1
|
Anthi AK, Kolderup A, Vaage EB, Bern M, Benjakul S, Tjärnhage E, Ruso-Julve F, Jensen KR, Lode HE, Vaysburd M, Nilsen J, Herigstad ML, Sakya SA, Tietze L, Pilati D, Nyquist-Andersen M, Dürkoop M, Gjølberg TT, Peng L, Foss S, Moe MC, Low BE, Wiles MV, Nemazee D, Jahnsen FL, Vaage JT, Howard KA, Sandlie I, James LC, Grødeland G, Lund-Johansen F, Andersen JT. An intranasal subunit vaccine induces protective systemic and mucosal antibody immunity against respiratory viruses in mouse models. Nat Commun 2025; 16:3999. [PMID: 40312392 PMCID: PMC12045997 DOI: 10.1038/s41467-025-59353-6] [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: 01/23/2024] [Accepted: 04/21/2025] [Indexed: 05/03/2025] Open
Abstract
Although vaccines are usually given intramuscularly, the intranasal delivery route may lead to better mucosal protection and limit the spread of respiratory virus while easing administration and improving vaccine acceptance. The challenge, however, is to achieve delivery across the selective epithelial cell barrier. Here we report on a subunit vaccine platform, in which the antigen is genetically fused to albumin to facilitate FcRn-mediated transport across the mucosal barrier in the presence of adjuvant. Intranasal delivery in conventional and transgenic mouse models induces both systemic and mucosal antigen-specific antibody responses that protect against challenge with SARS-CoV-2 or influenza A. When benchmarked against an intramuscularly administered mRNA vaccine or an intranasally administered antigen fused to an alternative carrier of similar size, only the albumin-based intranasal vaccine yields robust mucosal IgA antibody responses. Our results thus suggest that this needle-free, albumin-based vaccine platform may be suited for vaccination against respiratory pathogens.
Collapse
MESH Headings
- Animals
- Administration, Intranasal
- Mice
- Immunity, Mucosal/immunology
- Antibodies, Viral/immunology
- Antibodies, Viral/blood
- SARS-CoV-2/immunology
- Vaccines, Subunit/immunology
- Vaccines, Subunit/administration & dosage
- COVID-19/prevention & control
- COVID-19/immunology
- Female
- COVID-19 Vaccines/immunology
- COVID-19 Vaccines/administration & dosage
- Humans
- Influenza A virus/immunology
- Disease Models, Animal
- Influenza Vaccines/immunology
- Influenza Vaccines/administration & dosage
- Immunoglobulin A/immunology
- Receptors, Fc/genetics
- Receptors, Fc/immunology
- Mice, Transgenic
- Orthomyxoviridae Infections/prevention & control
- Orthomyxoviridae Infections/immunology
- Mice, Inbred C57BL
- Albumins/immunology
- mRNA Vaccines/immunology
- mRNA Vaccines/administration & dosage
- Histocompatibility Antigens Class I
Collapse
Affiliation(s)
- Aina Karen Anthi
- Department of Immunology, Oslo University Hospital Rikshospitalet, 0372, Oslo, Norway
- Institute of Clinical Medicine and Department of Pharmacology, University of Oslo and Oslo University Hospital Rikshospitalet, 0372, Oslo, Norway
- Precision Immunotherapy Alliance (PRIMA), University of Oslo, 0372, Oslo, Norway
| | - Anette Kolderup
- Department of Immunology, Oslo University Hospital Rikshospitalet, 0372, Oslo, Norway
- Institute of Clinical Medicine and Department of Pharmacology, University of Oslo and Oslo University Hospital Rikshospitalet, 0372, Oslo, Norway
- Precision Immunotherapy Alliance (PRIMA), University of Oslo, 0372, Oslo, Norway
| | - Eline Benno Vaage
- Department of Immunology, Oslo University Hospital Rikshospitalet, 0372, Oslo, Norway
- Precision Immunotherapy Alliance (PRIMA), University of Oslo, 0372, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, 0372, Oslo, Norway
| | - Malin Bern
- Department of Immunology, Oslo University Hospital Rikshospitalet, 0372, Oslo, Norway
- Institute of Clinical Medicine and Department of Pharmacology, University of Oslo and Oslo University Hospital Rikshospitalet, 0372, Oslo, Norway
| | - Sopisa Benjakul
- Department of Immunology, Oslo University Hospital Rikshospitalet, 0372, Oslo, Norway
- Institute of Clinical Medicine and Department of Pharmacology, University of Oslo and Oslo University Hospital Rikshospitalet, 0372, Oslo, Norway
- Precision Immunotherapy Alliance (PRIMA), University of Oslo, 0372, Oslo, Norway
| | - Elias Tjärnhage
- Department of Immunology, Oslo University Hospital Rikshospitalet, 0372, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, 0372, Oslo, Norway
| | - Fulgencio Ruso-Julve
- Department of Immunology, Oslo University Hospital Rikshospitalet, 0372, Oslo, Norway
- Institute of Clinical Medicine and Department of Pharmacology, University of Oslo and Oslo University Hospital Rikshospitalet, 0372, Oslo, Norway
- Precision Immunotherapy Alliance (PRIMA), University of Oslo, 0372, Oslo, Norway
| | - Kjell-Rune Jensen
- Department of Immunology, Oslo University Hospital Rikshospitalet, 0372, Oslo, Norway
- Institute of Clinical Medicine and Department of Pharmacology, University of Oslo and Oslo University Hospital Rikshospitalet, 0372, Oslo, Norway
- Precision Immunotherapy Alliance (PRIMA), University of Oslo, 0372, Oslo, Norway
| | - Heidrun Elisabeth Lode
- Department of Immunology, Oslo University Hospital Rikshospitalet, 0372, Oslo, Norway
- Institute of Clinical Medicine and Department of Pharmacology, University of Oslo and Oslo University Hospital Rikshospitalet, 0372, Oslo, Norway
- Precision Immunotherapy Alliance (PRIMA), University of Oslo, 0372, Oslo, Norway
- Center of Eye Research, Department of Ophthalmology, Oslo University Hospital Ullevål and University of Oslo, 0450, Oslo, Norway
| | - Marina Vaysburd
- Protein and Nucleic Acid Chemistry Division, Medical Research Council, Laboratory of Molecular Biology, Cambridge, CB2 0QH, United Kingdom
| | - Jeannette Nilsen
- Department of Immunology, Oslo University Hospital Rikshospitalet, 0372, Oslo, Norway
- Institute of Clinical Medicine and Department of Pharmacology, University of Oslo and Oslo University Hospital Rikshospitalet, 0372, Oslo, Norway
- Precision Immunotherapy Alliance (PRIMA), University of Oslo, 0372, Oslo, Norway
| | - Marie Leangen Herigstad
- Department of Immunology, Oslo University Hospital Rikshospitalet, 0372, Oslo, Norway
- Institute of Clinical Medicine and Department of Pharmacology, University of Oslo and Oslo University Hospital Rikshospitalet, 0372, Oslo, Norway
- Precision Immunotherapy Alliance (PRIMA), University of Oslo, 0372, Oslo, Norway
| | - Siri Aastedatter Sakya
- Department of Immunology, Oslo University Hospital Rikshospitalet, 0372, Oslo, Norway
- Institute of Clinical Medicine and Department of Pharmacology, University of Oslo and Oslo University Hospital Rikshospitalet, 0372, Oslo, Norway
- Precision Immunotherapy Alliance (PRIMA), University of Oslo, 0372, Oslo, Norway
| | - Lisa Tietze
- Department of Immunology, Oslo University Hospital Rikshospitalet, 0372, Oslo, Norway
- Precision Immunotherapy Alliance (PRIMA), University of Oslo, 0372, Oslo, Norway
| | - Diego Pilati
- Interdisciplinary Nanoscience Center (iNANO), Department of Molecular Biology and Genetics, Aarhus University, DK-8000, Aarhus C, Denmark
| | - Mari Nyquist-Andersen
- Department of Immunology, Oslo University Hospital Rikshospitalet, 0372, Oslo, Norway
- Institute of Clinical Medicine and Department of Pharmacology, University of Oslo and Oslo University Hospital Rikshospitalet, 0372, Oslo, Norway
- Precision Immunotherapy Alliance (PRIMA), University of Oslo, 0372, Oslo, Norway
| | - Mirjam Dürkoop
- Department of Immunology, Oslo University Hospital Rikshospitalet, 0372, Oslo, Norway
- Institute of Clinical Medicine and Department of Pharmacology, University of Oslo and Oslo University Hospital Rikshospitalet, 0372, Oslo, Norway
- Precision Immunotherapy Alliance (PRIMA), University of Oslo, 0372, Oslo, Norway
| | - Torleif Tollefsrud Gjølberg
- Department of Immunology, Oslo University Hospital Rikshospitalet, 0372, Oslo, Norway
- Institute of Clinical Medicine and Department of Pharmacology, University of Oslo and Oslo University Hospital Rikshospitalet, 0372, Oslo, Norway
- Precision Immunotherapy Alliance (PRIMA), University of Oslo, 0372, Oslo, Norway
- Center of Eye Research, Department of Ophthalmology, Oslo University Hospital Ullevål and University of Oslo, 0450, Oslo, Norway
| | - Linghang Peng
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Stian Foss
- Department of Immunology, Oslo University Hospital Rikshospitalet, 0372, Oslo, Norway
- Institute of Clinical Medicine and Department of Pharmacology, University of Oslo and Oslo University Hospital Rikshospitalet, 0372, Oslo, Norway
- Precision Immunotherapy Alliance (PRIMA), University of Oslo, 0372, Oslo, Norway
| | - Morten C Moe
- Center of Eye Research, Department of Ophthalmology, Oslo University Hospital Ullevål and University of Oslo, 0450, Oslo, Norway
| | | | | | - David Nemazee
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Frode L Jahnsen
- Institute of Clinical Medicine, University of Oslo, 0372, Oslo, Norway
- Department of Pathology, Oslo University Hospital Rikshospitalet, 0372, Oslo, Norway
| | - John Torgils Vaage
- Department of Immunology, Oslo University Hospital Rikshospitalet, 0372, Oslo, Norway
| | - Kenneth A Howard
- Interdisciplinary Nanoscience Center (iNANO), Department of Molecular Biology and Genetics, Aarhus University, DK-8000, Aarhus C, Denmark
| | - Inger Sandlie
- Department of Biosciences, University of Oslo, 0371, Oslo, Norway
| | - Leo C James
- Protein and Nucleic Acid Chemistry Division, Medical Research Council, Laboratory of Molecular Biology, Cambridge, CB2 0QH, United Kingdom
| | - Gunnveig Grødeland
- Department of Immunology, Oslo University Hospital Rikshospitalet, 0372, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, 0372, Oslo, Norway
| | - Fridtjof Lund-Johansen
- Department of Immunology, Oslo University Hospital Rikshospitalet, 0372, Oslo, Norway
- Precision Immunotherapy Alliance (PRIMA), University of Oslo, 0372, Oslo, Norway
| | - Jan Terje Andersen
- Department of Immunology, Oslo University Hospital Rikshospitalet, 0372, Oslo, Norway.
- Institute of Clinical Medicine and Department of Pharmacology, University of Oslo and Oslo University Hospital Rikshospitalet, 0372, Oslo, Norway.
- Precision Immunotherapy Alliance (PRIMA), University of Oslo, 0372, Oslo, Norway.
| |
Collapse
|
2
|
Sasset L, Angioni R, Presa N, Sánchez-Rodríguez R, Cozzolino C, Bertoldi N, Marinello S, Loy M, Mazzitelli M, Rea F, Cattelan A, Molon B. Immune Modulation and Efficacy of Tixagevimab/Cilgavimab Pre-Exposure Prophylaxis in Lung Transplant Recipients During the Omicron Wave. Int J Mol Sci 2025; 26:3696. [PMID: 40332340 PMCID: PMC12027920 DOI: 10.3390/ijms26083696] [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: 01/30/2025] [Revised: 04/04/2025] [Accepted: 04/08/2025] [Indexed: 05/08/2025] Open
Abstract
Lung transplant recipients are at increased risk of severe COVID-19 due to lifelong immunosuppressive therapy, which impairs both innate and adaptive immune responses. Identifying effective supportive therapies is essential for mitigating the heightened vulnerability of this population. This study investigated the effects of tixagevimab/cilgavimab, a monoclonal antibody therapy, as pre-exposure prophylaxis (PrEP) in this population. A prospective study was conducted on 19 lung transplant recipients at Padua University Hospital, Italy, during the Omicron variant wave (May-June 2022). Participants received tixagevimab/cilgavimab intramuscularly and were monitored for 180 days. SARS-CoV-2-specific antibody levels were measured at baseline (T0), one month (T1), and three months (T3) post-treatment. Cytokine profiles and clinical outcomes, including SARS-CoV-2 infections, were also assessed. At baseline, 50% of patients had negative antibody responses, but one-month post-treatment, all patients exceeded 700 kBAU/mL (median 3870 kBAU/mL), with levels decreasing but remaining positive at three months (median 1670 kBAU/mL). Remarkably, a higher level of circulating IL-18 was found at T3 in comparison to T0 in patients who did not experience COVID-19 after PrEP. This finding aligns with IL-18's primary role in stimulating type-1 T helper (Th1) cell responses, necessary for the induction of virus-specific cytotoxic T lymphocytes (CTLs). These results suggest that tixagevimab/cilgavimab may induce a systemic immune signature that could contribute to priming the immune response against SARS-CoV-2, potentially mediated by interactions with immune cell subsets.
Collapse
Affiliation(s)
- Lolita Sasset
- Infectious and Tropical Diseases Unit, University Hospital of Padua, 35128 Padua, Italy; (L.S.); (A.C.)
| | - Roberta Angioni
- Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy; (R.S.-R.); (B.M.)
- Fondazione Istituto di Ricerca Pediatrica-Città della Speranza, 35127 Padua, Italy
| | - Nicolò Presa
- Infectious and Tropical Diseases Unit, University Hospital of Padua, 35128 Padua, Italy; (L.S.); (A.C.)
| | - Ricardo Sánchez-Rodríguez
- Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy; (R.S.-R.); (B.M.)
- Fondazione Istituto di Ricerca Pediatrica-Città della Speranza, 35127 Padua, Italy
| | - Claudia Cozzolino
- Department of Cardiac, Thoracic, Vascular Sciences, and Public Health, University Hospital of Padua, 35128 Padua, Italy; (C.C.)
| | - Nicole Bertoldi
- Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy; (R.S.-R.); (B.M.)
