1
|
Inoue K, Tomomasa D, Nakagama Y, Takeuchi H, Tanaka Y, Tanimoto K, Kamiya T, Isoda T, Takagi M, Tanaka K, Yoshifuji K, Miwa Y, Ohnishi H, Okada S, Mori T, Yasuda S, Kido Y, Morio T, Kanegane H. Real-world experience of tixagevimab/cilgavimab prophylaxis in Japanese patients with immunodeficiency. J Infect Chemother 2024:S1341-321X(24)00324-6. [PMID: 39645037 DOI: 10.1016/j.jiac.2024.12.006] [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: 08/28/2024] [Revised: 11/30/2024] [Accepted: 12/04/2024] [Indexed: 12/09/2024]
Abstract
BACKGROUND Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes severe illness and mortality in patients with immunodeficiency. Although vaccination has been recommended, the induction of protective antibodies by immunization, and thus the disease-preventive effect, has proven insufficient in immunodeficient patients, especially in those with predominantly antibody deficiency. A monoclonal antibody combination of tixagevimab and cilgavimab (TIX/CIL) was developed as a pre-exposure prophylaxis (PrEP). In this study, we investigated the post-PrEP increase in antiviral antibody titers and detailed the breakthrough infections that occurred despite PrEP in Japanese immunodeficient patients who had received TIX/CIL. METHODS Blood samples were collected before and after TIX/CIL administration between November 2022 and August 2023. Antibody titers against the S-protein of SARS-CoV-2 were measured to evaluate TIX/CIL-induced protection. Information regarding breakthrough infection, as evidenced by positive antigen and/or PCR tests, was collected. RESULTS A significant increase in the anti-S antibody titer was observed in all 89 immunodeficient patients who had received TIX/CIL. However, 14 (16 %) patients experienced breakthrough SARS-CoV-2 infections, of which one died of respiratory failure. CONCLUSION The shift in the SARS-CoV-2 circulating strain might have reduced the efficacy of TIX/CIL, leading to an increased number of breakthrough infections.
Collapse
Affiliation(s)
- Kento Inoue
- Department of Pediatrics and Developmental Biology, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, 1-5-45 Yushima, Bunkyo-ku, 113-8519, Tokyo, Japan
| | - Dan Tomomasa
- Department of Pediatrics and Developmental Biology, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, 1-5-45 Yushima, Bunkyo-ku, 113-8519, Tokyo, Japan
| | - Yu Nakagama
- Department of Virology and Parasitology, Graduate School of Medicine, Osaka Metropolitan University, 1-4-3 Asahi-machi, Abeno-ku, 545-8585, Osaka, Japan; Research Center for Infectious Disease Sciences, Graduate School of Medicine, Osaka Metropolitan University, 1-4-3 Asahi-machi, Abeno-ku, 545-8585, Osaka, Japan
| | - Hiroaki Takeuchi
- Department of High-risk Infectious Disease Control, Institute of Science Tokyo, 1-5-45 Yushima, Bunkyo-ku, 113-8519, Tokyo, Japan
| | - Yukie Tanaka
- Department of Molecular Microbiology and Immunology, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, 1-5-45 Yushima, Bunkyo-ku, 113-8519, Tokyo, Japan
| | - Kousuke Tanimoto
- Research Core, Institute of Research, Institute of Science Tokyo, 1-5-45 Yushima, Bunkyo-ku, 113-8519, Tokyo, Japan
| | - Takahiro Kamiya
- Department of Clinical Research Center, Institute of Science Tokyo, 1-5-45 Yushima, Bunkyo-ku, 113-8519, Tokyo, Japan
| | - Takeshi Isoda
- Department of Pediatrics and Developmental Biology, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, 1-5-45 Yushima, Bunkyo-ku, 113-8519, Tokyo, Japan
| | - Masatoshi Takagi
- Department of Pediatrics and Developmental Biology, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, 1-5-45 Yushima, Bunkyo-ku, 113-8519, Tokyo, Japan
| | - Keisuke Tanaka
- Department of Hematology, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, 1-5-45 Yushima, Bunkyo-ku, 113-8519, Tokyo, Japan
| | - Kota Yoshifuji
- Department of Hematology, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, 1-5-45 Yushima, Bunkyo-ku, 113-8519, Tokyo, Japan
| | - Yuki Miwa
- Department of Pediatrics, Gifu University Graduate School of Medicine, Yanagido 1-1, Gifu, 501-1194, Gifu, Japan
| | - Hidenori Ohnishi
- Department of Pediatrics, Gifu University Graduate School of Medicine, Yanagido 1-1, Gifu, 501-1194, Gifu, Japan
| | - Satoshi Okada
- Department of Pediatrics, Hiroshima University Graduate School of Biomedical and Health Sciences, Kasumi 1-2-3 Minami-ku, Hiroshima, 734-0037, Hiroshima, Japan
| | - Takehiko Mori
- Department of Hematology, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, 1-5-45 Yushima, Bunkyo-ku, 113-8519, Tokyo, Japan
| | - Shinsuke Yasuda
- Department of Rheumatology, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, 1-5-45 Yushima, Bunkyo-ku, 113-8519, Tokyo, Japan
| | - Yasutoshi Kido
- Department of Virology and Parasitology, Graduate School of Medicine, Osaka Metropolitan University, 1-4-3 Asahi-machi, Abeno-ku, 545-8585, Osaka, Japan; Research Center for Infectious Disease Sciences, Graduate School of Medicine, Osaka Metropolitan University, 1-4-3 Asahi-machi, Abeno-ku, 545-8585, Osaka, Japan
| | - Tomohiro Morio
- Department of Pediatrics and Developmental Biology, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, 1-5-45 Yushima, Bunkyo-ku, 113-8519, Tokyo, Japan
| | - Hirokazu Kanegane
- Department of Child Health and Development, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, 1-5-45 Yushima, Bunkyo-ku, 113-8519, Tokyo, Japan.
| |
Collapse
|
2
|
Lindahl H, Kahn F, Nilsdotter-Augustinsson Å, Fredrikson M, Hedberg P, Killander Möller I, Hansson L, Blixt L, Eketorp Sylvan S, Österborg A, Aleman S, Carlander C, Nyström S, Bergman P. Inborn errors of immunity are associated with increased COVID-19-related hospitalization and intensive care compared to the general population. J Allergy Clin Immunol 2024:S0091-6749(24)01125-4. [PMID: 39447887 DOI: 10.1016/j.jaci.2024.10.013] [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: 07/18/2024] [Revised: 09/14/2024] [Accepted: 10/08/2024] [Indexed: 10/26/2024]
Abstract
BACKGROUND It is thought that patients with inborn errors of immunity (IEI) are more susceptible to severe coronavirus disease 2019 (COVID-19) than the general population, but a quantification of this potential risk is largely missing. OBJECTIVE We assessed the impact of COVID-19 on patients with IEI. METHODS A nationwide cohort study was performed to estimate the relative risk (RR) for hospitalization, intensive care, and death within 30 days after a positive severe acute respiratory syndrome coronavirus 2 test result in an IEI population (n = 2392) compared to the general population (n = 8,270,705) using data from Swedish national registries. Three time periods were studied: the prevaccination period, and the Alpha/Delta and Omicron periods. Adjustment was made for demographics, income, comorbidities, and vaccination status. RESULTS During the prevaccination period, 25.2% of the IEI population was hospitalized, compared to 17.5% and 5.2% during the Alpha/Delta and Omicron periods, respectively. For the 3 time periods, the adjusted RR [95% confidence interval] for hospitalization in the IEI population compared to the general population was 3.1 [2.1-4.2], 3.5 [2.4-4.8], and 4.3 [2.5-6.7], respectively. The respective values for intensive care after COVID-19 were 5.6 [2.6-10.8], 4.7 [1.7-10.1], and 4.7 [1.7-10.1] for the 3 periods. Five patients (0.6%) in the IEI population died within 30 days of a positive PCR test result compared to 18,773 (0.2%) in the general population during the 3 study periods. CONCLUSION Patients with IEI had a 3 to 4 times higher risk for hospitalization and a 5 times higher risk for intensive care during COVID-19 compared to the general population.
