 23:312. [PMID: 40437463 PMCID: PMC12121195 DOI: 10.1186/s12916-025-04157-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Collaborators] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 05/20/2025] [Indexed: 06/01/2025] Open
Abstract
BACKGROUND Little data exist on the COVID-19 vaccine response in African countries who despite having high disease burden, have low COVID-19 mortality rates. We investigated the longitudinal immune response in a West-African urban population upon COVID-19 vaccination, two years after the start of the pandemic. METHODS The HERITAGE study is a prospective cohort study of 301 residents of Accra, Ghana. Participants received two doses of a COVID-19 vaccine (AZD1222 or BNT162b2) from December 2021 and were followed-up for 12 months. COVID-19 status was determined by RT-PCR at seven time points. Serological responses, including anti-Nucleocapsid IgG, anti-Spike IgG and live-virus neutralisation were determined at four time points during the 12 months follow-up. RESULTS COVID-19 positivity was 19.3% at baseline and reduced rapidly upon vaccination. Serological analyses indicated previous exposure to SARS-CoV-2 in 80.5% of the HERITAGE participants. After vaccination, neutralising antibody titres (NAbTs) against six different SARS-CoV-2 variants significantly (p < 0.001) increased, with fold changes (FC) ranging from 1.87 to 4.59. Highest NAbTs were recorded in the previously exposed group. Participants without prior exposure showed a continues increase in NAbTs between months 3 and 12 for circulating variants (Omicron B.A2 (FC 2.44, p < 0.001) and XBB.1.5 (FC 1.91, p = 0.05)). By comparison a matched cohort from the UK-based LEGACY study showed generally lower NAbTs at baseline (HERITAGE vs LEGACY for Wild-type: 250.3 vs 141.3, p < 0.0001, for A.27 84.6 vs 43.2, p = 0.0129, for Eta 159.7 vs 118.1, p = 0.3428, for Delta 158.6 vs 10.0, p < 0.0001, for Omicron B.A2 153.7 vs 10.0, p < 0.0001) and after receiving the vaccine (HERITAGE vs LEGACY for Wild-type: 882.6 vs 337.7, p < 0.0001, for A.27 552.0 vs 227.7, p = 0.0001, for Eta 682.2 vs 295.3, p < 0.0001, for Delta 557.6 vs 165.1, p < 0.0001, for Omicron B.A2 283.3 vs 124.2, p < 0.0001). NAbTs kinetics between the two cohorts were more similar when analysis was restricted to previously unexposed participants when adjusted for circulating variants during the sampling period. CONCLUSIONS Two doses of AZD1222 or BNT162b2 significantly increased existing NAbTs against SARS-CoV-2 in a highly exposed population, showing durable boosting of pre-existing infection-induced immunity. This indicates the importance of considering local population exposure in vaccination design and deployment.
Collapse
Affiliation(s)
| | - Yakubu Alhassan
- Yemaachi Biotech, Accra, Ghana
- Department of Biostatistics, School of Public Health, University of Ghana, Accra, Ghana
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Joyce M Ngoi
- Yemaachi Biotech, Accra, Ghana
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), University of Ghana, Accra, Ghana
| | | | | | | | - Edward J Carr
- The Francis Crick Institute, London, UK
- University College London, Gower St, London, UK
| | - Mary Y Wu
- The Francis Crick Institute, London, UK
| | | | - Emma C Wall
- The Francis Crick Institute, London, UK
- University College London, Gower St, London, UK
- National Institute for Health Research (NIHR) University College London Hospitals (UCLH) Biomedical Research Centre, London, UK
- NIHR UCLH Clinical Research Facility, London, UK
| | | | - Dzifa Dey
- University of Ghana Medical School, Korle Bu Teaching Hospital, Accra, Ghana
| | | | | | | | | | - Gordon Awandare
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), University of Ghana, Accra, Ghana
| | - Peter K Quashie
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), University of Ghana, Accra, Ghana
| | - Yaw Bediako
- Yemaachi Biotech, Accra, Ghana.
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), University of Ghana, Accra, Ghana.
| |
Collapse
Collaborators
Emmanuel Agbeli, Wisdom Akotia, Susan Amoako, Apetsi Ampiah, Charles Ansong, Seyram B Atukpa, Wisdom Aveey, Frank Danquah, Stephen L Darkoh, Patricia Kaba, Ruth Kiome, Esmy Kotey, Silas Lawer,
Collapse
|
2
|
Fabricius D, Ludwig C, Proffen M, Hägele J, Scholz J, Vieweg C, Rode I, Hoffmann S, Körper S, Schrezenmeier H, Jahrsdörfer B. Effective cellular and neutralizing immunity against SARS-CoV-2 after mRNA booster vaccination is associated with pDC and B cell activation. Front Immunol 2025; 16:1580448. [PMID: 40421016 PMCID: PMC12104250 DOI: 10.3389/fimmu.2025.1580448] [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: 02/20/2025] [Accepted: 04/14/2025] [Indexed: 05/28/2025] Open
Abstract
Introduction The emergence of SARS-CoV-2 variants of concern (VOCs), particularly Omicron, has challenged the efficacy of initial COVID-19 vaccination strategies. Booster immunizations, especially with mRNA vaccines, were introduced to enhance and prolong immune protection. However, the underlying mechanisms of humoral and cellular immunity induced by homologous versus heterologous vaccination regimens remain incompletely understood. This study aimed to elucidate the immune responses, including B cell, plasmacytoid dendritic cell (pDC), and T cell activation, following mRNA booster vaccination. Methods In a longitudinal cohort study, 136 individuals received three different vaccination regimens: homologous mRNA, heterologous vector-mRNA-mRNA, or heterologous vector-vector-mRNA vaccinations. Serum and peripheral blood mononuclear cells (PBMCs) were collected at multiple time points up to 64 weeks after initial vaccination. Anti-SARS-CoV-2 IgG titers and neutralization capacity against the wildtype virus and Omicron variant were measured using ELISA and cPass assays. Cellular immunity was assessed by IFN-γ release assays, and flow cytometry was employed to analyze B cell and pDC frequencies, viability, and activation markers. Functional pDC-mediated T cell activation was evaluated in mixed lymphocyte cultures. Results mRNA booster vaccination stabilized high anti-SARS-CoV-2 IgG titers and neutralizing activity against wildtype virus across all regimens, with the homologous mRNA group showing the highest antibody titers and Omicron neutralization capacity. Peripheral B cell frequencies and activation markers (MHC class I/II, CD86) were significantly upregulated post-booster. pDCs demonstrated enhanced antigen-presenting capacity and significantly promoted SARS-CoV-2-specific T cell IFN-γ responses in vitro. Despite differences in humoral responses between regimens, breakthrough infection rates up to 25 weeks post-booster were comparable across cohorts, suggesting compensatory mechanisms via cellular immunity. Discussion Our findings highlight the pivotal role of pDCs and T cells in sustaining effective immunity following mRNA booster vaccination. While homologous mRNA regimens induce superior humoral responses, robust cellular immunity in heterologous regimens may balance protection levels against breakthrough infections. The study underscores the importance of integrated humoral and cellular immune responses, suggesting potential for optimized booster strategies and pDC-targeted vaccine designs to enhance long-term protection against SARS-CoV-2 and emerging variants.
Collapse
Affiliation(s)
- Dorit Fabricius
- Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany
| | - Carolin Ludwig
- Department of Transfusion Medicine, Ulm University, Ulm, Germany
- Institute for Clinical Transfusion Medicine and Immunogenetics, German Red Cross Blood Transfusion Service Baden-Württemberg – Hessen and University Hospital Ulm, Ulm, Germany
| | - Matthias Proffen
- Department of Transfusion Medicine, Ulm University, Ulm, Germany
- Institute for Clinical Transfusion Medicine and Immunogenetics, German Red Cross Blood Transfusion Service Baden-Württemberg – Hessen and University Hospital Ulm, Ulm, Germany
| | - Janina Hägele
- Department of Transfusion Medicine, Ulm University, Ulm, Germany
- Institute for Clinical Transfusion Medicine and Immunogenetics, German Red Cross Blood Transfusion Service Baden-Württemberg – Hessen and University Hospital Ulm, Ulm, Germany
| | - Judith Scholz
- Department of Transfusion Medicine, Ulm University, Ulm, Germany
- Institute for Clinical Transfusion Medicine and Immunogenetics, German Red Cross Blood Transfusion Service Baden-Württemberg – Hessen and University Hospital Ulm, Ulm, Germany
| | - Christiane Vieweg
- Department of Transfusion Medicine, Ulm University, Ulm, Germany
- Institute for Clinical Transfusion Medicine and Immunogenetics, German Red Cross Blood Transfusion Service Baden-Württemberg – Hessen and University Hospital Ulm, Ulm, Germany
| | - Immanuel Rode
- Department of Transfusion Medicine, Ulm University, Ulm, Germany
- Institute for Clinical Transfusion Medicine and Immunogenetics, German Red Cross Blood Transfusion Service Baden-Württemberg – Hessen and University Hospital Ulm, Ulm, Germany
| | - Simone Hoffmann
- Department of Transfusion Medicine, Ulm University, Ulm, Germany
- Institute for Clinical Transfusion Medicine and Immunogenetics, German Red Cross Blood Transfusion Service Baden-Württemberg – Hessen and University Hospital Ulm, Ulm, Germany
| | - Sixten Körper
- Department of Transfusion Medicine, Ulm University, Ulm, Germany
- Institute for Clinical Transfusion Medicine and Immunogenetics, German Red Cross Blood Transfusion Service Baden-Württemberg – Hessen and University Hospital Ulm, Ulm, Germany
| | - Hubert Schrezenmeier
- Department of Transfusion Medicine, Ulm University, Ulm, Germany
- Institute for Clinical Transfusion Medicine and Immunogenetics, German Red Cross Blood Transfusion Service Baden-Württemberg – Hessen and University Hospital Ulm, Ulm, Germany
| | - Bernd Jahrsdörfer
- Department of Transfusion Medicine, Ulm University, Ulm, Germany
- Institute for Clinical Transfusion Medicine and Immunogenetics, German Red Cross Blood Transfusion Service Baden-Württemberg – Hessen and University Hospital Ulm, Ulm, Germany
| |
Collapse
|
3
|
Mcconney CS, Kenney D, Ennis CS, Smith-Mahoney EL, Ayuso MJ, Zhong J, Douam F, Sagar M, Snyder-Cappione JE. Individuals Infected with SARS-CoV-2 Prior to COVID-19 Vaccination Maintain Vaccine-Induced RBD-Specific Antibody Levels and Viral Neutralization Activity for One Year. Viruses 2025; 17:640. [PMID: 40431652 PMCID: PMC12115583 DOI: 10.3390/v17050640] [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: 02/06/2025] [Revised: 03/30/2025] [Accepted: 04/23/2025] [Indexed: 05/29/2025] Open
Abstract
The effectiveness of multiple COVID-19 vaccinations in individuals with a history of SARS-CoV-2 infection remains unclear; specifically, elucidation of the durability of anti-viral antibody responses could provide important insights for epidemiological applications. We utilized the BU ELISA protocol to measure the circulating SARS-CoV-2 receptor-binding domain (RBD) and nucleocapsid (N) specific IgG and IgA antibody levels in a cohort of individuals infected with SARS-CoV-2 in the spring of 2020, with the sample collection spanning six months to two years post-symptom onset. Further, we interrogated the neutralization activity of these samples against the ancestral SARS-CoV-2 (WA-1) and Delta and Omicron (BA.1) variants. Consistent with previous studies, we found a more rapid waning of anti-N compared to anti-RBD antibodies in months prior to the first vaccinations. Vaccine-induced antibody responses in individuals previously infected with SARS-CoV-2 were elevated and sustained for more than one year post-vaccination. Similarly, neutralization activity against WA-1, Delta, and Omicron increased and remained higher than pre-vaccination levels for one year after the first COVID-19 vaccine dose. Collectively, these results indicate that infection followed by vaccination yields robust antibody responses against SARS-CoV-2 that endure for one year. These results suggest that an annual booster would stably boost anti-SARS-CoV-2 antibody responses, preventing infection and disease.
Collapse
Affiliation(s)
- Christina S. Mcconney
- Department of Virology, Immunology, and Microbiology, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA 02118, USA; (C.S.M.); (D.K.); (M.J.A.); (J.Z.); (F.D.)
| | - Devin Kenney
- Department of Virology, Immunology, and Microbiology, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA 02118, USA; (C.S.M.); (D.K.); (M.J.A.); (J.Z.); (F.D.)
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA 02118, USA
| | - Christina S. Ennis
- Cancer Center, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA 02118, USA;
| | - Erika L. Smith-Mahoney
- Department of Virology, Immunology, and Microbiology, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA 02118, USA; (C.S.M.); (D.K.); (M.J.A.); (J.Z.); (F.D.)
| | - Maria Jose Ayuso
- Department of Virology, Immunology, and Microbiology, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA 02118, USA; (C.S.M.); (D.K.); (M.J.A.); (J.Z.); (F.D.)
| | - Jiabao Zhong
- Department of Virology, Immunology, and Microbiology, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA 02118, USA; (C.S.M.); (D.K.); (M.J.A.); (J.Z.); (F.D.)
| | - Florian Douam
- Department of Virology, Immunology, and Microbiology, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA 02118, USA; (C.S.M.); (D.K.); (M.J.A.); (J.Z.); (F.D.)
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA 02118, USA
| | - Manish Sagar
- Department of Medicine, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA 02118, USA;
| | - Jennifer E. Snyder-Cappione
- Department of Virology, Immunology, and Microbiology, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA 02118, USA; (C.S.M.); (D.K.); (M.J.A.); (J.Z.); (F.D.)
| |
Collapse
|
4
|
Castro-Trujillo S, Castro-Meneses J, Rojas MC, Castro-Amaya M, Lastra G, Narváez CF. Regulatory cytokines modulate early isotype-specific response associated with COVID-19 survival. Front Immunol 2025; 16:1543626. [PMID: 40342417 PMCID: PMC12058664 DOI: 10.3389/fimmu.2025.1543626] [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: 02/07/2025] [Accepted: 04/01/2025] [Indexed: 05/11/2025] Open
Abstract
Identifying immune markers driving early and effective antibody response in patients with severe coronavirus disease 2019 (COVID-19) is critical due to the threat of future coronavirus pandemics, incomplete global vaccination, and suboptimal booster coverage. Patients with life-threatening severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection are characterized by dysregulated thromboinflammation and cytokine storm that could influence the isotype virus-specific antibody response and the subsequent clinical outcome. We investigated the association between COVID-19-related mortality with the dynamics, magnitude, and relative avidity of nucleoprotein (N), spike (S), and receptor-binding domain (RBD)-specific IgM, IgA, and IgG in circulation. We also assessed the relationship between the virus-specific antibody responses and cytokine patterns, as well as systemic and pulmonary thromboinflammation markers. This multicenter study included COVID-19 patients hospitalized early in the pandemic, classified as survivors (n=62) and non-survivors (n=17). We developed indirect enzyme-linked immunosorbent assays (ELISAs) to evaluate each virus-specific isotype using well-characterized outpatient COVID-19 (n=180) and pre-pandemic cohorts (n=111). The pro-inflammatory interleukin (IL)-6 and tumor necrosis factor (TNF)-α, as well as the regulatory IL-10, transforming growth factor (TGF)-β1, and soluble tumor necrosis factor receptor I (sTNFRI) levels were evaluated. The ELISAs performed highly for all virus-specific isotypes, although modest for IgM-N. Non-survivors increased N-specific, but no S-specific, IgM and IgA responses throughout the disease course and, more notably, a delayed class switching to IgG-S and IgG-RBD compared to survivors. No differences were observed in the virus-specific IgG relative avidity. Survivors exhibited an antibody response proportional to the degree of systemic and pulmonary thromboinflammation, whereas non-survivors showed those dissociated because of their uncontrolled severe thromboinflammation. Only the survivors showed a dominant regulatory cytokine pattern in the early phase of infection (<10 days after symptoms onset), which strongly correlated with developing IgG-S and IgG-RBD protective antibodies. We developed easy-to-use immune assays that enable patient monitoring and identify at-risk populations in low- to middle-income regions. Non-survivors displayed an ineffective N-mediated antibody response, marked by an inability to control inflammation and a compromised time-dependent class switching toward S and RBD-specific IgG. The regulatory cytokine axis, including TGF-β1, maybe a critical immune correlate of effective antibody-mediated immunity in COVID-19.
Collapse
Affiliation(s)
- Sebastián Castro-Trujillo
- División de Inmunología, Programa de Medicina, Facultad de Ciencias de la Salud, Universidad Surcolombiana, Neiva, Huila, Colombia
| | - Juanita Castro-Meneses
- División de Inmunología, Programa de Medicina, Facultad de Ciencias de la Salud, Universidad Surcolombiana, Neiva, Huila, Colombia
- Programa de Biología Aplicada, Facultad de Ciencias Exactas y Naturales, Universidad Surcolombiana, Neiva, Huila, Colombia
| | - María Clemencia Rojas
- Dirección Laboratorio de Salud Pública, Secretaría de Salud Departamental, Gobernación del Huila, Neiva, Huila, Colombia
| | - Marcela Castro-Amaya
- Departamento de Medicina Interna, E.S.E. Hospital Universitario de Neiva. Programa de Medicina, Universidad Surcolombiana, Neiva, Huila, Colombia
| | - Giovani Lastra
- Departamento de Medicina Interna, E.S.E. Hospital Universitario de Neiva. Programa de Medicina, Universidad Surcolombiana, Neiva, Huila, Colombia
- Servicio de Neumología, E.S.E. Hospital Universitario de Neiva. Programa de Medicina, Universidad Surcolombiana, Neiva, Huila, Colombia
| | - Carlos F. Narváez
- División de Inmunología, Programa de Medicina, Facultad de Ciencias de la Salud, Universidad Surcolombiana, Neiva, Huila, Colombia
| |
Collapse
|
5
|
Çolak SM, İlgar T, Bahçeci İ, Özkaya E, Hüner Yiğit M, Durmuş H, Atiş F, Ertürk A, Yazıcı ZA. Clinical Significance of Neutralizing Antibodies in COVID-19: Implications for Disease Prognosis. Life (Basel) 2025; 15:429. [PMID: 40141774 PMCID: PMC11944107 DOI: 10.3390/life15030429] [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: 01/21/2025] [Revised: 02/23/2025] [Accepted: 03/05/2025] [Indexed: 03/28/2025] Open
Abstract
The pathogenesis of COVID-19 highlights a complex relationship between disease severity and neutralizing antibodies (NAbs). We aimed to investigate the relationships among the total NAb (tNAb) levels, the presence of potential neutralization antibodies (pNAbs), and the clinical outcomes of COVID-19 patients. Patients aged ≥18 years diagnosed between October 2021 and September 2022 were grouped by symptom severity. Blood samples were taken at two time points and data on demographics, epidemiology, and vaccination were recorded. The tNAbs and pNAbs were measured by an enhanced chemiluminescence assay and a surrogate virus neutralization test, respectively. The tNAbs of 68 and the pNAbs of 52 patients were analyzed. Twenty-six (38.2%) patients had severe infection. The 28-day mortality rate was 16.2% (n = 11). The tNAb levels in the control blood samples (CBSs) were significantly higher than those of the admission blood samples (ABSs) (p < 0.001). The statistical analysis showed no relationship between disease severity and pNAbs. Vaccinated patients had significantly higher tNAbs in the ABSs and CBSs (p < 0.001 and p < 0.001, respectively). The presence of pNAbs in the ABSs was correlated with a lower 28-day mortality (p = 0.026) and a milder disease course (p = 0.041). Although these findings support a correlation between tNAbs and disease severity and mortality, their presence seems to be unrelated to favorable clinical outcomes.
Collapse
Affiliation(s)
- Sudem Mahmutoğlu Çolak
- Departments of Infectious Diseases and Clinical Microbiology, Faculty of Medicine, Recep Tayyip Erdogan University, 53100 Rize, Türkiye; (S.M.Ç.); (T.İ.); (A.E.)
| | - Tuba İlgar
- Departments of Infectious Diseases and Clinical Microbiology, Faculty of Medicine, Recep Tayyip Erdogan University, 53100 Rize, Türkiye; (S.M.Ç.); (T.İ.); (A.E.)
| | - İlkay Bahçeci
- Departments of Medical Microbiology, Faculty of Medicine, Recep Tayyip Erdogan University, 53100 Rize, Türkiye; (İ.B.); (H.D.); (F.A.)
| | - Esra Özkaya
- Department of Microbiology, Faculty of Medicine, Karadeniz Technical University, 61080 Trabzon, Türkiye;
| | - Merve Hüner Yiğit
- Departments of Medical Biochemistry, Faculty of Medicine, Recep Tayyip Erdogan University, 53100 Rize, Türkiye;
| | - Hilal Durmuş
- Departments of Medical Microbiology, Faculty of Medicine, Recep Tayyip Erdogan University, 53100 Rize, Türkiye; (İ.B.); (H.D.); (F.A.)
| | - Feyza Atiş
- Departments of Medical Microbiology, Faculty of Medicine, Recep Tayyip Erdogan University, 53100 Rize, Türkiye; (İ.B.); (H.D.); (F.A.)
| | - Ayşe Ertürk
- Departments of Infectious Diseases and Clinical Microbiology, Faculty of Medicine, Recep Tayyip Erdogan University, 53100 Rize, Türkiye; (S.M.Ç.); (T.İ.); (A.E.)
| | - Zihni Acar Yazıcı
- Departments of Medical Microbiology, Faculty of Medicine, Recep Tayyip Erdogan University, 53100 Rize, Türkiye; (İ.B.); (H.D.); (F.A.)
| |
Collapse
|
6
|
Venturi G, Gallinaro A, Fortuna C, Pirillo MF, Scoglio A, Di Carlo B, Marsili G, Michelini Z, Amendola A, Carocci A, Dispinseri S, Borghi M, Canitano A, Falce C, Zappitelli A, Scarlatti G, Lixi ML, Aste A, Masala L, Baroncelli S, Cara A, Negri D. Viral and immune profiles during the first wave of SARS-CoV-2 infection in hospitalized patients in Sardinia, Italy. Sci Rep 2025; 15:6660. [PMID: 39994243 PMCID: PMC11850715 DOI: 10.1038/s41598-025-90324-5] [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: 04/05/2024] [Accepted: 02/12/2025] [Indexed: 02/26/2025] Open
Abstract
We performed a retrospective immunological analysis of the antibody response in serum and in nasopharyngeal swabs (NPS) obtained from 46 individuals infected with ancestral SARS-CoV-2 Wuhan-Hu-1 strain during the first COVID-19 wave in Cagliari (Sardinia, Italy), with a 4-month follow-up after the hospital admission. We implemented a comprehensive antibody response in serum and in mucosal samples using assays established in our laboratories. In NPS we evaluated the viral load by real time PCR, presence and kinetics of anti-Spike IgG and IgA by ELISA as well as their anti-Wuhan neutralization activity, showing induction and persistence of anti-viral immunity at the mucosal level. Neutralizing antibodies were measured in serum and NPS using a safe pseudovirus-based assay validated after comparison with a standard neutralization test using live SARS-CoV-2. We evaluated cross-neutralizing antibodies against all the major early variants of concerns (VoC) in sera. Of note, we detected a remarkable reduction of neutralizing activity against BA.1 compared to BA.2 and BA.5 Omicron subvariants, which was confirmed in sera from an analogous cohort of patients at the San Raffaele hospital in Milan, a geographically distant region of Italy, infected with the ancestral virus during the same period of time.