- Fondazione Istituto di Ricerca Pediatrica-Città della Speranza, 35127 Padua, Italy
| | - Serena Marinello
- Infectious and Tropical Diseases Unit, University Hospital of Padua, 35128 Padua, Italy; (L.S.); (A.C.)
| | - Monica Loy
- Department of Cardiac, Thoracic, Vascular Sciences, and Public Health, University Hospital of Padua, 35128 Padua, Italy; (C.C.)
| | - Maria Mazzitelli
- Infectious and Tropical Diseases Unit, University Hospital of Padua, 35128 Padua, Italy; (L.S.); (A.C.)
| | - Federico Rea
- Department of Cardiac, Thoracic, Vascular Sciences, and Public Health, University Hospital of Padua, 35128 Padua, Italy; (C.C.)
| | - Annamaria Cattelan
- Infectious and Tropical Diseases Unit, University Hospital of Padua, 35128 Padua, Italy; (L.S.); (A.C.)
- Department of Molecular Medicine, University of Padua, 35131 Padua, Italy
| | - Barbara Molon
- Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy; (R.S.-R.); (B.M.)
- Fondazione Istituto di Ricerca Pediatrica-Città della Speranza, 35127 Padua, Italy
| |
Collapse
|
3
|
Olofsson A, Humbert M, Rekha RS, Frankling MH, Lund-Johansen F, Bergman P, Björkhem-Bergman L, Karlsson AC. Adaptive immune responses against common viruses are sustained and functional in end-of-life patients. iScience 2025; 28:112082. [PMID: 40124502 PMCID: PMC11930376 DOI: 10.1016/j.isci.2025.112082] [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: 08/14/2024] [Revised: 11/22/2024] [Accepted: 02/18/2025] [Indexed: 03/25/2025] Open
Abstract
Viral infections occur with increased frequency in patients in palliative care, impacting their quality of life and increasing mortality rates. Still, the function of the immune system has never been thoroughly studied at the end of life. We investigated virus-specific humoral and cellular immune responses in elderly end-of-life patients (n = 38) and controls (n = 28). Virus-specific T cell responses were characterized using high-parameter flow cytometry, after stimulation with cytomegalovirus (CMV) and human coronavirus OC43 peptides. Although some virus-specific T cells from patients exhibited elevated expression of costimulatory and coinhibitory molecules, their functional profile remained largely intact compared to controls. The expression of the cytotoxic markers Granzyme B, CD107a, and 2B4 on CMV-specific T cells correlated closely with survival time. Significantly, our data demonstrate that both humoral and cellular immunity remain responsive and functional against common viruses in end-of-life patients.
Collapse
Affiliation(s)
- Anna Olofsson
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Marion Humbert
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Rokeya S. Rekha
- Division of Clinical Immunology, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Maria Helde Frankling
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Theme Cancer, Karolinska University Hospital, Stockholm, Sweden
| | - Fridtjof Lund-Johansen
- Institute of Clinical Medicine, University of Oslo, Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Department of Immunology, Oslo University Hospital, ImmunoLingo Convergence Center, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Peter Bergman
- Division of Clinical Immunology, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Linda Björkhem-Bergman
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Research and Development Unit/Palliative Care, Stockholms Sjukhem, Mariebergsgatan 22, Stockholm, Sweden
| | - Annika C. Karlsson
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
4
|
Alirezaylavasani A, Egner IM, Dahl B, Chopra A, de Matos Kasahara T, Goll GL, Jahnsen J, Grødeland G, Vaage JT, Lund-Johansen F, Holter JC, Halvorsen B, Jørgensen KK, Munthe LA, Kared H. Deficient SARS-CoV-2 hybrid immunity during inflammatory bowel disease. Clin Immunol 2025; 271:110404. [PMID: 39645159 DOI: 10.1016/j.clim.2024.110404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 10/28/2024] [Accepted: 11/19/2024] [Indexed: 12/09/2024]
Abstract
Patients with Inflammatory Bowel Disease (IBD) undergoing immunosuppressive therapies face heightened susceptibility to severe COVID-19. An in-depth understanding of systemic inflammation and cellular immune responses after SARS-CoV-2 vaccination and breakthrough infections (BTI) is required for optimizing vaccine strategies in this population. While the prevalence of high serological responders post- third COVID-19 vaccine dose was lower, and the antibody waning was higher in IBD patients than in healthy donors (HD), IBD patients showed an increase in anti-RBD Wild Type IgG levels and cross-reactive Spike -specific memory B cells following BTI. However, there was no significant enhancement in cellular immune responses against anti-SARS-CoV-2 post-BTI, with responses instead characterized by activation of SARS-CoV-2 specific and also bystander CD8 T cells. These results suggest a complex interaction between chronic inflammation in IBD and the generation of new immune responses, highlighting the need for tailored vaccine regimens and anti-inflammatory therapies to boost cellular immunity against SARS-CoV-2.
Collapse
Affiliation(s)
- Amin Alirezaylavasani
- Department of Immunology, Oslo University Hospital, Oslo, Norway; KG Jebsen Centre for B cell Malignancies, Institute of Clinical Medicine, University of Oslo, Norway; Precision Immunotherapy Alliance, University of Oslo, Oslo, Norway
| | - Ingrid Marie Egner
- Department of Immunology, Oslo University Hospital, Oslo, Norway; KG Jebsen Centre for B cell Malignancies, Institute of Clinical Medicine, University of Oslo, Norway; Precision Immunotherapy Alliance, University of Oslo, Oslo, Norway
| | - Børresdatter Dahl
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
| | - Adity Chopra
- Department of Immunology, Oslo University Hospital, Oslo, Norway; Precision Immunotherapy Alliance, University of Oslo, Oslo, Norway; ImmunoLingo Convergence Center, University of Oslo, Oslo, Norway
| | | | - Guro Løvik Goll
- Division of Rheumatology and Research, Diakonhjemmet Hospital, Oslo, Norway; Institute of Health and Society, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Jørgen Jahnsen
- Department of Gastroenterology, Akershus University Hospital, Lørenskog, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Gunnveig Grødeland
- Department of Immunology, Oslo University Hospital, Oslo, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - John Torgils Vaage
- Department of Immunology, Oslo University Hospital, Oslo, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Fridtjof Lund-Johansen
- Department of Immunology, Oslo University Hospital, Oslo, Norway; Precision Immunotherapy Alliance, University of Oslo, Oslo, Norway; ImmunoLingo Convergence Center, University of Oslo, Oslo, Norway
| | - Jan Cato Holter
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.; Department of Microbiology, Oslo University Hospital, Oslo, Norway
| | - Bente Halvorsen
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | | | - Ludvig A Munthe
- Department of Immunology, Oslo University Hospital, Oslo, Norway; KG Jebsen Centre for B cell Malignancies, Institute of Clinical Medicine, University of Oslo, Norway; Precision Immunotherapy Alliance, University of Oslo, Oslo, Norway.
| | - Hassen Kared
- Department of Immunology, Oslo University Hospital, Oslo, Norway; KG Jebsen Centre for B cell Malignancies, Institute of Clinical Medicine, University of Oslo, Norway; Precision Immunotherapy Alliance, University of Oslo, Oslo, Norway.
| |
Collapse
|
5
|
Viermyr HK, Tonby K, Ponzi E, Trouillet-Assant S, Poissy J, Arribas JR, Dyon-Tafani V, Bouscambert-Duchamp M, Assoumou L, Halvorsen B, Tekin NB, Diallo A, De Gastines L, Munthe LA, Murphy SL, Ueland T, Michelsen AE, Lund-Johansen F, Aukrust P, Mootien J, Dervieux B, Zerbib Y, Richard JC, Prével R, Malvy D, Timsit JF, Peiffer-Smadja N, Roux D, Piroth L, Ait-Oufella H, Vieira C, Dalgard O, Heggelund L, Müller KE, Møller JH, Kildal AB, Skogen V, Aballi S, Sjøberg Øgaard JD, Dyrhol-Riise AM, Tveita A, Alirezaylavasani A, Costagliola D, Yazdanpanah Y, Olsen IC, Dahl TB, Kared H, Holten AR, Trøseid M. Safety of baricitinib in vaccinated patients with severe and critical COVID-19 sub study of the randomised Bari-SolidAct trial. EBioMedicine 2025; 111:105511. [PMID: 39731852 PMCID: PMC11743795 DOI: 10.1016/j.ebiom.2024.105511] [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: 07/17/2024] [Revised: 12/04/2024] [Accepted: 12/05/2024] [Indexed: 12/30/2024] Open
Abstract
BACKGROUND The Bari-SolidAct randomized controlled trial compared baricitinib with placebo in patients with severe COVID-19. A post hoc analysis revealed a higher incidence of serious adverse events (SAEs) among SARS-CoV-2-vaccinated participants who had received baricitinib. This sub-study aimed to investigate whether vaccination influences the safety profile of baricitinib in patients with severe COVID-19. METHODS Biobanked samples from 146 participants (55 vaccinated vs. 91 unvaccinated) were analysed longitudinally for inflammation markers, humoral responses, tissue viral loads, and plasma viral antigens on days 1, 3, and 8. High-dimensional analyses, including RNA sequencing and flow cytometry, were performed on available samples. Mediation analyses were used to assess relationships between SAEs, baseline-adjusted biomarkers, and treatment-vaccination status. FINDINGS Vaccinated participants were older, more frequently hospitalized, had more comorbidities, and exhibited higher nasopharyngeal viral loads. Baricitinib treatment did not affect antibody responses or viral clearance, but reduced markers of T-cell and monocyte activation compared to placebo (sCD25, sCD14, sCD163, sTIM-3). Age, baseline levels of plasma viral antigen, and several inflammatory markers, as well as IL-2, IL-6, Neopterin, CXCL16, sCD14, and suPAR on day 8 were associated with the occurrence of SAEs. However, mediation analyses of markers linked to SAEs, baricitinib treatment, or vaccination status did not reveal statistically significant interactions between vaccination status and SAEs. INTERPRETATION This sub-study did not identify any virus- or host-related biomarkers significantly associated with the interaction between SARS-CoV-2 vaccination status and the safety of baricitinib. However, caution should be exercised due to the moderate sample size. FUNDING EU Horizon 2020 (grant number 101015736).
Collapse
Affiliation(s)
- Hans-Kittil Viermyr
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway; Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Section for Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Kristian Tonby
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Infectious Diseases, Oslo University Hospital Ullevål, Oslo, Norway
| | - Erica Ponzi
- Department of Research Support for Clinical Trials, Oslo University Hospital, Oslo, Norway
| | - Sophie Trouillet-Assant
- Centre International de Recherche en Infectiologie (CIRI), Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR5308, ENS Lyon, Lyon, France; Joint Research Unit Hospices Civils de Lyon-bioMerieux S.A., Hôpital Lyon Sud, Pierre-Bénite, France
| | - Julien Poissy
- Université Lille, Inserm U1285, CHU Lille, Pôle de Médecine Intensive-Réanimatin, CNRS, UMR 8576, France; Université Lille, Unité de Glycobiologie Structurale et Fonctionnelle (UGSF), F-59000, Lille, France
| | - José R Arribas
- Infectious Diseases Unit, Internal Medicine Department, La Paz University Hospital, IdiPAZ, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
| | - Virginie Dyon-Tafani
- Centre International de Recherche en Infectiologie (CIRI), Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR5308, ENS Lyon, Lyon, France
| | - Maude Bouscambert-Duchamp
- Hospices Civils de Lyon, Laboratoire de Virologie, Institut des Agents Infectieux de Lyon, Centre National de Référence des Virus Respiratoires France Sud, F-69317, Lyon, France; Université Claude Bernard Lyon 1, Virpath, CIRI, INSERM U1111, CNRS UMR5308, ENS Lyon, F-69372, Lyon, France
| | - Lambert Assoumou
- Sorbonne Université, INSERM, Institut Pierre Louis d'Épidémiologie et de Santé Publique (IPLESP), Paris, France
| | - Bente Halvorsen
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway; Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Nuriye Basdag Tekin
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway; Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Alpha Diallo
- ANRS, Maladies Infectieuses Emergentes, F-75015, Paris, France; Institut National de la Santé et de la Recherche Médicale, INSERM, F-75013, Paris, France
| | - Lucie De Gastines
- ANRS, Maladies Infectieuses Emergentes, F-75015, Paris, France; Institut National de la Santé et de la Recherche Médicale, INSERM, F-75013, Paris, France
| | - Ludvig A Munthe
- Department of Immunology, Oslo University Hospital, Oslo, Norway; KG Jebsen Centre for B Cell Malignancies, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Precision Immunotherapy Alliance, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Sarah Louise Murphy
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway; Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Thor Ueland
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway; Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Thrombosis Research Center (TREC), Division of Internal Medicine, University Hospital of North Norway, Tromsø, Norway
| | - Annika E Michelsen
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway; Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Fridtjof Lund-Johansen
- Department of Immunology, Oslo University Hospital, Oslo, Norway; Precision Immunotherapy Alliance, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Pål Aukrust
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway; Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Joy Mootien
- Intensive Care Unit, Antibiotic Stewardship Team, Groupe Hospitalier Région Mulhouse et Sud Alsace, Mulhouse, France
| | - Benjamin Dervieux
- Infectious Diseases Unit, Groupe Hospitalier Région Mulhouse et Sud Alsace, Mulhouse, France
| | - Yoann Zerbib
- Intensive Care Department, Amiens-Picardie University Hospital, Amiens, France
| | - Jean-Christophe Richard
- Medical Intensive Care Unit, Hospices Civils de Lyon, Croix-Rousse Hospital - Université Lyon 1, Lyon, France; CREATIS INSERM 1044 CNRS 5220, Villeurbanne, France
| | - Renaud Prével
- CHU Bordeaux, Medical Intensive Care Unit, Bordeaux, France; Univ Bordeaux, Centre de Recherche Cardio-Thoracique de Bordeaux, Inserm UMR 1045, Bordeaux, France
| | - Denis Malvy
- Department of Infectious and Tropical Diseases, University Hospital, UMR 1219 Inserm/EMR 271 IRD, University of Bordeaux, Bordeaux, France
| | - Jean-François Timsit
- AP-HP, Bichat Hospital, Medical and Infectious Diseases ICU (MI2), F-75018, Paris, France; Université Paris-Cité, INSERM, F-75018, Paris, France; OUCTOME REA Research Network, France
| | - Nathan Peiffer-Smadja
- Université Paris Cité, Inserm, IAME, Paris, 75018, France; Service de Maladies Infectieuses et Tropicales, Hôpital Bichat Claude Bernard, AP-HP, Paris, 75018, France
| | - Damien Roux
- Université Paris Cité, AP-HP, Hôpital Louis Mourier, DMU ESPRIT, Service de Médecine Intensive Réanimation, Colombes, France
| | - Lionel Piroth
- Infectious Diseases Department, University Hospital, Dijon, France; INSERM CIC 1432, University of Burgundy, Dijon, France
| | - Hafid Ait-Oufella
- Service de Médecine Intensive-Réanimation, Hôpital Saint-Antoine, AP-HP, Sorbonne Université, Paris, F-75012, France
| | - Cesar Vieira
- Centro Hospitalar Universitário de Lisboa Central, Hospital Curry Cabral, Department of Intensive Care Medicine - Lisbon, Portugal
| | - Olav Dalgard
- Department of Infectious Diseases, Oslo University Hospital Ullevål, Oslo, Norway; Department of Infectious Diseases, Akershus University Hospital, Norway
| | - Lars Heggelund
- Department of Internal Medicine, Drammen Hospital, Vestre Viken Hospital Trust, Norway; Department of Clinical Science, Bergen Integrated Diagnostic Stewardship Cluster, Faculty of Medicine, University of Bergen, Bergen, Norway
| | - Karl Erik Müller
- Department of Internal Medicine, Drammen Hospital, Vestre Viken Hospital Trust, Norway; Department of Clinical Science, Bergen Integrated Diagnostic Stewardship Cluster, Faculty of Medicine, University of Bergen, Bergen, Norway
| | | | - Anders Benjamin Kildal
- Department of Anesthesiology and Intensive Care, University Hospital of North Norway, Tromsø, Norway; Faculty of Health Sciences, UIT The Arctic University of Norway, Norway
| | - Vegard Skogen
- Department of Infectious Diseases, University Hospital of North Norway, Tromsø, Norway; Faculty of Health Sciences, UIT The Arctic University of Norway, Norway
| | - Saad Aballi
- Department of Infectious Diseases, Østfold Hospital Kalnes, Grålum, Norway
| | - Jonas Daniel Sjøberg Øgaard
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway; Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Anne Ma Dyrhol-Riise
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Infectious Diseases, Oslo University Hospital Ullevål, Oslo, Norway
| | - Anders Tveita
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway; Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Section for Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet, Oslo, Norway; Department of Internal Medicine, Bærum Hospital, Vestre Viken Hospital Trust, 1346, Gjettum, Norway
| | | | - Dominique Costagliola
- Sorbonne Université, INSERM, Institut Pierre Louis d'Épidémiologie et de Santé Publique (IPLESP), Paris, France
| | - Yazdan Yazdanpanah
- Infectious and Tropical Diseases Department, Bichat - Claude Bernard Hospital, AP-HP Nord-Université Paris Cité, Paris, France; IAME INSERM UMR 1137, Université Paris Cité, Paris, France; ANRS, Maladies Infectieuses Emergentes, F-75015, Paris, France; Institut National de la Santé et de la Recherche Médicale, INSERM, F-75013, Paris, France
| | - Inge Christoffer Olsen
- Department of Research Support for Clinical Trials, Oslo University Hospital, Oslo, Norway
| | - Tuva Børresdatter Dahl
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway; Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Hassen Kared
- Department of Immunology, Oslo University Hospital, Oslo, Norway; KG Jebsen Centre for B Cell Malignancies, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Precision Immunotherapy Alliance, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Aleksander Rygh Holten
- Department of Infectious Diseases, Oslo University Hospital Ullevål, Oslo, Norway; Department of Acute Medicine, Oslo University Hospital, Oslo, Norway
| | - Marius Trøseid
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway; Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Section for Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet, Oslo, Norway.