Collapse
Affiliation(s)
- Hannes Lindahl
- Department of Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden; Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Fredrik Kahn
- Department of Clinical Sciences Lund, Section for Infection Medicine, Lund University, Lund, Sweden; Department of Infectious Diseases, Skåne University Hospital, Lund, Sweden
| | - Åsa Nilsdotter-Augustinsson
- Division of Inflammation and Infection, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Mats Fredrikson
- Department of Biomedical and Clinical Sciences, Faculty of Medicine, Linköping University, Linköping, Sweden
| | - Pontus Hedberg
- Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | | | - Lotta Hansson
- Department of Hematology, Karolinska University Hospital, Stockholm, Sweden; Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Lisa Blixt
- Department of Hematology, Karolinska University Hospital, Stockholm, Sweden; Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | | | - Anders Österborg
- Department of Hematology, Karolinska University Hospital, Stockholm, Sweden; Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Soo Aleman
- Division of Infectious Diseases and Dermatology, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden; Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Christina Carlander
- Division of Infectious Diseases and Dermatology, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden; Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden; Department of Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Sofia Nyström
- Department of Clinical Immunology and Transfusion Medicine, Linköping University, Linköping, Sweden; Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Peter Bergman
- Department of Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden; Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden; Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
3
|
Neth O, Mahlaoui N, Cunningham-Rundles C. Protecting children and adults with primary antibody deficiencies against common and emergent pathogens and non-infectious complications. Clin Exp Immunol 2024; 218:136-150. [PMID: 39011978 PMCID: PMC11482499 DOI: 10.1093/cei/uxae059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 05/15/2024] [Accepted: 07/05/2024] [Indexed: 07/17/2024] Open
Abstract
Prevention and treatment of infections are primary goals of treatment of children and adults with primary immune deficiencies due to decreased antibody production. Approaches to these goals include immunoglobulin replacement therapy, vaccination, and prophylactic treatment with antimicrobials. In this review, the infectious and non-infectious complications of antibody deficiencies will be discussed along with the limited number of studies that support the effective use of the available therapies and to drive the development of new therapies. Some illustrative case studies will be presented and the outlook for additional controlled clinical trials and potential for therapies driven by the underlying disease genetics will be considered.
Collapse
Affiliation(s)
- Olaf Neth
- Pediatric Infectious Diseases, Rheumatology and Immunology, University Hospital Virgen del Rocío, Institute of Biomedicine of Seville (IBiS), Sevilla, Spain
| | - Nizar Mahlaoui
- French National Reference Center for Primary Immunodeficiencies (CEREDIH), Necker-Enfants Malades University Hospital, Assistance Publique-Hôpitaux de Paris (APHP), Paris, France
- Pediatric Immuno-Hematology and Rheumatology Unit, Necker-Enfants Malades University Hospital, Assistance Publique-Hôpitaux de Paris (APHP), Paris, France
| | - Charlotte Cunningham-Rundles
- Department of Medicine, Icahn School of Medicine at Mount-Sinai, New York, NY, USA
- Department of Pediatrics, Icahn School of Medicine at Mount-Sinai, New York, NY, USA
| |
Collapse
|
4
|
Fink DL, Idilli O, Shields A, Richter A, Burns SO. Prevalence of Anti-Interferon α Auto-Antibodies in Patients with Antibody Deficiency. J Clin Immunol 2024; 44:161. [PMID: 38990439 DOI: 10.1007/s10875-024-01761-2] [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: 04/08/2024] [Accepted: 06/27/2024] [Indexed: 07/12/2024]
Affiliation(s)
- Douglas L Fink
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, London, UK.
- Royal Free London NHS Foundation Trust, London, UK.
| | - Orest Idilli
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, London, UK
| | - Adrian Shields
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Alex Richter
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Siobhan O Burns
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, London, UK
- Royal Free London NHS Foundation Trust, London, UK
| |
Collapse
|
5
|
Chang-Rabley E, van Zelm MC, Ricotta EE, Edwards ESJ. An Overview of the Strategies to Boost SARS-CoV-2-Specific Immunity in People with Inborn Errors of Immunity. Vaccines (Basel) 2024; 12:675. [PMID: 38932404 PMCID: PMC11209597 DOI: 10.3390/vaccines12060675] [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: 05/03/2024] [Revised: 06/09/2024] [Accepted: 06/12/2024] [Indexed: 06/28/2024] Open
Abstract
The SARS-CoV-2 pandemic has heightened concerns about immunological protection, especially for individuals with inborn errors of immunity (IEI). While COVID-19 vaccines elicit strong immune responses in healthy individuals, their effectiveness in IEI patients remains unclear, particularly against new viral variants and vaccine formulations. This uncertainty has led to anxiety, prolonged self-isolation, and repeated vaccinations with uncertain benefits among IEI patients. Despite some level of immune response from vaccination, the definition of protective immunity in IEI individuals is still unknown. Given their susceptibility to severe COVID-19, strategies such as immunoglobulin replacement therapy (IgRT) and monoclonal antibodies have been employed to provide passive immunity, and protection against both current and emerging variants. This review examines the efficacy of COVID-19 vaccines and antibody-based therapies in IEI patients, their capacity to recognize viral variants, and the necessary advances required for the ongoing protection of people with IEIs.