Collapse
Affiliation(s)
- Giulietta Venturi
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | | | - Claudia Fortuna
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | | | - Arianna Scoglio
- National Center for the Control and Evaluation of Medicines, Istituto Superiore di Sanità, Rome, Italy
| | - Beatrice Di Carlo
- National Center for the Control and Evaluation of Medicines, Istituto Superiore di Sanità, Rome, Italy
| | - Giulia Marsili
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Zuleika Michelini
- National Center for Global Health, Istituto Superiore di Sanità, Rome, Italy
| | - Antonello Amendola
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Alberto Carocci
- National Center for the Control and Evaluation of Medicines, Istituto Superiore di Sanità, Rome, Italy
| | - Stefania Dispinseri
- Viral Evolution and Transmission Unit, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Martina Borghi
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Andrea Canitano
- National Center for Global Health, Istituto Superiore di Sanità, Rome, Italy
| | - Chiara Falce
- National Center for Global Health, Istituto Superiore di Sanità, Rome, Italy
| | - Alice Zappitelli
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
- Center for Gender-Specific Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Gabriella Scarlatti
- Viral Evolution and Transmission Unit, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Maria Luisa Lixi
- Laboratory Medicine, Santissima Trinità Hospital, Cagliari, Italy
| | - Alessandra Aste
- Laboratory Medicine, Santissima Trinità Hospital, Cagliari, Italy
| | - Laura Masala
- Laboratory Medicine, Santissima Trinità Hospital, Cagliari, Italy
| | - Silvia Baroncelli
- National Center for Global Health, Istituto Superiore di Sanità, Rome, Italy
| | - Andrea Cara
- National Center for Global Health, Istituto Superiore di Sanità, Rome, Italy.
| | - Donatella Negri
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy.
| |
Collapse
|
7
|
Gallinaro A, Falce C, Pirillo MF, Borghi M, Grasso F, Canitano A, Cecchetti S, Baratella M, Michelini Z, Mariotti S, Chiantore MV, Farina I, Di Virgilio A, Tinari A, Scarlatti G, Negri D, Cara A. Simian Immunodeficiency Virus-Based Virus-like Particles Are an Efficient Tool to Induce Persistent Anti-SARS-CoV-2 Spike Neutralizing Antibodies and Specific T Cells in Mice. Vaccines (Basel) 2025; 13:216. [PMID: 40266067 PMCID: PMC11945333 DOI: 10.3390/vaccines13030216] [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: 12/30/2024] [Revised: 02/06/2025] [Accepted: 02/13/2025] [Indexed: 04/24/2025] Open
Abstract
Background/Objectives: Virus-like particles (VLPs) represent an attractive platform for delivering vaccine formulations, combining a high biosafety profile with a potent immune-stimulatory ability. VLPs are non-infectious, non-replicating, self-assembling nanostructures that can be exploited to efficiently expose membrane-tethered glycoproteins such as the SARS-CoV-2 Spike (S) protein, the main target of approved preventive vaccines. Here, we describe the development and preclinical validation of Simian Immunodeficiency Virus (SIV)-based GFP-labeled VLPs displaying S from the B.1.617.2 (Delta) variant (VLP/S-Delta) for inducing persistent anti-SARS-CoV-2 neutralizing antibodies (nAbs) and S-specific T cell responses in mice. Methods: SIV-derived VLP/S-Delta were produced by co-transfecting a plasmid expressing SIVGag-GFP, required for VLP assembly and quantification by flow virometry, a plasmid encoding the Delta S protein deleted in the cytoplasmic tail (CT), to improve membrane binding, and a VSV.G-expressing plasmid, to enhance VLP uptake. Recovered VLPs were titrated by flow virometry and characterized in vitro by transmission electron microscopy (TEM) and confocal microscopy (CLSM). BALB/c mice were immunized intramuscularly with VLP/S-Delta following a prime-boost regimen, and humoral and cellular immune responses were assessed. Results: VLP/S-Delta were efficiently pseudotyped with CT-truncated S-Delta. After BALB/c priming, VLP/S-Delta elicited both specific anti-RBD IgGs and anti-Delta nAbs that significantly increased after the boost and were maintained over time. The prime-boost vaccination induced similar levels of cross-nAbs against the ancestral Wuhan-Hu-1 strain as well as cross-nAbs against Omicron BA.1, BA.2 and BA.4/5 VoCs, albeit at lower levels. Moreover, immunization with VLP/S-Delta induced S-specific IFNγ-producing T cells. Conclusions: These data suggest that SIV-based VLPs are an appropriate delivery system for the elicitation of efficient and sustained humoral and cellular immunity in mice, paving the way for further improvements in the immunogen design to enhance the quality and breadth of immune responses against different viral glycoproteins.
Collapse
Affiliation(s)
- Alessandra Gallinaro
- National Center for Global Health, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.G.); (C.F.); (M.F.P.); (A.C.); (Z.M.)
| | - Chiara Falce
- National Center for Global Health, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.G.); (C.F.); (M.F.P.); (A.C.); (Z.M.)
| | - Maria Franca Pirillo
- National Center for Global Health, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.G.); (C.F.); (M.F.P.); (A.C.); (Z.M.)
| | - Martina Borghi
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy; (M.B.); (F.G.); (S.M.); (M.V.C.); (I.F.)
| | - Felicia Grasso
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy; (M.B.); (F.G.); (S.M.); (M.V.C.); (I.F.)
| | - Andrea Canitano
- National Center for Global Health, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.G.); (C.F.); (M.F.P.); (A.C.); (Z.M.)
| | - Serena Cecchetti
- Confocal Microscopy Unit NMR, Confocal Microscopy Area Core Facilities, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Marco Baratella
- Viral Evolution and Transmission Unit, IRCCS Ospedale San Raffaele, 20132 Milan, Italy; (M.B.); (G.S.)
| | - Zuleika Michelini
- National Center for Global Health, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.G.); (C.F.); (M.F.P.); (A.C.); (Z.M.)
| | - Sabrina Mariotti
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy; (M.B.); (F.G.); (S.M.); (M.V.C.); (I.F.)
| | - Maria Vincenza Chiantore
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy; (M.B.); (F.G.); (S.M.); (M.V.C.); (I.F.)
| | - Iole Farina
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy; (M.B.); (F.G.); (S.M.); (M.V.C.); (I.F.)
| | - Antonio Di Virgilio
- Center for Animal Research and Welfare, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Antonella Tinari
- Center for Gender Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Gabriella Scarlatti
- Viral Evolution and Transmission Unit, IRCCS Ospedale San Raffaele, 20132 Milan, Italy; (M.B.); (G.S.)
| | - Donatella Negri
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy; (M.B.); (F.G.); (S.M.); (M.V.C.); (I.F.)
| | - Andrea Cara
- National Center for Global Health, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.G.); (C.F.); (M.F.P.); (A.C.); (Z.M.)
| |
Collapse
|
8
|
Patel RS, Duque D, Bavananthasivam J, Hewitt M, Sandhu JK, Kumar R, Tran A, Agrawal B. Mixed lipopeptide-based mucosal vaccine candidate induces cross-variant immunity and protects against SARS-CoV-2 infection in hamsters. Immunohorizons 2025; 9:vlae011. [PMID: 39849995 PMCID: PMC11841972 DOI: 10.1093/immhor/vlae011] [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: 11/07/2024] [Accepted: 11/14/2024] [Indexed: 01/25/2025] Open
Abstract
The global dissemination of SARS-CoV-2 led to a worldwide pandemic in March 2020. Even after the official downgrading of the COVID-19 pandemic, infection with SARS-CoV-2 variants continues. The rapid development and deployment of SARS-CoV-2 vaccines helped to mitigate the pandemic to a great extent. However, the current vaccines are suboptimal; they elicit incomplete and short-lived protection and are ineffective against evolving virus variants. Updating the spike antigen according to the prevailing variant and repeated boosters is not the long-term solution. We have designed a lipopeptide-based, mucosal, pan-coronavirus vaccine candidate, derived from highly conserved and/or functional regions of the SARS-CoV-2 spike, nucleocapsid, and membrane proteins. Our studies demonstrate that the designed lipopeptides (LPMix) induced both cellular and humoral (mucosal and systemic) immune responses upon intranasal immunization in mice. Furthermore, the antibodies bound to the wild-type and mutated S proteins of SARS-CoV-2 variants of concern, including Alpha, Beta, Delta and Omicron, and also led to efficient neutralization in a surrogate viral neutralization assay. Our sequence alignment and 3-dimensional molecular modeling studies demonstrated that spike-derived epitopes, P1 and P2, are sequentially and/or structurally conserved among the SARS-CoV-2 variants. The addition of a novel mucosal adjuvant, heat-killed Caulobacter crescentus, to the lipopeptide vaccine significantly bolstered mucosal antibody responses. Finally, the lipopeptide-based intranasal vaccine demonstrated significant improvement in lung pathologies in a hamster model of SARS-CoV-2 infection. These studies are fundamentally important and open new avenues in the investigation of an innovative, broadly protective intranasal vaccine platform for SARS-CoV-2 and its variants.
Collapse
Affiliation(s)
- Raj S Patel
- Department of Surgery, Faculty of Medicine and Dentistry, College of Health Sciences, University of Alberta, Edmonton, AB, Canada
| | - Diana Duque
- Infectious Diseases, Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada
| | - Jegarubee Bavananthasivam
- Infectious Diseases, Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada
| | - Melissa Hewitt
- Preclinical Imaging, Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada
| | - Jagdeep K Sandhu
- Preclinical Imaging, Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada
| | | | - Anh Tran
- Infectious Diseases, Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - Babita Agrawal
- Department of Surgery, Faculty of Medicine and Dentistry, College of Health Sciences, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
9
|
Krishna B, Metaxaki M, Perera M, Wills M, Sithole N. Comparison of different T cell assays for the retrospective determination of SARS-CoV-2 infection. J Gen Virol 2024; 105. [PMID: 39704047 DOI: 10.1099/jgv.0.002055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2024] Open
Abstract
It is important to be able to retrospectively determine severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections with high accuracy, both for post-coronavirus disease 2019 (COVID-19) epidemiological studies, and to distinguish between Long COVID and other multi-syndromic diseases that have overlapping symptoms. Although serum antibody levels can be measured to retrospectively diagnose SARS-CoV-2 infections, peptide stimulation of memory T cell responses is a more sensitive approach. This is because robust memory T cells are generated after SARS-CoV-2 infection and persist even after antibodies wane below detectability thresholds. In this study, we compare T cell responses using FluoroSpot-based methods and overnight stimulation of whole blood with SARS-CoV-2 peptides followed by an ELISA. Both approaches have comparable sensitivity and specificity but require different equipment and samples to be used. Furthermore, the elimination of peptides that cross-react with other coronaviruses increases the assay specificity but trades off some sensitivity. Finally, this approach can be used on archival, cryopreserved PBMCs. This work shows comparative advantages for several methods to measure SARS-CoV-2 T cell responses that could be utilized by any laboratory studying the effects of the coronavirus disease 2019 pandemic.
Collapse
Affiliation(s)
- Benjamin Krishna
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, UK
| | - Marina Metaxaki
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, UK
| | - Marianne Perera
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, UK
- Division of Virology, Department of Pathology, University of Cambridge, Cambridge, UK
| | - Mark Wills
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, UK
| | - Nyarie Sithole
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, UK
| |
Collapse
|
10
|
Barbon S, Armellin F, Passerini V, De Angeli S, Primerano S, Del Pup L, Durante E, Macchi V, De Caro R, Parnigotto PP, Veronesi A, Porzionato A. Innate immune response in COVID-19: single-cell multi-omics profile of NK lymphocytes in a clinical case series. Cell Commun Signal 2024; 22:496. [PMID: 39407208 PMCID: PMC11476714 DOI: 10.1186/s12964-024-01867-5] [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: 07/23/2024] [Accepted: 10/02/2024] [Indexed: 10/20/2024] Open
Abstract
BACKGROUND COVID-19 pandemic caused by the Severe Acute Respiratory Syndrome-Coronavirus-2 (SARS-CoV-2) represents the biggest global health emergency in recent decades. The host immune response to SARS-CoV-2 seems to play a key role in disease pathogenesis and clinical manifestations, with Natural Killer (NK) lymphocytes being among the targets of virus-induced regulation. METHODS This study performed a single-cell multi-omics analysis of transcripts and proteins of NK lymphocytes in COVID-19 patients, for the characterization of the innate immunological response to infection. NK cells were isolated from peripheral blood samples collected from adult subjects divided into 3 study groups: (1) non-infected subjects (Naïve group, n = 3), (2) post COVID-19 convalescent subjects (Healed group, n = 3) and (3) patients that were vaccinated against SARS-CoV-2 (Vaccine group, n = 3). Cells were then analysed by the BD Rhapsody System for the single-cell multi-omics investigation of transcriptome and membrane proteins. RESULTS The bioinformatic analysis identified 5 cell clusters which differentially expressed gene/protein markers, defining NK cell subsets as "Active NK cells" and "Mature NK cells". Calculating the relative proportion of each cluster within patient groups, more than 40% of the Naïve group cell population was found to belong to Mature NKs, whereas more than 75% of the Vaccine group cell population belonged to the cluster of Active NKs. Regarding the Healed group, it seemed to show intermediate phenotype between Active and Mature NK cells. Differential expression of specific genes, proteins and signaling pathways was detected comparing the profile of the 3 experimental groups, revealing a more activated NK cell phenotype in vaccinated patients versus recovered individuals. CONCLUSIONS The present study detected differential expression of NK cell markers in relation to SARS-CoV-2 infection and vaccine administration, suggesting the possibility to identify key molecular targets for clinical-diagnostic use of the individual response to viral infection and/or re-infection.
Collapse
Affiliation(s)
- Silvia Barbon
- Section of Human Anatomy, Department of Neuroscience, University of Padova, Via Gabelli 65, 35121, Padova, Italy.
- Foundation for Biology and Regenerative Medicine, Tissue Engineering and Signaling - T.E.S. Onlus, Padova, Italy.
| | - Fabrizio Armellin
- Complex Operative Unit of Transfusion Medicine - Marca Trevigiana Local Unit of Health and Social Services 2, Treviso Hospital, Piazzale dell'Ospedale 1, 31100, Treviso, Italy
| | - Verena Passerini
- Foundation for Biology and Regenerative Medicine, Tissue Engineering and Signaling - T.E.S. Onlus, Padova, Italy
| | - Sergio De Angeli
- Complex Operative Unit of Transfusion Medicine - Marca Trevigiana Local Unit of Health and Social Services 2, Treviso Hospital, Piazzale dell'Ospedale 1, 31100, Treviso, Italy
| | - Simona Primerano
- Complex Operative Unit of Transfusion Medicine - Marca Trevigiana Local Unit of Health and Social Services 2, Treviso Hospital, Piazzale dell'Ospedale 1, 31100, Treviso, Italy
| | - Laura Del Pup
- Complex Operative Unit of Transfusion Medicine - Marca Trevigiana Local Unit of Health and Social Services 2, Treviso Hospital, Piazzale dell'Ospedale 1, 31100, Treviso, Italy
| | - Elisabetta Durante
- Complex Operative Unit of Transfusion Medicine - Marca Trevigiana Local Unit of Health and Social Services 2, Treviso Hospital, Piazzale dell'Ospedale 1, 31100, Treviso, Italy
| | - Veronica Macchi
- Section of Human Anatomy, Department of Neuroscience, University of Padova, Via Gabelli 65, 35121, Padova, Italy
| | - Raffaele De Caro
- Section of Human Anatomy, Department of Neuroscience, University of Padova, Via Gabelli 65, 35121, Padova, Italy
- Foundation for Biology and Regenerative Medicine, Tissue Engineering and Signaling - T.E.S. Onlus, Padova, Italy
| | - Pier Paolo Parnigotto
- Foundation for Biology and Regenerative Medicine, Tissue Engineering and Signaling - T.E.S. Onlus, Padova, Italy
| | - Arianna Veronesi
- Complex Operative Unit of Transfusion Medicine - Marca Trevigiana Local Unit of Health and Social Services 2, Treviso Hospital, Piazzale dell'Ospedale 1, 31100, Treviso, Italy.
| | - Andrea Porzionato
- Section of Human Anatomy, Department of Neuroscience, University of Padova, Via Gabelli 65, 35121, Padova, Italy
- Foundation for Biology and Regenerative Medicine, Tissue Engineering and Signaling - T.E.S. Onlus, Padova, Italy
| |
Collapse
|
11
|
Kumar S, Nan L, Kalodimou G, Jany S, Freudenstein A, Brandmüller C, Müller K, Girl P, Ehmann R, Guggemos W, Seilmaier M, Wendtner CM, Volz A, Sutter G, Fux R, Tscherne A. Implementation of an Immunoassay Based on the MVA-T7pol-Expression System for Rapid Identification of Immunogenic SARS-CoV-2 Antigens: A Proof-of-Concept Study. Int J Mol Sci 2024; 25:10898. [PMID: 39456680 PMCID: PMC11508112 DOI: 10.3390/ijms252010898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 10/01/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
The emergence of hitherto unknown viral pathogens presents a great challenge for researchers to develop effective therapeutics and vaccines within a short time to avoid an uncontrolled global spread, as seen during the coronavirus disease 2019 (COVID-19) pandemic. Therefore, rapid and simple methods to identify immunogenic antigens as potential therapeutical targets are urgently needed for a better pandemic preparedness. To address this problem, we chose the well-characterized Modified Vaccinia virus Ankara (MVA)-T7pol expression system to establish a workflow to identify immunogens when a new pathogen emerges, generate candidate vaccines, and test their immunogenicity in an animal model. By using this system, we detected severe acute respiratory syndrome (SARS) coronavirus 2 (SARS-CoV-2) nucleoprotein (N)-, and spike (S)-specific antibodies in COVID-19 patient sera, which is in line with the current literature and our observations from previous immunogenicity studies. Furthermore, we detected antibodies directed against the SARS-CoV-2-membrane (M) and -ORF3a proteins in COVID-19 patient sera and aimed to generate recombinant MVA candidate vaccines expressing either the M or ORF3a protein. When testing our candidate vaccines in a prime-boost immunization regimen in humanized HLA-A2.1-/HLA-DR1-transgenic H-2 class I-/class II-knockout mice, we were able to demonstrate M- and ORF3a-specific cellular and humoral immune responses. Hence, the established workflow using the MVA-T7pol expression system represents a rapid and efficient tool to identify potential immunogenic antigens and provides a basis for future development of candidate vaccines.
Collapse
Affiliation(s)
- Satendra Kumar
- Division of Virology, Department of Veterinary Sciences, Ludwig Maximilians University Munich (LMU Munich), 85764 Oberschleißheim, Germany; (S.K.); (L.N.); (G.K.)
| | - Liangliang Nan
- Division of Virology, Department of Veterinary Sciences, Ludwig Maximilians University Munich (LMU Munich), 85764 Oberschleißheim, Germany; (S.K.); (L.N.); (G.K.)
| | - Georgia Kalodimou
- Division of Virology, Department of Veterinary Sciences, Ludwig Maximilians University Munich (LMU Munich), 85764 Oberschleißheim, Germany; (S.K.); (L.N.); (G.K.)
- German Center for Infection Research, Partner Site Munich, 85764 Oberschleißheim, Germany (R.E.)
| | - Sylvia Jany
- Division of Virology, Department of Veterinary Sciences, Ludwig Maximilians University Munich (LMU Munich), 85764 Oberschleißheim, Germany; (S.K.); (L.N.); (G.K.)
| | - Astrid Freudenstein
- Division of Virology, Department of Veterinary Sciences, Ludwig Maximilians University Munich (LMU Munich), 85764 Oberschleißheim, Germany; (S.K.); (L.N.); (G.K.)
| | - Christine Brandmüller
- Division of Virology, Department of Veterinary Sciences, Ludwig Maximilians University Munich (LMU Munich), 85764 Oberschleißheim, Germany; (S.K.); (L.N.); (G.K.)
| | - Katharina Müller
- German Center for Infection Research, Partner Site Munich, 85764 Oberschleißheim, Germany (R.E.)
- Bundeswehr Institute of Microbiology, 80937 Munich, Germany
| | - Philipp Girl
- German Center for Infection Research, Partner Site Munich, 85764 Oberschleißheim, Germany (R.E.)
- Bundeswehr Institute of Microbiology, 80937 Munich, Germany
- Chair of Bacteriology and Mycology, Department of Veterinary Sciences, Ludwig Maximilians University Munich (LMU Munich), 85764 Oberschleißheim, Germany
| | - Rosina Ehmann
- German Center for Infection Research, Partner Site Munich, 85764 Oberschleißheim, Germany (R.E.)
- Bundeswehr Institute of Microbiology, 80937 Munich, Germany
| | - Wolfgang Guggemos
- Munich Clinic Schwabing, Academic Teaching Hospital, Ludwig Maximilians University Munich (LMU Munich), 80804 Munich, Germany; (W.G.); (M.S.)
| | - Michael Seilmaier
- Munich Clinic Schwabing, Academic Teaching Hospital, Ludwig Maximilians University Munich (LMU Munich), 80804 Munich, Germany; (W.G.); (M.S.)
| | - Clemens-Martin Wendtner
- Medical Clinic III, University Hospital, Ludwig Maximilians University Munich (LMU Munich), 80336 Munich, Germany;
| | - Asisa Volz
- Institute of Virology, University of Veterinary Medicine Hannover, 30559 Hannover, Germany;
- German Center for Infection Research, Partner Site Hannover-Braunschweig, 30559 Hannover, Germany
| | - Gerd Sutter
- Division of Virology, Department of Veterinary Sciences, Ludwig Maximilians University Munich (LMU Munich), 85764 Oberschleißheim, Germany; (S.K.); (L.N.); (G.K.)
- German Center for Infection Research, Partner Site Munich, 85764 Oberschleißheim, Germany (R.E.)
| | - Robert Fux
- Division of Virology, Department of Veterinary Sciences, Ludwig Maximilians University Munich (LMU Munich), 85764 Oberschleißheim, Germany; (S.K.); (L.N.); (G.K.)
| | - Alina Tscherne
- Division of Virology, Department of Veterinary Sciences, Ludwig Maximilians University Munich (LMU Munich), 85764 Oberschleißheim, Germany; (S.K.); (L.N.); (G.K.)
- German Center for Infection Research, Partner Site Munich, 85764 Oberschleißheim, Germany (R.E.)
| |
Collapse
|
12
|
Dragotakes Q, Johnson PW, Buras MR, Carter RE, Joyner MJ, Bloch E, Gebo KA, Hanley DF, Henderson JP, Pirofski LA, Shoham S, Senefeld JW, Tobian AA, Wiggins CC, Wright RS, Paneth NS, Sullivan DJ, Casadevall A. Estimates of actual and potential lives saved in the United States from the use of COVID-19 convalescent plasma. Proc Natl Acad Sci U S A 2024; 121:e2414957121. [PMID: 39352932 PMCID: PMC11474081 DOI: 10.1073/pnas.2414957121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 08/23/2024] [Indexed: 10/04/2024] Open
Abstract
In the Spring of 2020, the United States of America (USA) deployed COVID-19 convalescent plasma (CCP) to treat hospitalized patients. Over 500,000 patients were treated with CCP during the first year of the pandemic. In this study, we estimated the number of actual inpatient lives saved by CCP treatment in the United States of America based on CCP weekly use, weekly national mortality data, and CCP mortality reduction data from meta-analyses of randomized controlled trials and real-world data. We also estimate the potential number of lives saved if CCP had been deployed for 100% of hospitalized patients or used in 15 to 75% of outpatients. Depending on the assumptions modeled in stratified analyses, we estimated that CCP saved between 16,476 and 66,296 lives. The CCP ideal use might have saved as many as 234,869 lives and prevented 1,136,133 hospitalizations. CCP deployment was a successful strategy for ameliorating the impact of the COVID-19 pandemic in the USA. This experience has important implications for convalescent plasma use in future infectious disease emergencies.