| |
Collapse
|
6
|
Wolf AS, Bjørlykke KH, Ørbo HS, Bhandari S, Solum G, Kjønstad IF, Jyssum I, Nygaard UC, Kristoffersen AB, Christensen IE, Josefsson SE, Lund KP, Chopra A, Osen JR, Chaban V, Tveter AT, Sexton J, Kvien TK, Jahnsen J, Haavardsholm EA, Grødeland G, Vaage JT, Provan SA, Kared H, Lund-Johansen F, Munthe LA, Syversen SW, Goll GL, Jørgensen KK, Mjaaland S. T cell responses to repeated SARS-CoV-2 vaccination and breakthrough infections in patients on TNF inhibitor treatment: a prospective cohort study. EBioMedicine 2024; 108:105317. [PMID: 39260039 PMCID: PMC11416219 DOI: 10.1016/j.ebiom.2024.105317] [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: 01/31/2024] [Revised: 07/29/2024] [Accepted: 08/15/2024] [Indexed: 09/13/2024] Open
Abstract
BACKGROUND Understanding cellular responses to SARS-CoV-2 immunisations is important for informing vaccine recommendations in patients with inflammatory bowel disease (IBD) and other vulnerable patients on immunosuppressive therapies. This study investigated the magnitude and quality of T cell responses after multiple SARS-CoV-2 vaccine doses and COVID-19 breakthrough infection. METHODS This prospective, observational study included patients with IBD and arthritis on tumour necrosis factor inhibitors (TNFi) receiving up to four SARS-CoV-2 vaccine doses. T cell responses to SARS-CoV-2 peptides were measured by flow cytometry before and 2-4 weeks after vaccinations and breakthrough infection to assess the frequency and polyfunctionality of responding cells, along with receptor-binding domain (anti-RBD) antibodies. FINDINGS Between March 2, 2021, and December 20, 2022, 143 patients (118 IBD, 25 arthritis) and 73 healthy controls were included. In patients with either IBD or arthritis, humoral immunity was attenuated compared to healthy controls (median anti-RBD levels 3391 vs. 6280 BAU/ml, p = 0.008) after three SARS-CoV-2 vaccine doses. Patients with IBD had comparable quantities (median CD4 0.11% vs. 0.11%, p = 0.26, CD8 0.031% vs. 0.047%, p = 0.33) and quality (polyfunctionality score: 0.403 vs. 0.371, p = 0.39; 0.105 vs. 0.101, p = 0.87) of spike-specific T cells to healthy controls. Patients with arthritis had lower frequencies but comparable quality of responding T cells to controls. Breakthrough infection increased spike-specific CD8 T cell quality and T cell responses against non-spike peptides. INTERPRETATION Patients with IBD on TNFi have T cell responses comparable to healthy controls despite attenuated humoral responses following three vaccine doses. Repeated vaccination and breakthrough infection increased the quality of T cell responses. Our study adds evidence that, in the absence of other risk factors, this group may in future be able to follow the general recommendations for COVID-19 vaccines. FUNDING South-Eastern Norway Regional Health Authority, Coalition for Epidemic Preparedness Innovations (CEPI), Norwegian Institute of Public Health, Akershus University Hospital, Diakonhjemmet Hospital.
Collapse
Affiliation(s)
- Asia-Sophia Wolf
- Division of Infection Control, Section for Immunology, Norwegian Institute of Public Health, Oslo, Norway.
| | - Kristin H Bjørlykke
- Department of Gastroenterology, Akershus University Hospital, Lørenskog, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Hilde S Ørbo
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Center for Treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway
| | - Sabin Bhandari
- Division of Infection Control, Section for Immunology, Norwegian Institute of Public Health, Oslo, Norway
| | - Guri Solum
- Division of Infection Control, Section for Immunology, Norwegian Institute of Public Health, Oslo, Norway
| | - Ingrid Fadum Kjønstad
- Division of Infection Control, Section for Immunology, Norwegian Institute of Public Health, Oslo, Norway
| | - Ingrid Jyssum
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Center for Treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway
| | - Unni C Nygaard
- Division of Infection Control, Section for Immunology, Norwegian Institute of Public Health, Oslo, Norway
| | - Anja Bråthen Kristoffersen
- Division of Infection Control, Section for Modelling and Bioinformatics, Norwegian Institute of Public Health, Oslo, Norway
| | - Ingrid E Christensen
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Center for Treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway
| | - Sarah E Josefsson
- Division of Infection Control, Section for Immunology, Norwegian Institute of Public Health, Oslo, Norway
| | - Katrine Persgård Lund
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Immunology, Oslo University Hospital, Oslo, Norway; KG Jebsen Centre for B Cell Malignancy, University of Oslo, Oslo, Norway
| | - Adity Chopra
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Immunology, Oslo University Hospital, Oslo, Norway
| | - Julie Røkke Osen
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Immunology, Oslo University Hospital, Oslo, Norway; KG Jebsen Centre for B Cell Malignancy, University of Oslo, Oslo, Norway
| | - Viktoriia Chaban
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Immunology, Oslo University Hospital, Oslo, Norway; KG Jebsen Centre for B Cell Malignancy, University of Oslo, Oslo, Norway
| | - Anne T Tveter
- Center for Treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway; Faculty of Health Sciences, Oslo Metropolitan University, Oslo, Norway
| | - Joseph Sexton
- Center for Treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway
| | - Tore K Kvien
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Center for Treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway
| | - Jørgen Jahnsen
- Department of Gastroenterology, Akershus University Hospital, Lørenskog, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Espen A Haavardsholm
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Center for Treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway
| | - Gunnveig Grødeland
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Immunology, Oslo University Hospital, Oslo, Norway
| | - John Torgils Vaage
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Immunology, Oslo University Hospital, Oslo, Norway
| | - Sella A Provan
- Center for Treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway; Section for Public Health, Inland Norway University of Applied Sciences, Norway
| | - Hassen Kared
- Department of Immunology, Oslo University Hospital, Oslo, Norway; KG Jebsen Centre for B Cell Malignancy, University of Oslo, Oslo, Norway
| | - Fridtjof Lund-Johansen
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Immunology, Oslo University Hospital, Oslo, Norway
| | - Ludvig A Munthe
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Immunology, Oslo University Hospital, Oslo, Norway; KG Jebsen Centre for B Cell Malignancy, University of Oslo, Oslo, Norway
| | - Silje Watterdal Syversen
- Center for Treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway; Institute of Health and Society, University of Oslo, Norway
| | - Guro Løvik Goll
- Center for Treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway; Institute of Health and Society, University of Oslo, Norway
| | | | - Siri Mjaaland
- Division of Infection Control, Section for Immunology, Norwegian Institute of Public Health, Oslo, Norway
| |
Collapse
|
7
|
Tunheim G, Fossum E, Robertson AH, Rø GØI, Chopra A, Vaage JT, Vikse EL, Kran AMB, Magnus P, Trogstad L, Mjaaland S, Hungnes O, Lund-Johansen F. Characterization of the SARS-CoV-2 antibody landscape in Norway in the late summer of 2022: high seroprevalence in all age groups with patterns of primary Omicron infection in children and hybrid immunity in adults. BMC Infect Dis 2024; 24:841. [PMID: 39164637 PMCID: PMC11334563 DOI: 10.1186/s12879-024-09670-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 07/25/2024] [Indexed: 08/22/2024] Open
Abstract
BACKGROUND According to Norwegian registries, 91% of individuals ≥ 16 years had received ≥ 1 dose of COVID-19 vaccine by mid-July 2022, whereas less than 2% of children < 12 years were vaccinated. Confirmed COVID-19 was reported for 27% of the population, but relaxation of testing lead to substantial underreporting. We have characterized the humoral immunity to SARS-CoV-2 in Norway in the late summer of 2022 by estimating the seroprevalence and identifying antibody profiles based on reactivity to Wuhan or Omicron-like viruses in a nationwide cross-sectional collection of residual sera, and validated our findings using cohort sera. METHODS 1,914 anonymized convenience sera and 243 NorFlu-cohort sera previously collected from the Oslo-area with reported infection and vaccination status were analyzed for antibodies against spike, the receptor-binding domain (RBD) of the ancestral Wuhan strain and Omicron BA.2 RBD, and nucleocapsid (N). Samples were also tested for antibodies inhibiting RBD-ACE2 interaction. Neutralization assays were performed on subsets of residual sera against B.1, BA.2, XBB.1.5 and BQ.1.1. RESULTS The national seroprevalence estimate from vaccination and/or infection was 99.1% (95% CrI 97.0-100.0%) based on Wuhan (spike_W and RBD_W) and RBD_BA2 antibodies. Sera from children < 12 years had 2.2 times higher levels of antibodies against RBD_BA2 than RBD_W and their seroprevalence estimate showed a 14.4 percentage points increase when also including anti-RBD_BA2 antibodies compared to Wuhan-antibodies alone. 50.3% (95% CI 45.0-55.5%) of residual sera from children and 38.1% (95% CI 36.0-40.4%) of all residual sera were positive for anti-N-antibodies. By combining measurements of binding- and ACE2-RBD-interaction-inhibiting antibodies, reactivity profiles indicative of infection and vaccination history were identified and validated using cohort sera. Residual sera with a profile indicative of hybrid immunity were able to neutralize newer Omicron variants XBB.1.5 and BQ.1.1. CONCLUSIONS By late summer of 2022, most of the Norwegian population had antibodies to SARS-CoV-2, and almost all children had been infected. Antibody profiles indicated that children mostly had experienced a primary Omicron infection, while hybrid immunity was common among adults. The finding that sera displaying hybrid immunity could neutralize newer Omicron variants indicates that Wuhan-like priming of the immune response did not have a harmful imprinting effect and that infections induce cross-reacting antibodies against future variants.