Collapse
Affiliation(s)
- Emma Chang-Rabley
- The Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Menno C. van Zelm
- Allergy and Clinical Immunology Laboratory, Department of Immunology, Central Clinical School, Monash University, Melbourne, VIC 3800, Australia
- The Jeffrey Modell Diagnostic and Research Centre for Primary Immunodeficiencies in Melbourne, Melbourne, VIC 3000, Australia
- Department of Immunology, Erasmus MC, University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Emily E. Ricotta
- The Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
- Department of Preventive Medicine and Biostatistics, Uniform Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Emily S. J. Edwards
- Allergy and Clinical Immunology Laboratory, Department of Immunology, Central Clinical School, Monash University, Melbourne, VIC 3800, Australia
- The Jeffrey Modell Diagnostic and Research Centre for Primary Immunodeficiencies in Melbourne, Melbourne, VIC 3000, Australia
| |
Collapse
|
6
|
Goodyear CS, Patel A, Barnes E, Willicombe M, Siebert S, de Silva TI, Snowden JA, Lim SH, Bowden SJ, Billingham L, Richter A, Carroll M, Carr EJ, Beale R, Rea D, Parry H, Pirrie S, Lim Z, Satsangi J, Dunachie SJ, Cook G, Miller P, Basu N, Gilmour A, Hodgkins AM, Evans L, Hughes A, Longet S, Meacham G, Yong KL, A'Hearne MJ, Koh MBC, Burns SO, Orchard K, Paterson C, McIlroy G, Murray SM, Thomson T, Dimitriadis S, Goulston L, Miller S, Keillor V, Prendecki M, Thomas D, Kirkham A, McInnes IB, Kearns P. Immunogenicity of third dose COVID-19 vaccine strategies in patients who are immunocompromised with suboptimal immunity following two doses (OCTAVE-DUO): an open-label, multicentre, randomised, controlled, phase 3 trial. THE LANCET. RHEUMATOLOGY 2024; 6:e339-e351. [PMID: 38734019 DOI: 10.1016/s2665-9913(24)00065-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 03/06/2024] [Accepted: 03/11/2024] [Indexed: 05/13/2024]
Abstract
BACKGROUND The humoral and T-cell responses to booster COVID-19 vaccine types in multidisease immunocompromised individuals who do not generate adequate antibody responses to two COVID-19 vaccine doses, is not fully understood. The OCTAVE DUO trial aimed to determine the value of third vaccinations in a wide range of patients with primary and secondary immunodeficiencies. METHODS OCTAVE-DUO was a prospective, open-label, multicentre, randomised, controlled, phase 3 trial investigating humoral and T-cell responses in patients who are immunocompromised following a third vaccine dose with BNT162b2 or mRNA-1273, and of NVX-CoV2373 for those with lymphoid malignancies. We recruited patients who were immunocompromised from 11 UK hospitals, aged at least 18 years, with previous sub-optimal responses to two doses of SARS-CoV-2 vaccine. Participants were randomly assigned 1:1 (1:1:1 for those with lymphoid malignancies), stratified by disease, previous vaccination type, and anti-spike antibody response following two doses. Individuals with lived experience of immune susceptibility were involved in the study design and implementation. The primary outcome was vaccine-specific immunity defined by anti-SARS-CoV-2 spike antibodies (Roche Diagnostics UK and Ireland, Burgess Hill, UK) and T-cell responses (Oxford Immunotec, Abingdon, UK) before and 21 days after the third vaccine dose analysed by a modified intention-to-treat analysis. The trial is registered with the ISRCTN registry, ISRCTN 15354495, and the EU Clinical Trials Register, EudraCT 2021-003632-87, and is complete. FINDINGS Between Aug 4, 2021 and Mar 31, 2022, 804 participants across nine disease cohorts were randomly assigned to receive BNT162b2 (n=377), mRNA-1273 (n=374), or NVX-CoV2373 (n=53). 356 (45%) of 789 participants were women, 433 (55%) were men, and 659 (85%) of 775 were White. Anti-SARS-CoV-2 spike antibodies measured 21 days after the third vaccine dose were significantly higher than baseline pre-third dose titres in the modified intention-to-treat analysis (median 1384 arbitrary units [AU]/mL [IQR 4·3-7990·0] compared with median 11·5 AU/mL [0·4-63·1]; p<0·001). Of participants who were baseline low responders, 380 (90%) of 423 increased their antibody concentrations to more than 400 AU/mL. Conversely, 166 (54%) of 308 baseline non-responders had no response after the third dose. Detectable T-cell responses following the third vaccine dose were seen in 494 (80%) of 616 participants. There were 24 serious adverse events (BNT612b2 eight [33%] of 24, mRNA-1273 12 [50%], NVX-CoV2373 four [17%]), two (8%) of which were categorised as vaccine-related. There were seven deaths (1%) during the trial, none of which were vaccine-related. INTERPRETATION A third vaccine dose improved the serological and T-cell response in the majority of patients who are immunocompromised. Individuals with chronic renal disease, lymphoid malignancy, on B-cell targeted therapies, or with no serological response after two vaccine doses are at higher risk of poor response to a third vaccine dose. FUNDING Medical Research Council, Blood Cancer UK.
Collapse
Affiliation(s)
- Carl S Goodyear
- College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, UK
| | - Amit Patel
- Cancer Research UK Clinical Trials Unit, University of Birmingham, Edgbaston, Birmingham, UK
| | - Eleanor Barnes
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Michelle Willicombe
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, Hammersmith Campus, London, UK
| | - Stefan Siebert
- College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, UK
| | - Thushan I de Silva
- Department of Infection, Immunity and Cardiovascular Disease, The Medical School, The University of Sheffield, Sheffield, UK
| | - John A Snowden
- Department of Haematology, Sheffield Teaching Hospitals NHS Foundation Trust, Royal Hallamshire Hospital, Sheffield, UK
| | - Sean H Lim
- Centre for Cancer Immunology, University of Southampton, Southampton, UK
| | - Sarah J Bowden
- Cancer Research UK Clinical Trials Unit, University of Birmingham, Edgbaston, Birmingham, UK
| | - Lucinda Billingham
- Cancer Research UK Clinical Trials Unit, University of Birmingham, Edgbaston, Birmingham, UK
| | - Alex Richter
- Clinical Immunology Service, University of Birmingham, Edgbaston, Birmingham, UK
| | - Miles Carroll
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | | | | | - Daniel Rea
- Cancer Research UK Clinical Trials Unit, University of Birmingham, Edgbaston, Birmingham, UK
| | - Helen Parry
- Department of Haematology, University Hospitals Birmingham NHS Foundations Trust, Birmingham, UK
| | - Sarah Pirrie
- Cancer Research UK Clinical Trials Unit, University of Birmingham, Edgbaston, Birmingham, UK
| | - Zixiang Lim
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Jack Satsangi
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | | | - Gordon Cook
- National Institute for Health Research Leeds MIC, University of Leeds, Leeds, UK
| | - Paul Miller
- British Society of Blood and Marrow Transplantation and Cellular Therapy, Guy's Hospital, London, UK
| | - Neil Basu
- College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, UK
| | - Ashley Gilmour
- College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, UK
| | - Anne-Marie Hodgkins
- Cancer Research UK Clinical Trials Unit, University of Birmingham, Edgbaston, Birmingham, UK
| | - Lili Evans
- Cancer Research UK Clinical Trials Unit, University of Birmingham, Edgbaston, Birmingham, UK
| | - Ana Hughes
- Cancer Research UK Clinical Trials Unit, University of Birmingham, Edgbaston, Birmingham, UK
| | - Stephanie Longet
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK; Centre International de Recherche en Infectiologie, Team GIMAP (Saint-Etienne), Université Claude Bernard Lyon 1, Inserm, CNRS, Lyon, France
| | - Georgina Meacham
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Kwee L Yong
- Cancer Institute, Department of Haematology, University College London, London, UK
| | | | - Mickey B C Koh
- Infection and Immunity Clinical Academic Group, St George's, University of London and Department of Haematology, St George's Hospital NHS Foundation Trust, London, UK
| | - Siobhan O Burns
- Clinical Immunology, Royal Free Hospital, Hampstead, London, UK; Institute of Immunity and Transplantation, University College London, Hampstead, London, UK
| | - Kim Orchard
- Department of Haematology, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Caron Paterson
- College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, UK
| | - Graham McIlroy
- Department of Haematology, University Hospitals Birmingham NHS Foundations Trust, Birmingham, UK
| | - Sam M Murray
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Tina Thomson
- Imperial College Healthcare NHS Trust, Hammersmith Hospital, London, UK
| | | | - Lyndsey Goulston
- National Institute of Health Research, Southampton Clinical Research Facility, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Samantha Miller
- College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, UK
| | - Victoria Keillor
- College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, UK
| | - Maria Prendecki
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, Hammersmith Campus, London, UK
| | - David Thomas
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, UK
| | - Amanda Kirkham
- Cancer Research UK Clinical Trials Unit, University of Birmingham, Edgbaston, Birmingham, UK
| | - Iain B McInnes
- College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, UK
| | - Pamela Kearns
- Cancer Research UK Clinical Trials Unit, University of Birmingham, Edgbaston, Birmingham, UK; National Institute for Health Research, Birmingham Biomedical Research Centre, Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK.