Collapse
Affiliation(s)
- Quigly Dragotakes
- Department of Molecular Microbiology and Immunology, Johns Hopkins School of Public Health, Baltimore, MD21205
| | - Patrick W. Johnson
- Division of Clinical Trials and Biostatistics, Mayo Clinic, Jacksonville, FL32224
| | - Matthew R. Buras
- Division of Biostatistics and Clinical Trials, Department of Quantitative Health Sciences, Scottsdale, AZ85259
| | - Rickey E. Carter
- Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, FL32224
| | - Michael J. Joyner
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN55905
| | - Evan Bloch
- Department of Pathology, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD21205
| | - Kelly A. Gebo
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD21205
| | - Daniel F. Hanley
- Department of Neurology, Brain Injury Outcomes Division, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD21205
| | - Jeffrey P. Henderson
- Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine, Louis, St. Louis, MO63110
| | - Liise-anne Pirofski
- Division of Infectious Diseases, Albert Einstein College of Medicine, New York, NY10461
| | - Shmuel Shoham
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD21205
| | - Jonathon W. Senefeld
- Department of Health and Kinesiology, University of Illinois Urbana-Champaign, Urbana, IL61801
| | - Aaron A.R. Tobian
- Department of Pathology, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD21205
| | - Chad C. Wiggins
- Department of Kinesiology, Michigan State University, East Lansing, MI48823
| | - R. Scott Wright
- Departments of Cardiovascular Medicine and Human Research Protection Program, Mayo Clinic, Rochester, MN55905
| | - Nigel S. Paneth
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI48823
- Department of Pediatrics and Human Development, Michigan State University, East Lansing, MI48823
| | - David J. Sullivan
- Department of Molecular Microbiology and Immunology, Johns Hopkins School of Public Health, Baltimore, MD21205
| | - Arturo Casadevall
- Department of Molecular Microbiology and Immunology, Johns Hopkins School of Public Health, Baltimore, MD21205
| |
Collapse
|
13
|
Macchia I, La Sorsa V, Ciervo A, Ruspantini I, Negri D, Borghi M, De Angelis ML, Luciani F, Martina A, Taglieri S, Durastanti V, Altavista MC, Urbani F, Mancini F. T Cell Peptide Prediction, Immune Response, and Host-Pathogen Relationship in Vaccinated and Recovered from Mild COVID-19 Subjects. Biomolecules 2024; 14:1217. [PMID: 39456150 PMCID: PMC11505848 DOI: 10.3390/biom14101217] [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: 07/29/2024] [Revised: 09/12/2024] [Accepted: 09/18/2024] [Indexed: 10/28/2024] Open
Abstract
COVID-19 remains a significant threat, particularly to vulnerable populations. The emergence of new variants necessitates the development of treatments and vaccines that induce both humoral and cellular immunity. This study aimed to identify potentially immunogenic SARS-CoV-2 peptides and to explore the intricate host-pathogen interactions involving peripheral immune responses, memory profiles, and various demographic, clinical, and lifestyle factors. Using in silico and experimental methods, we identified several CD8-restricted SARS-CoV-2 peptides that are either poorly studied or have previously unreported immunogenicity: fifteen from the Spike and three each from non-structural proteins Nsp1-2-3-16. A Spike peptide, LA-9, demonstrated a 57% response rate in ELISpot assays using PBMCs from 14 HLA-A*02:01 positive, vaccinated, and mild-COVID-19 recovered subjects, indicating its potential for diagnostics, research, and multi-epitope vaccine platforms. We also found that younger individuals, with fewer vaccine doses and longer intervals since infection, showed lower anti-Spike (ELISA) and anti-Wuhan neutralizing antibodies (pseudovirus assay), higher naïve T cells, and lower central memory, effector memory, and CD4hiCD8low T cells (flow cytometry) compared to older subjects. In our cohort, a higher prevalence of Vδ2-γδ and DN T cells, and fewer naïve CD8 T cells, seemed to correlate with strong cellular and lower anti-NP antibody responses and to associate with Omicron infection, absence of confusional state, and habitual sporting activity.
Collapse
Affiliation(s)
- Iole Macchia
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (I.M.); (M.L.D.A.); (S.T.)
| | - Valentina La Sorsa
- Research Promotion and Coordination Service, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Alessandra Ciervo
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.C.); (D.N.); (M.B.); (F.M.)
| | - Irene Ruspantini
- Core Facilities, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Donatella Negri
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.C.); (D.N.); (M.B.); (F.M.)
| | - Martina Borghi
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.C.); (D.N.); (M.B.); (F.M.)
| | - Maria Laura De Angelis
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (I.M.); (M.L.D.A.); (S.T.)
| | - Francesca Luciani
- National Center for the Control and Evaluation of Medicines, Istituto Superiore di Sanità, 00161 Rome, Italy; (F.L.); (A.M.)
| | - Antonio Martina
- National Center for the Control and Evaluation of Medicines, Istituto Superiore di Sanità, 00161 Rome, Italy; (F.L.); (A.M.)
| | - Silvia Taglieri
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (I.M.); (M.L.D.A.); (S.T.)
| | - Valentina Durastanti
- Neurology Unit, San Filippo Neri Hospital, ASL RM1, 00135 Rome, Italy; (V.D.); (M.C.A.)
| | | | - Francesca Urbani
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (I.M.); (M.L.D.A.); (S.T.)
| | - Fabiola Mancini
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.C.); (D.N.); (M.B.); (F.M.)
| |
Collapse
|
14
|
Trinh THK, Tran TD, Pham DL, Nguyen VN, Vu QTT, Pham TD, Nguyen PH, Le MK, Truong DDK, Hoang VA, Huynh N, Ngo DQ, Vuong LN. Characteristics of Immunogenicity against SARS-CoV-2 in a Community-Based Model of Care during the Fourth Wave of COVID-19 Outbreak in Ho Chi Minh City. Yonsei Med J 2024; 65:501-510. [PMID: 39193758 PMCID: PMC11359602 DOI: 10.3349/ymj.2023.0567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 02/20/2024] [Accepted: 03/25/2024] [Indexed: 08/29/2024] Open
Abstract
PURPOSE Although some immune protection from close contact with individuals who have coronavirus disease 2019 (COVID-19) has been documented, there is limited data on the seroprevalence of antibodies against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in individuals who were in lockdown with confirmed COVID-19 cases. This study investigated immunogenicity against SARS-CoV-2 in household members and people who lived near home-quarantined patients with COVID-19. MATERIALS AND METHODS This cross-sectional study was conducted during the community-based care that took place during lockdowns in District 10, Ho Chi Minh City, Vietnam from July to September 2021. SARS-CoV-2 antibody levels were determined in index cases of COVID-19, household contacts, and a no-contact group from the same area. RESULTS A total of 770 participants were included (355 index cases, 103 household contacts, and 312 no contacts). All index cases were unvaccinated, but >90% of individuals in the household and no-contact groups had received ≥1 vaccine dose. SARS-CoV-2 neutralizing antibodies (Nabs) were present in >77% of unvaccinated index cases versus 64%/65.4% in the household/no-contact groups (p=0.001). Antibody concentrations in unvaccinated index cases were significantly higher than those in household contacts and no contacts, with no difference between the latter groups. In all cases, antibody levels declined markedly ≥6 weeks after infection, and failed to persist beyond this time in the household and no-contact groups. CONCLUSION Community-based care may have helped to create community immunogenicity, but Nabs did not persist, highlighting a need for vaccination for all individuals before, or from 6 weeks after, infection with SARS-CoV-2.
Collapse
Affiliation(s)
- Tu Hoang Kim Trinh
- University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh, Vietnam
| | - Tuan Diep Tran
- University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh, Vietnam
| | - Duy Le Pham
- University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh, Vietnam
| | - Vinh Nhu Nguyen
- University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh, Vietnam
| | - Quan Tran Thien Vu
- University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh, Vietnam
| | | | - Phong Hoai Nguyen
- University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh, Vietnam
| | - Minh Kieu Le
- University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh, Vietnam
| | | | - Vu Anh Hoang
- University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh, Vietnam
| | - Nghia Huynh
- University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh, Vietnam
| | - Dat Quoc Ngo
- University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh, Vietnam
| | - Lan Ngoc Vuong
- University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh, Vietnam.
| |
Collapse
|
15
|
Spanjaart AM, Ljungman P, Tridello G, Schwartz J, Martinez-Cibrián N, Barba P, Kwon M, Lopez-Corral L, Martinez-Lopez J, Ferra C, Di Blasi R, Ghesquieres H, Mutsaers P, Calkoen F, Jak M, van Doesum J, Vermaat JSP, van der Poel M, Maertens J, Gambella M, Metafuni E, Ciceri F, Saccardi R, Nicholson E, Tholouli E, Matthew C, Potter V, Bloor A, Besley C, Roddie C, Wilson K, Nagler A, Campos A, Petersen SL, Folber F, Bader P, Finke J, Kroger N, Knelange N, de La Camara R, Kersten MJ, Mielke S. Improved outcome of COVID-19 over time in patients treated with CAR T-cell therapy: Update of the European COVID-19 multicenter study on behalf of the European Society for Blood and Marrow Transplantation (EBMT) Infectious Diseases Working Party (IDWP) and the European Hematology Association (EHA) Lymphoma Group. Leukemia 2024; 38:1985-1991. [PMID: 39043963 PMCID: PMC11347385 DOI: 10.1038/s41375-024-02336-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 06/04/2024] [Accepted: 07/02/2024] [Indexed: 07/25/2024]
Abstract
COVID-19 has been associated with high mortality in patients treated with Chimeric Antigen Receptor (CAR) T-cell therapy for hematologic malignancies. Here, we investigated whether the outcome has improved over time with the primary objective of assessing COVID-19-attributable mortality in the Omicron period of 2022 compared to previous years. Data for this multicenter study were collected using the MED-A and COVID-19 report forms developed by the EBMT. One-hundred-eighty patients were included in the analysis, 39 diagnosed in 2020, 35 in 2021 and 106 in 2022. The median age was 58.9 years (min-max: 5.2-78.4). There was a successive decrease in COVID-19-related mortality over time (2020: 43.6%, 2021: 22.9%, 2022: 7.5%) and in multivariate analysis year of infection was the strongest predictor of survival (p = 0.0001). Comparing 2022 with 2020-2021, significantly fewer patients had lower respiratory symptoms (21.7% vs 37.8%, p = 0.01), needed oxygen support (25.5% vs 43.2%, p = 0.01), or were admitted to ICU (5.7% vs 33.8%, p = 0.0001). Although COVID-19-related mortality has decreased over time, CAR T-cell recipients remain at higher risk for complications than the general population. Consequently, vigilant monitoring for COVID-19 in patients undergoing B-cell-targeting CAR T-cell treatment is continuously recommended ensuring optimal prevention of infection and advanced state-of-the art treatment when needed.
Collapse
Affiliation(s)
- Anne Mea Spanjaart
- Department of Hematology, Amsterdam UMC location University of Amsterdam, Cancer Center Amsterdam and LYMMCARE, Amsterdam, The Netherlands
| | - Per Ljungman
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation, Karolinska University Hospital Huddinge and Karolinska Comprehensive Cancer Center, Stockholm, Sweden
| | - Gloria Tridello
- European Society for Blood and Marrow Transplantation (EBMT) Data Office, Department of Medical Statistics & Bioinformatics, Leiden, Netherlands
| | - Juana Schwartz
- European Society for Blood and Marrow Transplantation (EBMT) Leiden Study Unit, European Society for Blood and Marrow Transplantation (EBMT) Data Office, Leiden, Netherlands
| | | | - Pere Barba
- Department of Hematology, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Mi Kwon
- Department of Hematology, Hospital G. Universitario Gregorio Marañon, Institute of Health Research Gregorio Marañon, Madrid, Spain
| | - Lucia Lopez-Corral
- Department of Hematology, Hospital Universitario de Salamanca and IBSAL, Salamanca, Spain
| | - Joaquin Martinez-Lopez
- Department of Hematology, Hospital Univ. 12 de Octubre, Complutense University, CNIO, Madrid, Spain
| | - Christelle Ferra
- Clinical Hematology Department, Catalan Institute of Oncology, Hospital Germans Trias i Pujol, Josep Carreras Research Institute, Barcelona, Spain
| | - Roberta Di Blasi
- Department of Hematology, Assistance Publique Hôpitaux de Paris-Hopital Saint-Louis, Paris, France
| | - Hervé Ghesquieres
- Department of Hematology, Hospices Civils de Lyon, Lyon Sud Hospital, Pierre-Bénite, France
| | - Pim Mutsaers
- Department of Hematology, Erasmus MC Cancer Center, Rotterdam, the Netherlands
| | - Friso Calkoen
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Margot Jak
- Department of Hematology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Jaap van Doesum
- Department of Hematology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Joost S P Vermaat
- Department of Hematology, Leiden University Medical Center, Leiden, Netherlands
| | - Marjolein van der Poel
- Department of Hematology, Department of Internal Medicine, Division of Hematology, GROW School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, Netherlands
| | - Johan Maertens
- Deptartment of Hematology, University Hospital Gasthuisberg, Leuven, Belgium
| | - Massimiliano Gambella
- Department of Hematology and Cellular Therapy, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Elisabetta Metafuni
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica e Ematologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Fabio Ciceri
- Hematology and BMT Unit IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Riccardo Saccardi
- Cell Therapy and Transfusion Medicine Unit Azienda Ospedaliero Universitaria Careggi, Firenze, Italy
| | - Emma Nicholson
- Department of Haematology, The Royal Marsden Hospital, London, United Kingdom
| | - Eleni Tholouli
- Department of Clinical Haematology, Manchester Royal Infirmary, Manchester University NHS Foundation Trust, Manchester, UK
| | - Collin Matthew
- Adult HSCT unit, Northern Centre for Bone Marrow Transplantation, Newcastle Tyne, UK
| | - Victoria Potter
- King's College Hospital NHS Foundation Trust, Department of Haematological Medicine, Denmark Hill, London, UK
| | - Adrian Bloor
- Adult Leukaemia and Bone Marrow Transplant Unit, Christie NHS Foundation Trust Hospital, University of Manchester, Manchester, UK
| | - Caroline Besley
- Department of Haematology, University Hospitals Bristol and Weston NHSFT, Bristol, UK
| | - Claire Roddie
- Department of Haematology, University College London Hospital, London, UK
| | - Keith Wilson
- Blood and Bone Marrow Transplantation Department, University Hospital of Cardiff, Cardiff, UK
| | - Arnon Nagler
- Chaim Sheba Medical Center, Tel Aviv University, Tel Hashomer, Israel
| | - Antonio Campos
- Celular Therapy Department, Instituto Portugués de Oncologia do Porto, Francisco Gentil, E.P.E, Porto, Portugal
| | - Soeren Lykke Petersen
- Department of Hematology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Frantisek Folber
- Department of internal Medicine, Hematology and Oncology, Masaryk University Hospital Brno, Brno, Czech Republic
| | - Peter Bader
- Department for Children and Adolescents, Division for Stem Cell Transplantation and Immunology, University Hospital Frankfurt, Goethe University, Frankfurt, Germany
| | - Jurgen Finke
- Department of Hematology/Oncology/Stem Cell Transplantation, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg im Breisgau, Germany
| | - Nicolaus Kroger
- Department of Stem cell Transplantation, University Hospital Eppendorf, Hamburg, Germany
| | - Nina Knelange
- European Society for Blood and Marrow Transplantation (EBMT) Leiden Study Unit, European Society for Blood and Marrow Transplantation (EBMT) Data Office, Leiden, Netherlands
| | - Rafael de La Camara
- Department of Hematology, Hospital Universitario de La Princesa, Madrid, Spain
| | - Marie José Kersten
- Department of Hematology, Amsterdam UMC location University of Amsterdam, Cancer Center Amsterdam and LYMMCARE, Amsterdam, The Netherlands
| | - Stephan Mielke
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Department of Laboratory Medicine, Karolinska Institutet and University Hospital, Karolinska Comprehensive Cancer Center, Karolinska ATMP Center, Stockholm, Sweden.
- Cellular Therapy and immunobiology working party (CTIWP) of EBMT, .
| |
Collapse
|
16
|
Póvoa da Costa F, de Sarges KML, da Silva R, dos Santos EF, do Nascimento MH, Rodrigues AM, Cantanhede MHD, Rodrigues FBB, Viana MDNDSDA, Leite MDM, de Oliveira CF, Neves PFMD, Pereira Neto GDS, de Brito MTFM, da Silva ALS, Henriques DF, Quaresma JAS, Falcão LFM, Queiroz MAF, Vallinoto IMVC, Vallinoto ACR, Viana GMR, dos Santos EJM. Genetic, Clinical, Epidemiological, and Immunological Profiling of IgG Response Duration after SARS-CoV-2 Infection. Int J Mol Sci 2024; 25:8740. [PMID: 39201427 PMCID: PMC11354850 DOI: 10.3390/ijms25168740] [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: 06/03/2024] [Revised: 08/06/2024] [Accepted: 08/08/2024] [Indexed: 09/02/2024] Open
Abstract
The IgG response against SARS-CoV-2 infection can persist for over six months (long response; LR). However, among 30% of those infected, the duration can be as short as three months or less (short response; SR). The present study assembled serological data on the anti-SARS-CoV-2 IgG response duration of two previous studies and integrated these results with the plasmatic cytokine levels and genetic profile of 10 immune-relevant SNPs that were also previously published, along with the plasmatic total IgG, IgA, and IgM levels, allowing for the genetic, clinical, immunological, and epidemiological aspects of the post-COVID-19 IgG response duration to be understood. The SR was associated with previous mild acute COVID-19 and with an SNP (rs2228145) in IL6R related to low gene expression. Additionally, among the SR subgroup, no statistically significant Spearman correlations were observed between the plasma levels of IL-17A and the Th17 regulatory cytokines IFN-γ (rs = 0.2399; p = 0.1043), IL-4 (rs = 0.0273; p = 0.8554), and IL-2 (rs = 0.2204; p = 0.1365), while among the LR subgroup, weaker but statistically significant Spearman correlations were observed between the plasma levels of IL-17A and IFN-γ (rs = 0.3873; p = 0.0016), IL-4 (rs = 0.2671; p = 0.0328), and IL-2 (rs = 0.3959; p = 0.0012). These results suggest that the Th17 response mediated by the IL-6 pathway has a role in the prolonged IgG response to SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Flávia Póvoa da Costa
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Pará, Brazil; (F.P.d.C.); (K.M.L.d.S.); (R.d.S.); (E.F.d.S.); (M.H.d.N.); (A.M.R.); (M.H.D.C.); (F.B.B.R.); (M.d.N.d.S.d.A.V.); (M.d.M.L.); (M.T.F.M.d.B.); (A.L.S.d.S.)
- Graduate Program in Biology of Infectious and Parasitic Agents, Federal University of Pará, Belém 66075-110, Pará, Brazil; (G.d.S.P.N.); (M.A.F.Q.); (I.M.V.C.V.); (A.C.R.V.); (G.M.R.V.)
| | - Kevin Matheus Lima de Sarges
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Pará, Brazil; (F.P.d.C.); (K.M.L.d.S.); (R.d.S.); (E.F.d.S.); (M.H.d.N.); (A.M.R.); (M.H.D.C.); (F.B.B.R.); (M.d.N.d.S.d.A.V.); (M.d.M.L.); (M.T.F.M.d.B.); (A.L.S.d.S.)
- Graduate Program in Biology of Infectious and Parasitic Agents, Federal University of Pará, Belém 66075-110, Pará, Brazil; (G.d.S.P.N.); (M.A.F.Q.); (I.M.V.C.V.); (A.C.R.V.); (G.M.R.V.)
| | - Rosilene da Silva
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Pará, Brazil; (F.P.d.C.); (K.M.L.d.S.); (R.d.S.); (E.F.d.S.); (M.H.d.N.); (A.M.R.); (M.H.D.C.); (F.B.B.R.); (M.d.N.d.S.d.A.V.); (M.d.M.L.); (M.T.F.M.d.B.); (A.L.S.d.S.)
- Graduate Program in Biology of Infectious and Parasitic Agents, Federal University of Pará, Belém 66075-110, Pará, Brazil; (G.d.S.P.N.); (M.A.F.Q.); (I.M.V.C.V.); (A.C.R.V.); (G.M.R.V.)
| | - Erika Ferreira dos Santos
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Pará, Brazil; (F.P.d.C.); (K.M.L.d.S.); (R.d.S.); (E.F.d.S.); (M.H.d.N.); (A.M.R.); (M.H.D.C.); (F.B.B.R.); (M.d.N.d.S.d.A.V.); (M.d.M.L.); (M.T.F.M.d.B.); (A.L.S.d.S.)
- Graduate Program in Biology of Infectious and Parasitic Agents, Federal University of Pará, Belém 66075-110, Pará, Brazil; (G.d.S.P.N.); (M.A.F.Q.); (I.M.V.C.V.); (A.C.R.V.); (G.M.R.V.)
| | - Matheus Holanda do Nascimento
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Pará, Brazil; (F.P.d.C.); (K.M.L.d.S.); (R.d.S.); (E.F.d.S.); (M.H.d.N.); (A.M.R.); (M.H.D.C.); (F.B.B.R.); (M.d.N.d.S.d.A.V.); (M.d.M.L.); (M.T.F.M.d.B.); (A.L.S.d.S.)
| | - Alice Maciel Rodrigues
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Pará, Brazil; (F.P.d.C.); (K.M.L.d.S.); (R.d.S.); (E.F.d.S.); (M.H.d.N.); (A.M.R.); (M.H.D.C.); (F.B.B.R.); (M.d.N.d.S.d.A.V.); (M.d.M.L.); (M.T.F.M.d.B.); (A.L.S.d.S.)
| | - Marcos Henrique Damasceno Cantanhede
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Pará, Brazil; (F.P.d.C.); (K.M.L.d.S.); (R.d.S.); (E.F.d.S.); (M.H.d.N.); (A.M.R.); (M.H.D.C.); (F.B.B.R.); (M.d.N.d.S.d.A.V.); (M.d.M.L.); (M.T.F.M.d.B.); (A.L.S.d.S.)
- Graduate Program in Biology of Infectious and Parasitic Agents, Federal University of Pará, Belém 66075-110, Pará, Brazil; (G.d.S.P.N.); (M.A.F.Q.); (I.M.V.C.V.); (A.C.R.V.); (G.M.R.V.)
| | - Fabíola Brasil Barbosa Rodrigues
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Pará, Brazil; (F.P.d.C.); (K.M.L.d.S.); (R.d.S.); (E.F.d.S.); (M.H.d.N.); (A.M.R.); (M.H.D.C.); (F.B.B.R.); (M.d.N.d.S.d.A.V.); (M.d.M.L.); (M.T.F.M.d.B.); (A.L.S.d.S.)
- Graduate Program in Biology of Infectious and Parasitic Agents, Federal University of Pará, Belém 66075-110, Pará, Brazil; (G.d.S.P.N.); (M.A.F.Q.); (I.M.V.C.V.); (A.C.R.V.); (G.M.R.V.)
| | - Maria de Nazaré do Socorro de Almeida Viana
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Pará, Brazil; (F.P.d.C.); (K.M.L.d.S.); (R.d.S.); (E.F.d.S.); (M.H.d.N.); (A.M.R.); (M.H.D.C.); (F.B.B.R.); (M.d.N.d.S.d.A.V.); (M.d.M.L.); (M.T.F.M.d.B.); (A.L.S.d.S.)
- Graduate Program in Biology of Infectious and Parasitic Agents, Federal University of Pará, Belém 66075-110, Pará, Brazil; (G.d.S.P.N.); (M.A.F.Q.); (I.M.V.C.V.); (A.C.R.V.); (G.M.R.V.)
| | - Mauro de Meira Leite
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Pará, Brazil; (F.P.d.C.); (K.M.L.d.S.); (R.d.S.); (E.F.d.S.); (M.H.d.N.); (A.M.R.); (M.H.D.C.); (F.B.B.R.); (M.d.N.d.S.d.A.V.); (M.d.M.L.); (M.T.F.M.d.B.); (A.L.S.d.S.)
- Graduate Program in Biology of Infectious and Parasitic Agents, Federal University of Pará, Belém 66075-110, Pará, Brazil; (G.d.S.P.N.); (M.A.F.Q.); (I.M.V.C.V.); (A.C.R.V.); (G.M.R.V.)
| | - Camille Ferreira de Oliveira
- Section of Arbovirology and Hemorrhagic Fevers, Evandro Chagas Institute, Health Surveillance Secretariat, Brazilian Ministry of Health, Ananindeua 67000-000, Pará, Brazil; (C.F.d.O.); (D.F.H.)
| | - Pablo Fabiano Moura das Neves
- Center of Biological and Health Sciences, State University of Pará, Belém 66050-540, Pará, Brazil; (P.F.M.d.N.); (J.A.S.Q.); (L.F.M.F.)
| | - Gabriel dos Santos Pereira Neto
- Graduate Program in Biology of Infectious and Parasitic Agents, Federal University of Pará, Belém 66075-110, Pará, Brazil; (G.d.S.P.N.); (M.A.F.Q.); (I.M.V.C.V.); (A.C.R.V.); (G.M.R.V.)