Collapse
Affiliation(s)
- Gro Tunheim
- Division of Infection Control, Norwegian Institute of Public Health (NIPH), Oslo, Norway.
| | - Even Fossum
- Division of Infection Control, Norwegian Institute of Public Health (NIPH), Oslo, Norway
| | - Anna Hayman Robertson
- Division of Infection Control, Norwegian Institute of Public Health (NIPH), Oslo, Norway
| | | | - Adity Chopra
- Department of Immunology, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - John T Vaage
- Department of Immunology, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Elisabeth Lea Vikse
- Division of Infection Control, Norwegian Institute of Public Health (NIPH), Oslo, Norway
| | - Anne-Marte Bakken Kran
- Division of Infection Control, Norwegian Institute of Public Health (NIPH), Oslo, Norway
| | - Per Magnus
- Center for Fertility and Health, Norwegian Institute of Public Health (NIPH), Oslo, Norway
| | - Lill Trogstad
- Division of Infection Control, Norwegian Institute of Public Health (NIPH), Oslo, Norway
| | - Siri Mjaaland
- Division of Infection Control, Norwegian Institute of Public Health (NIPH), Oslo, Norway
| | - Olav Hungnes
- Division of Infection Control, Norwegian Institute of Public Health (NIPH), Oslo, Norway
| | | |
Collapse
|
8
|
Ahmed MI, Einhauser S, Peiter C, Senninger A, Baranov O, Eser TM, Huth M, Olbrich L, Castelletti N, Rubio-Acero R, Carnell G, Heeney J, Kroidl I, Held K, Wieser A, Janke C, Hoelscher M, Hasenauer J, Wagner R, Geldmacher C, on behalf of the KoCo19/ORCHESTRA working group. Evolution of protective SARS-CoV-2-specific B and T cell responses upon vaccination and Omicron breakthrough infection. iScience 2024; 27:110138. [PMID: 38974469 PMCID: PMC11225850 DOI: 10.1016/j.isci.2024.110138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 03/21/2024] [Accepted: 05/27/2024] [Indexed: 07/09/2024] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) Omicron breakthrough infection (BTI) induced better protection than triple vaccination. To address the underlying immunological mechanisms, we studied antibody and T cell response dynamics during vaccination and after BTI. Each vaccination significantly increased peak neutralization titers with simultaneous increases in circulating spike-specific T cell frequencies. Neutralization titers significantly associated with a reduced hazard rate for SARS-CoV-2 infection. Yet, 97% of triple vaccinees became SARS-CoV-2 infected. BTI further boosted neutralization magnitude and breadth, broadened virus-specific T cell responses to non-vaccine-encoded antigens, and protected with an efficiency of 88% from further infections by December 2022. This effect was then assessed by utilizing mathematical modeling, which accounted for time-dependent infection risk, the antibody, and T cell concentration at any time point after BTI. Our findings suggest that cross-variant protective hybrid immunity induced by vaccination and BTI was an important contributor to the reduced virus transmission observed in Bavaria in late 2022 and thereafter.
Collapse
Affiliation(s)
- Mohamed I.M. Ahmed
- Division of Infectious Diseases and Tropical Medicine, University Hospital, LMU Munich, 80799 Munich, Germany
- German Centre for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| | - Sebastian Einhauser
- Institute for Medical Microbiology and Hygiene, University of Regensburg, 93053 Regensburg, Germany
| | - Clemens Peiter
- Faculty of Mathematics and Natural Sciences, University of Bonn, 53113 Bonn, Germany
| | - Antonia Senninger
- Institute for Medical Microbiology and Hygiene, University of Regensburg, 93053 Regensburg, Germany
| | - Olga Baranov
- Division of Infectious Diseases and Tropical Medicine, University Hospital, LMU Munich, 80799 Munich, Germany
- German Centre for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| | - Tabea M. Eser
- Division of Infectious Diseases and Tropical Medicine, University Hospital, LMU Munich, 80799 Munich, Germany
- German Centre for Infection Research (DZIF), Partner Site Munich, Munich, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology, Infection and Pandemic Research, 80799 Munich, Germany
| | - Manuel Huth
- Faculty of Mathematics and Natural Sciences, University of Bonn, 53113 Bonn, Germany
- Institute of Computational Biology, Helmholtz Zentrum München – German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Laura Olbrich
- Division of Infectious Diseases and Tropical Medicine, University Hospital, LMU Munich, 80799 Munich, Germany
- German Centre for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| | - Noemi Castelletti
- Division of Infectious Diseases and Tropical Medicine, University Hospital, LMU Munich, 80799 Munich, Germany
| | - Raquel Rubio-Acero
- Division of Infectious Diseases and Tropical Medicine, University Hospital, LMU Munich, 80799 Munich, Germany
| | - George Carnell
- Lab of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Jonathan Heeney
- Lab of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Inge Kroidl
- Division of Infectious Diseases and Tropical Medicine, University Hospital, LMU Munich, 80799 Munich, Germany
- German Centre for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| | - Kathrin Held
- Division of Infectious Diseases and Tropical Medicine, University Hospital, LMU Munich, 80799 Munich, Germany
- German Centre for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| | - Andreas Wieser
- Division of Infectious Diseases and Tropical Medicine, University Hospital, LMU Munich, 80799 Munich, Germany
- German Centre for Infection Research (DZIF), Partner Site Munich, Munich, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology, Infection and Pandemic Research, 80799 Munich, Germany
| | - Christian Janke
- Division of Infectious Diseases and Tropical Medicine, University Hospital, LMU Munich, 80799 Munich, Germany
| | - Michael Hoelscher
- Division of Infectious Diseases and Tropical Medicine, University Hospital, LMU Munich, 80799 Munich, Germany
- German Centre for Infection Research (DZIF), Partner Site Munich, Munich, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology, Infection and Pandemic Research, 80799 Munich, Germany
- Unit Global Health, Helmholtz Zentrum München, German Research Center for Environmental Health (HMGU), 85764 Neuherberg, Germany
| | - Jan Hasenauer
- Faculty of Mathematics and Natural Sciences, University of Bonn, 53113 Bonn, Germany
- Institute of Computational Biology, Helmholtz Zentrum München – German Research Center for Environmental Health, 85764 Neuherberg, Germany
- Center for Mathematics, Technische Universität München, 85748 Garching, Germany
| | - Ralf Wagner
- Institute for Medical Microbiology and Hygiene, University of Regensburg, 93053 Regensburg, Germany
- Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Christof Geldmacher
- Division of Infectious Diseases and Tropical Medicine, University Hospital, LMU Munich, 80799 Munich, Germany
- German Centre for Infection Research (DZIF), Partner Site Munich, Munich, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology, Infection and Pandemic Research, 80799 Munich, Germany
| | - on behalf of the KoCo19/ORCHESTRA working group
- Division of Infectious Diseases and Tropical Medicine, University Hospital, LMU Munich, 80799 Munich, Germany
- German Centre for Infection Research (DZIF), Partner Site Munich, Munich, Germany
- Institute for Medical Microbiology and Hygiene, University of Regensburg, 93053 Regensburg, Germany
- Faculty of Mathematics and Natural Sciences, University of Bonn, 53113 Bonn, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology, Infection and Pandemic Research, 80799 Munich, Germany
- Institute of Computational Biology, Helmholtz Zentrum München – German Research Center for Environmental Health, 85764 Neuherberg, Germany
- Lab of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
- Unit Global Health, Helmholtz Zentrum München, German Research Center for Environmental Health (HMGU), 85764 Neuherberg, Germany
- Center for Mathematics, Technische Universität München, 85748 Garching, Germany
- Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, 93053 Regensburg, Germany
| |
Collapse
|
9
|
Wüsthoff LEC, Lund-Johansen F, Henriksen K, Wildendahl G, Jacobsen JA, Gomes L, Anjum HS, Barlinn R, Kran AMB, Munthe LA, Vaage JT. Seroprevalence of SARS-CoV-2 and humoral immune responses to COVID-19 mRNA vaccines among people who use drugs - in the light of tailored mitigating strategies. Harm Reduct J 2024; 21:120. [PMID: 38890611 PMCID: PMC11186241 DOI: 10.1186/s12954-024-01023-9] [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: 05/16/2023] [Accepted: 05/14/2024] [Indexed: 06/20/2024] Open
Abstract
BACKGROUND During the initial wave of the COVID-19 pandemic, there was a surprisingly low incidence of SARS-CoV-2 among People Who Use Drugs (PWUD) in Oslo, Norway, despite their heightened vulnerability regarding risk of infection and severe courses of the disease.This study aims to investigate the seroprevalence of SARS-CoV-2 antibodies among PWUD, their antibody responses to relevant virus infections and COVID-19 mRNA vaccines, and their vaccination coverage compared to the general population. METHODS Conducted as a prospective cohort study, data was collected from residents in six institutions for homeless PWUD and users of a low-threshold clinic for opioid agonist treatment. Ninety-seven participants were recruited for SARS-CoV-2 seroprevalence analysis. Additional two participants with known positive SARS-CoV-2 test results were recruited for further analyses. Twenty-five participants completed follow-up. Data included questionnaires, nasal swabs and blood samples. Data on vaccination coverage was obtained from the National Vaccine Register. Serologic methods included detection of antibodies to relevant virus proteins, neutralizing antibodies to SARS-CoV-2, antibodies to the full-length spike protein, and receptor-binding domain from SARS-CoV-2. RESULTS Among PWUD, antibodies to SARS-CoV-2 were detected in 2 out of 97 samples before vaccines against SARS-CoV-2 were available, comparable to a 2.8% frequency in population-based screening. Levels of serum antibodies to seasonal coronaviruses and Epstein-Barr-Virus (EBV) in PWUD were similar to population-based levels. After the second vaccine dose, binding and neutralizing antibody levels to SARS-CoV-2 in PWUD were comparable to controls. Eighty-four of PWUD received at least one dose of COVID-19 mRNA vaccine, compared to 89% in the general population. CONCLUSION Results indicate that PWUD did not exhibit increased SARS-CoV-2 seroprevalence or elevated serum antibodies to seasonal coronaviruses and EBV. Moreover, vaccine responses in PWUD were comparable to controls, suggesting that vaccination is effective in conferring protection against SARS-CoV-2 also in this population.
Collapse
Affiliation(s)
- Linda Elise Couëssurel Wüsthoff
- Unit for Clinical Research on Addictions, Oslo University Hospital, PO Box 4959 Nydalen, Oslo, 0424, Norway.
- Norwegian Centre for Addiction Reasearch, Institute of Clinical Medicine, University of Oslo, PO Box 1039 Blindern, Oslo, 0315, Norway.
| | - Fridtjof Lund-Johansen
- Department of Immunology, Oslo University Hospital, PO Box 4950 Nydalen, Oslo, 0424, Norway
- ImmunoLingo Convergence Center, Institute of Clinical Medicine, University of Oslo, Gaustadalleen 21, Oslo, 0349, Norway
- Institute of Clinical Medicine, University of Oslo, PO Box 1171 Blindern, Oslo, 0318, Norway
- Precision Immunotherapy Alliance, Institute of Clinical Medicine, University of Oslo, PO Box 1171 Blindern, 0318, Oslo, Norway
| | - Kathleen Henriksen
- Agency for Social and Welfare Services, Oslo Municipality, PO Box 30 Sentrum, Oslo, 0101, Norway
- Student Health Services, Student Welfare Services in Oslo, Problemveien 9, Oslo, 0313, Norway
| | - Gull Wildendahl
- Agency for Social and Welfare Services, Oslo Municipality, PO Box 30 Sentrum, Oslo, 0101, Norway
| | - Jon-Aksel Jacobsen
- Agency for Social and Welfare Services, Oslo Municipality, PO Box 30 Sentrum, Oslo, 0101, Norway
| | - Leni Gomes
- Agency for Social and Welfare Services, Oslo Municipality, PO Box 30 Sentrum, Oslo, 0101, Norway
| | - Hina Sarwar Anjum
- Agency for Social and Welfare Services, Oslo Municipality, PO Box 30 Sentrum, Oslo, 0101, Norway
| | - Regine Barlinn
- Department of Microbiology, Oslo University Hospital, PO Box 4950 Nydalen, Oslo, 0424, Norway
| | - Anne-Marte Bakken Kran
- Division of Infection Control, Norwegian Institute of Public Health, PO Box 222 Skøyen, Oslo, 0213, Norway
| | - Ludvig Andre Munthe
- Department of Immunology, Oslo University Hospital, PO Box 4950 Nydalen, Oslo, 0424, Norway
- KG Jebsen Centre for B cell Malignancies, and Precision Immunotherapy Alliance, Institute of Clinical Medicine, University of Oslo, PO Box 4950 Nydalen, Oslo, 0424, Norway
| | - John T Vaage
- Department of Immunology, Oslo University Hospital, PO Box 4950 Nydalen, Oslo, 0424, Norway
- Institute of Clinical Medicine, University of Oslo, PO Box 1171 Blindern, Oslo, 0318, Norway
| |
Collapse
|
10
|
Chen CH, Chao DY, Kor CT, Kuo SF, Lin JS, Lai HW, Liu YT, Lin CH, Chen MK. A cross-sectional study of SARS-CoV-2 antibodies among healthcare workers in a tertiary care hospital in Taiwan: implications for protection against the Omicron variants. BMC Infect Dis 2024; 24:529. [PMID: 38802771 PMCID: PMC11129381 DOI: 10.1186/s12879-024-09411-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 05/16/2024] [Indexed: 05/29/2024] Open
Abstract
BACKGROUND Taiwan, deeply impacted by the 2003 SARS outbreak, promptly implemented rigorous infection control and prevention (ICP) measures in January 2020 to combat the global COVID-19 pandemic. This cross-sectional serologic study was conducted among healthcare workers (HCWs) in a tertiary care hospital in Taiwan from August 1, 2022, to February 28, 2023. The study aimed to assess HCWs' antibody responses to COVID-19 vaccination against Omicron subvariants BA.1, BA.4, and BA.5, considering variations in prior infection. Additionally, it evaluated the effectiveness of ICP and vaccination policies within the hospital setting in Taiwan. METHODS A cross-sectional serology study was conducted in Taiwan to investigate the seroprevalence rates of Omicron subvariants BA.1, BA.4, and BA.5 among HCWs. A total of 777 HCWs participated in this study. A structured questionnaire was collected to obtain the epidemiological characteristics and risk factors for potential exposure. Enzyme-linked immunosorbent assay was used to detect antibody responses. Serum samples were selected for protection against Omicron subvariants BA.1, BA.4, and BA.5 by using a pseudotyped-based neutralization assay. RESULTS More than 99% of the participants had received SARS-CoV-2 vaccination. Overall, 57.7% had been infected with SARS-CoV-2, with some being asymptomatic. The SARS-CoV-2 Anti-Spike S1 protein IgG (Anti-S) distribution was 40,000 AU/mL for 20.2% (157/777) of participants, with a mean ± standard deviation of 23,442 ± 22,086. The decay curve for Anti-S was less than 20,000 AU/ml after 120 days. The probability curve of 50% neutralization showed an Anti-S of 55,000 AU/ml. The optimum Anti-S was 41,328 AU/mL (equal to 5,869 WHO's standard BAU/mL), with 86.1% sensitivity and 63.5% specificity. CONCLUSIONS In this significant study, 20.2% of HCWs achieved seroprotection against Omicron subvariants BA.1, BA.4, and BA.5. Their immunity against Omicron subvariants was further reinforced through recommended vaccinations and the development of natural immunity from SARS-CoV-2 exposure, collectively enhancing their protection against Omicron.