| |
Collapse
|
7
|
Karbiener M, Kindle G, Meyts I, Seppänen MRJ, Candotti F, Kamieniak M, Ilk R, Kreil TR, Seidel MG. Clinical efficacy of SARS-CoV-2 Omicron-neutralizing antibodies in immunoglobulin preparations for the treatment of agammaglobulinemia in patients with primary antibody deficiency. J Med Virol 2024; 96:e29738. [PMID: 38884390 DOI: 10.1002/jmv.29738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 05/24/2024] [Accepted: 06/04/2024] [Indexed: 06/18/2024]
Abstract
Immunocompromised individuals are at significantly elevated risk for severe courses of coronavirus disease 2019 (COVID-19). In addition to vaccination, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) neutralizing antibodies (nAbs) have been applied throughout the pandemic, with time of treatment onset and potency against the currently prevailing virus variant identified as relevant factors for medical benefit. Using data from the European Society for Immunodeficiencies (ESID) registry, the present study evaluated COVID-19 cases in three groups of patients with inborn errors of immunity (IEI; 981 agammaglobulinemia patients on immunoglobulin replacement therapy (IGRT); 8960 non-agammaglobulinemia patients on IGRT; 14 428 patients without IGRT), and the neutralizing capacity of 1100 immunoglobulin lots against SARS-CoV-2 ("Wuhan" and Omicron strains), throughout 3 years. From the first (2020/2021) to the second (2021/2022) cold season, i.e., during the virus drift to the more contagious Omicron variants, an increase in case numbers was recorded that was comparable (~2- to 3-fold) for all three study groups. During the same period, immunoglobulin lots showed a profound nAb increase against the archetypal SARS-CoV-2 strain, yet only low levels of Omicron nAbs. Notably, shortly before the third (2022/2023) cold season, Omicron-neutralizing capacity of released immunoglobulin lots had plateaued at high levels. From the second to the third cold season, COVID-19 cases dropped markedly. While a ~6-fold case reduction was recorded for the groups of non-agammaglobulinemia patients on IGRT and IEI patients not receiving IGRT, the decline was ~30-fold for the group of agammaglobulinemia patients on IGRT. These findings suggest a substantial COVID-19-protective effect of IGRT, at least for distinct groups of antibody-deficient patients.
Collapse
Affiliation(s)
- Michael Karbiener
- Global Pathogen Safety, Takeda Manufacturing Austria AG, Vienna, Austria
| | - Gerhard Kindle
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Centre for Biobanking FREEZE, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Isabelle Meyts
- Department of Pediatrics, University Hospitals Leuven, KU Leuven, Leuven, Belgium
- Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Mikko R J Seppänen
- Adult Immunodeficiency Unit, Infectious Diseases, Inflammation Center, University of Helsinki and HUS Helsinki University Hospital, Helsinki, Finland
- ERN-RITA Core Center, RITAFIN, Helsinki, Finland
- Rare Disease Center and Pediatric Research Center, Children and Adolescents, University of Helsinki and HUS Helsinki University Hospital, Helsinki, Finland
| | - Fabio Candotti
- Division of Immunology and Allergy, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Marta Kamieniak
- Takeda Development Center Americas, Inc., Cambridge, Massachusetts, USA
| | - Reinhard Ilk
- Global Manufacturing Sciences, Takeda Manufacturing Austria AG, Vienna, Austria
| | - Thomas R Kreil
- Global Pathogen Safety, Takeda Manufacturing Austria AG, Vienna, Austria
| | - Markus G Seidel
- on behalf of the ESID Registry Working Party; Styrian Children's Cancer Research Unit for Cancer and Inborn Errors of the Blood and Immunity in Children, Medical University of Graz, Graz, Austria
- Division of Pediatric Hematology and Oncology, Department of Pediatric and Adolescent Medicine, Medical University of Graz, Graz, Austria
| |
Collapse
|
8
|
van Leeuwen LPM, Grobben M, GeurtsvanKessel CH, Ellerbroek PM, de Bree GJ, Potjewijd J, Rutgers A, Jolink H, van de Veerdonk FL, van Gils MJ, de Vries RD, Dalm VASH. Immunogenicity of COVID-19 booster vaccination in IEI patients and their one year clinical follow-up after start of the COVID-19 vaccination program. Front Immunol 2024; 15:1390022. [PMID: 38698851 PMCID: PMC11063285 DOI: 10.3389/fimmu.2024.1390022] [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: 02/22/2024] [Accepted: 04/05/2024] [Indexed: 05/05/2024] Open
Abstract
Purpose Previous studies have demonstrated that the majority of patients with an inborn error of immunity (IEI) develop a spike (S)-specific IgG antibody and T-cell response after two doses of the mRNA-1273 COVID-19 vaccine, but little is known about the response to a booster vaccination. We studied the immune responses 8 weeks after booster vaccination with mRNA-based COVID-19 vaccines in 171 IEI patients. Moreover, we evaluated the clinical outcomes in these patients one year after the start of the Dutch COVID-19 vaccination campaign. Methods This study was embedded in a large prospective multicenter study investigating the immunogenicity of COVID-19 mRNA-based vaccines in IEI (VACOPID study). Blood samples were taken from 244 participants 8 weeks after booster vaccination. These participants included 171 IEI patients (X-linked agammaglobulinemia (XLA;N=11), combined immunodeficiency (CID;N=4), common variable immunodeficiency (CVID;N=45), isolated or undefined antibody deficiencies (N=108) and phagocyte defects (N=3)) and 73 controls. SARS-CoV-2-specific IgG titers, neutralizing antibodies, and T-cell responses were evaluated. One year after the start of the COVID-19 vaccination program, 334 study participants (239 IEI patients and 95 controls) completed a questionnaire to supplement their clinical data focusing on SARS-CoV-2 infections. Results After booster vaccination, S-specific IgG titers increased in all COVID-19 naive IEI cohorts and controls, when compared to titers at 6 months after the priming regimen. The fold-increases did not differ between controls and IEI cohorts. SARS-CoV-2-specific T-cell responses also increased equally in all cohorts after booster vaccination compared to 6 months after the priming regimen. Most SARS-CoV-2 infections during the study period occurred in the period when the Omicron variant had become dominant. The clinical course of these infections was mild, although IEI patients experienced more frequent fever and dyspnea compared to controls and their symptoms persisted longer. Conclusion Our study demonstrates that mRNA-based booster vaccination induces robust recall of memory B-cell and T-cell responses in most IEI patients. One-year clinical follow-up demonstrated that SARS-CoV-2 infections in IEI patients were mild. Given our results, we support booster campaigns with newer variant-specific COVID-19 booster vaccines to IEI patients with milder phenotypes.