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Pará, Brazil
| | - Mioni Thieli Figueiredo Magalhães de Brito
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Pará, Brazil; (F.P.d.C.); (K.M.L.d.S.); (R.d.S.); (E.F.d.S.); (M.H.d.N.); (A.M.R.); (M.H.D.C.); (F.B.B.R.); (M.d.N.d.S.d.A.V.); (M.d.M.L.); (M.T.F.M.d.B.); (A.L.S.d.S.)
| | - Andréa Luciana Soares da Silva
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Pará, Brazil; (F.P.d.C.); (K.M.L.d.S.); (R.d.S.); (E.F.d.S.); (M.H.d.N.); (A.M.R.); (M.H.D.C.); (F.B.B.R.); (M.d.N.d.S.d.A.V.); (M.d.M.L.); (M.T.F.M.d.B.); (A.L.S.d.S.)
| | - Daniele Freitas Henriques
- Section of Arbovirology and Hemorrhagic Fevers, Evandro Chagas Institute, Health Surveillance Secretariat, Brazilian Ministry of Health, Ananindeua 67000-000, Pará, Brazil; (C.F.d.O.); (D.F.H.)
| | - Juarez Antônio Simões Quaresma
- Center of Biological and Health Sciences, State University of Pará, Belém 66050-540, Pará, Brazil; (P.F.M.d.N.); (J.A.S.Q.); (L.F.M.F.)
| | - Luiz Fábio Magno Falcão
- Center of Biological and Health Sciences, State University of Pará, Belém 66050-540, Pará, Brazil; (P.F.M.d.N.); (J.A.S.Q.); (L.F.M.F.)
| | - Maria Alice Freitas Queiroz
- Graduate Program in Biology of Infectious and Parasitic Agents, Federal University of Pará, Belém 66075-110, Pará, Brazil; (G.d.S.P.N.); (M.A.F.Q.); (I.M.V.C.V.); (A.C.R.V.); (G.M.R.V.)
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Pará, Brazil
| | - Izaura Maria Vieira Cayres Vallinoto
- Graduate Program in Biology of Infectious and Parasitic Agents, Federal University of Pará, Belém 66075-110, Pará, Brazil; (G.d.S.P.N.); (M.A.F.Q.); (I.M.V.C.V.); (A.C.R.V.); (G.M.R.V.)
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Pará, Brazil
| | - Antonio Carlos Rosário Vallinoto
- Graduate Program in Biology of Infectious and Parasitic Agents, Federal University of Pará, Belém 66075-110, Pará, Brazil; (G.d.S.P.N.); (M.A.F.Q.); (I.M.V.C.V.); (A.C.R.V.); (G.M.R.V.)
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Pará, Brazil
| | - Giselle Maria Rachid Viana
- Graduate Program in Biology of Infectious and Parasitic Agents, Federal University of Pará, Belém 66075-110, Pará, Brazil; (G.d.S.P.N.); (M.A.F.Q.); (I.M.V.C.V.); (A.C.R.V.); (G.M.R.V.)
- Malaria Basic Research Laboratory, Parasitology Section, Evandro Chagas Institute, Health Surveillance Secretariat, Brazilian Ministry of Health, Ananindeua 67000-000, Pará, Brazil
| | - Eduardo José Melo dos Santos
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Pará, Brazil; (F.P.d.C.); (K.M.L.d.S.); (R.d.S.); (E.F.d.S.); (M.H.d.N.); (A.M.R.); (M.H.D.C.); (F.B.B.R.); (M.d.N.d.S.d.A.V.); (M.d.M.L.); (M.T.F.M.d.B.); (A.L.S.d.S.)
- Graduate Program in Biology of Infectious and Parasitic Agents, Federal University of Pará, Belém 66075-110, Pará, Brazil; (G.d.S.P.N.); (M.A.F.Q.); (I.M.V.C.V.); (A.C.R.V.); (G.M.R.V.)
| |
Collapse
|
17
|
Shoham S. Convalescent Plasma for Immunocompromised Patients. Curr Top Microbiol Immunol 2024. [PMID: 39117848 DOI: 10.1007/82_2024_272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/10/2024]
Abstract
COVID-19 convalescent plasma (CCP) is an important therapeutic option for immunocompromised patients with COVID-19. Such patients are at increased risk for serious complications of infection and may also develop a unique syndrome of persistent infection. This article reviews the rationale for CCP utilization in immunocompromised patients and the evidence for its value in immunosuppressed patients with both acute and persistent COVID-19. Both historical precedence and understanding of the mechanisms of action of antibody treatment support this use, as do several lines of evidence derived from case series, comparative studies, randomized trials, and systematic reviews of the literature. A summary of recommendations from multiple practice guidelines is also provided.
Collapse
Affiliation(s)
- Shmuel Shoham
- Department of Medicine, Johns Hopkins School of Medicine, 1830 East Monument St., Room 447, Baltimore, MD, 21205, USA.
| |
Collapse
|
18
|
Roohi A, Gharagozlou S. Vitamin D supplementation and calcium: Many-faced gods or nobody in fighting against Corona Virus Disease 2019. Clin Nutr ESPEN 2024; 62:172-184. [PMID: 38901939 DOI: 10.1016/j.clnesp.2024.05.015] [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: 11/15/2023] [Revised: 05/07/2024] [Accepted: 05/22/2024] [Indexed: 06/22/2024]
Abstract
In December 2019, Corona Virus Disease 2019 (COVID-19) was first identified and designated as a pandemic in March 2020 due to rapid spread of the virus globally. At the beginning of the pandemic, only a few treatment options, mainly focused on supportive care and repurposing medications, were available. Due to its effects on immune system, vitamin D was a topic of interest during the pandemic, and researchers investigated its potential impact on COVID-19 outcomes. However, the results of studies about the impact of vitamin D on the disease are inconclusive. In the present narrative review, different roles of vitamin D regarding the COVID-19 have been discussed to show that vitamin D supplementation should be recommended carefully.
Collapse
Affiliation(s)
- Azam Roohi
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran.
| | | |
Collapse
|
19
|
Lapadula G, Mezzadri L, Lo Cascio G, Antolini L, Malandrin S, Ranzani A, Limonta S, Cavallero A, Bonfanti P. Anti-spike antibody level is associated with the risk of clinical progression among subjects hospitalized with COVID-19 pneumonia: results from a retrospective cohort study. Infection 2024; 52:1499-1509. [PMID: 38652224 PMCID: PMC11289057 DOI: 10.1007/s15010-024-02250-9] [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: 12/19/2023] [Accepted: 03/25/2024] [Indexed: 04/25/2024]
Abstract
PURPOSE Antibodies against SARS-CoV-2 spike (anti-S) may confer protection against symptomatic COVID-19. Whether their level predicts progression among those with COVID-19 pneumonia remains unclear. METHODS We conducted a retrospective cohort study to assess predictors of anti-S levels and whether anti-S titer is associated with death or mechanical ventilation (MV). Adults hospitalized for COVID-19 pneumonia between July 2021 and July 2022 were enrolled if anti-S had been measured within 72 h of admission. Predictors of anti-S level were explored using multivariable quantile regression. The association between anti-S levels and 30-day death/MV was investigated via multivariable logistic regression. Analyses were stratified by vaccine status. RESULTS The median anti-S level was 1370 BAU/ml in 328 vaccinated and 15.5 BAU/ml in 206 unvaccinated individuals. Among the vaccinated, shorter symptom duration (p = 0.001), hematological malignancies (p = 0.002), and immunosuppressive therapy (p = 0.004) were associated with lower anti-S levels. In the unvaccinated group, symptom duration was the only predictor of anti-S levels (p < 0.001). After 30 days, 134 patients experienced death or MV. Among vaccinated individuals, higher anti-S levels correlated significantly with lower death/MV risk (per log2 increase, OR 0.88, 95%CI 0.81-0.97), irrespective of age and solid malignancies. Among unvaccinated, a marginally protective effect was observed (OR 0.86, 95%CI 0.73-1.01), independent of age, immunosuppressive therapy, and diabetes. Adjustment for monoclonal antibody treatment strengthened the association (OR 0.81, 95%CI 0.68-0.96). CONCLUSION This study suggests that levels of anti-S antibodies can predict critical or fatal outcomes in COVID-19 pneumonia patients, regardless of vaccination. Whether anti-S Ab could guide risk assessment and vaccination boosting merits further evaluation.
Collapse
Affiliation(s)
- Giuseppe Lapadula
- Infectious Diseases Unit, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy.
- School of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy.
| | - Luca Mezzadri
- School of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy
| | - Giustina Lo Cascio
- School of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy
| | - Laura Antolini
- Bicocca Bioinformatics Biostatistics and Bioimaging Center-B4, University of Milano-Bicocca, Milan, Italy
| | - Sergio Malandrin
- Microbiology Unit, Fondazione IRCCS San Gerardo Dei Tintori, Monza, Italy
| | - Alice Ranzani
- Infectious Diseases Unit, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Silvia Limonta
- Infectious Diseases Unit, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Annalisa Cavallero
- Microbiology Unit, Fondazione IRCCS San Gerardo Dei Tintori, Monza, Italy
| | - Paolo Bonfanti
- Infectious Diseases Unit, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
- School of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy
| |
Collapse
|
20
|
Noviello M, De Lorenzo R, Chimienti R, Maugeri N, De Lalla C, Siracusano G, Lorè NI, Rancoita PMV, Cugnata F, Tassi E, Dispinseri S, Abbati D, Beretta V, Ruggiero E, Manfredi F, Merolla A, Cantarelli E, Tresoldi C, Pastori C, Caccia R, Sironi F, Marzinotto I, Saliu F, Ghezzi S, Lampasona V, Vicenzi E, Cinque P, Manfredi AA, Scarlatti G, Dellabona P, Lopalco L, Di Serio C, Malnati M, Ciceri F, Rovere-Querini P, Bonini C. The longitudinal characterization of immune responses in COVID-19 patients reveals novel prognostic signatures for disease severity, patients' survival and long COVID. Front Immunol 2024; 15:1381091. [PMID: 39136010 PMCID: PMC11317765 DOI: 10.3389/fimmu.2024.1381091] [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: 02/02/2024] [Accepted: 06/07/2024] [Indexed: 08/15/2024] Open
Abstract
Introduction SARS-CoV-2 pandemic still poses a significant burden on global health and economy, especially for symptoms persisting beyond the acute disease. COVID-19 manifests with various degrees of severity and the identification of early biomarkers capable of stratifying patient based on risk of progression could allow tailored treatments. Methods We longitudinally analyzed 67 patients, classified according to a WHO ordinal scale as having Mild, Moderate, or Severe COVID-19. Peripheral blood samples were prospectively collected at hospital admission and during a 6-month follow-up after discharge. Several subsets and markers of the innate and adaptive immunity were monitored as putative factors associated with COVID-19 symptoms. Results More than 50 immunological parameters were associated with disease severity. A decision tree including the main clinical, laboratory, and biological variables at admission identified low NK-cell precursors and CD14+CD91+ monocytes, and high CD8+ Effector Memory T cell frequencies as the most robust immunological correlates of COVID-19 severity and reduced survival. Moreover, low regulatory B-cell frequency at one month was associated with the susceptibility to develop long COVID at six months, likely due to their immunomodulatory ability. Discussion These results highlight the profound perturbation of the immune response during COVID-19. The evaluation of specific innate and adaptive immune-cell subsets allows to distinguish between different acute and persistent COVID-19 symptoms.
Collapse
Affiliation(s)
- Maddalena Noviello
- Experimental Hematology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
- Cell Therapy Immunomonitoring Laboratory Monitoraggio Immunologico Terapie Cellulari (MITiCi), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Rebecca De Lorenzo
- Vita-Salute San Raffaele University, Milan, Italy
- Internal Medicine Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Raniero Chimienti
- Viral Evolution and Transmission Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Norma Maugeri
- Autoimmunity and Vascular Inflammation Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Claudia De Lalla
- Experimental Immunology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Gabriel Siracusano
- Immunobiology of Human Immunodeficiency Virus (HIV) Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Nicola Ivan Lorè
- Emerging Bacterial Pathogens Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Paola Maria Vittoria Rancoita
- University Centre for Statistics in the Biomedical Sciences (CUSSB), Vita-Salute San Raffaele University, Milan, Italy
| | - Federica Cugnata
- University Centre for Statistics in the Biomedical Sciences (CUSSB), Vita-Salute San Raffaele University, Milan, Italy
| | - Elena Tassi
- Experimental Hematology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
- Cell Therapy Immunomonitoring Laboratory Monitoraggio Immunologico Terapie Cellulari (MITiCi), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Stefania Dispinseri
- Viral Evolution and Transmission Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Danilo Abbati
- Experimental Hematology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Valeria Beretta
- Experimental Hematology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
- Cell Therapy Immunomonitoring Laboratory Monitoraggio Immunologico Terapie Cellulari (MITiCi), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Eliana Ruggiero
- Experimental Hematology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Francesco Manfredi
- Experimental Hematology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Aurora Merolla
- Vita-Salute San Raffaele University, Milan, Italy
- Internal Medicine Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Elisa Cantarelli
- Biological Resource Center Centro Risorse Biologiche-Ospedale San Raffaele (CRB-OSR), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Cristina Tresoldi
- Biological Resource Center Centro Risorse Biologiche-Ospedale San Raffaele (CRB-OSR), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Claudia Pastori
- Immunobiology of Human Immunodeficiency Virus (HIV) Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Roberta Caccia
- Neurovirology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Francesca Sironi
- Viral Evolution and Transmission Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Ilaria Marzinotto
- Diabetes Research Institute, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale San Raffaele, Milan, Italy
| | - Fabio Saliu
- Emerging Bacterial Pathogens Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Silvia Ghezzi
- Viral Pathogenesis and Biosafety Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Vito Lampasona
- Diabetes Research Institute, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale San Raffaele, Milan, Italy
| | - Elisa Vicenzi
- Viral Pathogenesis and Biosafety Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Paola Cinque
- Neurovirology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Angelo Andrea Manfredi
- Autoimmunity and Vascular Inflammation Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Gabriella Scarlatti
- Viral Evolution and Transmission Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Paolo Dellabona
- Experimental Immunology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Lucia Lopalco
- Immunobiology of Human Immunodeficiency Virus (HIV) Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Clelia Di Serio
- University Centre for Statistics in the Biomedical Sciences (CUSSB), Vita-Salute San Raffaele University, Milan, Italy
| | - Mauro Malnati
- Viral Evolution and Transmission Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Fabio Ciceri
- Vita-Salute San Raffaele University, Milan, Italy
- Hematology and Bone Marrow Transplant Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Patrizia Rovere-Querini
- Vita-Salute San Raffaele University, Milan, Italy
- Internal Medicine Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Chiara Bonini
- Experimental Hematology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
- Cell Therapy Immunomonitoring Laboratory Monitoraggio Immunologico Terapie Cellulari (MITiCi), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
21
|
Stahl-Hennig C, Peter AS, Cordsmeier A, Stolte-Leeb N, Vestweber R, Socher E, Merida SA, Sauermann U, Bleyer M, Fraedrich K, Grunwald T, Winkler TH, Ensser A, Jäck HM, Überla K. Genetic barrier to resistance: a critical parameter for efficacy of neutralizing monoclonal antibodies against SARS-CoV-2 in a nonhuman primate model. J Virol 2024; 98:e0062824. [PMID: 38899895 PMCID: PMC11265388 DOI: 10.1128/jvi.00628-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 05/12/2024] [Indexed: 06/21/2024] Open
Abstract
The potency of antibody neutralization in cell culture has been used as the key criterion for selection of antibodies against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) for clinical development. As other aspects may also influence the degree of protection in vivo, we compared the efficacy of two neutralizing monoclonal antibodies (TRES6 and 4C12) targeting different epitopes of the receptor binding domain (RBD) of SARS-CoV-2 in a prophylactic setting in rhesus monkeys. All four animals treated with TRES6 had reduced viral loads in the upper respiratory tract 2 days after naso-oropharyngeal challenge with the Alpha SARS-CoV-2 variant. Starting 2 days after challenge, mutations conferring resistance to TRES6 were dominant in two of the rhesus monkeys, with both animals failing to maintain reduced viral loads. Consistent with its lower serum neutralization titer at the day of challenge, prophylaxis with 4C12 tended to suppress viral load at day 2 less efficiently than TRES6. However, a week after challenge, mean viral loads in the lower respiratory tract in 4C12-treated animals were lower than in the TRES6 group and no mutations conferring resistance to 4C12 could be detected in viral isolates from nasal or throat swabs. Thus, genetic barrier to resistance seems to be a critical parameter for the efficacy of prophylaxis with monoclonal antibodies against SARS-CoV-2. Furthermore, comparison of antibody concentrations in respiratory secretions to those in serum shows reduced distribution of the 4C12 antibody into respiratory secretions and a delay in the appearance of antibodies in bronchoalveolar lavage fluid compared to their appearance in secretions of the upper respiratory tract.IMPORTANCEMonoclonal antibodies are a powerful tool for the prophylaxis and treatment of acute viral infections. Hence, they were one of the first therapeutic agents licensed for the treatment of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Oftentimes, the main criterion for the selection of antibodies for clinical development is their potency of neutralization in cell culture. By comparing two antibodies targeting the Spike protein of SARS-CoV-2, we now observed that the antibody that neutralized SARS-CoV-2 more efficiently in cell culture suppressed viral load in challenged rhesus monkeys to a lesser extent. Extraordinary rapid emergence of mutants of the challenge virus, which had lost their sensitivity to the antibody, was identified as the major reason for the reduced efficacy of the antibody in rhesus monkeys. Therefore, the viral genetic barrier to resistance to antibodies also affects their efficacy.
Collapse
Affiliation(s)
| | - Antonia Sophia Peter
- Institute of Clinical and Molecular Virology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Arne Cordsmeier
- Institute of Clinical and Molecular Virology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | | | - Ramona Vestweber
- Unit of Infection Models, German Primate Center, Göttingen, Germany
| | - Eileen Socher
- Institute of Anatomy, Functional and Clinical Anatomy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | | | - Ulrike Sauermann
- Unit of Infection Models, German Primate Center, Göttingen, Germany
| | - Martina Bleyer
- Unit of Infection Models, German Primate Center, Göttingen, Germany
| | - Kirsten Fraedrich
- Institute of Clinical and Molecular Virology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Thomas Grunwald
- Department of Vaccines and Infection Models, Fraunhofer Institute for Cell Therapy and Immunology IZI, Leipzig, Germany
| | - Thomas H. Winkler
- Division of Genetics, Department Biology, Nikolaus-Fiebiger-Center of Molecular Medicine, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Armin Ensser
- Institute of Clinical and Molecular Virology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Hans-Martin Jäck
- Division of Molecular Immunology, Internal Medicine III, Nikolaus-Fiebiger-Center of Molecular Medicine, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Klaus Überla
- Institute of Clinical and Molecular Virology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
22
|
Wagner JT, Müller-Schmucker SM, Wang W, Arnold P, Uhlig N, Issmail L, Eberlein V, Damm D, Roshanbinfar K, Ensser A, Oltmanns F, Peter AS, Temchura V, Schrödel S, Engel FB, Thirion C, Grunwald T, Wuhrer M, Grimm D, Überla K. Influence of AAV vector tropism on long-term expression and Fc-γ receptor binding of an antibody targeting SARS-CoV-2. Commun Biol 2024; 7:865. [PMID: 39009807 PMCID: PMC11250830 DOI: 10.1038/s42003-024-06529-3] [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: 11/17/2023] [Accepted: 07/02/2024] [Indexed: 07/17/2024] Open
Abstract
Long-acting passive immunization strategies are needed to protect immunosuppressed vulnerable groups from infectious diseases. To further explore this concept for COVID-19, we constructed Adeno-associated viral (AAV) vectors encoding the human variable regions of the SARS-CoV-2 neutralizing antibody, TRES6, fused to murine constant regions. An optimized vector construct was packaged in hepatotropic (AAV8) or myotropic (AAVMYO) AAV capsids and injected intravenously into syngeneic TRIANNI-mice. The highest TRES6 serum concentrations (511 µg/ml) were detected 24 weeks after injection of the myotropic vector particles and mean TRES6 serum concentrations remained above 100 µg/ml for at least one year. Anti-drug antibodies or TRES6-specific T cells were not detectable. After injection of the AAV8 particles, vector mRNA was detected in the liver, while the AAVMYO particles led to high vector mRNA levels in the heart and skeletal muscle. The analysis of the Fc-glycosylation pattern of the TRES6 serum antibodies revealed critical differences between the capsids that coincided with different binding activities to murine Fc-γ-receptors. Concomitantly, the vector-based immune prophylaxis led to protection against SARS-CoV-2 infection in K18-hACE2 mice. High and long-lasting expression levels, absence of anti-drug antibodies and favourable Fc-γ-receptor binding activities warrant further exploration of myotropic AAV vector-based delivery of antibodies and other biologicals.
Collapse
Affiliation(s)
- Jannik T Wagner
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Sandra M Müller-Schmucker
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Wenjun Wang
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, Netherlands
| | - Philipp Arnold
- Institute of Functional and Clinical Anatomy, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Nadja Uhlig
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Preclinical Validation, Leipzig, Germany
| | - Leila Issmail
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Preclinical Validation, Leipzig, Germany
| | - Valentina Eberlein
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Preclinical Validation, Leipzig, Germany
| | - Dominik Damm
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Kaveh Roshanbinfar
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Armin Ensser
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Friederike Oltmanns
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Antonia Sophia Peter
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Vladimir Temchura
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | | | - Felix B Engel
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | | | - Thomas Grunwald
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Preclinical Validation, Leipzig, Germany
| | - Manfred Wuhrer
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, Netherlands
| | - Dirk Grimm
- Department of Infectious Diseases/Virology, Section Viral Vector Technologies, Medical Faculty and Faculty of Engineering Sciences, University of Heidelberg; BioQuant Center, BQ0030, University of Heidelberg; German Center for Infection Research (DZIF), German Center for Cardiovascular Research (DZHK), partner site, Heidelberg, Germany
| | - Klaus Überla
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany.
| |
Collapse
|
23
|
Tong Z, Tong J, Lei W, Xie Y, Cui Y, Jia G, Li S, Zhang Z, Cheng Z, Xing X, Ma H, Deng L, Zhang R, Zhao X, Liu K, Wang Q, Qi J, Huang H, Song R, Su Z, Wu G, Lou J, Gao GF. Deciphering a reliable synergistic bispecific strategy of rescuing antibodies for SARS-CoV-2 escape variants, including BA.2.86, EG.5.1, and JN.1. Cell Rep 2024; 43:114338. [PMID: 38850530 DOI: 10.1016/j.celrep.2024.114338] [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: 01/25/2024] [Revised: 05/07/2024] [Accepted: 05/23/2024] [Indexed: 06/10/2024] Open
Abstract
The game between therapeutic monoclonal antibodies (mAbs) and continuously emerging severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants has favored the virus, as most therapeutic mAbs have been evaded. Addressing this challenge, we systematically explored a reproducible bispecific antibody (bsAb)-dependent synergistic effect in this study. It could effectively restore the neutralizing activity of the bsAb when any of its single mAbs is escaped by variants. This synergy is primarily attributed to the binding angle of receptor-binding domain (RBD)-5, facilitating inter-spike cross-linking and promoting cryptic epitope exposure that classical antibody cocktails cannot achieve. Furthermore, RBD-5 with RBD-2, RBD-6, and RBD-7, alongside RBD-8, also exhibit significantly enhanced effects. This study not only shifts the paradigm in understanding antibody interactions but paves the way for developing more effective therapeutic antibodies against rapidly mutating SARS-CoV-2, with Dia-19 already showing promise against emerging variants like BA.2.86, EG.5.1, and JN.1.