Collapse
Affiliation(s)
- Chang-Hua Chen
- Division of Infectious Diseases, Department of Internal Medicine, Changhua Christian Hospital, Changhua; No.135, Nanxiao St., Changhua City, Changhua County, 50006, Taiwan.
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung-Hsing University, No. 145 Xingda Rd., South Dist, Taichung City, 40227, Taiwan.
- Doctoral Program in Microbial Genomics, National Chung Hsing University and Academia Sinica, No. 145 Xingda Rd., South Dist., Taichung City, 40227, Taiwan.
| | - Day-Yu Chao
- Graduate Institute of Microbiology and Public Health, College of Veterinary Medicine, National Chung-Hsing University, No. 145 Xingda Rd., South Dist, Taichung City, 40227, Taiwan
| | - Chew-Teng Kor
- Big Data Center, Changhua Christian Hospital, No.135, Nanxiao St., Changhua City, Changhua County, 50006, Taiwan
- Institute of Statistics and Information Science, National Changhua University of Education, Changhua County, No.1, Jinde Rd, Changhua City, Changhua County, 50074, Taiwan
| | - Su-Feng Kuo
- Clinical Microbiology Laboratory, Changhua Christian Hospital, No.135, Nanxiao St., Changhua City, Changhua County, 50006, Taiwan
| | - Jen-Shiou Lin
- Clinical Microbiology Laboratory, Changhua Christian Hospital, No.135, Nanxiao St., Changhua City, Changhua County, 50006, Taiwan
| | - Huei-Wen Lai
- Center for Infection Prevention and Control, Changhua Christian Hospital, No.135, Nanxiao St., Changhua City, Changhua County, 50006, Taiwan
| | - Yen-Tze Liu
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung-Hsing University, No. 145 Xingda Rd., South Dist, Taichung City, 40227, Taiwan
- Department of Family Medicine, Changhua Christian Hospital, No.135, Nanxiao St., Changhua City, Changhua County, 50006, Taiwan
| | - Ching-Hsiung Lin
- Division of Chest Medicine, Department of Internal Medicine, Changhua Christian Hospital, No.135, Nanxiao St., Changhua City, Changhua County, 50006, Taiwan
| | - Mu-Kuan Chen
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung-Hsing University, No. 145 Xingda Rd., South Dist, Taichung City, 40227, Taiwan.
- Department Of Otorhinolaryngology - Head & Neck Surgery, Changhua Christian Hospital, No.135, Nanxiao St., Changhua City, Changhua County, 50006, Taiwan.
| |
Collapse
|
11
|
Campagna R, Dominelli F, Zingaropoli MA, Ciurluini F, Grilli G, Amoroso A, De Domenico A, Amatore D, Lia MS, Cortesi E, Picone V, Mastroianni CM, Ciardi MR, De Santis R, Lista F, Antonelli G, Turriziani O. COVID-19 vaccination in cancer patients: Immune responses one year after the third dose. Vaccine 2024; 42:2687-2694. [PMID: 38499458 DOI: 10.1016/j.vaccine.2024.03.017] [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: 01/05/2024] [Revised: 03/06/2024] [Accepted: 03/06/2024] [Indexed: 03/20/2024]
Abstract
Cancer patients (CPs), being immunosuppressed due to the treatment received or to the disease itself, are more susceptible to infections and their potential complications, showing therefore an increased risk of developing severe COVID-19 compared to the general population. We evaluated the immune responses to anti-SARS-CoV-2 vaccination in patients with solid tumors one year after the administration of the third dose and the effect of cancer treatment on vaccine immunogenicity was assessed. Healthy donors (HDs) were enrolled. Binding and neutralizing antibody (Ab) titers were evaluated using chemiluminescence immunoassay (CLIA) and Plaque Reduction Neutralization Test (PRNT) respectively. T-cell response was analyzed using multiparametric flow cytometry. CPs who were administered three vaccine doses showed lower Ab titers than CPs with four doses and HDs. Overall, a lower cell-mediated response was found in CPs, with a predominance of monofunctional T-cells producing TNFα. Lower Ab titers and a weaker T-cell response were observed in CPs without prior SARS-CoV-2 infection when compared to those with a previous infection. While no differences in the humoral response were found comparing immunotherapy and non-immunotherapy patients, a stronger T-cell response in CPs treated with immunotherapy was observed. Our results emphasize the need of booster doses in cancer patients to achieve a level of protection similar to that observed in healthy donors and underlines the importance of considering the treatment received to reach a proper immune response.
Collapse
Affiliation(s)
- Roberta Campagna
- Department of Molecular Medicine Sapienza University of Rome, Viale dell'Università, 33, 000185 Rome, Italy.
| | - Federica Dominelli
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Piazzale Aldo Moro, 5, 00185 Rome, Italy.
| | - Maria Antonella Zingaropoli
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Piazzale Aldo Moro, 5, 00185 Rome, Italy.
| | - Fabio Ciurluini
- Department of Radiological, Oncological and Pathological Science, Sapienza University of Rome, 00185 Rome, Italy.
| | - Giorgia Grilli
- Defence Institute for Biomedical Sciences, 00184 Rome, Italy.
| | | | | | | | | | - Enrico Cortesi
- Department of Radiological, Oncological and Pathological Science, Sapienza University of Rome, 00185 Rome, Italy.
| | - Vincenzo Picone
- Department of Radiological, Oncological and Pathological Science, Sapienza University of Rome, 00185 Rome, Italy.
| | - Claudio Maria Mastroianni
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Piazzale Aldo Moro, 5, 00185 Rome, Italy.
| | - Maria Rosa Ciardi
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Piazzale Aldo Moro, 5, 00185 Rome, Italy.
| | - Riccardo De Santis
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Piazzale Aldo Moro, 5, 00185 Rome, Italy; Defence Institute for Biomedical Sciences, 00184 Rome, Italy.
| | - Florigio Lista
- Defence Institute for Biomedical Sciences, 00184 Rome, Italy.
| | - Guido Antonelli
- Department of Molecular Medicine Sapienza University of Rome, Viale dell'Università, 33, 000185 Rome, Italy.
| | - Ombretta Turriziani
- Department of Molecular Medicine Sapienza University of Rome, Viale dell'Università, 33, 000185 Rome, Italy.
| |
Collapse
|
12
|
Ørbo HS, Bjørlykke KH, Sexton J, Jyssum I, Tveter AT, Christensen IE, Mjaaland S, Kvien TK, Grødeland G, Kro GB, Jahnsen J, Haavardsholm EA, Munthe LA, Provan SA, Vaage JT, Goll GL, Jørgensen KK, Syversen SW. Incidence and outcome of COVID-19 following vaccine and hybrid immunity in patients on immunosuppressive therapy: identification of protective post-immunisation anti-RBD antibody levels in a prospective cohort study. RMD Open 2024; 10:e003545. [PMID: 38599653 PMCID: PMC11015197 DOI: 10.1136/rmdopen-2023-003545] [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: 07/28/2023] [Accepted: 02/15/2024] [Indexed: 04/12/2024] Open
Abstract
OBJECTIVES To assess incidence, severity and predictors of COVID-19, including protective post-vaccination levels of antibodies to the receptor-binding domain of SARS-CoV-2 spike protein (anti-RBD), informing further vaccine strategies for patients with immune-mediated inflammatory diseases (IMIDs) on immunosuppressive medication. METHODS IMIDs on immunosuppressives and healthy controls (HC) receiving SARS-CoV-2 vaccines were included in this prospective observational study. COVID-19 and outcome were registered and anti-RBD antibodies measured 2-5 weeks post-immunisation. RESULTS Between 15 February 2021 and 15 February 2023, 1729 IMIDs and 350 HC provided blood samples and self-reported COVID-19. The incidence of COVID-19 was 66% in patients and 67% in HC, with re-infection occurring in 12% of patients. Severe COVID-19 was recorded in 22 (2%) patients and no HC. No COVID-19-related deaths occurred. Vaccine-induced immunity gave higher risk of COVID-19 (HR 5.89 (95% CI 4.45 to 7.80)) than hybrid immunity. Post-immunisation anti-RBD levels <6000 binding antibody units/mL were associated with an increased risk of COVID-19 following three (HR 1.37 (95% CI 1.08 to 1.74)) and four doses (HR 1.28 (95% CI 1.02 to 1.62)), and of COVID-19 re-infection (HR 4.47 (95% CI 1.87 to 10.67)). CONCLUSION Vaccinated patients with IMID have a low risk of severe COVID-19. Hybrid immunity lowers the risk of infection. High post-immunisation anti-RBD levels protect against COVID-19. These results suggest that knowledge on COVID-19 history, and assessment of antibody levels post-immunisation can help individualise vaccination programme series in high-risk individuals. TRIAL REGISTRATION NUMBER NCT04798625.
Collapse
Affiliation(s)
- Hilde S Ørbo
- Center for treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Kristin H Bjørlykke
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Gastroenterology, Akershus University Hospital, Lørenskog, Norway
| | - Joseph Sexton
- Center for treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway
| | - Ingrid Jyssum
- Center for treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Anne T Tveter
- Center for treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway
| | - Ingrid E Christensen
- Center for treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Siri Mjaaland
- Division of Infection Control, Section for Immunology, Norwegian Institute of Public Health, Oslo, Norway
| | - Tore K Kvien
- Center for treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Gunnveig Grødeland
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Immunology, Oslo University Hospital, Oslo, Norway
| | - Grete B Kro
- Department of Microbiology, Oslo University Hospital, Oslo, Norway
| | - Jørgen Jahnsen
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Gastroenterology, Akershus University Hospital, Lørenskog, Norway
| | - Espen A Haavardsholm
- Center for treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Ludvig A Munthe
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- KG Jebsen Centre for B cell Malignancies, University of Oslo, Oslo, Norway
| | - Sella A Provan
- Center for treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway
- Section for Public Health, Inland Norway University of Applied Sciences, Elverum, Norway
| | - John T Vaage
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Immunology, Oslo University Hospital, Oslo, Norway
| | - Guro Løvik Goll
- Center for treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway
- Institute of Health and Society, University of Oslo, Oslo, Norway
| | | | - Silje Watterdal Syversen
- Center for treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway
- Institute of Health and Society, University of Oslo, Oslo, Norway
| |
Collapse
|
13
|
Sehgal ANA, Safran J, Kratzer B, Gattinger P, Stieger RB, Musiejovsky L, Trapin D, Ettel P, Körmöczi U, Rottal A, Borochova K, Dorofeeva Y, Tulaeva I, Weber M, Grabmeier-Pfistershammer K, Perkmann T, Wiedermann U, Valenta R, Pickl WF. Flow Cytometry-Based Measurement of Antibodies Specific for Cell Surface-Expressed Folded SARS-CoV-2 Receptor-Binding Domains. Vaccines (Basel) 2024; 12:377. [PMID: 38675759 PMCID: PMC11053794 DOI: 10.3390/vaccines12040377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 03/20/2024] [Accepted: 03/25/2024] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND COVID-19, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has now become endemic and is currently one of the important respiratory virus infections regularly affecting mankind. The assessment of immunity against SARS-CoV-2 and its variants is important for guiding active and passive immunization and SARS-CoV-2-specific treatment strategies. METHODS We here devised a novel flow cytometry-based diagnostic platform for the assessment of immunity against cell-bound virus antigens. This platform is based on a collection of HEK-293T cell lines which, as exemplified in our study, stably express the receptor-binding domains (RBDs) of the SARS-CoV-2 S-proteins of eight major SARS-CoV-2 variants, ranging from Wuhan-Hu-1 to Omicron. RESULTS RBD-expressing cell lines stably display comparable levels of RBD on the surface of HEK-293T cells, as shown with anti-FLAG-tag antibodies directed against a N-terminally introduced 3x-FLAG sequence while the functionality of RBD was proven by ACE2 binding. We exemplify the usefulness and specificity of the cell-based test by direct binding of IgG and IgA antibodies of SARS-CoV-2-exposed and/or vaccinated individuals in which the assay shows a wide linear performance range both at very low and very high serum antibody concentrations. In another application, i.e., antibody adsorption studies, the test proved to be a powerful tool for measuring the ratios of individual variant-specific antibodies. CONCLUSION We have established a toolbox for measuring SARS-CoV-2-specific immunity against cell-bound virus antigens, which may be considered as an important addition to the armamentarium of SARS-CoV-2-specific diagnostic tests, allowing flexible and quick adaptation to new variants of concern.
Collapse
Affiliation(s)
- Al Nasar Ahmed Sehgal
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria (J.S.); (R.B.S.)
| | - Jera Safran
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria (J.S.); (R.B.S.)
| | - Bernhard Kratzer
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria (J.S.); (R.B.S.)
| | - Pia Gattinger
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Robert B. Stieger
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria (J.S.); (R.B.S.)
| | - Laszlo Musiejovsky
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria (J.S.); (R.B.S.)
| | - Doris Trapin
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria (J.S.); (R.B.S.)
| | - Paul Ettel
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria (J.S.); (R.B.S.)
| | - Ulrike Körmöczi
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria (J.S.); (R.B.S.)
| | - Arno Rottal
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria (J.S.); (R.B.S.)
| | - Kristina Borochova
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Yulia Dorofeeva
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Inna Tulaeva
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
- Laboratory for Immunopathology, Department of Clinical Immunology and Allergology, Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | - Milena Weber
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Katharina Grabmeier-Pfistershammer
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria (J.S.); (R.B.S.)
| | - Thomas Perkmann
- Department of Laboratory Medicine, Medical University of Vienna, 1090 Vienna, Austria;
| | - Ursula Wiedermann
- Institute of Specific Prophylaxis and Tropical Medicine, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Rudolf Valenta
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
- Laboratory for Immunopathology, Department of Clinical Immunology and Allergology, Sechenov First Moscow State Medical University, 119991 Moscow, Russia
- NRC Institute of Immunology FMBA of Russia, 115478 Moscow, Russia
- Karl Landsteiner University of Health Sciences, 3500 Krems, Austria
| | - Winfried F. Pickl
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria (J.S.); (R.B.S.)