Collapse
Affiliation(s)
- Leanne P. M. van Leeuwen
- Department of Viroscience, Erasmus MC University Medical Center Rotterdam, Rotterdam, Netherlands
- Travel Clinic, Erasmus MC University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Marloes Grobben
- Department of Medical Microbiology and Infection Prevention, Amsterdam Institute for Infection and Immunity, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | | | - Pauline M. Ellerbroek
- Department of Internal Medicine, Infectious Diseases, University Medical Center Utrecht, Utrecht, Netherlands
| | | | - Judith Potjewijd
- Department of Internal Medicine, Division Clinical Immunology, Maastricht UMC, Maastricht, Netherlands
| | - Abraham Rutgers
- Department of Rheumatology and Clinical Immunology, UMC Groningen, Groningen, Netherlands
| | - Hetty Jolink
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Frank L. van de Veerdonk
- Department of Internal Medicine, Radboud University Medical Center Nijmegen, Nijmegen, Netherlands
| | - Marit J. van Gils
- Department of Medical Microbiology and Infection Prevention, Amsterdam Institute for Infection and Immunity, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Rory D. de Vries
- Department of Viroscience, Erasmus MC University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Virgil A. S. H. Dalm
- Department of Internal Medicine, Division of Allergy & Clinical Immunology, Erasmus MC University Medical Center Rotterdam, Rotterdam, Netherlands
- Department of Immunology, Erasmus MC University Medical Center Rotterdam, Rotterdam, Netherlands
| |
Collapse
|
9
|
Cusa G, Sardella G, Garzi G, Firinu D, Milito C. SARS-CoV-2 vaccination in primary antibody deficiencies: an overview on efficacy, immunogenicity, durability of immune response and safety. Curr Opin Allergy Clin Immunol 2024; 24:37-43. [PMID: 37962877 DOI: 10.1097/aci.0000000000000955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
PURPOSE OF REVIEW This review aims to summarize the current best knowledge on the efficacy of COVID-19 vaccination in vulnerable patients affected by primary antibody deficiencies (PADs), both in patients previously infected and vaccine-immunized, focusing also on the durability, on the need for multiple booster doses and on the safety of anti-SARS-CoV-2 vaccines. RECENT FINDINGS Patients vaccinated for SARS-CoV2 have variable humoral response, still showing a tendency towards an increase in antibody titers, with factors such as booster doses, previous infections, age and specific genetic mutations influencing the outcome. Long-lasting cellular responses to SARS-CoV-2 vaccination instead, mostly of the T-cell type, have been observed. Overall, the duration of protection given by vaccinations is sufficient and increased upon further simulations. Furthermore, the safety profile in PID patients is excellent, with most adverse events being transient and mild and no major adverse event reported. SUMMARY Several studies have emphasized the benefit of vaccinating patients with PADs against the SARS-CoV-2 virus and the necessity of administering booster doses. This review, by gathering the most recent and significant data from the scientific literature, could be helpful in clinical practice in the management of disease prevention in patients affected by primary immunodeficiency and also serve as inspiration for further in-depth clinical research.
Collapse
Affiliation(s)
- Gabriella Cusa
- Department of Molecular Medicine, "Sapienza" University of Rome, Rome
| | - Germano Sardella
- Department of Molecular Medicine, "Sapienza" University of Rome, Rome
| | - Giulia Garzi
- Department of Molecular Medicine, "Sapienza" University of Rome, Rome
| | - Davide Firinu
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Cinzia Milito
- Department of Molecular Medicine, "Sapienza" University of Rome, Rome
| |
Collapse
|
10
|
Costanzo GAML, Deiana CM, Sanna G, Perra A, Campagna M, Ledda AG, Coghe F, Palmas V, Cappai R, Manzin A, Chessa L, Del Giacco S, Firinu D. Impact of Exposure to Vaccination and Infection on Cellular and Antibody Response to SARS-CoV-2 in CVID Patients Through COVID-19 Pandemic. J Clin Immunol 2023; 44:12. [PMID: 38129351 DOI: 10.1007/s10875-023-01616-2] [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: 08/08/2023] [Accepted: 11/09/2023] [Indexed: 12/23/2023]
Abstract
PURPOSE The purpose of this study is to investigate the kinetics of response against SARS-CoV-2 elicited by vaccination and/or breakthrough infection (occurred after 3 doses of BNT162b2) in a cohort CVID patients. METHODS We measured humoral and cellular immunity using quantitative anti-spike antibody (anti-S-IgG) and neutralization assay and specific interferon-gamma release assay (IGRA) before and after the third or fourth dose of BNT162b2 and/or after COVID-19. RESULTS In CVID, 58.3% seroconverted after 2 doses that increased to 77.8% after 3 doses. Between the second and third dose, there was a decline in humoral compartment that led to titers below the cutoff of 1:10 (MNA90%) in CVID. This was paralleled by a significantly lower proportion (30%) and reduced magnitude of the residual cellular response among CVID. The third dose achieved a lower titer of anti-S and nAb against the Wuhan strain than HC and significantly decreased the rate of those showing solely a positive neutralizing activity and those with simultaneous negativity of IGRA and nAbs; the differences in IGRA were overall reduced with respect to HC. At further sampling after breakthrough SARS-COV-2 infection, mostly in the omicron era, or fourth dose, 6 months after the last event, the residual nAb titer to Wuhan strain was still significantly higher in HC, while there was no significant difference of nAbs to BA.1. The rate of IGRA responders was 65.5% in CVID and 90.5% in HC (p=0.04), while the magnitude of response was similar. None of CVID had double negativity to nAbs and IGRA at the last sampling. CONCLUSION This data shows an increase of adaptive immunity in CVID after mRNA vaccination in parallel to boosters, accrual number of exposures and formation of hybrid immunity.
Collapse
Affiliation(s)
| | - Carla Maria Deiana
- Department of Medical Sciences and Public Health, University of Cagliari, 09100, Cagliari, Italy
| | - Giuseppina Sanna
- Microbiology and Virology Unit, Department of Biomedical Sciences, University of Cagliari, 09042, Monserrato, Italy
| | - Andrea Perra
- Oncology and Molecular Pathology Unit, Department of Biomedical Sciences, University of Cagliari, 09100, Cagliari, Italy
| | - Marcello Campagna
- Department of Medical Sciences and Public Health, University of Cagliari, 09100, Cagliari, Italy
| | - Andrea Giovanni Ledda
- Department of Medical Sciences and Public Health, University of Cagliari, 09100, Cagliari, Italy
| | - Ferdinando Coghe
- Laboratory Clinical Chemical Analysis and Microbiology, University Hospital of Cagliari, 09042, Monserrato, Italy
| | - Vanessa Palmas
- Microbiology and Virology Unit, Department of Biomedical Sciences, University of Cagliari, 09042, Monserrato, Italy
| | - Riccardo Cappai
- Laboratory Clinical Chemical Analysis and Microbiology, University Hospital of Cagliari, 09042, Monserrato, Italy
| | - Aldo Manzin
- Microbiology and Virology Unit, Department of Biomedical Sciences, University of Cagliari, 09042, Monserrato, Italy
| | - Luchino Chessa
- Department of Medical Sciences and Public Health, University of Cagliari, 09100, Cagliari, Italy
| | - Stefano Del Giacco
- Department of Medical Sciences and Public Health, University of Cagliari, 09100, Cagliari, Italy
| | - Davide Firinu
- Department of Medical Sciences and Public Health, University of Cagliari, 09100, Cagliari, Italy.
- Unit of Internal Medicine, Policlinico Universitario - AOU di Cagliari, Cagliari, Italy.