Collapse
Affiliation(s)
- Zhou Tong
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China; Shanxi Academy of Advanced Research and Innovation, Xinhua Road, Taiyuan, Shanxi 030032, China
| | - Jianyu Tong
- Shanxi Academy of Advanced Research and Innovation, Xinhua Road, Taiyuan, Shanxi 030032, China
| | - Wenwen Lei
- NHC Key Laboratory of Biosafety, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Yufeng Xie
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Yingzi Cui
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Guowen Jia
- The State Key Laboratory of Biotherapy, Frontiers Medical Center of Tianfu Jincheng Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan 610044, China
| | - Shihua Li
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Zezhong Zhang
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Zhimin Cheng
- Shanxi Academy of Advanced Research and Innovation, Xinhua Road, Taiyuan, Shanxi 030032, China
| | - Xiao Xing
- Shanxi Academy of Advanced Research and Innovation, Xinhua Road, Taiyuan, Shanxi 030032, China
| | - Haiyun Ma
- The State Key Laboratory of Biotherapy, Frontiers Medical Center of Tianfu Jincheng Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan 610044, China
| | - Lan Deng
- Sunshine Guojian Pharmaceutical (Shanghai) Co., Ltd., a 3SBio, Inc., company, 399 Libing Road, Shanghai 201203, China
| | - Rong Zhang
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Xin Zhao
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Kefang Liu
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Qihui Wang
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Jianxun Qi
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Haomin Huang
- Sunshine Guojian Pharmaceutical (Shanghai) Co., Ltd., a 3SBio, Inc., company, 399 Libing Road, Shanghai 201203, China
| | - Rui Song
- Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| | - Zhaoming Su
- The State Key Laboratory of Biotherapy, Frontiers Medical Center of Tianfu Jincheng Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan 610044, China
| | - Guizhen Wu
- NHC Key Laboratory of Biosafety, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Jing Lou
- Sunshine Guojian Pharmaceutical (Shanghai) Co., Ltd., a 3SBio, Inc., company, 399 Libing Road, Shanghai 201203, China.
| | - George Fu Gao
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China.
| |
Collapse
|
24
|
Theel ES, Kirby JE, Pollock NR. Testing for SARS-CoV-2: lessons learned and current use cases. Clin Microbiol Rev 2024; 37:e0007223. [PMID: 38488364 PMCID: PMC11237512 DOI: 10.1128/cmr.00072-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2024] Open
Abstract
SUMMARYThe emergence and worldwide dissemination of SARS-CoV-2 required both urgent development of new diagnostic tests and expansion of diagnostic testing capacity on an unprecedented scale. The rapid evolution of technologies that allowed testing to move out of traditional laboratories and into point-of-care testing centers and the home transformed the diagnostic landscape. Four years later, with the end of the formal public health emergency but continued global circulation of the virus, it is important to take a fresh look at available SARS-CoV-2 testing technologies and consider how they should be used going forward. This review considers current use case scenarios for SARS-CoV-2 antigen, nucleic acid amplification, and immunologic tests, incorporating the latest evidence for analytical/clinical performance characteristics and advantages/limitations for each test type to inform current debates about how tests should or should not be used.
Collapse
Affiliation(s)
- Elitza S. Theel
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - James E. Kirby
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Nira R. Pollock
- Harvard Medical School, Boston, Massachusetts, USA
- Department of Laboratory Medicine, Boston Children’s Hospital, Boston, Massachusetts, USA
| |
Collapse
|
25
|
Bezanovic MR, Obradovic ZB, Bujandric N, Kocic N, Milanovic MK, Majkic M, Obrovski B, Grujic J. Reactivity of anti-SARS-CoV-2 antibodies in Serbian voluntary blood donors. Transfus Med 2024; 34:200-210. [PMID: 38561316 DOI: 10.1111/tme.13034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 02/07/2024] [Accepted: 03/12/2024] [Indexed: 04/04/2024]
Abstract
BACKGROUND The COVID-19 pandemic has major implications on the entire blood supply system worldwide. Seroepidemiological studies are certainly necessary for better understanding the global burden that the COVID-19 pandemic represents. OBJECTIVES In this study, we analysed the association between demographic factors, COVID-19 severity, vaccination status and the reactivity of anti-SARS-CoV-2 IgG antibodies in Serbian blood donors. MATERIALS AND METHODS In a prospective study, demographic data and data related to previous SARS-CoV-2 infection, COVID-19 severity and vaccination status among whole blood donors were analysed, from February 10 to August 10, 2022, at the Blood Transfusion Institute of Vojvodina, Serbia. The detection and determination of the level of anti-SARS-CoV-2 IgG antibodies were performed using LIAISON® SARS-CoV-2 TrimericS IgG immunoassay. RESULTS A total of 1190 blood donors were included, 24.5% were female and 75.5% were male while their average age was 41 years. Anti-SARS-CoV-2 antibody values ranged from 2.40 to 3120 BAU/ml with a mean value of 1354.56 BAU/ml. Statistical analysis showed that COVID-19 severity and vaccination status are linked with reactivity of anti-SARS-CoV-2 antibodies, while gender and age of voluntary blood donors are not related to the values of anti-SARS-CoV-2 antibodies. CONCLUSION The values of anti-SARS-CoV-2 antibodies in voluntary blood donors in Serbia are kept relatively high, especially in blood donors who have overcome the severe COVID-19, as well as in donors who have been vaccinated against COVID-19. Further SARS-CoV-2 seroprevalence studies in our country are certainly still necessary so global strategies to fight against COVID-19 would be adequately evaluated.
Collapse
Affiliation(s)
- Milomir Radoslav Bezanovic
- Department for Blood Collection, Testing and Production of Blood Products, Blood Transfusion Institute of Vojvodina, Novi Sad, Serbia
| | - Zorana Budakov Obradovic
- Department for Blood Collection, Testing and Production of Blood Products, Blood Transfusion Institute of Vojvodina, Novi Sad, Serbia
- Department of Transfusiology, Faculty of Medicine in Novi Sad, University of Novi Sad, Novi Sad, Serbia
| | - Nevenka Bujandric
- Department for Blood Collection, Testing and Production of Blood Products, Blood Transfusion Institute of Vojvodina, Novi Sad, Serbia
- Department of Transfusiology, Faculty of Medicine in Novi Sad, University of Novi Sad, Novi Sad, Serbia
| | - Neda Kocic
- Department for Blood Collection, Testing and Production of Blood Products, Blood Transfusion Institute of Vojvodina, Novi Sad, Serbia
| | - Mirjana Krga Milanovic
- Department for Blood Collection, Testing and Production of Blood Products, Blood Transfusion Institute of Vojvodina, Novi Sad, Serbia
| | - Milan Majkic
- Clinic for Orthopedic Surgery and Traumatology, Clinical Center of Vojvodina, Novi Sad, Serbia
| | - Boris Obrovski
- Department of Environmental Engineering and Occupational Health and Safety, Faculty of Technical Sciences in Novi Sad, University of Novi Sad, Novi Sad, Serbia
| | - Jasmina Grujic
- Department for Blood Collection, Testing and Production of Blood Products, Blood Transfusion Institute of Vojvodina, Novi Sad, Serbia
- Department of Transfusiology, Faculty of Medicine in Novi Sad, University of Novi Sad, Novi Sad, Serbia
| |
Collapse
|
26
|
Chao AS, Lin CY, Chiang MH, Lu KY, Tsai CK, Chen KJ, Chien CW, Wu TS, Chang YL, Chao A, Lin G, Chiu CY. Metabolomic profiling of maternal plasma identifies inverse associations of acetate and urea with anti-SARS-CoV-2 antibody titers following COVID-19 vaccination during pregnancy. J Mol Med (Berl) 2024; 102:819-830. [PMID: 38568327 DOI: 10.1007/s00109-024-02438-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 02/19/2024] [Accepted: 03/12/2024] [Indexed: 05/21/2024]
Abstract
We conducted a comprehensive metabolomic analysis of plasma samples obtained from pregnant women who displayed varying post-vaccination antibody titers after receiving mRNA-1273-SARS-CoV-2 vaccines. The study involved 62 pregnant women, all of whom had been vaccinated after reaching 24 weeks of gestation. To quantify post-vaccination plasma antibody titers, we employed binding antibody units (BAU) in accordance with the World Health Organization International Standard. Subsequently, we classified the study participants into three distinct BAU/mL categories: those with high titers (above 2000), medium titers (ranging from 1000 to 2000), and low titers (below 1000). Plasma metabolomic profiling was conducted using 1H nuclear magnetic resonance spectroscopy, and the obtained data were correlated with the categorized antibody titers. Notably, in pregnant women exhibiting elevated anti-SARS-CoV-2 antibody titers, reduced plasma concentrations of acetate and urea were observed. A significant negative correlation between these compounds and antibody titers was also evident. An analysis of metabolomics pathways revealed significant inverse associations between antibody titers and four distinct amino acid metabolic pathways: (1) biosynthesis of phenylalanine, tyrosine, and tryptophan; (2) biosynthesis of valine, leucine, and isoleucine; (3) phenylalanine metabolism; and (4) degradation of valine, leucine, and isoleucine. Additionally, an association between the synthesis and degradation pathways of ketone bodies was evident. In conclusion, we identified different metabolic pathways that underlie the diverse humoral responses triggered by COVID-19 mRNA vaccines during pregnancy. Our data hold significant implications for refining COVID-19 vaccination approaches in expectant mothers. KEY MESSAGES : Anti-SARS-CoV-2 antibody titers decline as the number of days since COVID-19 vaccination increases. Anti-SARS-CoV-2 antibody titers are inversely associated with acetate, a microbial-derived metabolite, and urea. Amino acid metabolism is significantly associated with SARS-CoV-2 antibody titers.
Collapse
Affiliation(s)
- An-Shine Chao
- Department of Obstetrics and Gynecology, New Taipei Municipal Tu Cheng Hospital, New Taipei City, Taiwan.
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital at Linkou and Chang Gung University College of Medicine, Taoyuan, Taiwan.
| | - Chiao-Yun Lin
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital at Linkou and Chang Gung University College of Medicine, Taoyuan, Taiwan
- Gynecologic Cancer Research Center, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Meng-Han Chiang
- Clinical Metabolomics Core Lab, Chang , Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Kuan-Ying Lu
- Clinical Metabolomics Core Lab, Chang , Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Cheng-Kun Tsai
- Clinical Metabolomics Core Lab, Chang , Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Kuan-Ju Chen
- Department of Obstetrics and Gynecology, New Taipei Municipal Tu Cheng Hospital, New Taipei City, Taiwan
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital at Linkou and Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Chih-Wei Chien
- Department of Obstetrics and Gynecology, New Taipei Municipal Tu Cheng Hospital, New Taipei City, Taiwan
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital at Linkou and Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Ting-Shu Wu
- Department of Infectious Control, Chang , Gung Memorial Hospital at Linkou and Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Yao-Lung Chang
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital at Linkou and Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Angel Chao
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital at Linkou and Chang Gung University College of Medicine, Taoyuan, Taiwan
- Gynecologic Cancer Research Center, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Gigin Lin
- Clinical Metabolomics Core Lab, Chang , Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
- Department of Medical Imaging and Intervention and Institute for Radiological Research, Chang Gung Memorial Hospital at Linkou and Chang Gung University, Taoyuan, Taiwan
| | - Chih-Yung Chiu
- Clinical Metabolomics Core Lab, Chang , Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.
- Division of Pediatric Pulmonology, Department of Pediatrics, Chang , Gung Memorial Hospital at Linkou and Chang Gung University College of Medicine, Taoyuan, Taiwan.
| |
Collapse
|
27
|
O'Mahoney C, Watt I, Fiedler S, Devenish S, Srikanth S, Justice E, Dover T, Dean D, Peng C. Microfluidic Diffusional Sizing (MDS) Measurements of Secretory Neutralizing Antibody Affinity Against SARS-CoV-2. Ann Biomed Eng 2024; 52:1653-1664. [PMID: 38459195 PMCID: PMC11082020 DOI: 10.1007/s10439-024-03478-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 02/19/2024] [Indexed: 03/10/2024]
Abstract
SARS-CoV-2 has rampantly spread around the globe and continues to cause unprecedented loss through ongoing waves of (re)infection. Increasing our understanding of the protection against infection with SARS-CoV-2 is critical to ending the pandemic. Serological assays have been widely used to assess immune responses, but secretory antibodies, the essential first line of defense, have been studied to only a limited extent. Of particular interest and importance are neutralizing antibodies, which block the binding of the spike protein of SARS-CoV-2 to the human receptor angiotensin-converting enzyme-2 (ACE2) and thus are essential for immune defense. Here, we employed Microfluidic Diffusional Sizing (MDS), an immobilization-free technology, to characterize neutralizing antibody affinity to SARS-CoV-2 spike receptor-binding domain (RBD) and spike trimer in saliva. Affinity measurement was obtained through a contrived sample and buffer using recombinant SARS-CoV-2 RBD and monoclonal antibody. Limited saliva samples demonstrated that MDS applies to saliva neutralizing antibody measurement. The ability to disrupt a complex of ACE2-Fc and spike trimer is shown. Using a quantitative assay on the patient sample, we determined the affinity and binding site concentration of the neutralizing antibodies.
Collapse
Affiliation(s)
- Cara O'Mahoney
- Department of Bioengineering, Clemson University, Clemson, SC, USA
| | - Ian Watt
- Fluidic Analytics, Cambridge, UK
| | | | | | - Sujata Srikanth
- Center for Innovative Medical Devices and Sensors, Clemson University, Clemson, SC, USA
| | - Erica Justice
- Center for Innovative Medical Devices and Sensors, Clemson University, Clemson, SC, USA
| | - Tristan Dover
- Center for Innovative Medical Devices and Sensors, Clemson University, Clemson, SC, USA
| | - Delphine Dean
- Department of Bioengineering, Clemson University, Clemson, SC, USA
- Center for Innovative Medical Devices and Sensors, Clemson University, Clemson, SC, USA
| | - Congyue Peng
- Department of Bioengineering, Clemson University, Clemson, SC, USA.
- Center for Innovative Medical Devices and Sensors, Clemson University, Clemson, SC, USA.
| |
Collapse
|
28
|
Mink S, Reimann P, Fraunberger P. Prognostic value of anti-SARS-CoV-2 antibodies: a systematic review. Clin Chem Lab Med 2024; 62:1029-1043. [PMID: 38349073 DOI: 10.1515/cclm-2023-1487] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 02/02/2024] [Indexed: 04/30/2024]
Abstract
OBJECTIVES Globally, over 772 million cases of COVID-19 have been reported. New variants of interest with corresponding spikes in case numbers continue to be identified. Vulnerable patients, including older adults or patients with severe comorbidities, continue to be at risk. A large body of evidence has been accumulated regarding anti-SARS-CoV-2-antibodies and COVID-19 but the usefulness of antibody measurements remains unclear. This systematic review aims to assess the prognostic value of anti-SARS-CoV-2-antibodies and their usefulness for guiding booster vaccinations. METHODS Studies in English and published between January 2020 and October 2023 were included. Studies that relied on multiparameter-models or comprised fewer than 100 participants were excluded. PubMed and via the WHO COVID-19 research database, Embase and Medline databases were searched. Study selection and quality assessment was conducted independently by two researchers. RESULTS After screening 1,160 studies, 33 studies comprising >30 million individuals were included. Anti-SARS-CoV-2-antibodies were strongly associated with reduced risk of SARS-CoV-2-infection and better outcomes, including mortality. Risk of infection and COVID-19 severity decreased with increasing antibody levels. CONCLUSIONS Anti-SARS-CoV-2-antibodies are useful for early identification of high-risk patients and timely adjustment of therapy. Protective thresholds may be applied to advise booster vaccinations but verification in separate cohorts is required.
Collapse
Affiliation(s)
- Sylvia Mink
- Central Medical Laboratories, Feldkirch, Austria
- Private University in the Principality of Liechtenstein, Triesen, Principality of Liechtenstein
| | - Patrick Reimann
- Private University in the Principality of Liechtenstein, Triesen, Principality of Liechtenstein
- Department of Internal Medicine, Academic Teaching Hospital Feldkirch, Feldkirch, Austria
| | - Peter Fraunberger
- Central Medical Laboratories, Feldkirch, Austria
- Private University in the Principality of Liechtenstein, Triesen, Principality of Liechtenstein
| |
Collapse
|
29
|
Reemann L, Kneidinger N, Sczepanski B, Koczulla AR. COVID-19 in Lung Transplant Recipients: A Report on 10 Recent Cases. Viruses 2024; 16:709. [PMID: 38793590 PMCID: PMC11126037 DOI: 10.3390/v16050709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 04/23/2024] [Accepted: 04/26/2024] [Indexed: 05/26/2024] Open
Abstract
Due to immunosuppression, transplant recipients are at higher risk of infections with SARS-CoV-2 and worse clinical outcomes than immunocompetent hosts. Furthermore, lung transplant patients represent a special group among solid organ recipients, since pneumonia is the main manifestation of COVID-19. However, data on the course of disease and the changes in morbidity and mortality during the course of the pandemic are limited. In our pulmonary rehabilitation clinic, we treat patients shortly after lung transplant as well as long-term transplant patients. Over the last almost 4 years of pandemic, we witnessed several COVID-19 infections in lung transplant patients in our clinic as well as patients who acquired an infection beforehand. In this paper, we aim at retrospectively describing a series of recent COVID-19 cases in our clinic, looking at the clinical course of disease and outcomes in lung transplant patients.
Collapse
Affiliation(s)
- Lea Reemann
- Institute for Pulmonary Rehabilitation Research, Schoen Klinik Berchtesgadener Land, 83471 Schoenau am Koenigssee, Germany; (B.S.); (A.R.K.)
| | - Nikolaus Kneidinger
- Department of Medicine V, Comprehensive Pneumology Center Munich (CPC-M), German Center for Lung Research (DZL), Ludwig-Maximilians University (LMU) University Hospital, 81377 Munich, Germany;
- Division of Pulmonology, Department of Internal Medicine, Medical University of Graz, 8010 Graz, Austria
| | - Bernd Sczepanski
- Institute for Pulmonary Rehabilitation Research, Schoen Klinik Berchtesgadener Land, 83471 Schoenau am Koenigssee, Germany; (B.S.); (A.R.K.)
| | - Andreas Rembert Koczulla
- Institute for Pulmonary Rehabilitation Research, Schoen Klinik Berchtesgadener Land, 83471 Schoenau am Koenigssee, Germany; (B.S.); (A.R.K.)
- Department of Pulmonary Rehabilitation, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Philipps-University of Marburg, 35043 Marburg, Germany
- Teaching Hospital, Paracelsus Medical University Salzburg, 5020 Salzburg, Austria
| |
Collapse
|
30
|
Kumar A, Tripathi P, Kumar P, Shekhar R, Pathak R. From Detection to Protection: Antibodies and Their Crucial Role in Diagnosing and Combatting SARS-CoV-2. Vaccines (Basel) 2024; 12:459. [PMID: 38793710 PMCID: PMC11125746 DOI: 10.3390/vaccines12050459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 04/20/2024] [Accepted: 04/22/2024] [Indexed: 05/26/2024] Open
Abstract
Understanding the antibody response to SARS-CoV-2, the virus responsible for COVID-19, is crucial to comprehending disease progression and the significance of vaccine and therapeutic development. The emergence of highly contagious variants poses a significant challenge to humoral immunity, underscoring the necessity of grasping the intricacies of specific antibodies. This review emphasizes the pivotal role of antibodies in shaping immune responses and their implications for diagnosing, preventing, and treating SARS-CoV-2 infection. It delves into the kinetics and characteristics of the antibody response to SARS-CoV-2 and explores current antibody-based diagnostics, discussing their strengths, clinical utility, and limitations. Furthermore, we underscore the therapeutic potential of SARS-CoV-2-specific antibodies, discussing various antibody-based therapies such as monoclonal antibodies, polyclonal antibodies, anti-cytokines, convalescent plasma, and hyperimmunoglobulin-based therapies. Moreover, we offer insights into antibody responses to SARS-CoV-2 vaccines, emphasizing the significance of neutralizing antibodies in order to confer immunity to SARS-CoV-2, along with emerging variants of concern (VOCs) and circulating Omicron subvariants. We also highlight challenges in the field, such as the risks of antibody-dependent enhancement (ADE) for SARS-CoV-2 antibodies, and shed light on the challenges associated with the original antigenic sin (OAS) effect and long COVID. Overall, this review intends to provide valuable insights, which are crucial to advancing sensitive diagnostic tools, identifying efficient antibody-based therapeutics, and developing effective vaccines to combat the evolving threat of SARS-CoV-2 variants on a global scale.
Collapse
Affiliation(s)
- Anoop Kumar
- Molecular Diagnostic Laboratory, National Institute of Biologicals, Noida 201309, India
| | - Prajna Tripathi
- Department of Microbiology and Immunology, Weill Cornell Medical College, New York, NY 10021, USA;
| | - Prashant Kumar
- R. Ken Coit College of Pharmacy, University of Arizona, Tucson, AZ 85721, USA
| | - Ritu Shekhar
- Department of Molecular Genetics and Microbiology, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Rajiv Pathak
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA
| |
Collapse
|
31
|
Huang L, Tong X, Cui J, Du X, Liao Y, Tan X, Ju Y, Zhong X, Zhou W, Xu X, Li Y. Recurrent and persistent fever after SARS-CoV-2 infection in patients with follicular lymphoma: A case series. Int J Infect Dis 2024; 141:106973. [PMID: 38395220 DOI: 10.1016/j.ijid.2024.02.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 02/15/2024] [Accepted: 02/16/2024] [Indexed: 02/25/2024] Open
Abstract
Although persistent or recurrent COVID-19 infection is well described in some immunosuppressed patient cohort, to date, there have been no reports of this phenomenon in the context of repeatedly negative SARS-CoV-2 testing in the upper respiratory tract. We reported six patients with follicular lymphoma who developed recurrent symptomatic COVID-19 infection. They tested persistently negative for SARS-CoV-2 on pharyngeal swabs and ultimately confirmed by bronchoalveolar lavage fluid metagenomics next-generation sequencing. All six patients presented with lymphopenia and B-cell depletion, and five of them received the anti-cluster of differentiation 20 treatment in the last year. Persistent fever was the most common symptom and bilateral ground-glass opacities were the primary pattern on chest computed tomography. A relatively long course of unnecessary and ineffective antibacterial and/or antifungal treatments was administered until the definitive diagnosis. Persistent fever subsided rapidly with nirmatrelvir/ritonavir treatment. Our case highlighted that recurrent COVID-19 infection should be suspected in immunocompromised patients with persistent fever despite negative pharyngeal swabs, and urgent bronchoalveolar lavage fluid testing is necessary. Treatment with nirmatrelvir/ritonavir appeared to be very effective in these patients.
Collapse
Affiliation(s)
- Lixue Huang
- Department of Pulmonary and Critical Care Medicine, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Science, Beijing, China
| | - Xunliang Tong
- Department of Pulmonary and Critical Care Medicine, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Science, Beijing, China
| | - Jia Cui
- Department of Pulmonary and Critical Care Medicine, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Science, Beijing, China
| | - Xiaoman Du
- Department of Pulmonary and Critical Care Medicine, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Science, Beijing, China
| | - Yixuan Liao
- Department of Pulmonary and Critical Care Medicine, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Science, Beijing, China
| | - Xiaoming Tan
- Department of Pulmonary and Critical Care Medicine, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Science, Beijing, China
| | - Yang Ju
- Department of Pulmonary and Critical Care Medicine, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Science, Beijing, China
| | - Xuefeng Zhong
- Department of Pulmonary and Critical Care Medicine, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Science, Beijing, China
| | - Wei Zhou
- Department of Pulmonary and Critical Care Medicine, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Science, Beijing, China
| | - Xiaomao Xu
- Department of Pulmonary and Critical Care Medicine, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Science, Beijing, China
| | - Yanming Li
- Department of Pulmonary and Critical Care Medicine, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Science, Beijing, China.
| |
Collapse
|
32
|
Sandeep, Shinde SH, Ahmed S, Sharma SS, Pande AH. Engineered polyspecific antibodies: A new frontier in the field of immunotherapeutics. Immunology 2024; 171:464-496. [PMID: 38140855 DOI: 10.1111/imm.13743] [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: 06/02/2023] [Accepted: 12/07/2023] [Indexed: 12/24/2023] Open
Abstract
The 21st-century beginning remarked with the huge success of monospecific MAbs, however, in the last couple of years, polyspecific MAbs (PsAbs) have been an interesting topic and show promise of being biobetter than monospecific MAbs. Polyspecificity, in which a single antibody serves multiple specific target binding, has been hypothesized to contribute to the development of a highly effective antibody repertoire for immune defence. This polyspecific MAb trend represents an explosion that is gripping the whole pharmaceutical industry. This review is concerned with the current development and quality enforcement of PsAbs. All provided literature on monospecific MAbs and polyspecific MAbs (PsAbs) were searched using various electronic databases such as PubMed, Google Scholar, Web of Science, Elsevier, Springer, ACS, Google Patent and books via the keywords Antibody engineering, Polyspecific antibody, Conventional antibody, non-conventional antibody, and Single domain antibody. In the literature, there are more than 100 different formats to construct PsAb by quadroma technology, chemical conjugation and genetic engineering. Till March 2023, nine PsAb have been approved around the world, and around 330 are in advanced developmental stages, showing the dominancy of PsAb in the growing health sector. Recent advancements in protein engineering techniques and the fusion of non-conventional antibodies have made it possible to create complex PsAbs that demonstrate higher stability and enhanced potency. This marks the most significant achievement for cancer immunotherapy, in which PsAbs have immense promise. It is worth mentioning that seven out of the nine PsAbs have been approved as anti-cancer therapy. As PsAbs continue to acquire prominence, they could pave the way for the development of novel immunotherapies for multiple diseases.