- Karl Landsteiner University of Health Sciences, 3500 Krems, Austria
| |
Collapse
|
14
|
Kared H, Jyssum I, Alirezaylavasani A, Egner IM, The Tran T, Tietze L, Lund KP, Tveter AT, Provan SA, Ørbo H, Haavardsholm EA, Vaage JT, Jørgensen K, Syversen SW, Lund-Johansen F, Goll GL, Munthe LA. Dynamics of SARS-CoV-2 immunity after vaccination and breakthrough infection in rituximab-treated rheumatoid arthritis patients: a prospective cohort study. Front Immunol 2024; 15:1296273. [PMID: 38455062 PMCID: PMC10917913 DOI: 10.3389/fimmu.2024.1296273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 02/05/2024] [Indexed: 03/09/2024] Open
Abstract
Background SARS-CoV-2 vaccination in rheumatoid arthritis (RA) patients treated with B cell-depleting drugs induced limited seroconversion but robust cellular response. We aimed to document specific T and B cell immunity in response to vaccine booster doses and breakthrough infection (BTI). Methods We included 76 RA patients treated with rituximab who received up to four SARS-CoV-2 vaccine doses or three doses plus BTI, in addition to vaccinated healthy donors (HD) and control patients treated with tumor necrosis factor inhibitor (TNFi). We quantified anti-SARS-CoV-2 receptor-binding domain (RBD) Spike IgG, anti-nucleocapsid (NC) IgG, 92 circulating inflammatory proteins, Spike-binding B cells, and Spike-specific T cells along with comprehensive high-dimensional phenotyping and functional assays. Findings The time since the last rituximab infusion, persistent inflammation, and age were associated with the anti-SARS-CoV-2 RBD IgG seroconversion. The vaccine-elicited serological response was accompanied by an incomplete induction of peripheral Spike-specific memory B cells but occurred independently of T cell responses. Vaccine- and BTI-elicited cellular immunity was similar between RA and HD ex vivo in terms of frequency or phenotype of Spike-specific cytotoxic T cells and in vitro in terms of the functionality and differentiation profile of Spike-specific T cells. Interpretation SARS-CoV-2 vaccination in RA can induce persistent effector T-cell responses that are reactivated by BTI. Paused rituximab medication allowed serological responses after a booster dose (D4), especially in RA with lower inflammation, enabling efficient humoral and cellular immunity after BTI, and contributed overall to the development of potential durable immunity.
Collapse
Affiliation(s)
- Hassen Kared
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- KG Jebsen Centre for B cell Malignancies, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Precision Immunotherapy Alliance, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Ingrid Jyssum
- Center for Treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Amin Alirezaylavasani
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- KG Jebsen Centre for B cell Malignancies, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Precision Immunotherapy Alliance, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Ingrid M. Egner
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- KG Jebsen Centre for B cell Malignancies, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Precision Immunotherapy Alliance, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Trung The Tran
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- Precision Immunotherapy Alliance, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- ImmunoLingo Convergence Center, University of Oslo, Oslo, Norway
| | - Lisa Tietze
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- Precision Immunotherapy Alliance, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- ImmunoLingo Convergence Center, University of Oslo, Oslo, Norway
| | - Katrine Persgård Lund
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- KG Jebsen Centre for B cell Malignancies, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Precision Immunotherapy Alliance, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Anne Therese Tveter
- Center for Treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway
| | - Sella A. Provan
- Center for Treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway
| | - Hilde Ørbo
- Center for Treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway
| | - Espen A. Haavardsholm
- Center for Treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway
| | - John Torgils Vaage
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- Center for Treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway
| | - Kristin Jørgensen
- Department of Gastroenterology, Akershus University Hospital, Lørenskog, Norway
| | - Silje Watterdal Syversen
- Center for Treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway
| | - Fridtjof Lund-Johansen
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- Precision Immunotherapy Alliance, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- ImmunoLingo Convergence Center, University of Oslo, Oslo, Norway
| | - Guro Løvik Goll
- Center for Treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway
| | - Ludvig A. Munthe
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- KG Jebsen Centre for B cell Malignancies, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
15
|
Gainullin M, Federico L, Røkke Osen J, Chaban V, Kared H, Alirezaylavasani A, Lund-Johansen F, Wildendahl G, Jacobsen JA, Sarwar Anjum H, Stratford R, Tennøe S, Malone B, Clancy T, Vaage JT, Henriksen K, Wüsthoff L, Munthe LA. People who use drugs show no increase in pre-existing T-cell cross-reactivity toward SARS-CoV-2 but develop a normal polyfunctional T-cell response after standard mRNA vaccination. Front Immunol 2024; 14:1235210. [PMID: 38299149 PMCID: PMC10827924 DOI: 10.3389/fimmu.2023.1235210] [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: 06/05/2023] [Accepted: 12/27/2023] [Indexed: 02/02/2024] Open
Abstract
People who use drugs (PWUD) are at a high risk of contracting and developing severe coronavirus disease 2019 (COVID-19) and other infectious diseases due to their lifestyle, comorbidities, and the detrimental effects of opioids on cellular immunity. However, there is limited research on vaccine responses in PWUD, particularly regarding the role that T cells play in the immune response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Here, we show that before vaccination, PWUD did not exhibit an increased frequency of preexisting cross-reactive T cells to SARS-CoV-2 and that, despite the inhibitory effects that opioids have on T-cell immunity, standard vaccination can elicit robust polyfunctional CD4+ and CD8+ T-cell responses that were similar to those found in controls. Our findings indicate that vaccination stimulates an effective immune response in PWUD and highlight targeted vaccination as an essential public health instrument for the control of COVID-19 and other infectious diseases in this group of high-risk patients.
Collapse
Affiliation(s)
- Murat Gainullin
- KG Jebsen Centre for B cell Malignancies, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- NEC OncoImmunity AS, Oslo, Norway
- Department of Immunology, Oslo University Hospital, Oslo, Norway
| | - Lorenzo Federico
- KG Jebsen Centre for B cell Malignancies, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Immunology, Oslo University Hospital, Oslo, Norway
| | - Julie Røkke Osen
- KG Jebsen Centre for B cell Malignancies, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Immunology, Oslo University Hospital, Oslo, Norway
| | - Viktoriia Chaban
- KG Jebsen Centre for B cell Malignancies, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Immunology, Oslo University Hospital, Oslo, Norway
| | - Hassen Kared
- KG Jebsen Centre for B cell Malignancies, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Immunology, Oslo University Hospital, Oslo, Norway
| | - Amin Alirezaylavasani
- KG Jebsen Centre for B cell Malignancies, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Immunology, Oslo University Hospital, Oslo, Norway
| | - Fridtjof Lund-Johansen
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- ImmunoLingo Convergence Center, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | | | | | | | | | | | | | | | - John T. Vaage
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Kathleen Henriksen
- Agency for Social and Welfare Services, Oslo, Norway
- Student Health Services, University of Oslo, Oslo, Norway
| | - Linda Wüsthoff
- Unit for Clinical Research on Addictions, Oslo University Hospital, Oslo, Norway
- Norwegian Centre for Addiction Reasearch, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Ludvig A. Munthe
- KG Jebsen Centre for B cell Malignancies, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Immunology, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
16
|
Su WY, Ho TS, Tsai TC, Du PX, Tsai PS, Keskin BB, Shizen MA, Lin PC, Lin WH, Shih HC, Syu GD. Developing magnetic barcode bead fluorescence assay for high throughput analyzing humoral responses against multiple SARS-CoV-2 variants. Biosens Bioelectron 2023; 241:115709. [PMID: 37776623 DOI: 10.1016/j.bios.2023.115709] [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: 07/04/2023] [Revised: 09/20/2023] [Accepted: 09/21/2023] [Indexed: 10/02/2023]
Abstract
The continuous mutation of SARS-CoV-2 highlights the need for rapid, cost-effective, and high-throughput detection methods. To better analyze the antibody levels against SARS-CoV-2 and its variants in vaccinated or infected subjects, we developed a multiplex detection named Barcode Bead Fluorescence (BBF) assay. These barcode beads were magnetic, characterized by 2-dimensional edges, highly multiplexed, and could be decrypted with visible light. We conjugated 12 magnetic barcode beads with corresponding nine spike proteins (wild-type, alpha, beta, gamma, delta, and current omicrons), two nucleocapsid proteins (wild-type and omicron), and one negative control. First, the conjugated beads underwent serial quality controls via fluorescence labeling, e.g., reproducibility (R square = 0.99) and detection limits (119 pg via anti-spike antibody). Next, we investigated serums from vaccinated subjects and COVID-19 patients for clinical applications. A significant reduction of antibody levels against all variant beads was observed in both vaccinated and COVID-19 studies. Subjects with two doses of mRNA-1273 exhibited the highest level of antibodies against all spike variants compared to two doses of AZD1222 and unvaccinated. We also found that COVID-19 patients showed higher antibody levels against spike beads from wild-type, alpha, beta, and delta. Finally, the nucleocapsid beads served as markers to distinguish infections from vaccinated subjects. Overall, this study developed the BBF assay for analyzing humoral immune responses, which has the advantages of robustness, automation, scalability, and cost-effectiveness.
Collapse
Affiliation(s)
- Wen-Yu Su
- Department of Biotechnology and Bioindustry Sciences, National Cheng Kung University, Tainan, 701, Taiwan, ROC
| | - Tzong-Shiann Ho
- Department of Pediatrics, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, 701, Taiwan, ROC; Center of Infectious Disease and Signaling Research, National Cheng Kung University, Tainan, 701, Taiwan, ROC; Department of Pediatrics, Tainan Hospital, Ministry of Health and Welfare, Tainan, 700, Taiwan, ROC; Department of Pediatrics, National Cheng Kung University Hospital Dou-Liou Branch, College of Medicine, National Cheng Kung University, Yunlin, 640, Taiwan, ROC
| | - Tien-Chun Tsai
- Core Facility Center, National Cheng Kung University, Tainan, 701, Taiwan, ROC
| | - Pin-Xian Du
- Department of Biotechnology and Bioindustry Sciences, National Cheng Kung University, Tainan, 701, Taiwan, ROC
| | - Pei-Shan Tsai
- Department of Biotechnology and Bioindustry Sciences, National Cheng Kung University, Tainan, 701, Taiwan, ROC
| | - Batuhan Birol Keskin
- Department of Biotechnology and Bioindustry Sciences, National Cheng Kung University, Tainan, 701, Taiwan, ROC
| | - Maulida Azizza Shizen
- Department of Biotechnology and Bioindustry Sciences, National Cheng Kung University, Tainan, 701, Taiwan, ROC
| | - Pei-Chun Lin
- Department of Biotechnology and Bioindustry Sciences, National Cheng Kung University, Tainan, 701, Taiwan, ROC
| | - Wei-Hsun Lin
- Department of Biotechnology and Bioindustry Sciences, National Cheng Kung University, Tainan, 701, Taiwan, ROC
| | - Hsi-Chang Shih
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Guan-Da Syu
- Department of Biotechnology and Bioindustry Sciences, National Cheng Kung University, Tainan, 701, Taiwan, ROC; International Center for Wound Repair and Regeneration, National Cheng Kung University, Tainan, 701, Taiwan, ROC; Medical Device Innovation Center, National Cheng Kung University, Tainan, 701, Taiwan, ROC.
| |
Collapse
|
17
|
Hueting D, Schriever K, Sun R, Vlachiotis S, Zuo F, Du L, Persson H, Hofström C, Ohlin M, Walldén K, Buggert M, Hammarström L, Marcotte H, Pan-Hammarström Q, Andréll J, Syrén PO. Design, structure and plasma binding of ancestral β-CoV scaffold antigens. Nat Commun 2023; 14:6527. [PMID: 37845250 PMCID: PMC10579346 DOI: 10.1038/s41467-023-42200-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 10/03/2023] [Indexed: 10/18/2023] Open
Abstract
We report the application of ancestral sequence reconstruction on coronavirus spike protein, resulting in stable and highly soluble ancestral scaffold antigens (AnSAs). The AnSAs interact with plasma of patients recovered from COVID-19 but do not bind to the human angiotensin-converting enzyme 2 (ACE2) receptor. Cryo-EM analysis of the AnSAs yield high resolution structures (2.6-2.8 Å) indicating a closed pre-fusion conformation in which all three receptor-binding domains (RBDs) are facing downwards. The structures reveal an intricate hydrogen-bonding network mediated by well-resolved loops, both within and across monomers, tethering the N-terminal domain and RBD together. We show that AnSA-5 can induce and boost a broad-spectrum immune response against the wild-type RBD as well as circulating variants of concern in an immune organoid model derived from tonsils. Finally, we highlight how AnSAs are potent scaffolds by replacing the ancestral RBD with the wild-type sequence, which restores ACE2 binding and increases the interaction with convalescent plasma.
Collapse
Affiliation(s)
- David Hueting
- School of Engineering Sciences in Chemistry, Biotechnology and Health, Department of Fibre and Polymer Technology, KTH Royal Institute of Technology, Stockholm, Sweden
- School of Engineering Sciences in Chemistry, Biotechnology and Health, Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Karen Schriever
- School of Engineering Sciences in Chemistry, Biotechnology and Health, Department of Fibre and Polymer Technology, KTH Royal Institute of Technology, Stockholm, Sweden
- School of Engineering Sciences in Chemistry, Biotechnology and Health, Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Rui Sun
- Division of Immunology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Stelios Vlachiotis
- Division of Immunology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Fanglei Zuo
- Division of Immunology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Likun Du
- Division of Immunology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Helena Persson
- School of Engineering Sciences in Chemistry, Biotechnology and Health, Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
- Drug Discovery and Development Platform, Science for Life Laboratory, Solna, Sweden
| | - Camilla Hofström
- School of Engineering Sciences in Chemistry, Biotechnology and Health, Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
- Drug Discovery and Development Platform, Science for Life Laboratory, Solna, Sweden
| | - Mats Ohlin
- Drug Discovery and Development Platform, Science for Life Laboratory, Solna, Sweden
- Department of Immunotechnology, Lund University, Lund, Sweden
| | - Karin Walldén
- Department of Biochemistry and Biophysics, Science for Life Laboratory, Stockholm University, Stockholm, Sweden
| | - Marcus Buggert
- Center for Infectious Disease, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Lennart Hammarström
- Division of Immunology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Harold Marcotte
- Division of Immunology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Qiang Pan-Hammarström
- Division of Immunology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Juni Andréll
- Department of Biochemistry and Biophysics, Science for Life Laboratory, Stockholm University, Stockholm, Sweden.
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden.
| | - Per-Olof Syrén
- School of Engineering Sciences in Chemistry, Biotechnology and Health, Department of Fibre and Polymer Technology, KTH Royal Institute of Technology, Stockholm, Sweden.