- Azienda Ospedaliero Universitaria, SS 554-Bivio Sestu, 09042, Monserrato, CA, Italy.
| |
Collapse
|
11
|
Lucane Z, Kursite M, Sablinskis K, Gailite L, Kurjane N. COVID-19 Vaccination Coverage and Factors Influencing Vaccine Hesitancy among Patients with Inborn Errors of Immunity in Latvia: A Mixed-Methods Study. Vaccines (Basel) 2023; 11:1637. [PMID: 38005969 PMCID: PMC10675738 DOI: 10.3390/vaccines11111637] [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: 09/30/2023] [Revised: 10/19/2023] [Accepted: 10/24/2023] [Indexed: 11/26/2023] Open
Abstract
BACKGROUND The European Society for Immunodeficiencies recommends that all patients with inborn errors of immunity (IEI) without contraindications should receive SARS-CoV-2 vaccination. The aim of this study was to investigate the reasons that discourage IEI patients from receiving the recommended vaccination and to assess vaccination coverage among IEI patients in Latvia. METHODS In this multicenter mixed-methods study, the vaccination status of all patients with IEI within two tertiary centers in Latvia was reviewed using electronic health records. Semi-structured interviews were conducted with 16 IEI patients who did not undergo vaccination, and a thematic analysis was performed. RESULTS A total of 341 patients (49.3% female; median age 19.7 years (IQR:17)) were included in the quantitative part. The proportion of fully vaccinated individuals aged ≥ 12 years was 66.8%-70.9% with patients with selective IgA deficiency and 58.8% with other IEI (χ² = 14.12, p < 0.001). The proportion of fully vaccinated individuals aged 5-11 years was 11.1%. Age was associated with vaccination status: younger patients were found to have a significantly lower likelihood of receiving vaccination (U = 8585, p < 0.001). The five main themes identified were as follows: (1) fear and uncertainty; (2) risk and benefit assessment: COVID-19 vaccine-is it worth it? (3) external influences: the dark horse of the decision-making-people around us; (4) individuals against the system; and (5) beliefs about vaccination and COVID-19. Under-representation of certain IEI groups and recall bias are possible limitations of this study. CONCLUSIONS While most reasons for hesitancy were similar to those previously described in the general population, disease-specific concerns were also identified.
Collapse
Affiliation(s)
- Zane Lucane
- Department of Biology and Microbiology, Riga Stradins University, LV-1007 Riga, Latvia
| | - Mirdza Kursite
- Department of Public Health and Epidemiology, Riga Stradiņš University, LV-1007 Riga, Latvia
| | - Kristaps Sablinskis
- Department of Internal Diseases, Riga Stradins University, LV-1007 Riga, Latvia
| | - Linda Gailite
- Scientific Laboratory of Molecular Genetics, Riga Stradins University, LV-1007 Riga, Latvia
| | - Natalja Kurjane
- Department of Biology and Microbiology, Riga Stradins University, LV-1007 Riga, Latvia
- Outpatient Clinic, Pauls Stradins Clinical University Hospital, LV-1002 Riga, Latvia
- Outpatient Clinic, Children’s Clinical University Hospital, LV-1004 Riga, Latvia
| |
Collapse
|
12
|
Zendt M, Bustos Carrillo FA, Kelly S, Saturday T, DeGrange M, Ginigeme A, Wu L, Callier V, Ortega-Villa A, Faust M, Chang-Rabley E, Bugal K, Kenney H, Khil P, Youn JH, Osei G, Regmi P, Anderson V, Bosticardo M, Daub J, DiMaggio T, Kreuzburg S, Pala F, Pfister J, Treat J, Ulrick J, Karkanitsa M, Kalish H, Kuhns DB, Priel DL, Fink DL, Tsang JS, Sparks R, Uzel G, Waldman MA, Zerbe CS, Delmonte OM, Bergerson JRE, Das S, Freeman AF, Lionakis MS, Sadtler K, van Doremalen N, Munster V, Notarangelo LD, Holland SM, Ricotta EE. Characterization of the antispike IgG immune response to COVID-19 vaccines in people with a wide variety of immunodeficiencies. SCIENCE ADVANCES 2023; 9:eadh3150. [PMID: 37824621 PMCID: PMC10569702 DOI: 10.1126/sciadv.adh3150] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 09/07/2023] [Indexed: 10/14/2023]
Abstract
Research on coronavirus disease 2019 vaccination in immune-deficient/disordered people (IDP) has focused on cancer and organ transplantation populations. In a prospective cohort of 195 IDP and 35 healthy volunteers (HV), antispike immunoglobulin G (IgG) was detected in 88% of IDP after dose 2, increasing to 93% by 6 months after dose 3. Despite high seroconversion, median IgG levels for IDP never surpassed one-third that of HV. IgG binding to Omicron BA.1 was lowest among variants. Angiotensin-converting enzyme 2 pseudo-neutralization only modestly correlated with antispike IgG concentration. IgG levels were not significantly altered by receipt of different messenger RNA-based vaccines, immunomodulating treatments, and prior severe acute respiratory syndrome coronavirus 2 infections. While our data show that three doses of coronavirus disease 2019 vaccinations induce antispike IgG in most IDP, additional doses are needed to increase protection. Because of the notably reduced IgG response to Omicron BA.1, the efficacy of additional vaccinations, including bivalent vaccines, should be studied in this population.