Collapse
Affiliation(s)
- Sandeep
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER), Mohali, Punjab, India
| | - Suraj H Shinde
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER), Mohali, Punjab, India
| | - Sakeel Ahmed
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Mohali, Punjab, India
| | - Shyam Sunder Sharma
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Mohali, Punjab, India
| | - Abhay H Pande
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER), Mohali, Punjab, India
| |
Collapse
|
33
|
Dababseh MMO, Sabaka P, Duraníková O, Horváthová S, Valkovič P, Straka I, Nagyová A, Boža V, Kravec M, Jurenka J, Koščálová A, Mihalov P, Marešová E, Bendžala M, Kušnírová A, Stankovič I. Delayed Antibody Response in the Acute Phase of Infection Is Associated with a Lower Mental Component of Quality of Life in Survivors of Severe and Critical COVID-19. J Clin Med 2024; 13:1938. [PMID: 38610703 PMCID: PMC11012816 DOI: 10.3390/jcm13071938] [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: 02/20/2024] [Revised: 03/19/2024] [Accepted: 03/22/2024] [Indexed: 04/14/2024] Open
Abstract
Background: The long-term sequelae of coronavirus disease 2019 (COVID-19) significantly affects quality of life (QoL) in disease survivors. Delayed development of the adaptive immune response is associated with more severe disease and a worse prognosis in COVID-19. The effects of delayed immune response on COVID-19 sequelae and QoL are unknown. Methods: We conducted a prospective study to assess the relationship between the delayed antibody response in the acute phase of infection in naïve unvaccinated patients suffering from severe or critical COVID-19 and their QoL 12 months after hospital discharge. The 12-item Short Form Survey (SF-12) questionnaire was used for assessment of QoL. The SF-12 evaluates both mental and physical components of QoL, incorporating a mental component score (MCS-12) and a physical component score (PCS-12). A delayed antibody response was defined as testing negative for anti-spike SARS-CoV-2 antibodies at the time of hospital admission. Results: The study included 274 patients (154 men and 120 women). Of the enrolled patients, 144 had a delayed immune response. These patients had a significantly lower MCS-12 (p = 0.002), but PCS-12 (p = 0.397) was not significantly different at the 12-month follow-up compared to patients with positive anti-spike SARS-CoV-2 antibodies. The MCS-12 at the time of follow-up was negatively associated with delayed antibody response irrespective of possible confounders (p = 0.006; B = 3.609; ηp2 = 0.035; 95% CI = 1.069-6.150). An MSC-12 below 50 points at the time of follow-up was positively associated with delayed antibody response (p = 0.001; B = 1.092; OR = 2.979; 95% CI = 1.554-5.711). Conclusions: This study confirmed that, in patients with severe and critical COVID-19, a negative result for anti-spike SARS-CoV-2 antibodies at the time of hospital admission is associated with a lower mental component of QoL in unvaccinated patients naïve to COVID-19 one year after hospital discharge.
Collapse
Affiliation(s)
- Mohammad Mahmud Otman Dababseh
- Department of Infectology and Geographical Medicine, Faculty of Medicine, Comenius University in Bratislava, 833 05 Bratislava, Slovakia; (M.M.O.D.); (A.N.); (J.J.); (P.M.); (E.M.); (M.B.)
| | - Peter Sabaka
- Department of Infectology and Geographical Medicine, Faculty of Medicine, Comenius University in Bratislava, 833 05 Bratislava, Slovakia; (M.M.O.D.); (A.N.); (J.J.); (P.M.); (E.M.); (M.B.)
| | - Oľga Duraníková
- 2nd Department of Neurology, Faculty of Medicine, Comenius University in Bratislava, 833 05 Bratislava, Slovakia; (O.D.); (S.H.); (P.V.); (I.S.)
| | - Simona Horváthová
- 2nd Department of Neurology, Faculty of Medicine, Comenius University in Bratislava, 833 05 Bratislava, Slovakia; (O.D.); (S.H.); (P.V.); (I.S.)
| | - Peter Valkovič
- 2nd Department of Neurology, Faculty of Medicine, Comenius University in Bratislava, 833 05 Bratislava, Slovakia; (O.D.); (S.H.); (P.V.); (I.S.)
- Institute of Normal and Pathological Physiology, Slovak Academy of Sciences, 814 38 Bratislava, Slovakia
| | - Igor Straka
- 2nd Department of Neurology, Faculty of Medicine, Comenius University in Bratislava, 833 05 Bratislava, Slovakia; (O.D.); (S.H.); (P.V.); (I.S.)
| | - Anna Nagyová
- Department of Infectology and Geographical Medicine, Faculty of Medicine, Comenius University in Bratislava, 833 05 Bratislava, Slovakia; (M.M.O.D.); (A.N.); (J.J.); (P.M.); (E.M.); (M.B.)
| | - Vladimír Boža
- Department of Applied Informatics, Faculty of Mathematics, Physics and Informatics, Comenius University in Bratislava, 842 48 Bratislava, Slovakia (M.K.)
| | - Marián Kravec
- Department of Applied Informatics, Faculty of Mathematics, Physics and Informatics, Comenius University in Bratislava, 842 48 Bratislava, Slovakia (M.K.)
| | - Ján Jurenka
- Department of Infectology and Geographical Medicine, Faculty of Medicine, Comenius University in Bratislava, 833 05 Bratislava, Slovakia; (M.M.O.D.); (A.N.); (J.J.); (P.M.); (E.M.); (M.B.)
| | - Alena Koščálová
- Department of Infectology, Slovak Medical University, 833 05 Bratislava, Slovakia;
| | - Peter Mihalov
- Department of Infectology and Geographical Medicine, Faculty of Medicine, Comenius University in Bratislava, 833 05 Bratislava, Slovakia; (M.M.O.D.); (A.N.); (J.J.); (P.M.); (E.M.); (M.B.)
| | - Eliška Marešová
- Department of Infectology and Geographical Medicine, Faculty of Medicine, Comenius University in Bratislava, 833 05 Bratislava, Slovakia; (M.M.O.D.); (A.N.); (J.J.); (P.M.); (E.M.); (M.B.)
| | - Matej Bendžala
- Department of Infectology and Geographical Medicine, Faculty of Medicine, Comenius University in Bratislava, 833 05 Bratislava, Slovakia; (M.M.O.D.); (A.N.); (J.J.); (P.M.); (E.M.); (M.B.)
| | - Alice Kušnírová
- 2nd Department of Neurology, Faculty of Medicine, Comenius University in Bratislava, 833 05 Bratislava, Slovakia; (O.D.); (S.H.); (P.V.); (I.S.)
| | - Igor Stankovič
- Department of Infectology and Geographical Medicine, Faculty of Medicine, Comenius University in Bratislava, 833 05 Bratislava, Slovakia; (M.M.O.D.); (A.N.); (J.J.); (P.M.); (E.M.); (M.B.)
| |
Collapse
|
34
|
Malik S, Asghar M, Waheed Y. Outlining recent updates on influenza therapeutics and vaccines: A comprehensive review. Vaccine X 2024; 17:100452. [PMID: 38328274 PMCID: PMC10848012 DOI: 10.1016/j.jvacx.2024.100452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 12/27/2023] [Accepted: 01/29/2024] [Indexed: 02/09/2024] Open
Abstract
Influenza virus has presented a considerable healthcare challenge during the past years, particularly in vulnerable groups with compromised immune systems. Therapeutics and vaccination have always been in research annals since the spread of influenza. Efforts have been going on to develop an antiviral therapeutic approach that could assist in better disease management and reduce the overall disease complexity, resistance development, and fatality rates. On the other hand, vaccination presents a chance for effective, long-term, cost-benefit, and preventive response against the morbidity and mortality associated with the influenza. However, the issues of resistance development, strain mutation, antigenic variability, and inability to cure wide-spectrum and large-scale strains of the virus by available vaccines remain there. The article gathers the updated data for the therapeutics and available influenza vaccines, their mechanism of action, shortcomings, and trials under clinical experimentation. A methodological approach has been adopted to identify the prospective therapeutics and available vaccines approved and within the clinical trials against the influenza virus. Review contains influenza therapeutics, including traditional and novel antiviral drugs and inhibitor therapies against influenza virus as well as research trials based on newer drug combinations and latest technologies such as nanotechnology and organic and plant-based natural products. Most recent development of influenza vaccine has been discussed including some updates on traditional vaccination protocols and discussion on next-generation and upgraded novel technologies. This review will help the readers to understand the righteous approach for dealing with influenza virus infection and for deducing futuristic approaches for novel therapeutic and vaccine trials against Influenza.
Collapse
Affiliation(s)
- Shiza Malik
- Bridging Health Foundation, Rawalpindi, Punjab 46000, Pakistan
| | - Muhammad Asghar
- Department of Biology, Lund University, Sweden
- Department of Healthcare Biotechnology, Atta-Ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), H-12, Islamabad, Pakistan
| | - Yasir Waheed
- Office of Research, Innovation, and Commercialization (ORIC), Shaheed Zulfiqar Ali Bhutto Medical University (SZABMU), Islamabad 44000, Pakistan
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Byblos 1401, Lebanon
| |
Collapse
|
35
|
Zhang D, Kukkar D, Kim KH, Bhatt P. A comprehensive review on immunogen and immune-response proteins of SARS-CoV-2 and their applications in prevention, diagnosis, and treatment of COVID-19. Int J Biol Macromol 2024; 259:129284. [PMID: 38211928 DOI: 10.1016/j.ijbiomac.2024.129284] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 01/03/2024] [Accepted: 01/04/2024] [Indexed: 01/13/2024]
Abstract
Exposure to severe acute respiratory syndrome-corona virus-2 (SARS-CoV-2) prompts humoral immune responses in the human body. As the auxiliary diagnosis of a current infection, the existence of viral proteins can be checked from specific antibodies (Abs) induced by immunogenic viral proteins. For people with a weakened immune system, Ab treatment can help neutralize viral antigens to resist and treat the disease. On the other hand, highly immunogenic viral proteins can serve as effective markers for detecting prior infections. Additionally, the identification of viral particles or the presence of antibodies may help establish an immune defense against the virus. These immunogenic proteins rather than SARS-CoV-2 can be given to uninfected people as a vaccination to improve their coping ability against COVID-19 through the generation of memory plasma cells. In this work, we review immunogenic and immune-response proteins derived from SARS-CoV-2 with regard to their classification, origin, and diverse applications (e.g., prevention (vaccine development), diagnostic testing, and treatment (via neutralizing Abs)). Finally, advanced immunization strategies against COVID-19 are discussed along with the contemporary circumstances and future challenges.
Collapse
Affiliation(s)
- Daohong Zhang
- College of Food Engineering, Ludong University, Yantai 264025, Shandong, China; Bio-Nanotechnology Research Institute, Ludong University, Yantai 264025, Shandong, China
| | - Deepak Kukkar
- Department of Biotechnology, Chandigarh University, Gharuan, Mohali 140413, Punjab, India; University Center for Research and Development, Chandigarh University, Gharuan, Mohali 140413, Punjab, India
| | - Ki-Hyun Kim
- Department of Civil & Environmental Engineering, Hanyang University, 222 Wangsimni-Ro, Seoul 04763, Republic of Korea.
| | - Poornima Bhatt
- Department of Biotechnology, Chandigarh University, Gharuan, Mohali 140413, Punjab, India; University Center for Research and Development, Chandigarh University, Gharuan, Mohali 140413, Punjab, India
| |
Collapse
|
36
|
Nedelcu I, Florian P, Ion D, Militaru E, Damalan A, Popescu CI, Hristea A. Dynamics of serum cross-neutralization capacity against SARS-CoV-2 Delta variant in convalescent COVID-19 patients. J Med Virol 2024; 96:e29448. [PMID: 38318776 DOI: 10.1002/jmv.29448] [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: 10/10/2023] [Revised: 01/17/2024] [Accepted: 01/23/2024] [Indexed: 02/07/2024]
Abstract
The magnitude and breadth of the neutralizing antibody response against variants of concern following natural infection would provide valuable insights regarding the immune response induced by severe acute respiratory syndrome-related coronavirus (SARS-CoV-2) infection. Herein, 25 patients were followed at 30 ±7 (Visit 1), 90± 15 (Visit 2), and 180 ± 15 (Visit 3) days post symptom onset (PSO). The neutralization titers against both Wuhan-Hu-1 (WT) and Delta variant were analyzed in parallel along with anti-Spike antibodies (anti-S1/S2 immunoglobulin G [IgG]). The median values of half-maximal neutralization titer (NT50 ) for the WT and Delta variants decreased by 75.8% and 82.2% at Visit 2 and by 85.4% and 81.4% at Visit 3, respectively. At Visit 1, the correlation between the anti-S1/S2 IgG and Nabs titers for the Delta variant was moderate for WT (r = 0.58) and weak for the Delta variant (r = 0.39). However, the correlation coefficient consistently remained above 0.7, with a very strong correlation at Visit 3 for both WT and Delta variants (r = 0.81). The dynamics of anti-S1/S2 IgG antibodies, NT50 , and cross-neutralization index correlated at different time points PSO. Longitudinal analysis of the cross-neutralization capacity of immune sera will inform upon the durability of the immune response against SARS CoV-2 variants.
Collapse
Affiliation(s)
- Iulia Nedelcu
- University of Medicine and Pharmacy "Carol Davila" Bucharest, Bucharest, Romania
- National Institute for Infectious Diseases "Prof. Dr. Matei Bals", Bucharest, Romania
| | - Paula Florian
- Institute of Biochemistry of the Romanian Academy, Bucharest, Romania
| | - Daniel Ion
- University of Medicine and Pharmacy "Carol Davila" Bucharest, Bucharest, Romania
| | - Eliza Militaru
- University of Medicine and Pharmacy "Carol Davila" Bucharest, Bucharest, Romania
- National Institute for Infectious Diseases "Prof. Dr. Matei Bals", Bucharest, Romania
| | - Anca Damalan
- Department of Prevention and Control of Nosocomial Infections, Fundeni Clinical Institute, Bucharest, Romania
| | - Costin-Ioan Popescu
- Institute of Biochemistry of the Romanian Academy, Bucharest, Romania
- Prothanor Biotech S.R.L, Bucharest, Romania
| | - Adriana Hristea
- University of Medicine and Pharmacy "Carol Davila" Bucharest, Bucharest, Romania
- National Institute for Infectious Diseases "Prof. Dr. Matei Bals", Bucharest, Romania
| |
Collapse
|
37
|
Lapuente D, Winkler TH, Tenbusch M. B-cell and antibody responses to SARS-CoV-2: infection, vaccination, and hybrid immunity. Cell Mol Immunol 2024; 21:144-158. [PMID: 37945737 PMCID: PMC10805925 DOI: 10.1038/s41423-023-01095-w] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Accepted: 10/13/2023] [Indexed: 11/12/2023] Open
Abstract
The emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in 2019 prompted scientific, medical, and biotech communities to investigate infection- and vaccine-induced immune responses in the context of this pathogen. B-cell and antibody responses are at the center of these investigations, as neutralizing antibodies (nAbs) are an important correlate of protection (COP) from infection and the primary target of SARS-CoV-2 vaccine modalities. In addition to absolute levels, nAb longevity, neutralization breadth, immunoglobulin isotype and subtype composition, and presence at mucosal sites have become important topics for scientists and health policy makers. The recent pandemic was and still is a unique setting in which to study de novo and memory B-cell (MBC) and antibody responses in the dynamic interplay of infection- and vaccine-induced immunity. It also provided an opportunity to explore new vaccine platforms, such as mRNA or adenoviral vector vaccines, in unprecedented cohort sizes. Combined with the technological advances of recent years, this situation has provided detailed mechanistic insights into the development of B-cell and antibody responses but also revealed some unexpected findings. In this review, we summarize the key findings of the last 2.5 years regarding infection- and vaccine-induced B-cell immunity, which we believe are of significant value not only in the context of SARS-CoV-2 but also for future vaccination approaches in endemic and pandemic settings.
Collapse
Affiliation(s)
- Dennis Lapuente
- Institut für klinische und molekulare Virologie, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossgarten 4, 91054, Erlangen, Germany
| | - Thomas H Winkler
- Department of Biology, Division of Genetics, Nikolaus-Fiebiger-Center for Molecular Medicine, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany.
- Medical Immunology Campus Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossplatz 1, 91054, Erlangen, Germany.
| | - Matthias Tenbusch
- Institut für klinische und molekulare Virologie, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossgarten 4, 91054, Erlangen, Germany
- Medical Immunology Campus Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossplatz 1, 91054, Erlangen, Germany
| |
Collapse
|
38
|
Lauritsen CJ, Trinh IV, Desai SP, Clancey E, Murrell AE, Rambaran S, Chandra S, Elliott DH, Smira AR, Mo Z, Stone AE, Agbodji A, Dugas CM, Satou R, Pridjian G, Longo S, Ley SH, Robinson JE, Norton EB, Piedimonte G, Gunn BM. Passive antibody transfer from pregnant women to their fetus are maximized after SARS-CoV-2 vaccination irrespective of prior infection. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. GLOBAL 2024; 3:100189. [PMID: 38268538 PMCID: PMC10805668 DOI: 10.1016/j.jacig.2023.100189] [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] [Received: 04/06/2023] [Revised: 07/19/2023] [Accepted: 08/14/2023] [Indexed: 01/26/2024]
Abstract
Background Pregnancy is associated with a higher risk of adverse symptoms and outcomes for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection for both mother and neonate. Antibodies can provide protection against SARS-CoV-2 infection and are induced in pregnant women after vaccination or infection. Passive transfer of these antibodies from mother to fetus in utero may provide protection to the neonate against infection. However, it is unclear whether the magnitude or quality and kinetics of maternally derived fetal antibodies differs in the context of maternal infection or vaccination. Objective We aimed to determine whether antibodies transferred from maternal to fetus differed in quality or quantity between infection- or vaccination-induced humoral immune responses. Methods We evaluated 93 paired maternal and neonatal umbilical cord blood plasma samples collected between October 2020 and February 2022 from a birth cohort of pregnant women from New Orleans, Louisiana, with histories of SARS-CoV-2 infection and/or vaccination. Plasma was profiled for the levels of spike-specific antibodies and induction of antiviral humoral immune functions, including neutralization and Fc-mediated innate immune effector functions. Responses were compared between 4 groups according to maternal infection and vaccination. Results We found that SARS-CoV-2 vaccination or infection during pregnancy increased the levels of antiviral antibodies compared to naive subjects. Vaccinated mothers and cord samples had the highest anti-spike antibody levels and antiviral function independent of the time of vaccination during pregnancy. Conclusions These results show that the most effective passive transfer of functional antibodies against SARS-CoV-2 in utero is achieved through vaccination, highlighting the importance of vaccination in pregnant women.
Collapse
Affiliation(s)
- Cody J. Lauritsen
- Paul G. Allen School of Global Health, Washington State University, Pullman, Wash
| | - Ivy V. Trinh
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, La
| | - Srushti P. Desai
- Department of Pediatrics, Tulane University School of Medicine, New Orleans, La
| | - Erin Clancey
- Paul G. Allen School of Global Health, Washington State University, Pullman, Wash
| | - Amelie E. Murrell
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, La
| | - Saraswatie Rambaran
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, La
| | - Sruti Chandra
- Department of Pediatrics, Tulane University School of Medicine, New Orleans, La
| | - Debra H. Elliott
- Department of Pediatrics, Tulane University School of Medicine, New Orleans, La
| | - Ashley R. Smira
- Department of Pediatrics, Tulane University School of Medicine, New Orleans, La
| | - Zhiyin Mo
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, La
| | - Addison E. Stone
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, La
| | - Ayitevi Agbodji
- Department of Pediatrics, Tulane University School of Medicine, New Orleans, La
| | - Courtney M. Dugas
- Department of Physiology, Tulane University School of Medicine, New Orleans, La
| | - Ryousuke Satou
- Department of Physiology, Tulane University School of Medicine, New Orleans, La
| | - Gabriella Pridjian
- Department of Obstetrics and Gynecology, Tulane University School of Medicine, New Orleans, La
| | | | - Sylvia H. Ley
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, La
| | - James E. Robinson
- Department of Pediatrics, Tulane University School of Medicine, New Orleans, La
| | - Elizabeth B. Norton
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, La
| | - Giovanni Piedimonte
- Department of Pediatrics, Tulane University School of Medicine, New Orleans, La
| | - Bronwyn M. Gunn
- Paul G. Allen School of Global Health, Washington State University, Pullman, Wash
| |
Collapse
|
39
|
Hill JA, Martens MJ, Young JAH, Bhavsar K, Kou J, Chen M, Lee LW, Baluch A, Dhodapkar MV, Nakamura R, Peyton K, Howard DS, Ibrahim U, Shahid Z, Armistead P, Westervelt P, McCarty J, McGuirk J, Hamadani M, DeWolf S, Hosszu K, Sharon E, Spahn A, Toor AA, Waldvogel S, Greenberger LM, Auletta JJ, Horowitz MM, Riches ML, Perales MA. SARS-CoV-2 vaccination in the first year after hematopoietic cell transplant or chimeric antigen receptor T cell therapy: A prospective, multicenter, observational study (BMT CTN 2101). MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.01.24.24301058. [PMID: 38343800 PMCID: PMC10854344 DOI: 10.1101/2024.01.24.24301058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/17/2024]
Abstract
Background The optimal timing of vaccination with SARS-CoV-2 vaccines after cellular therapy is incompletely understood. Objective To describe humoral and cellular responses after SARS-CoV-2 vaccination initiated <4 months versus 4-12 months after cellular therapy. Design Multicenter prospective observational study. Setting 34 centers in the United States. Participants 466 allogeneic hematopoietic cell transplant (HCT; n=231), autologous HCT (n=170), or chimeric antigen receptor T cell (CAR-T cell) therapy (n=65) recipients enrolled between April 2021 and June 2022. Interventions SARS-CoV-2 vaccination as part of routine care. Measurements We obtained blood prior to and after vaccinations at up to five time points and tested for SARS-CoV-2 spike (anti-S) IgG in all participants and neutralizing antibodies for Wuhan D614G, Delta B.1.617.2, and Omicron B.1.1.529 strains, as well as SARS-CoV-2-specific T cell receptors (TCRs), in a subgroup. Results Anti-S IgG and neutralizing antibody responses increased with vaccination in HCT recipients irrespective of vaccine initiation timing but were unchanged in CAR-T cell recipients initiating vaccines within 4 months. Anti-S IgG ≥2,500 U/mL was correlated with high neutralizing antibody titers and attained by the last time point in 70%, 69%, and 34% of allogeneic HCT, autologous HCT, and CAR-T cell recipients, respectively. SARS-CoV-2-specific T cell responses were attained in 57%, 83%, and 58%, respectively. Humoral and cellular responses did not significantly differ among participants initiating vaccinations <4 months vs 4-12 months after cellular therapy. Pre-cellular therapy SARS-CoV-2 infection or vaccination were key predictors of post-cellular therapy anti-S IgG levels. Limitations The majority of participants were adults and received mRNA vaccines. Conclusions These data support starting mRNA SARS-CoV-2 vaccination three to four months after allogeneic HCT, autologous HCT, and CAR-T cell therapy. Funding National Marrow Donor Program, Leukemia and Lymphoma Society, Multiple Myeloma Research Foundation, Novartis, LabCorp, American Society for Transplantation and Cellular Therapy, Adaptive Biotechnologies, and the National Institutes of Health.