- School of Engineering Sciences in Chemistry, Biotechnology and Health, Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden.
| |
Collapse
|
18
|
Åsberg A, Hovd M, Kjellevold SA, Stenehjem AE, Wien TN, Broch LU, Reier-Nielsen M, Qvale TH, Marti HP, Heldal K, Bitter J, Hagelsteen Kvien E, Vaage JT, Lund-Johansen F, Midtvedt K. Humoral Response After 6 or More Successive Doses of SARS-CoV-2 mRNA Vaccines in Kidney Transplant Recipients-Should We Keep Vaccinating? Transplantation 2023; 107:e279-e280. [PMID: 37496122 PMCID: PMC10519284 DOI: 10.1097/tp.0000000000004732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 06/14/2023] [Indexed: 07/28/2023]
Affiliation(s)
- Anders Åsberg
- Department of Transplantation Medicine, Oslo University Hospital-Rikshospitalet, Oslo, Norway
- Norwegian Renal Registry, Oslo University Hospital-Rikshospitalet, Oslo, Norway
- Department of Pharmacy, University of Oslo, Oslo, Norway
| | - Markus Hovd
- Department of Transplantation Medicine, Oslo University Hospital-Rikshospitalet, Oslo, Norway
- Norwegian Renal Registry, Oslo University Hospital-Rikshospitalet, Oslo, Norway
| | | | - Aud-E. Stenehjem
- Department of Nephrology, Oslo University Hospital, Oslo, Norway
| | - Tale Norbye Wien
- Department of Internal Medicine and Department of Medical Research, Bærum Hospital, Vestre Viken Hospital Trust, Norway
| | | | | | - Tor Hatlestad Qvale
- Department of Internal Medicine, Haugesund Hospital, Helse Fonna, Haugesund, Norway
| | - Hans-Peter Marti
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Kristian Heldal
- Department of Transplantation Medicine, Oslo University Hospital-Rikshospitalet, Oslo, Norway
- Institute of Health and Society, University of Oslo, Norway
| | - Jan Bitter
- Department for Internal Medicine, Sørlandet Sykehus Kristiansand Hospital Trust, Kristiandsand, Norway
| | | | | | | | - Karsten Midtvedt
- Department of Transplantation Medicine, Oslo University Hospital-Rikshospitalet, Oslo, Norway
| |
Collapse
|
19
|
Woelfel S, Dütschler J, König M, Dulovic A, Graf N, Junker D, Oikonomou V, Krieger C, Truniger S, Franke A, Eckhold A, Forsch K, Koller S, Wyss J, Krupka N, Oberholzer M, Frei N, Geissler N, Schaub P, Albrich WC, Friedrich M, Schneiderhan-Marra N, Misselwitz B, Korte W, Bürgi JJ, Brand S. STAR SIGN study: Evaluation of COVID-19 vaccine efficacy against the SARS-CoV-2 variants BQ.1.1 and XBB.1.5 in patients with inflammatory bowel disease. Aliment Pharmacol Ther 2023; 58:678-691. [PMID: 37571863 DOI: 10.1111/apt.17661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 07/06/2023] [Accepted: 07/26/2023] [Indexed: 08/13/2023]
Abstract
BACKGROUND Vaccine-elicited immune responses are impaired in patients with inflammatory bowel disease (IBD) treated with anti-TNF biologics. AIMS To assess vaccination efficacy against the novel omicron sublineages BQ.1.1 and XBB.1.5 in immunosuppressed patients with IBD. METHODS This prospective multicentre case-control study included 98 biologic-treated patients with IBD and 48 healthy controls. Anti-spike IgG concentrations and surrogate neutralisation against SARS-CoV-2 wild-type, BA.1, BA.5, BQ.1.1, and XBB.1.5 were measured at two different time points (2-16 weeks and 22-40 weeks) following third dose vaccination. Surrogate neutralisation was based on antibody-mediated blockage of ACE2-spike protein-protein interaction. Primary outcome was surrogate neutralisation against tested SARS-CoV-2 sublineages. Secondary outcomes were proportions of participants with insufficient surrogate neutralisation, impact of breakthrough infection, and correlation of surrogate neutralisation with anti-spike IgG concentration. RESULTS Surrogate neutralisation against all tested sublineages was reduced in patients with IBD who were treated with anti-TNF biologics compared to patients treated with non-anti-TNF biologics and healthy controls (each p ≤ 0.001) at visit 1. Anti-TNF therapy (odds ratio 0.29 [95% CI 0.19-0.46]) and time since vaccination (0.85 [0.72-1.00]) were associated with low, and mRNA-1273 vaccination (1.86 [1.12-3.08]) with high wild-type surrogate neutralisation in a β-regression model. Accordingly, higher proportions of patients treated with anti-TNF biologics had insufficient surrogate neutralisation against omicron sublineages at visit 1 compared to patients treated with non-anti-TNF biologics and healthy controls (each p ≤ 0.015). Surrogate neutralisation against all tested sublineages decreased over time but was increased by breakthrough infection. Anti-spike IgG concentrations correlated with surrogate neutralisation. CONCLUSIONS Patients with IBD who are treated with anti-TNF biologics show impaired neutralisation against novel omicron sublineages BQ.1.1 and XBB.1.5 and may benefit from prioritisation for future variant-adapted vaccines.
Collapse
Affiliation(s)
- Simon Woelfel
- Max von Pettenkofer Institute of Hygiene and Medical Microbiology, Faculty of Medicine, Ludwig Maximilian University of Munich (LMU Munich), Munich, Germany
- Department of Gastroenterology and Hepatology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
| | - Joel Dütschler
- Department of Gastroenterology and Hepatology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
- Outpatient Clinic, Ambulatory Services Rorschach, Rorschach, Switzerland
| | - Marius König
- Department of Gastroenterology and Hepatology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
| | - Alex Dulovic
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
| | - Nicole Graf
- Clinical Trials Unit, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
| | - Daniel Junker
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
| | - Vasileios Oikonomou
- Department of Visceral Surgery and Medicine, Inselspital Bern University Hospital, University of Bern, Bern, Switzerland
| | - Claudia Krieger
- Department of Gastroenterology and Hepatology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
| | - Samuel Truniger
- Department of Gastroenterology and Hepatology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
- Outpatient Clinic, Ambulatory Services Rorschach, Rorschach, Switzerland
| | - Annett Franke
- Department of Gastroenterology and Hepatology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
- Outpatient Clinic, Ambulatory Services Rorschach, Rorschach, Switzerland
| | - Annika Eckhold
- Department of Gastroenterology and Hepatology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
| | - Kristina Forsch
- Department of Gastroenterology and Hepatology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
| | - Seraina Koller
- Department of Gastroenterology and Hepatology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
| | - Jacqueline Wyss
- Department of Visceral Surgery and Medicine, Inselspital Bern University Hospital, University of Bern, Bern, Switzerland
| | - Niklas Krupka
- Department of Visceral Surgery and Medicine, Inselspital Bern University Hospital, University of Bern, Bern, Switzerland
| | | | - Nicola Frei
- Department of Gastroenterology and Hepatology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
| | - Nora Geissler
- Department of Gastroenterology and Hepatology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
| | - Peter Schaub
- Department of Gastroenterology and Hepatology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
| | - Werner C Albrich
- Division of Infectious Diseases & Hospital Epidemiology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
| | - Matthias Friedrich
- Translational Gastroenterology Unit, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | | | - Benjamin Misselwitz
- Department of Visceral Surgery and Medicine, Inselspital Bern University Hospital, University of Bern, Bern, Switzerland
| | | | | | - Stephan Brand
- Department of Gastroenterology and Hepatology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
| |
Collapse
|
20
|
Clairon Q, Prague M, Planas D, Bruel T, Hocqueloux L, Prazuck T, Schwartz O, Thiébaut R, Guedj J. Modeling the kinetics of the neutralizing antibody response against SARS-CoV-2 variants after several administrations of Bnt162b2. PLoS Comput Biol 2023; 19:e1011282. [PMID: 37549192 PMCID: PMC10434962 DOI: 10.1371/journal.pcbi.1011282] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 08/17/2023] [Accepted: 06/20/2023] [Indexed: 08/09/2023] Open
Abstract
Because SARS-CoV-2 constantly mutates to escape from the immune response, there is a reduction of neutralizing capacity of antibodies initially targeting the historical strain against emerging Variants of Concern (VoC)s. That is why the measure of the protection conferred by vaccination cannot solely rely on the antibody levels, but also requires to measure their neutralization capacity. Here we used a mathematical model to follow the humoral response in 26 individuals that received up to three vaccination doses of Bnt162b2 vaccine, and for whom both anti-S IgG and neutralization capacity was measured longitudinally against all main VoCs. Our model could identify two independent mechanisms that led to a marked increase in measured humoral response over the successive vaccination doses. In addition to the already known increase in IgG levels after each dose, we identified that the neutralization capacity was significantly increased after the third vaccine administration against all VoCs, despite large inter-individual variability. Consequently, the model projects that the mean duration of detectable neutralizing capacity against non-Omicron VoC is between 348 days (Beta variant, 95% Prediction Intervals PI [307; 389]) and 587 days (Alpha variant, 95% PI [537; 636]). Despite the low neutralization levels after three doses, the mean duration of detectable neutralizing capacity against Omicron variants varies between 173 days (BA.5 variant, 95% PI [142; 200]) and 256 days (BA.1 variant, 95% PI [227; 286]). Our model shows the benefit of incorporating the neutralization capacity in the follow-up of patients to better inform on their level of protection against the different SARS-CoV-2 variants. Trial registration: This clinical trial is registered with ClinicalTrials.gov, Trial IDs NCT04750720 and NCT05315583.
Collapse
Affiliation(s)
- Quentin Clairon
- Université de Bordeaux, Inria Bordeaux Sud-Ouest, Bordeaux, France
- Inserm, Bordeaux Population Health Research Center, SISTM Team, UMR1219, Bordeaux, France
- Vaccine Research Institute, Créteil, France
| | - Mélanie Prague
- Université de Bordeaux, Inria Bordeaux Sud-Ouest, Bordeaux, France
- Inserm, Bordeaux Population Health Research Center, SISTM Team, UMR1219, Bordeaux, France
- Vaccine Research Institute, Créteil, France
| | - Delphine Planas
- Vaccine Research Institute, Créteil, France
- Virus and Immunity Unit, Institut Pasteur, Université de Paris Cité, CNRS UMR3569, Paris, France
| | - Timothée Bruel
- Vaccine Research Institute, Créteil, France
- Virus and Immunity Unit, Institut Pasteur, Université de Paris Cité, CNRS UMR3569, Paris, France
| | - Laurent Hocqueloux
- Service des Maladies Infectieuses et Tropicales, Centre Hospitalier Régional, Orléans, France
| | - Thierry Prazuck
- Service des Maladies Infectieuses et Tropicales, Centre Hospitalier Régional, Orléans, France
| | - Olivier Schwartz
- Vaccine Research Institute, Créteil, France
- Virus and Immunity Unit, Institut Pasteur, Université de Paris Cité, CNRS UMR3569, Paris, France
| | - Rodolphe Thiébaut
- Université de Bordeaux, Inria Bordeaux Sud-Ouest, Bordeaux, France
- Inserm, Bordeaux Population Health Research Center, SISTM Team, UMR1219, Bordeaux, France
- Vaccine Research Institute, Créteil, France
| | | |
Collapse
|
21
|
Federico L, Tvedt THA, Gainullin M, Osen JR, Chaban V, Lund KP, Tietze L, Tran TT, Lund-Johansen F, Kared H, Lind A, Vaage JT, Stratford R, Tennøe S, Malone B, Clancy T, Myhre AEL, Gedde-Dahl T, Munthe LA. Robust spike-specific CD4 + and CD8 + T cell responses in SARS-CoV-2 vaccinated hematopoietic cell transplantation recipients: a prospective, cohort study. Front Immunol 2023; 14:1210899. [PMID: 37503339 PMCID: PMC10369799 DOI: 10.3389/fimmu.2023.1210899] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 06/13/2023] [Indexed: 07/29/2023] Open
Abstract
Poor overall survival of hematopoietic stem cell transplantation (HSCT) recipients who developed COVID-19 underlies the importance of SARS-CoV-2 vaccination. Previous studies of vaccine efficacy have reported weak humoral responses but conflicting results on T cell immunity. Here, we have examined the relationship between humoral and T cell response in 48 HSCT recipients who received two doses of Moderna's mRNA-1273 or Pfizer/BioNTech's BNT162b2 vaccines. Nearly all HSCT patients had robust T cell immunity regardless of protective humoral responses, with 18/48 (37%, IQR 8.679-5601 BAU/mL) displaying protective IgG anti-receptor binding domain (RBD) levels (>2000 BAU/mL). Flow cytometry analysis of activation induced markers (AIMs) revealed that 90% and 74% of HSCT patients showed reactivity towards immunodominant spike peptides in CD8+ and CD4+ T cells, respectively. The response rate increased to 90% for CD4+ T cells as well when we challenged the cells with a complete set of overlapping peptides spanning the entire spike protein. T cell response was detectable as early as 3 months after transplant, but only CD4+ T cell reactivity correlated with IgG anti-RBD level and time after transplantation. Boosting increased seroconversion rate, while only one patient developed COVID-19 requiring hospitalization. Our data suggest that HSCT recipients with poor serological responses were protected from severe COVID-19 by vaccine-induced T cell responses.
Collapse
Affiliation(s)
- Lorenzo Federico
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- KG Jebsen Centre for B Cell Malignancies, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | | | - Murat Gainullin
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- KG Jebsen Centre for B Cell Malignancies, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Julie Røkke Osen
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- KG Jebsen Centre for B Cell Malignancies, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Viktoriia Chaban
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- KG Jebsen Centre for B Cell Malignancies, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Katrine Persgård Lund
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- KG Jebsen Centre for B Cell Malignancies, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Lisa Tietze
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- ImmunoLingo Convergence Center, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Trung The Tran
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- ImmunoLingo Convergence Center, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Fridtjof Lund-Johansen
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- ImmunoLingo Convergence Center, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Hassen Kared
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- KG Jebsen Centre for B Cell Malignancies, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Andreas Lind
- Department of Microbiology, Oslo University Hospital, Oslo, Norway
| | - John Torgils Vaage
- ImmunoLingo Convergence Center, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | | | | | | | | | - Anders Eivind Leren Myhre
- Department of Haematology, Oslo University Hospital, Oslo, Norway
- ImmunoLingo Convergence Center, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | | | - Ludvig André Munthe
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- KG Jebsen Centre for B Cell Malignancies, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
22
|
Long J, Soni M, Muranski P, Miller MJ, Conry-Cantilena C, De Giorgi V. Case Report: Kinetics and durability of humoral and cellular response of SARS-CoV-2 messenger RNA vaccine in a lung and kidney transplant recipient. Front Immunol 2023; 14:1207638. [PMID: 37465681 PMCID: PMC10350526 DOI: 10.3389/fimmu.2023.1207638] [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: 04/17/2023] [Accepted: 06/09/2023] [Indexed: 07/20/2023] Open
Abstract
We present a case report of a 63-year-old female health care worker who is 15 years status post double lung transplant and six years status post living related donor kidney transplant who is healthy on a chronic immunosuppression regimen including prednisone, mycophenolate, and tacrolimus who received the SARS-CoV-2 mRNA vaccine (Pfizer-BioNTech BNT162b2) primary series and had poor initial humoral response to the COVID-19 mRNA vaccine, then demonstrated a robust, sustained immune response against S1 and S2 antigens for over seven months after receiving the recommended vaccine doses, including booster dose, without developing COVID-19 or other serious adverse events. Her immune response to vaccination indicates effective formation of anti-spike T cell memory despite chronic immunosuppression. This case report provides a comprehensive characterization of her immune response to this SARS-CoV-2 vaccination series. As vaccine effectiveness data is updated, and as better understanding of immune response including hybrid immunity emerges, these findings may reassure that recipients of SOTs may be capable of durable immune responses to emerging variants of SARS-CoV-2.