Collapse
Affiliation(s)
- Mackenzie Zendt
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research (DIR), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Fausto A. Bustos Carrillo
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research (DIR), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
- Office of Data Science and Emerging Technologies, Office of Science Management and Operations, NIAID, NIH, Rockville, MD, USA
| | - Sophie Kelly
- Trans-NIH Shared Resource on Biomedical Engineering and Physical Science, National Institute of Biomedical Imaging and Bioengineering (NIBIB), NIH, Bethesda, MD, USA
| | | | - Maureen DeGrange
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research (DIR), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
- Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Anita Ginigeme
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research (DIR), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
- Medical Science and Computing LLC, Rockville, MD, USA
| | - Lurline Wu
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research (DIR), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Viviane Callier
- Clinical Monitoring Research Program Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Ana Ortega-Villa
- Biostatistics Research Branch, Division of Clinical Research, NIAID, NIH, Rockville, MD, USA
| | | | - Emma Chang-Rabley
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research (DIR), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Kara Bugal
- Division of Laboratory Medicine, NIH Clinical Center, Bethesda, MD,USA
| | - Heather Kenney
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research (DIR), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Pavel Khil
- Division of Laboratory Medicine, NIH Clinical Center, Bethesda, MD,USA
| | - Jung-Ho Youn
- Division of Laboratory Medicine, NIH Clinical Center, Bethesda, MD,USA
| | - Gloria Osei
- Division of Laboratory Medicine, NIH Clinical Center, Bethesda, MD,USA
| | - Pravesh Regmi
- Division of Laboratory Medicine, NIH Clinical Center, Bethesda, MD,USA
| | - Victoria Anderson
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research (DIR), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Marita Bosticardo
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research (DIR), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Janine Daub
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research (DIR), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Thomas DiMaggio
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research (DIR), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Samantha Kreuzburg
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research (DIR), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Francesca Pala
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research (DIR), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Justina Pfister
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research (DIR), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Jennifer Treat
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research (DIR), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Jean Ulrick
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research (DIR), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | | | - Heather Kalish
- Trans-NIH Shared Resource on Biomedical Engineering and Physical Science, National Institute of Biomedical Imaging and Bioengineering (NIBIB), NIH, Bethesda, MD, USA
| | - Douglas B. Kuhns
- Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Debra L. Priel
- Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Danielle L. Fink
- Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - John S. Tsang
- Department of Immunobiology and Yale Center for Systems and Engineering Immunology, Yale School of Medicine, New Haven, CT, USA
- Department of Biomedical Engineering, Yale University, New Haven, CT,USA
| | - Rachel Sparks
- Laboratory of Immune System Biology, DIR, NIAID, NIH, Bethesda, MD,USA
| | - Gulbu Uzel
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research (DIR), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Meryl A. Waldman
- Kidney Disease Section, Kidney Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD, USA
| | - Christa S. Zerbe
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research (DIR), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Ottavia M. Delmonte
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research (DIR), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Jenna R. E. Bergerson
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research (DIR), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Sanchita Das
- Division of Laboratory Medicine, NIH Clinical Center, Bethesda, MD,USA
| | - Alexandra F. Freeman
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research (DIR), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Michail S. Lionakis
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research (DIR), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Kaitlyn Sadtler
- Section for Immunoengineering, NIBIB, NIH, Bethesda, MD, USA
| | | | | | - Luigi D. Notarangelo
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research (DIR), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Steven M. Holland
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research (DIR), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Emily E. Ricotta
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research (DIR), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| |
Collapse
|
13
|
Upasani V, Townsend K, Wu MY, Carr EJ, Hobbs A, Dowgier G, Ragno M, Herman LS, Sharma S, Shah D, Lee SFK, Chauhan N, Glanville JM, Neave L, Hanson S, Ravichandran S, Tynan A, O’Sullivan M, Moreira F, Workman S, Symes A, Burns SO, Tadros S, Hart JCL, Beale RCL, Gandhi S, Wall EC, McCoy L, Lowe DM. Commercial Immunoglobulin Products Contain Neutralizing Antibodies Against Severe Acute Respiratory Syndrome Coronavirus 2 Spike Protein. Clin Infect Dis 2023; 77:950-960. [PMID: 37338118 PMCID: PMC10552578 DOI: 10.1093/cid/ciad368] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 05/05/2023] [Accepted: 06/16/2023] [Indexed: 06/21/2023] Open
Abstract
BACKGROUND Patients with antibody deficiency respond poorly to coronavirus disease 2019 (COVID-19) vaccination and are at risk of severe or prolonged infection. They are given long-term immunoglobulin replacement therapy (IRT) prepared from healthy donor plasma to confer passive immunity against infection. Following widespread COVID-19 vaccination alongside natural exposure, we hypothesized that immunoglobulin preparations will now contain neutralizing severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike antibodies, which confer protection against COVID-19 disease and may help to treat chronic infection. METHODS We evaluated anti-SARS-CoV-2 spike antibody in a cohort of patients before and after immunoglobulin infusion. Neutralizing capacity of patient samples and immunoglobulin products was assessed using in vitro pseudovirus and live-virus neutralization assays, the latter investigating multiple batches against current circulating Omicron variants. We describe the clinical course of 9 patients started on IRT during treatment of COVID-19. RESULTS In 35 individuals with antibody deficiency established on IRT, median anti-spike antibody titer increased from 2123 to 10 600 U/mL postinfusion, with corresponding increase in pseudovirus neutralization titers to levels comparable to healthy donors. Testing immunoglobulin products directly in the live-virus assay confirmed neutralization, including of BQ1.1 and XBB variants, but with variation between immunoglobulin products and batches.Initiation of IRT alongside remdesivir in patients with antibody deficiency and prolonged COVID-19 infection (median 189 days, maximum >900 days with an ancestral viral strain) resulted in clearance of SARS-CoV-2 at a median of 20 days. CONCLUSIONS Immunoglobulin preparations now contain neutralizing anti-SARS-CoV-2 antibodies that are transmitted to patients and help to treat COVID-19 in individuals with failure of humoral immunity.
Collapse
Affiliation(s)
- Vinit Upasani
- Institute of Immunity and Transplantation, University College London (UCL), London, United Kingdom
| | - Katie Townsend
- Department of Clinical Immunology, Royal Free London National Health Service (NHS) Foundation Trust, London, United Kingdom
| | - Mary Y Wu
- COVID Surveillance Unit, Francis Crick Institute, London, United Kingdom
| | - Edward J Carr
- Francis Crick Institute, London, United Kingdom
- Department of Renal Medicine, Royal Free London NHS Foundation Trust, London, United Kingdom
| | - Agnieszka Hobbs
- COVID Surveillance Unit, Francis Crick Institute, London, United Kingdom
| | - Giulia Dowgier
- COVID Surveillance Unit, Francis Crick Institute, London, United Kingdom
| | - Martina Ragno
- COVID Surveillance Unit, Francis Crick Institute, London, United Kingdom
| | - Lou S Herman
- COVID Surveillance Unit, Francis Crick Institute, London, United Kingdom
| | - Sonal Sharma
- Department of Elderly Medicine, Barnet Hospital, Royal Free London NHS Foundation Trust, London, United Kingdom
| | - Devesh Shah
- Department of Elderly Medicine, Barnet Hospital, Royal Free London NHS Foundation Trust, London, United Kingdom