Collapse
Affiliation(s)
- Joshua A Hill
- Vaccine and Infectious Disease, Fred Hutchinson Cancer Center, and Department of Medicine, University of Washington, Seattle, WA, USA
| | - Michael J Martens
- Center for International Blood and Marrow Transplantation Research, Medical College of Wisconsin, Milwaukee, WI, USA
- Division of Biostatistics, Medical College of Wisconsin, Milwaukee, WI, USA
| | | | - Kavita Bhavsar
- Center for International Blood and Marrow Transplantation Research, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Jianqun Kou
- Center for International Blood and Marrow Transplantation Research, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Min Chen
- Center for International Blood and Marrow Transplantation Research, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Lik Wee Lee
- Adaptive Biotechnologies Corp, Seattle, WA, USA
| | - Aliyah Baluch
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | | | | | | | | | | | - Zainab Shahid
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Paul Armistead
- University of North Carolina Medical Center, Chapel Hill, NC, USA
| | - Peter Westervelt
- Barnes-Jewish Hospital, Washington University, St. Louis, MO, USA
| | - John McCarty
- Virginia Commonwealth University, Richmond, VA, USA
| | | | | | - Susan DeWolf
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kinga Hosszu
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Elad Sharon
- National Cancer Institute, Bethesda, MD, USA
| | - Ashley Spahn
- National Marrow Donor Program/Center for International Blood and Marrow Transplant Research, Minneapolis, MN, USA
| | - Amir A Toor
- Lehigh Valley Health Network, Allentown, PA, USA
| | - Stephanie Waldvogel
- National Marrow Donor Program/Center for International Blood and Marrow Transplant Research, Minneapolis, MN, USA
| | | | - Jeffery J Auletta
- National Marrow Donor Program/Center for International Blood and Marrow Transplant Research, Minneapolis, MN, USA
- Nationwide Children's Hospital, Columbus, OH, USA
| | - Mary M Horowitz
- Center for International Blood and Marrow Transplantation Research, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Marcie L Riches
- Center for International Blood and Marrow Transplantation Research, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Miguel-Angel Perales
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
40
|
Izac JR, Kwee EJ, Gaigalas A, Wang L. Quantitative and Standardized Pseudovirus Neutralization Assay for COVID-19. Methods Mol Biol 2024; 2779:259-271. [PMID: 38526789 DOI: 10.1007/978-1-0716-3738-8_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/27/2024]
Abstract
COVID-19 is a global pandemic caused by the highly infectious SARS-CoV-2 virus. Efforts to combat SARS-CoV-2 infection include mass vaccination and development of monoclonal and convalescent plasma therapeutics that require precise measurements of correlative, functional neutralizing antibodies that prevent virus infection. Developing rapid, safe, easy-to-use, and high-quality neutralization assays are essential for the success of the massive effort. Here, we developed a vesicular stomatitis virus-based neutralization assay that was capable of quantifying varying degrees of neutralization in patient serum samples. This assay has two detection readouts, flow cytometry and live cell imaging. The two readout methods produced consistent values of all 50% neutralization titers, further enhancing measurement confidence on the assay. Moreover, the use of available reference standards such as the World Health Organization International Standard (NIBSC code 20/136) enables quantification and standardization of the pseudovirus neutralization assay with neutralizing antibody titers measured in International Units/mL. Quantitative and standardized neutralization assays are critical for reliable efficacy evaluation and comparison of numerous vaccines and therapeutics.
Collapse
Affiliation(s)
- Jerilyn R Izac
- Biosystem and Biomaterials Division, National Institute of Standards and Technology (NIST), Gaithersburg, MD, USA
| | - Edward J Kwee
- Biosystem and Biomaterials Division, National Institute of Standards and Technology (NIST), Gaithersburg, MD, USA
| | - Adolfas Gaigalas
- Biosystem and Biomaterials Division, National Institute of Standards and Technology (NIST), Gaithersburg, MD, USA
| | - Lili Wang
- Biosystem and Biomaterials Division, National Institute of Standards and Technology (NIST), Gaithersburg, MD, USA.
| |
Collapse
|
41
|
Lodz NA, Mat Tamizi NF, Abd Mutalip MH, Ganapathy SS, Lin CZ, Ismail R, Ahmad NA. Anti-S Antibodies Against SARS-CoV-2 Infection Among Four Types of Vaccines in Malaysia. Asia Pac J Public Health 2024; 36:96-103. [PMID: 38166431 DOI: 10.1177/10105395231223332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2024]
Abstract
Monitoring SARS-CoV-2 antibody levels can provide insights into a person's immunity to COVID-19 and inform decisions about vaccination and public health measures. Anti-S may be useful as an indicator of an effective immune response. Thus, we conducted this study that aimed to determine the immune response of anti-S antibodies against SARS-CoV-2 for all the vaccine types over time among adult recipients in Malaysia and to determine the associated factors. This study was a cohort that recruited 2513 respondents aged 18 years and above from June to December 2021. Each participant was followed-up for 1-year period from the initial vaccine dose (baseline). We found that the anti-S antibody generally increased for all vaccine types and peaked at two weeks after the second dose vaccination, with Pfizer recipients having the highest median of 100 (100.00-100.00). During the third-month follow-up, the seropositivity of anti-S antibody and the median level decreased for all vaccines. We found that type of vaccines, comorbid status, infection, and booster status were significantly associated with the anti-S antibody level after one year.
Collapse
Affiliation(s)
- Noor Aliza Lodz
- Institute for Public Health, National Institute of Health, Ministry of Health Malaysia, Setia Alam, Malaysia
| | | | - Mohd Hatta Abd Mutalip
- Institute for Public Health, National Institute of Health, Ministry of Health Malaysia, Setia Alam, Malaysia
| | - Shubash Shander Ganapathy
- Institute for Public Health, National Institute of Health, Ministry of Health Malaysia, Setia Alam, Malaysia
| | - Chong Zhuo Lin
- Institute for Public Health, National Institute of Health, Ministry of Health Malaysia, Setia Alam, Malaysia
| | - Rohani Ismail
- Vaccine-Borne Disease Sector, Ministry of Health Malaysia, Putrajaya, Malaysia
| | - Noor Ani Ahmad
- Institute for Public Health, National Institute of Health, Ministry of Health Malaysia, Setia Alam, Malaysia
| |
Collapse
|
42
|
AlSereidi A, Salih SQM, Mohammed RT, Zaidan A, Albayati H, Pamucar D, Albahri A, Zaidan B, Shaalan K, Al-Obaidi J, Albahri O, Alamoodi A, Abdul Majid N, Garfan S, Al-Samarraay M, Jasim A, Baqer M. Novel Federated Decision Making for Distribution of Anti-SARS-CoV-2 Monoclonal Antibody to Eligible High-Risk Patients. INTERNATIONAL JOURNAL OF INFORMATION TECHNOLOGY & DECISION MAKING 2024; 23:197-268. [DOI: 10.1142/s021962202250050x] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
Abstract
Context: When the epidemic first broke out, no specific treatment was available for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The urgent need to end this unusual situation has resulted in many attempts to deal with SARS-CoV-2. In addition to several types of vaccinations that have been created, anti-SARS-CoV-2 monoclonal antibodies (mAbs) have added a new dimension to preventative and treatment efforts. This therapy also helps prevent severe symptoms for those at a high risk. Therefore, this is one of the most promising treatments for mild to moderate SARS-CoV-2 cases. However, the availability of anti-SARS-CoV-2 mAb therapy is limited and leads to two main challenges. The first is the privacy challenge of selecting eligible patients from the distribution hospital networking, which requires data sharing, and the second is the prioritization of all eligible patients amongst the distribution hospitals according to dose availability. To our knowledge, no research combined the federated fundamental approach with multicriteria decision-making methods for the treatment of SARS-COV-2, indicating a research gap. Objective: This paper presents a unique sequence processing methodology that distributes anti-SARS-CoV-2 mAbs to eligible high-risk patients with SARS-CoV-2 based on medical requirements by using a novel federated decision-making distributor. Method: This paper proposes a novel federated decision-making distributor (FDMD) of anti-SARS-CoV-2 mAbs for eligible high-risk patients. FDMD is implemented on augmented data of 49,152 cases of patients with SARS-CoV-2 with mild and moderate symptoms. For proof of concept, three hospitals with 16 patients each are enrolled. The proposed FDMD is constructed from the two sides of claim sequencing: central federated server (CFS) and local machine (LM). The CFS includes five sequential phases synchronised with the LMs, namely, the preliminary criteria setting phase that determines the high-risk criteria, calculates their weights using the newly formulated interval-valued spherical fuzzy and hesitant 2-tuple fuzzy-weighted zero-inconsistency (IVSH2-FWZIC), and allocates their values. The subsequent phases are federation, dose availability confirmation, global prioritization of eligible patients and alerting the hospitals with the patients most eligible for receiving the anti-SARS-CoV-2 mAbs according to dose availability. The LM independently performs all local prioritization processes without sharing patients’ data using the provided criteria settings and federated parameters from the CFS via the proposed Federated TOPSIS (F-TOPSIS). The sequential processing steps are coherently performed at both sides. Results and Discussion: (1) The proposed FDMD efficiently and independently identifies the high-risk patients most eligible for receiving anti-SARS-CoV-2 mAbs at each local distribution hospital. The final decision at the CFS relies on the indexed patients’ score and dose availability without sharing the patients’ data. (2) The IVSH2-FWZIC effectively weighs the high-risk criteria of patients with SARS-CoV-2. (3) The local and global prioritization ranks of the F-TOPSIS for eligible patients are subjected to a systematic ranking validated by high correlation results across nine scenarios by altering the weights of the criteria. (4) A comparative analysis of the experimental results with a prior study confirms the effectiveness of the proposed FDMD. Conclusion: The proposed FDMD has the benefits of centrally distributing anti-SARS-CoV-2 mAbs to high-risk patients prioritized based on their eligibility and dose availability, and simultaneously protecting their privacy and offering an effective cure to prevent progression to severe SARS-CoV-2 hospitalization or death.
Collapse
Affiliation(s)
- Abeer AlSereidi
- Faculty of Engineering & IT, The British university in Dubia, United Arab Emirates
| | | | - R. T. Mohammed
- Department of Computing Science, College of Science, Komar University of Science and Technology (KUST), Sulaymaniyah, Iraq
| | - A. A. Zaidan
- Faculty of Engineering & IT, The British university in Dubia, United Arab Emirates
| | - Hassan Albayati
- Department of Business Administration, College of Administrative Science, The University of Mashreq, 10021 Baghdad, Iraq
- Department of Computing, Faculty of Arts, Computing and Creative Industry, Universiti Pendidikan Sultan Idris, Tanjung Malim, Malaysia
| | - Dragan Pamucar
- University of Defence in Belgrade, Department of Logistic, Pavla Jurisica Sturma 33, 11000 Belgrade, Serbia
| | - A. S. Albahri
- Informatics Institute for Postgraduate Studies (IIPS), Iraqi Commission for Computers and Informatics (ICCI), Baghdad, Iraq
- University of Information Technology and Communications (UOITC), Baghdad, Iraq
| | - B. B. Zaidan
- Future Technology Research Center, National Yunlin University of Science and Technology, 123 University Road, Section 3, Douliou, Yunlin 64002, Taiwan
| | - Khaled Shaalan
- Faculty of Engineering & IT, The British university in Dubia, United Arab Emirates
| | - Jameel Al-Obaidi
- Department of Biology, Faculty of Science and Mathematics, Universiti Pendidikan Sultan Idris, Tanjong Malim 35900, Perak, Malaysia
| | - O. S. Albahri
- Computer Techniques Engineering Department Mazaya University College, Thi-Qar, Nassiriya, Iraq
| | - Abdulah Alamoodi
- Department of Computing, Faculty of Arts, Computing and Creative Industry, Universiti Pendidikan Sultan Idris, Tanjung Malim, Malaysia
| | - Nazia Abdul Majid
- Institute of Biological Sciences, Faculty of Science, Universiti Malaya, Kuala Lumpur, 50603, Malaysia
| | - Salem Garfan
- Department of Computing, Faculty of Arts, Computing and Creative Industry, Universiti Pendidikan Sultan Idris, Tanjung Malim, Malaysia
| | - M. S. Al-Samarraay
- Department of Computing, Faculty of Arts, Computing and Creative Industry, Universiti Pendidikan Sultan Idris, Tanjung Malim, Malaysia
| | | | | |
Collapse
|
43
|
Holdenrieder S, Dos Santos Ferreira CE, Izopet J, Theel ES, Wieser A. Clinical and laboratory considerations: determining an antibody-based composite correlate of risk for reinfection with SARS-CoV-2 or severe COVID-19. Front Public Health 2023; 11:1290402. [PMID: 38222091 PMCID: PMC10788057 DOI: 10.3389/fpubh.2023.1290402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 11/30/2023] [Indexed: 01/16/2024] Open
Abstract
Much of the global population now has some level of adaptive immunity to SARS-CoV-2 induced by exposure to the virus (natural infection), vaccination, or a combination of both (hybrid immunity). Key questions that subsequently arise relate to the duration and the level of protection an individual might expect based on their infection and vaccination history. A multi-component composite correlate of risk (CoR) could inform individuals and stakeholders about protection and aid decision making. This perspective evaluates the various elements that need to be accommodated in the development of an antibody-based composite CoR for reinfection with SARS-CoV-2 or development of severe COVID-19, including variation in exposure dose, transmission route, viral genetic variation, patient factors, and vaccination status. We provide an overview of antibody dynamics to aid exploration of the specifics of SARS-CoV-2 antibody testing. We further discuss anti-SARS-CoV-2 immunoassays, sample matrices, testing formats, frequency of sampling and the optimal time point for such sampling. While the development of a composite CoR is challenging, we provide our recommendations for each of these key areas and highlight areas that require further work to be undertaken.
Collapse
Affiliation(s)
- Stefan Holdenrieder
- Institute of Laboratory Medicine, German Heart Centre Munich, Technical University Munich, Munich, Germany
| | | | - Jacques Izopet
- Laboratory of Virology, Toulouse University Hospital and INFINITY Toulouse Institute for Infections and Inflammatory Diseases, INSERM UMR 1291 CNRS UMR 5051, University Toulouse III, Toulouse, France
| | - Elitza S. Theel
- Division of Clinical Microbiology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States
| | - Andreas Wieser
- Division of Infectious Diseases and Tropical Medicine, University Hospital, LMU Munich, Munich, Germany
- German Centre for Infection Research (DZIF), Munich, Germany
- Faculty of Medicine, Max Von Pettenkofer Institute, LMU Munich, Munich, Germany
- Immunology, Infection and Pandemic Research, Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Munich, Germany
| |
Collapse
|
44
|
Bakari M, Aboud S, Kasubi M, Mmbando BP, Ntinginya NE, Sichalwe A, Ubuguyu OS, Magesa A, Rutananukwa NL, Nyawale H, Kisinda A, Beyanga M, Horumpende PG, Mhame PS, Vumilia LM, Mziray LS, Mkala R, Shao E, Makubi A, Mshana SE, Kishimba R. Humoral Immune Responses following COVID-19 Vaccinations among Adults in Tanzania. Vaccines (Basel) 2023; 12:22. [PMID: 38250835 PMCID: PMC10819524 DOI: 10.3390/vaccines12010022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 12/07/2023] [Accepted: 12/21/2023] [Indexed: 01/23/2024] Open
Abstract
COVID-19 vaccination remains to be the most important intervention in the fight against the pandemic. The immunity among the vaccinated population and its durability can significantly vary due to various factors. This study investigated the humoral immune responses among individuals who received any of the COVID-19 vaccines approved for use in Tanzania. A total of 1048 randomly selected adults who received COVID-19 vaccines at different time points were enrolled and humoral immune responses (IR) were tested at baseline and three months later (960, 91.6%). The level of SARS-CoV-2 anti-spike/receptor binding domain (RBD) IgG, anti-nucleocapsid IgG, and IgM antibodies were determined using a commercially available chemiluminescent microparticle immunoassay. Descriptive data analysis was performed using STATA version 18 and R. At baseline, serum IgG against anti-spike/RBD was detected in 1010/1048 (96.4%) participants (95%CI: 94.9-97.5) and 98.3% (95%CI: 97.3-99) three months later. The IgG against the SARS-CoV-2 nucleocapsid proteins were detected in 40.8% and 45.3% of participants at baseline and follow-up, respectively. The proportion of seroconverters following vaccination and mean titers of anti-spike/RBD antibodies were significantly more among those who had past SARS-CoV-2 infection than in those with no evidence of past infection, (p < 0.001). Only 0.5% of those who had detectable anti-spike/RBD antibodies at baseline were negative after three months of follow-up and 1.5% had breakthrough infections. The majority of participants (99.5%) had detectable anti-spike/RBD antibodies beyond 6 months post-vaccination. The proportion of Tanzanians who mounted humoral IR following COVID-19 vaccination was very high. Seroconversions, as well as the mean titers and durability of humoral IR, were significantly enhanced by exposure to natural SARS-CoV-2 infection. In view of the limited availability of COVID-19 vaccines as well as challenges to completing subsequent doses, booster doses could only be suggested to high-risk groups.
Collapse
Affiliation(s)
- Muhammad Bakari
- School of Medicine, Muhimbili University of Health and Allied Sciences (MUHAS), Dar es Salaam P.O. Box 65001, Tanzania; (M.B.); (S.A.)
| | - Said Aboud
- School of Medicine, Muhimbili University of Health and Allied Sciences (MUHAS), Dar es Salaam P.O. Box 65001, Tanzania; (M.B.); (S.A.)
- National Institute for Medical Research (NIMR), Dar es Salaam P.O. Box 9653, Tanzania; (B.P.M.); (N.E.N.); (N.L.R.); (A.K.)
| | - Mabula Kasubi
- Muhimbili National Hospital (MNH), Dar es Salaam P.O. Box 65000, Tanzania;
| | - Bruno P. Mmbando
- National Institute for Medical Research (NIMR), Dar es Salaam P.O. Box 9653, Tanzania; (B.P.M.); (N.E.N.); (N.L.R.); (A.K.)
| | - Nyanda Elias Ntinginya
- National Institute for Medical Research (NIMR), Dar es Salaam P.O. Box 9653, Tanzania; (B.P.M.); (N.E.N.); (N.L.R.); (A.K.)
| | - Aifello Sichalwe
- Ministry of Health (MoH), Dodoma P.O. Box 743, Tanzania; (A.S.); (O.S.U.); (A.M.); (M.B.); (P.G.H.); (P.S.M.); (L.M.V.); (L.S.M.); (A.M.); (R.K.)
| | - Omary S. Ubuguyu
- Ministry of Health (MoH), Dodoma P.O. Box 743, Tanzania; (A.S.); (O.S.U.); (A.M.); (M.B.); (P.G.H.); (P.S.M.); (L.M.V.); (L.S.M.); (A.M.); (R.K.)
| | - Alex Magesa
- Ministry of Health (MoH), Dodoma P.O. Box 743, Tanzania; (A.S.); (O.S.U.); (A.M.); (M.B.); (P.G.H.); (P.S.M.); (L.M.V.); (L.S.M.); (A.M.); (R.K.)
| | - Nancy Ladislaus Rutananukwa
- National Institute for Medical Research (NIMR), Dar es Salaam P.O. Box 9653, Tanzania; (B.P.M.); (N.E.N.); (N.L.R.); (A.K.)
| | - Helmut Nyawale
- Department of Microbiology and Immunology, Weill Bugando School of Medicine, Catholic University of Health and Allied Sciences (CUHAS), Mwanza P.O. Box 1464, Tanzania;
| | - Abisai Kisinda
- National Institute for Medical Research (NIMR), Dar es Salaam P.O. Box 9653, Tanzania; (B.P.M.); (N.E.N.); (N.L.R.); (A.K.)
| | - Medard Beyanga
- Ministry of Health (MoH), Dodoma P.O. Box 743, Tanzania; (A.S.); (O.S.U.); (A.M.); (M.B.); (P.G.H.); (P.S.M.); (L.M.V.); (L.S.M.); (A.M.); (R.K.)
| | - Pius G. Horumpende
- Ministry of Health (MoH), Dodoma P.O. Box 743, Tanzania; (A.S.); (O.S.U.); (A.M.); (M.B.); (P.G.H.); (P.S.M.); (L.M.V.); (L.S.M.); (A.M.); (R.K.)
| | - Paulo S. Mhame
- Ministry of Health (MoH), Dodoma P.O. Box 743, Tanzania; (A.S.); (O.S.U.); (A.M.); (M.B.); (P.G.H.); (P.S.M.); (L.M.V.); (L.S.M.); (A.M.); (R.K.)
| | - Liggle M. Vumilia
- Ministry of Health (MoH), Dodoma P.O. Box 743, Tanzania; (A.S.); (O.S.U.); (A.M.); (M.B.); (P.G.H.); (P.S.M.); (L.M.V.); (L.S.M.); (A.M.); (R.K.)
| | - Lucy S. Mziray
- Ministry of Health (MoH), Dodoma P.O. Box 743, Tanzania; (A.S.); (O.S.U.); (A.M.); (M.B.); (P.G.H.); (P.S.M.); (L.M.V.); (L.S.M.); (A.M.); (R.K.)
| | - Reuben Mkala
- Benjamin Mkapa Hospital (BMH), Dodoma P.O. Box 11088, Tanzania;
| | - Elichilia Shao
- Kilimanjaro Christian Medical Centre (KCMC), Moshi P.O. Box 3010, Tanzania;
- Faculty of Medicine, Department of Internal Medicine, Kilimanjaro Christian Medical University College (KCMUCo), Moshi P.O. Box 2240, Tanzania
| | - Abel Makubi
- Ministry of Health (MoH), Dodoma P.O. Box 743, Tanzania; (A.S.); (O.S.U.); (A.M.); (M.B.); (P.G.H.); (P.S.M.); (L.M.V.); (L.S.M.); (A.M.); (R.K.)
- Muhimbili Orthopaedics Institute (MOI), Dar es Salaam P.O. Box 65474, Tanzania
| | - Stephen E. Mshana
- Department of Microbiology and Immunology, Weill Bugando School of Medicine, Catholic University of Health and Allied Sciences (CUHAS), Mwanza P.O. Box 1464, Tanzania;
| | - Rogath Kishimba
- Ministry of Health (MoH), Dodoma P.O. Box 743, Tanzania; (A.S.); (O.S.U.); (A.M.); (M.B.); (P.G.H.); (P.S.M.); (L.M.V.); (L.S.M.); (A.M.); (R.K.)
| |
Collapse
|
45
|
de Almeida DV, Cezar PA, Fernandes TFB, Schwarz MGA, Mendonça-Lima L, Giacoia-Gripp CBW, Côrtes FH, Lindenmeyer Guimarães M, Pilotto JH, De Sá NBR, Cazote ADS, Gomes LR, Quintana MDSB, Ribeiro-Alves M, Coelho LE, Geraldo KM, Ribeiro MPD, Cardoso SW, Grinsztejn B, Veloso VG, Morgado MG. The impact of early anti-SARS-CoV-2 antibody production on the length of hospitalization stay among COVID-19 patients. Microbiol Spectr 2023; 11:e0095923. [PMID: 37811977 PMCID: PMC10715214 DOI: 10.1128/spectrum.00959-23] [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: 03/03/2023] [Accepted: 08/23/2023] [Indexed: 10/10/2023] Open
Abstract
IMPORTANCE The study provides valuable insights into the sociodemographic characteristics, clinical outcomes, and humoral immune response of those affected by the virus that has devastated every field of human life since 2019; the COVID-19 patients. Firstly, the association among clinical manifestations, comorbidities, and the production of neutralizing antibodies (Nabs) against SARS-CoV-2 is explored. Secondly, varying levels of Nabs among patients are revealed, and a significant correlation between the presence of Nabs and a shorter duration of hospitalization is identified, which highlights the potential role of Nabs in predicting clinical outcomes. Lastly, a follow-up conducted 7 months later demonstrates the progression and persistence of Nabs production in recovered unvaccinated individuals. The study contributes essential knowledge regarding the characteristics of the study population, the early humoral immune response, and the dynamics of Nabs production over time. These findings have significant implications for understanding the immune response to COVID-19 and informing clinical management approaches.