Collapse
Affiliation(s)
- James Long
- Infectious Diseases Section, Department of Transfusion Medicine, National Institutes of Health Clinical Center, Bethesda, MD, United States
| | - Mithil Soni
- Columbia Center for Translational Immunology, Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, United States
| | - Pawel Muranski
- Columbia Center for Translational Immunology, Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, United States
| | - Maureen J. Miller
- Infectious Diseases Section, Department of Transfusion Medicine, National Institutes of Health Clinical Center, Bethesda, MD, United States
| | - Cathleen Conry-Cantilena
- Infectious Diseases Section, Department of Transfusion Medicine, National Institutes of Health Clinical Center, Bethesda, MD, United States
| | - Valeria De Giorgi
- Infectious Diseases Section, Department of Transfusion Medicine, National Institutes of Health Clinical Center, Bethesda, MD, United States
| |
Collapse
|
23
|
Hovd M, Åsberg A, Munthe LA, Heldal K, Reisæter AV, Vaage JT, Lund-Johansen F, Midtvedt K. Humoral vaccine response and breakthrough infections in kidney transplant recipients during the COVID-19 pandemic: a nationwide cohort study. EClinicalMedicine 2023; 60:102035. [PMID: 37362086 PMCID: PMC10242148 DOI: 10.1016/j.eclinm.2023.102035] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 05/18/2023] [Accepted: 05/19/2023] [Indexed: 06/28/2023] Open
Abstract
Background Kidney transplant recipients (KTRs) experienced reduced SARS-CoV-2 vaccine response and were at increased risk of severe COVID-19. It is unknown if level of vaccine induced anti-receptor binding domain IgG (anti-RBD IgG) correlates with protection from and survival following COVID-19. We aimed to evaluate the effect of vaccine response on risk of breakthrough infections (BTI) and COVID-19 death in KTRs. Methods We performed a nationwide study, examining the competing risk of SARS-CoV-2 infection, COVID-19 related/unrelated death, and vaccine efficacy as assessed by level of anti-RBD IgG response 4-10 weeks after each vaccination. The study included all KTR in Norway alive and with a functioning graft on February 20th, 2020, and events after November 11th, 2022 were right-censored. A pre-pandemic reference-cohort from January 1st 2019 to January 1st 2020 was included to evaluate excess mortality. The study was conducted at Oslo University Hospital, Rikshospitalet, Norway. Findings The study included 3607 KTRs (59 [48-70] years) with a functioning graft at February 20th, 2020, who received (median [IQR]) 4 [3-4] vaccines (range 2-6, 99% mRNA). Anti-RBD IgG was measured in 12 701 serum samples provided by 3213 KTRs. Vaccine response was assessed 41 [31-57] days after vaccination. A total of 1090 KTRs were infected with SARS-CoV-2, 1005 (92%) were BTI, and vaccine response did not protect against BTI. The hazard ratio for COVID-19 related death 40 days post-infection was 1.71 (95% CI: 1.14, 2.56) comparing vaccine response levels (≥5 vs. ≥5000 BAU/mL). No excess non-COVID-19 mortality was registered in KTRs surviving SARS-CoV-2 infection compared to a 2019 pre-pandemic reference. Interpretation Our findings suggested that SARS-CoV-2 mRNA vaccine response did not predict protection against infection, but prevention of fatal disease progression in KTRs and greater vaccine response further reduced the risk of COVID-19 death. No excess non-COVID-19 mortality was seen during the pandemic. Funding CEPI and internal funds.
Collapse
Affiliation(s)
- Markus Hovd
- Department of Transplantation Medicine, Oslo University Hospital, Norway
- Department of Pharmacy, University of Oslo, Norway
- The Norwegian Renal Registry, Department of Transplantation Medicine, Oslo University Hospital, Norway
| | - Anders Åsberg
- Department of Transplantation Medicine, Oslo University Hospital, Norway
- Department of Pharmacy, University of Oslo, Norway
- The Norwegian Renal Registry, Department of Transplantation Medicine, Oslo University Hospital, Norway
| | - Ludvig A Munthe
- Institute of Clinical Medicine, University of Oslo, Norway
- KG Jebsen Centre for B Cell Malignancies, Institute of Clinical Medicine, University of Oslo, Norway
| | - Kristian Heldal
- Department of Transplantation Medicine, Oslo University Hospital, Norway
- Institute of Health and Society, University of Oslo, Norway
| | - Anna V Reisæter
- Department of Transplantation Medicine, Oslo University Hospital, Norway
- The Norwegian Renal Registry, Department of Transplantation Medicine, Oslo University Hospital, Norway
| | - John T Vaage
- Institute of Clinical Medicine, University of Oslo, Norway
- Department of Immunology, Oslo University Hospital, Norway
| | - Fridtjof Lund-Johansen
- Department of Immunology, Oslo University Hospital, Norway
- ImmunoLingo Convergence Center, Institute of Clinical Medicine, University of Oslo, Norway
| | - Karsten Midtvedt
- Department of Transplantation Medicine, Oslo University Hospital, Norway
| |
Collapse
|
24
|
Ravussin A, Robertson AH, Wolf AS, Blix K, Kjønstad IF, Solum G, Feiring B, Strand BH, Lund-Johansen F, Munthe LA, Magnus P, Trogstad L, Mjaaland S. Determinants of humoral and cellular immune responses to three doses of mRNA SARS-CoV-2 vaccines in older adults: a longitudinal cohort study. THE LANCET. HEALTHY LONGEVITY 2023; 4:e188-e199. [PMID: 37148891 PMCID: PMC10156136 DOI: 10.1016/s2666-7568(23)00055-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 03/09/2023] [Accepted: 03/09/2023] [Indexed: 05/08/2023] Open
Abstract
BACKGROUND Older age is associated with poorer outcomes to COVID-19 infection. The Norwegian Institute of Public Health established a longitudinal cohort of adults aged 65-80 years to study the effects of the COVID-19 pandemic. Here we describe the characteristics of the cohort in general, and specifically the immune responses at baseline and after primary and booster vaccination in a subset of longitudinal blood samples, and the epidemiological factors affecting these responses. METHODS 4551 participants were recruited, with humoral (n=299) and cellular (n=90) responses measured before vaccination and after two and three vaccine doses. Information on general health, infections, and vaccinations were obtained from questionnaires and national health registries. FINDINGS Half of the participants had a chronic condition. 849 (18·7%) of 4551 were prefrail and 184 (4%) of 4551 were frail. 483 (10·6%) of 4551 had general activity limitations (scored with the Global Activity Limitation Index). After dose two, 295 (98·7%) of 299 participants were seropositive for anti-receptor binding domain IgG, and 210 (100%) of 210 participants after dose three. Spike-specific CD4 and CD8 T cell responses showed high heterogeneity after vaccination and responded to the alpha (B.1.1.7), delta (B.1.617.2), and omicron (B.1.1.529 or BA.1) variants of concern. Cellular responses to seasonal coronaviruses increased after SARS-CoV-2 vaccination. Heterologous prime boosting with mRNA vaccines was associated with the highest antibody (p=0·019) and CD4 T cell responses (p=0·003), and hypertension with lower antibody levels after three doses (p=0·04). INTERPRETATION Most older adults, including those with comorbidities, generated good serological and cellular responses after two vaccine doses. Responses further improved after three doses, particularly after heterologous boosting. Vaccination also generated cross-reactive T cells against variants of concern and seasonal coronaviruses. Frailty was not associated with impaired immune responses, but hypertension might indicate reduced responsiveness to vaccines even after three doses. Individual differences identified through longitudinal sampling enables better prediction of the variability of vaccine responses, which can help guide future policy on the need for subsequent doses and their timing. FUNDING Norwegian Institute of Public Health, Norwegian Ministry of Health, Research Council of Norway, and Coalition for Epidemic Preparedness Innovations.
Collapse
Affiliation(s)
- Anthony Ravussin
- Division of Infection Control, Section for Immunology, Norwegian Institute of Public Health, Oslo, Norway
| | - Anna Hayman Robertson
- Division of Infection Control, Section for Vaccine Epidemiology and Population Studies, Norwegian Institute of Public Health, Oslo, Norway.
| | - Asia-Sophia Wolf
- Division of Infection Control, Section for Immunology, Norwegian Institute of Public Health, Oslo, Norway
| | - Kristine Blix
- Division of Infection Control, Section for Vaccine Epidemiology and Population Studies, Norwegian Institute of Public Health, Oslo, Norway
| | - Ingrid Fadum Kjønstad
- Division of Infection Control, Section for Immunology, Norwegian Institute of Public Health, Oslo, Norway
| | - Guri Solum
- Division of Infection Control, Section for Immunology, Norwegian Institute of Public Health, Oslo, Norway
| | - Berit Feiring
- Division of Infection Control, Section for Vaccine Epidemiology and Population Studies, Norwegian Institute of Public Health, Oslo, Norway
| | - Bjørn Heine Strand
- Division of Mental and Physical Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Fridtjof Lund-Johansen
- Department of Immunology, Oslo University Hospital, Oslo, Norway; ImmunoLingo Convergence Center, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Ludvig A Munthe
- Department of Immunology, Oslo University Hospital, Oslo, Norway; KG Jebsen Centre for B cell Malignancies, University of Oslo, Oslo, Norway
| | - Per Magnus
- Centre for Fertility and Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Lill Trogstad
- Division of Infection Control, Section for Vaccine Epidemiology and Population Studies, Norwegian Institute of Public Health, Oslo, Norway
| | - Siri Mjaaland
- Division of Infection Control, Section for Immunology, Norwegian Institute of Public Health, Oslo, Norway
| |
Collapse
|
25
|
Puccini L, Fantini M, Biagetti C, Angelini R, Dirani G, Grumiro L, Schiavone P, Sparacino M, Semprini S, Sambri V, Cricca M. Kinetics of dried blood spot-measured anti-SARS-CoV2 Spike IgG in mRNA-vaccinated healthcare workers. Front Microbiol 2023; 14:1130677. [PMID: 36937271 PMCID: PMC10014594 DOI: 10.3389/fmicb.2023.1130677] [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: 12/23/2022] [Accepted: 01/27/2023] [Indexed: 03/05/2023] Open
Abstract
Introduction One of the major criticisms facing the research community during SARS-CoV2 pandemic was the lack of large-scale, longitudinal data on the efficacy of the SARS-CoV2 mRNA vaccines. Currently, even if COVID-19 antiviral treatments have been authorized by European Medicine Agency, prevention through approved specific vaccines is the best approach available in order to contain the ongoing pandemic. Objectives Here, we studied the antibody kinetic over a one-year period from vaccination with the Pfizer-BioNTech (Pfizer) vaccines and subsequent boosting with either the BioNTech or Moderna (Spikevax) vaccines in a large cohort of 8,071 healthcare workers (HCW). We also described the impact of SARS-CoV2 infection on antibody kinetic over the same period. Methods We assessed the anti SARS-CoV2 Spike IgG antibody kinetic by the high throughput dried blood spot (DBS) collection method and the GSP®/DELFIA® Anti-SARS-CoV2 IgG assay (PerkinElmer®). Results Our data support existing models showing that SARS-CoV2 vaccination elicits strong initial antibodies responses that decline with time but are transitorily increased by administering a vaccine booster. We also showed that using heterologous vaccine/booster combinations a stronger antibody response was elicited than utilizing a booster from the same vaccine manufacturer. Furthermore, by considering the impact of SARS-CoV2 infection occurrence in proximity to the scheduled booster administration, we confirmed that booster dose did not contribute significantly to elicit higher antibody responses. Conclusion DBS sampling in our large population of HCWs was fundamental to collect a large number of specimens and to clarify the effective mRNA vaccine-induced antibody kinetic and the role of both heterologous boosters and SARS-CoV2 infection in modulating antibody responses.
Collapse
Affiliation(s)
- Lucrezia Puccini
- Operative Unit of Microbiology, The Great Romagna Hub Laboratory, Pievesestina, Italy
| | - Michela Fantini
- Health Services Research, Evaluation and Policy Unit, AUSL Romagna, Rimini, Italy
| | - Carlo Biagetti
- Operative Unit of Infectious Disease, Ospedale Infermi, Rimini, Italy
| | | | - Giorgio Dirani
- Operative Unit of Microbiology, The Great Romagna Hub Laboratory, Pievesestina, Italy
| | - Laura Grumiro
- Operative Unit of Microbiology, The Great Romagna Hub Laboratory, Pievesestina, Italy
| | - Pasqua Schiavone
- Operative Unit of Microbiology, The Great Romagna Hub Laboratory, Pievesestina, Italy
| | - Monica Sparacino
- Operative Unit of Microbiology, The Great Romagna Hub Laboratory, Pievesestina, Italy
| | - Simona Semprini
- Operative Unit of Microbiology, The Great Romagna Hub Laboratory, Pievesestina, Italy
| | - Vittorio Sambri
- Operative Unit of Microbiology, The Great Romagna Hub Laboratory, Pievesestina, Italy
- Department of Experimental, Diagnostic and Specialty Medicine-DIMES, University of Bologna, Bologna, Italy
| | - Monica Cricca
- Operative Unit of Microbiology, The Great Romagna Hub Laboratory, Pievesestina, Italy
- Department of Experimental, Diagnostic and Specialty Medicine-DIMES, University of Bologna, Bologna, Italy
| |
Collapse
|