Collapse
Affiliation(s)
- Dalziza Victalina de Almeida
- Laboratório de Aids e Imunologia Molecular, Instituto Oswaldo Cruz, FUNDAÇÃO OSWALDO CRUZ, Rio de Janeiro, Brazil
| | - Priscila Alves Cezar
- Laboratório de Aids e Imunologia Molecular, Instituto Oswaldo Cruz, FUNDAÇÃO OSWALDO CRUZ, Rio de Janeiro, Brazil
| | | | - Marcos Gustavo Araujo Schwarz
- Laboratório de Genômica Funcional e Bioinformática, Instituto Oswaldo Cruz, FUNDAÇÃO OSWALDO CRUZ, Rio de Janeiro, Brazil
| | - Leila Mendonça-Lima
- Laboratório de Genômica Funcional e Bioinformática, Instituto Oswaldo Cruz, FUNDAÇÃO OSWALDO CRUZ, Rio de Janeiro, Brazil
| | | | - Fernanda Heloise Côrtes
- Laboratório de Aids e Imunologia Molecular, Instituto Oswaldo Cruz, FUNDAÇÃO OSWALDO CRUZ, Rio de Janeiro, Brazil
| | - Monick Lindenmeyer Guimarães
- Laboratório de Aids e Imunologia Molecular, Instituto Oswaldo Cruz, FUNDAÇÃO OSWALDO CRUZ, Rio de Janeiro, Brazil
| | - Jose Henrique Pilotto
- Laboratório de Aids e Imunologia Molecular, Instituto Oswaldo Cruz, FUNDAÇÃO OSWALDO CRUZ, Rio de Janeiro, Brazil
| | - Nathalia Beatriz Ramos De Sá
- Laboratório de Aids e Imunologia Molecular, Instituto Oswaldo Cruz, FUNDAÇÃO OSWALDO CRUZ, Rio de Janeiro, Brazil
| | - Andressa da Silva Cazote
- Laboratório de Aids e Imunologia Molecular, Instituto Oswaldo Cruz, FUNDAÇÃO OSWALDO CRUZ, Rio de Janeiro, Brazil
| | - Larissa Rodrigues Gomes
- Centro de Desenvolvimento Tecnológico em Saúde (CDTS)/Instituto Nacional de Ciência e Tecnologia de Inovação em Doenças Negligenciadas da População (INCT-IDPN), FUNDAÇÃO OSWALDO CRUZ, Rio de Janeiro, Brazil
| | | | - Marcelo Ribeiro-Alves
- Instituto Nacional de Infectologia Evandro Chagas, FUNDAÇÃO OSWALDO CRUZ, Rio de Janeiro, Brazil
| | - Lara Esteves Coelho
- Instituto Nacional de Infectologia Evandro Chagas, FUNDAÇÃO OSWALDO CRUZ, Rio de Janeiro, Brazil
| | - Kim Mattos Geraldo
- Instituto Nacional de Infectologia Evandro Chagas, FUNDAÇÃO OSWALDO CRUZ, Rio de Janeiro, Brazil
| | - Maria Pia Diniz Ribeiro
- Instituto Nacional de Infectologia Evandro Chagas, FUNDAÇÃO OSWALDO CRUZ, Rio de Janeiro, Brazil
| | - Sandra Wagner Cardoso
- Instituto Nacional de Infectologia Evandro Chagas, FUNDAÇÃO OSWALDO CRUZ, Rio de Janeiro, Brazil
| | - Beatriz Grinsztejn
- Instituto Nacional de Infectologia Evandro Chagas, FUNDAÇÃO OSWALDO CRUZ, Rio de Janeiro, Brazil
| | - Valdiléa G Veloso
- Instituto Nacional de Infectologia Evandro Chagas, FUNDAÇÃO OSWALDO CRUZ, Rio de Janeiro, Brazil
| | - Mariza Gonçalves Morgado
- Laboratório de Aids e Imunologia Molecular, Instituto Oswaldo Cruz, FUNDAÇÃO OSWALDO CRUZ, Rio de Janeiro, Brazil
| |
Collapse
|
46
|
Wu J, Yang H, Yu D, Yang X. Blood-derived product therapies for SARS-CoV-2 infection and long COVID. MedComm (Beijing) 2023; 4:e426. [PMID: 38020714 PMCID: PMC10651828 DOI: 10.1002/mco2.426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 10/15/2023] [Accepted: 10/25/2023] [Indexed: 12/01/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is capable of large-scale transmission and has caused the coronavirus disease 2019 (COVID-19) pandemic. Patients with COVID-19 may experience persistent long-term health issues, known as long COVID. Both acute SARS-CoV-2 infection and long COVID have resulted in persistent negative impacts on global public health. The effective application and development of blood-derived products are important strategies to combat the serious damage caused by COVID-19. Since the emergence of COVID-19, various blood-derived products that target or do not target SARS-CoV-2 have been investigated for therapeutic applications. SARS-CoV-2-targeting blood-derived products, including COVID-19 convalescent plasma, COVID-19 hyperimmune globulin, and recombinant anti-SARS-CoV-2 neutralizing immunoglobulin G, are virus-targeting and can provide immediate control of viral infection in the short term. Non-SARS-CoV-2-targeting blood-derived products, including intravenous immunoglobulin and human serum albumin exhibit anti-inflammatory, immunomodulatory, antioxidant, and anticoagulatory properties. Rational use of these products can be beneficial to patients with SARS-CoV-2 infection or long COVID. With evidence accumulated since the pandemic began, we here summarize the progress of blood-derived product therapies for COVID-19, discuss the effective methods and scenarios regarding these therapies, and provide guidance and suggestions for clinical treatment.
Collapse
Affiliation(s)
- Junzheng Wu
- Chengdu Rongsheng Pharmaceuticals Co., Ltd.ChengduChina
| | | | - Ding Yu
- Chengdu Rongsheng Pharmaceuticals Co., Ltd.ChengduChina
- Beijing Tiantan Biological Products Co., Ltd.BeijingChina
| | | |
Collapse
|
47
|
Huang PC, Lin TY, Chen CC, Wang SW, Tsai BY, Tsai PJ, Tu YF, Ko WC, Cheng CM, Shieh CC, Liu CC, Shen CF. Age and prior vaccination determine the antibody level in children with primary SARS-CoV-2 Omicron infection. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2023; 56:1187-1197. [PMID: 37739902 DOI: 10.1016/j.jmii.2023.08.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 08/09/2023] [Accepted: 08/27/2023] [Indexed: 09/24/2023]
Abstract
BACKGROUND Protection against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) reinfection relies on immunity generated after primary infection. However, humoral immunity following primary infection with the Omicron variant is not well understood. METHODS We prospectively recruited children <19 years with virologically-confirmed SARS-CoV-2 infection at National Cheng Kung University Hospital from February 2022 to September 2022 during the first wave of Omicron BA.2 outbreak in Taiwan. Serum samples were collected one month after acute infection to measure anti-spike protein receptor binding domain antibody levels and surrogate virus neutralizing antibody (NAb) levels against wild type disease and variants. RESULTS Of the 164 patients enrolled, most were under 5 years (65.2%) with a diagnosis of upper respiratory tract infection. Children under 6 months with maternal coronavirus disease 2019 (COVID-19) vaccination had higher levels of both anti-SARS-CoV-2 spike antibody (119.0 vs 27.4 U/ml, p < 0.05) and anti-wild type NAb (56.9% vs 27.6% inhibition, p = 0.001) than those without. Children aged 5-12 years with prior vaccination had higher anti-spike antibody, anti-wild type, and anti-Omicron BA.2 NAb levels than those without (all p < 0.05). In previously naïve children without maternal or self-vaccination, those 6 months to 2 years had the highest antibody levels. Multivariable linear regression analysis showed age was the only independent factor associated with antibody level. CONCLUSIONS In our study, children aged 6 months to 2 years have the highest antibody responses to SARS-CoV-2 Omicron variant infection. Age and prior vaccination are the main factors influencing the immunogenicity of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Pin-Chen Huang
- Department of Pediatrics, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan, ROC
| | - Ting-Yu Lin
- Department of Pediatrics, Kuo General Hospital, Tainan, Taiwan
| | - Chih-Chia Chen
- Department of Pediatrics, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan, ROC; Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan, ROC
| | - Shih-Wei Wang
- Department of Pediatrics, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan, ROC
| | - Bo-Yang Tsai
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan, ROC
| | - Pei-Jane Tsai
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan, ROC; Center of Infectious Disease and Signaling Research, National Cheng Kung University, Tainan, 70101, Taiwan, ROC; Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan, ROC
| | - Yi-Fang Tu
- Department of Pediatrics, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan, ROC
| | - Wen-Chien Ko
- Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan, ROC
| | - Chao-Min Cheng
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan, ROC
| | - Chi-Chang Shieh
- Department of Pediatrics, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan, ROC; Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan, ROC
| | - Ching-Chuan Liu
- Department of Pediatrics, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan, ROC; Center of Infectious Disease and Signaling Research, National Cheng Kung University, Tainan, 70101, Taiwan, ROC
| | - Ching-Fen Shen
- Department of Pediatrics, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan, ROC; Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan, ROC.
| |
Collapse
|
48
|
Mazzaracchio V, Rios Maciel M, Porto Santos T, Toda-Peters K, Shen AQ. Duplex Electrochemical Microfluidic Sensor for COVID-19 Antibody Detection: Natural versus Vaccine-Induced Humoral Response. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2207731. [PMID: 36916701 DOI: 10.1002/smll.202207731] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 02/03/2023] [Indexed: 06/18/2023]
Abstract
The rapid transmission and resilience of coronavirus disease 2019 (COVID-19) have led to urgent demands in monitoring humoral response for effective vaccine development, thus a multiplex co-detection platform to discriminate infection-induced from vaccine-induced antibodies is needed. Here a duplex electrochemical immunosensor for co-detection of anti-nucleocapsid IgG (N-IgG) and anti-spike IgG (S-IgG) is developed by using a two-working electrode system, via an indirect immunoassay, with antibody quantification obtained by differential pulse voltammetry. The screen-printed electrodes (SPEs) are modified by carbon black and electrodeposited gold nanoflowers for maximized surface areas, enabling the construction of an immunological chain for S-IgG and N-IgG electrochemical detection with enhanced performance. Using an optimized immunoassay protocol, a wide linear range between 30-750 and 20-1000 ng mL-1 , and a limit of detection of 28 and 15 ng mL-1 are achieved to detect N-IgG and S-IgG simultaneously in serum samples. This duplex immunosensor is then integrated in a microfluidic device to obtain significantly reduced detection time (≤ 7 min) while maintaining its analytical performance. The duplex microfluidic immunosensor can be easily expanded into multiplex format to achieve high throughput screening for the sero-surveillance of COVID-19 and other infectious diseases.
Collapse
Affiliation(s)
- Vincenzo Mazzaracchio
- Micro/Bio/Nanofluidics Unit, Okinawa Institute of Science and Technology Graduate University, Onna-son, Okinawa, 904-0495, Japan
- Department of Chemical Science and Technologies, University of Rome "Tor Vergata,", Via della Ricerca Scientifica, 00133, Rome, Italy
| | - Mauricio Rios Maciel
- Micro/Bio/Nanofluidics Unit, Okinawa Institute of Science and Technology Graduate University, Onna-son, Okinawa, 904-0495, Japan
| | - Tatiana Porto Santos
- Micro/Bio/Nanofluidics Unit, Okinawa Institute of Science and Technology Graduate University, Onna-son, Okinawa, 904-0495, Japan
| | - Kazumi Toda-Peters
- Micro/Bio/Nanofluidics Unit, Okinawa Institute of Science and Technology Graduate University, Onna-son, Okinawa, 904-0495, Japan
| | - Amy Q Shen
- Micro/Bio/Nanofluidics Unit, Okinawa Institute of Science and Technology Graduate University, Onna-son, Okinawa, 904-0495, Japan
| |
Collapse
|
49
|
O’Reilly S, Kenny G, Alrawahneh T, Francois N, Gu L, Angeliadis M, de Masson d’Autume V, Garcia Leon A, Feeney ER, Yousif O, Cotter A, de Barra E, Horgan M, Mallon PWG, Gautier V. Development of a novel medium throughput flow-cytometry based micro-neutralisation test for SARS-CoV-2 with applications in clinical vaccine trials and antibody screening. PLoS One 2023; 18:e0294262. [PMID: 38033116 PMCID: PMC10688860 DOI: 10.1371/journal.pone.0294262] [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: 11/16/2022] [Accepted: 10/27/2023] [Indexed: 12/02/2023] Open
Abstract
Quantifying neutralising capacity of circulating SARS-COV-2 antibodies is critical in evaluating protective humoral immune responses generated post-infection/post-vaccination. Here we describe a novel medium-throughput flow cytometry-based micro-neutralisation test to evaluate Neutralising Antibody (NAb) responses against live SARS-CoV-2 Wild Type and Variants of Concern (VOC) in convalescent/vaccinated populations. Flow Cytometry-Based Micro-Neutralisation Test (Micro-NT) was performed in 96-well plates using clinical isolates WT-B, WT-B.1.177.18 and/or VOCs Beta and Omicron. Plasma samples (All Ireland Infectious Diseases (AIID) Cohort) were serially diluted (8 points, half-log) from 1:20 and pre-incubated with SARS-CoV-2 (1h, 37°C). Virus-plasma mixture were added onto Vero E6 or Vero E6/TMPRSS2 cells for 18h. Percentage infected cells was analysed by automated flow cytometry following trypsinisation, fixation and SARS-CoV-2 Nucleoprotein intracellular staining. Half-maximal Neutralisation Titres (NT50) were determined using non-linear regression. Our assay was compared to Plaque Reduction Neutralisation Test (PRNT) and validated against the First WHO International Standard for anti-SARS-CoV-2 immunoglobulin. Both Micro-NT and PRNT achieved comparable NT50 values. Further validation showed adequate correlation with PRNT using a panel of secondary standards of clinical convalescent and vaccinated plasma samples. We found the assay to be reproducible through measuring both repeatability and intermediate precision. Screening 190 convalescent samples and 11 COVID-19 naive controls (AIID cohort) we demonstrated that Micro-NT has broad dynamic range differentiating NT50s <1/20 to >1/5000. We could also characterise immune-escape VOC Beta and Omicron BA.5, achieving fold-reductions in neutralising capacity similar to those published. Our flow cytometry-based Micro-NT is a robust and reliable assay to quantify NAb titres, and has been selected as an endpoint in clinical trials.
Collapse
Affiliation(s)
- Sophie O’Reilly
- Centre for Experimental Pathogen Host Research (CEPHR), University College Dublin, Belfield, Dublin, Ireland
- School of Medicine, University College Dublin, Belfield, Dublin, Ireland
| | - Grace Kenny
- Centre for Experimental Pathogen Host Research (CEPHR), University College Dublin, Belfield, Dublin, Ireland
- School of Medicine, University College Dublin, Belfield, Dublin, Ireland
- Department of Infectious Diseases, St Vincent’s University Hospital, Elm Park, Dublin, Ireland
| | - Tamara Alrawahneh
- Centre for Experimental Pathogen Host Research (CEPHR), University College Dublin, Belfield, Dublin, Ireland
- School of Medicine, University College Dublin, Belfield, Dublin, Ireland
| | - Nathan Francois
- Centre for Experimental Pathogen Host Research (CEPHR), University College Dublin, Belfield, Dublin, Ireland
- School of Medicine, University College Dublin, Belfield, Dublin, Ireland
| | - Lili Gu
- Centre for Experimental Pathogen Host Research (CEPHR), University College Dublin, Belfield, Dublin, Ireland
- School of Medicine, University College Dublin, Belfield, Dublin, Ireland
| | - Matthew Angeliadis
- Centre for Experimental Pathogen Host Research (CEPHR), University College Dublin, Belfield, Dublin, Ireland
- School of Medicine, University College Dublin, Belfield, Dublin, Ireland
| | - Valentin de Masson d’Autume
- Centre for Experimental Pathogen Host Research (CEPHR), University College Dublin, Belfield, Dublin, Ireland
- School of Medicine, University College Dublin, Belfield, Dublin, Ireland
| | - Alejandro Garcia Leon
- Centre for Experimental Pathogen Host Research (CEPHR), University College Dublin, Belfield, Dublin, Ireland
- School of Medicine, University College Dublin, Belfield, Dublin, Ireland
| | - Eoin R. Feeney
- Centre for Experimental Pathogen Host Research (CEPHR), University College Dublin, Belfield, Dublin, Ireland
- School of Medicine, University College Dublin, Belfield, Dublin, Ireland
- Department of Infectious Diseases, St Vincent’s University Hospital, Elm Park, Dublin, Ireland
| | - Obada Yousif
- Endocrinology Department, Wexford General Hospital, Carricklawn, Wexford, Ireland
| | - Aoife Cotter
- Centre for Experimental Pathogen Host Research (CEPHR), University College Dublin, Belfield, Dublin, Ireland
- School of Medicine, University College Dublin, Belfield, Dublin, Ireland
- Department of Infectious Diseases, Mater Misericordiae University Hospital, Eccles St, Dublin, Ireland
| | - Eoghan de Barra
- Department of Infectious Diseases, Beaumont Hospital, Beaumont, Dublin, Ireland
- Department of International Health and Tropical Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Mary Horgan
- Department of Infectious Diseases, Cork University Hospital, Wilton, Cork, Ireland
| | - Patrick W. G. Mallon
- Centre for Experimental Pathogen Host Research (CEPHR), University College Dublin, Belfield, Dublin, Ireland
- School of Medicine, University College Dublin, Belfield, Dublin, Ireland
- Department of Infectious Diseases, St Vincent’s University Hospital, Elm Park, Dublin, Ireland
| | - Virginie Gautier
- Centre for Experimental Pathogen Host Research (CEPHR), University College Dublin, Belfield, Dublin, Ireland
- School of Medicine, University College Dublin, Belfield, Dublin, Ireland
- Conway Institute of Biomedical and Biomolecular Research, University College Dublin, Belfield, Dublin 4, Ireland
| |
Collapse
|
50
|
Kenny G, O'Reilly S, Wrigley Kelly N, Negi R, Gaillard C, Alalwan D, Saini G, Alrawahneh T, Francois N, Angeliadis M, Garcia Leon AA, Tinago W, Feeney ER, Cotter AG, de Barra E, Yousif O, Horgan M, Doran P, Stemler J, Koehler P, Cox RJ, O'Shea D, Olesen OF, Landay A, Hogan AE, Lelievre JD, Gautier V, Cornely OA, Mallon PWG. Distinct receptor binding domain IgG thresholds predict protective host immunity across SARS-CoV-2 variants and time. Nat Commun 2023; 14:7015. [PMID: 37919289 PMCID: PMC10622572 DOI: 10.1038/s41467-023-42717-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 10/19/2023] [Indexed: 11/04/2023] Open
Abstract
SARS-CoV-2 neutralising antibodies provide protection against COVID-19. Evidence from early vaccine trials suggested binding antibody thresholds could serve as surrogate markers of neutralising capacity, but whether these thresholds predict sufficient neutralising capacity against variants of concern (VOCs), and whether this is impacted by vaccine or infection history remains unclear. Here we analyse individuals recovered from, vaccinated or with hybrid immunity against SARS-CoV-2. An NT50 ≥ 100 IU confers protection in vaccine trials, however, as VOC induce a reduction in NT50, we use NT50 ≥ 1000 IU as a cut off for WT NT50 that would retain neutralisation against VOC. In unvaccinated convalescent participants, a receptor binding domain (RBD) IgG of 456 BAU/mL predicts an NT50 against WT of 1000 IU with an accuracy of 80% (95%CI 73-86%). This threshold maintains accuracy in determining loss of protective immunity against VOC in two vaccinated cohorts. It predicts an NT50 < 100 IU against Beta with an accuracy of 80% (95%CI 67-89%) in 2 vaccine dose recipients. In booster vaccine recipients with a history of COVID-19 (hybrid immunity), accuracy is 87% (95%CI 77-94%) in determining an NT50 of <100 IU against BA.5. This analysis provides a discrete threshold that could be used in future clinical studies.
Collapse
Affiliation(s)
- Grace Kenny
- Centre for Experimental Pathogen Host Research (CEPHR), University College Dublin, Belfield, Dublin 4, Ireland.
| | - Sophie O'Reilly
- Centre for Experimental Pathogen Host Research (CEPHR), University College Dublin, Belfield, Dublin 4, Ireland
| | - Neil Wrigley Kelly
- St Vincent's University Hospital, Elm Park, Dublin 4, Ireland
- Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Co Kildare, Ireland
| | - Riya Negi
- Centre for Experimental Pathogen Host Research (CEPHR), University College Dublin, Belfield, Dublin 4, Ireland
| | - Colette Gaillard
- Centre for Experimental Pathogen Host Research (CEPHR), University College Dublin, Belfield, Dublin 4, Ireland
| | - Dana Alalwan
- Centre for Experimental Pathogen Host Research (CEPHR), University College Dublin, Belfield, Dublin 4, Ireland
| | - Gurvin Saini
- Centre for Experimental Pathogen Host Research (CEPHR), University College Dublin, Belfield, Dublin 4, Ireland
| | - Tamara Alrawahneh
- Centre for Experimental Pathogen Host Research (CEPHR), University College Dublin, Belfield, Dublin 4, Ireland
| | - Nathan Francois
- Centre for Experimental Pathogen Host Research (CEPHR), University College Dublin, Belfield, Dublin 4, Ireland
| | - Matthew Angeliadis
- Centre for Experimental Pathogen Host Research (CEPHR), University College Dublin, Belfield, Dublin 4, Ireland
| | - Alejandro Abner Garcia Leon
- Centre for Experimental Pathogen Host Research (CEPHR), University College Dublin, Belfield, Dublin 4, Ireland
| | - Willard Tinago
- Centre for Experimental Pathogen Host Research (CEPHR), University College Dublin, Belfield, Dublin 4, Ireland
| | - Eoin R Feeney
- Centre for Experimental Pathogen Host Research (CEPHR), University College Dublin, Belfield, Dublin 4, Ireland
- St Vincent's University Hospital, Elm Park, Dublin 4, Ireland
| | - Aoife G Cotter
- Centre for Experimental Pathogen Host Research (CEPHR), University College Dublin, Belfield, Dublin 4, Ireland
- Department of Infectious Diseases, Mater Misericordiae University Hospital, Eccles St, Dublin 7, Ireland
| | - Eoghan de Barra
- Department of Infectious Diseases, Beaumont Hospital, Beaumont, Dublin 9, Ireland
- Department of International Health and Tropical Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Obada Yousif
- Endocrinology Department, Wexford General Hospital, Carricklawn, Wexford, Ireland
| | - Mary Horgan
- Department of Infectious Diseases, Cork University Hospital, Wilton, Co Cork, Ireland
| | - Peter Doran
- School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| | - Jannik Stemler
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Internal Medicine and University of Cologne, Faculty of Medicine Institute of Translational Research, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany
- German Centre for Infection Research (DZIF), Partner Site Bonn-Cologne Department Cologne, Cologne, Germany
| | - Philipp Koehler
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Internal Medicine and University of Cologne, Faculty of Medicine Institute of Translational Research, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany
- German Centre for Infection Research (DZIF), Partner Site Bonn-Cologne Department Cologne, Cologne, Germany
| | - Rebecca Jane Cox
- Influenza Centre, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Donal O'Shea
- St Vincent's University Hospital, Elm Park, Dublin 4, Ireland
| | - Ole F Olesen
- European Vaccine Initiative, Heidelberg, Germany
| | - Alan Landay
- Department of internal Medicine, Rush University, Chicago, IL, USA
| | - Andrew E Hogan
- Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Co Kildare, Ireland
- National Children's Research Centre, Dublin 12, Ireland
| | | | - Virginie Gautier
- Centre for Experimental Pathogen Host Research (CEPHR), University College Dublin, Belfield, Dublin 4, Ireland
| | - Oliver A Cornely
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Internal Medicine and University of Cologne, Faculty of Medicine Institute of Translational Research, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany
- German Centre for Infection Research (DZIF), Partner Site Bonn-Cologne Department Cologne, Cologne, Germany
| | - Patrick W G Mallon
- Centre for Experimental Pathogen Host Research (CEPHR), University College Dublin, Belfield, Dublin 4, Ireland
- St Vincent's University Hospital, Elm Park, Dublin 4, Ireland
| |
Collapse
|