| | - Simon F K Lee
- Department of Infectious Diseases, Royal Free London NHS Foundation Trust, London, United Kingdom
| | - Neil Chauhan
- Department of Haematology, Royal Free London NHS Foundation Trust, London, United Kingdom
| | - Julie M Glanville
- Department of Haematology, Royal Free London NHS Foundation Trust, London, United Kingdom
| | - Lucy Neave
- Department of Haematology, Royal Free London NHS Foundation Trust, London, United Kingdom
| | - Steven Hanson
- Department of Haematology, Royal Free London NHS Foundation Trust, London, United Kingdom
| | - Sriram Ravichandran
- Department of Haematology, Royal Free London NHS Foundation Trust, London, United Kingdom
| | - Aoife Tynan
- Department of Pharmacy, Royal Free London NHS Foundation Trust, London, United Kingdom
| | - Mary O’Sullivan
- Department of Clinical Immunology, Royal Free London National Health Service (NHS) Foundation Trust, London, United Kingdom
| | - Fernando Moreira
- Department of Clinical Immunology, Royal Free London National Health Service (NHS) Foundation Trust, London, United Kingdom
| | - Sarita Workman
- Department of Clinical Immunology, Royal Free London National Health Service (NHS) Foundation Trust, London, United Kingdom
| | - Andrew Symes
- Department of Clinical Immunology, Royal Free London National Health Service (NHS) Foundation Trust, London, United Kingdom
| | - Siobhan O Burns
- Institute of Immunity and Transplantation, University College London (UCL), London, United Kingdom
- Department of Clinical Immunology, Royal Free London National Health Service (NHS) Foundation Trust, London, United Kingdom
| | - Susan Tadros
- Department of Clinical Immunology, Royal Free London National Health Service (NHS) Foundation Trust, London, United Kingdom
| | - Jennifer C L Hart
- Department of Virology, Royal Free London NHS Foundation Trust, London, United Kingdom
| | - Rupert C L Beale
- COVID Surveillance Unit, Francis Crick Institute, London, United Kingdom
- Department of Renal Medicine, Royal Free London NHS Foundation Trust, London, United Kingdom
| | - Sonia Gandhi
- COVID Surveillance Unit, Francis Crick Institute, London, United Kingdom
- UCL Hospitals Biomedical Research Centre, London, United Kingdom
| | - Emma C Wall
- COVID Surveillance Unit, Francis Crick Institute, London, United Kingdom
- UCL Hospitals Biomedical Research Centre, London, United Kingdom
| | - Laura McCoy
- Institute of Immunity and Transplantation, University College London (UCL), London, United Kingdom
| | - David M Lowe
- Institute of Immunity and Transplantation, University College London (UCL), London, United Kingdom
- Department of Clinical Immunology, Royal Free London National Health Service (NHS) Foundation Trust, London, United Kingdom
| |
Collapse
|
14
|
Senefeld JW, Joyner MJ. Severe Acute Respiratory Syndrome Coronavirus 2 Antibody Replacement Therapy for Immunocompromised Patients. Clin Infect Dis 2023; 77:961-963. [PMID: 37337905 DOI: 10.1093/cid/ciad367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 06/17/2023] [Indexed: 06/21/2023] Open
Affiliation(s)
- Jonathon W Senefeld
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
| | - Michael J Joyner
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
15
|
KARABİBER E, ATİK Ö, TEPETAM F, ERGAN B, İLKİ A, KARAKOÇ AYDINER E, ÖZEN A, ÖZYER F, BARIŞ S. Clinical and immunological outcomes of SARS-CoV-2 infection in patients with inborn errors of immunity receiving different brands and doses of COVID-19 vaccines. Tuberk Toraks 2023; 71:236-249. [PMID: 37740627 PMCID: PMC10912874 DOI: 10.5578/tt.20239705] [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: 07/26/2023] [Accepted: 08/17/2023] [Indexed: 09/24/2023] Open
Abstract
Introduction Vaccines against severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2) provide successful control of the coronavirus-2019 (COVID-19) pandemic. The safety and immunogenicity studies are encouraging in patients with inborn errors of immunity (IEI); however, data about mortality outcomes and severe disease after vaccination still need to be fully addressed. Therefore, we aimed to determine the clinical and immunological outcomes of SARS-CoV-2 infection in patients with IEI who have received vaccination. Materials and Methods Eighty-eight patients with a broad range of molecular etiologies were studied; 45 experienced SARS-CoV-2 infection. Infection outcomes were analyzed in terms of genetic etiology, background clinical characteristics, and immunization history, including the type and number of doses received and the time elapsed since vaccination. In addition, anti-SARS-CoV-2 antibodies were quantified using electrochemiluminescent immunoassay. Results Patients were immunized using one of the three regimens: inactivated (Sinovac, Coronavac®), mRNA (BNT162b2, Comirnaty®, Pfizer-Biontech), and a combination. All three regimens induced comparable anti-SARS-CoV-2 IgG levels, with no differences in the adverse events. Among 45 patients with COVID-19, 26 received a full course of vaccination, while 19 were vaccine-naive or received incomplete dosing. No patients died due to COVID-19 infection. The fully immunized group had a lower hospitalization rate (23% vs. 31.5%) and a shorter symptomatic phase than the others. Among the fully vaccinated patients, serum IgM and E levels were significantly lower in hospitalized patients than non-hospitalized patients. Conclusion COVID-19 vaccines were well-tolerated by the IEI patients, and a full course of immunization was associated with lower hospitalization rates and a shorter duration of COVID-19 symptoms.
Collapse
Affiliation(s)
- E. KARABİBER
- Division of Adult Immunology and Allergy, Department of Chest Diseases,
Marmara University Pendik Training and Research Hospital, İstanbul, Türkiye
- Division of Adult Immunology and Allergy, Department of Chest Diseases,
Süreyyapaşa Training and Research Hospital, İstanbul, Türkiye
- Department of Medical Microbiology, Marmara University Faculty of
Medicine, İstanbul, Türkiye
- Department of Pediatric Allergy and Immunology, Marmara University
Faculty of Medicine, İstanbul, Türkiye
- İstanbul Jeffrey Modell Diagnostic and Research Center for Primary
Immunodeficiencies, İstanbul, Türkiye
- Işıl Berat Barlan Center for Translational Medicine, İstanbul, Türkiye
| | - Ö. ATİK
- Division of Adult Immunology and Allergy, Department of Chest Diseases,
Süreyyapaşa Training and Research Hospital, İstanbul, Türkiye
| | - F.M. TEPETAM
- Division of Adult Immunology and Allergy, Department of Chest Diseases,
Süreyyapaşa Training and Research Hospital, İstanbul, Türkiye
| | - B. ERGAN
- Department of Medical Microbiology, Marmara University Faculty of
Medicine, İstanbul, Türkiye
| | - A. İLKİ
- Department of Medical Microbiology, Marmara University Faculty of
Medicine, İstanbul, Türkiye
| | - E. KARAKOÇ AYDINER
- Department of Pediatric Allergy and Immunology, Marmara University
Faculty of Medicine, İstanbul, Türkiye
- İstanbul Jeffrey Modell Diagnostic and Research Center for Primary
Immunodeficiencies, İstanbul, Türkiye
- Işıl Berat Barlan Center for Translational Medicine, İstanbul, Türkiye
| | - A. ÖZEN
- Department of Pediatric Allergy and Immunology, Marmara University
Faculty of Medicine, İstanbul, Türkiye
- İstanbul Jeffrey Modell Diagnostic and Research Center for Primary
Immunodeficiencies, İstanbul, Türkiye
- Işıl Berat Barlan Center for Translational Medicine, İstanbul, Türkiye
| | - F. ÖZYER
- Division of Adult Immunology and Allergy, Department of Chest Diseases,
Marmara University Pendik Training and Research Hospital, İstanbul, Türkiye
| | - S. BARIŞ
- Department of Pediatric Allergy and Immunology, Marmara University
Faculty of Medicine, İstanbul, Türkiye
- İstanbul Jeffrey Modell Diagnostic and Research Center for Primary
Immunodeficiencies, İstanbul, Türkiye
- Işıl Berat Barlan Center for Translational Medicine, İstanbul, Türkiye
| |
Collapse
|
16
|
Koehm M, Klippstein M, Dauth S, Hallmann K, Kohmer N, Burkhardt H, Ciesek S, Geisslinger G, Rabenau HF, Behrens F. Impact of different classes of immune-modulating treatments on B cell-related and T cell-related immune response before and after COVID-19 booster vaccination in patients with immune-mediated diseases and primary immunodeficiency: a cohort study. RMD Open 2023; 9:e003094. [PMID: 37652553 PMCID: PMC10476126 DOI: 10.1136/rmdopen-2023-003094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 06/23/2023] [Indexed: 09/02/2023] Open
Abstract
OBJECTIVES To evaluate the potential of immunosuppressed patients to mount B-cell and T-cell responses to COVID-19 booster vaccination (third vaccination). METHODS Patients with primary immunodeficiency (PID), immune-mediated inflammatory diseases (IMIDs) on CD20-depleting treatment with rituximab (RTX), or IMIDs treated with conventional synthetic disease-modifying antirheumatic drugs (csDMARDs) or biological disease-modifying antirheumatic drug (bDMARDs) were included and assessed before (baseline visit (BL)) and 2, 4 and 8 weeks after COVID-19 booster vaccination. Serum B-cell responses were assessed by antibody levels against SARS-CoV-2 spike protein (anti-spike IgG antibody (S-AB)) and a surrogate virus neutralisation test (sVNT). T-cell responses were assessed by an interferon gamma release assay (IGRA). RESULTS Fifty patients with PID (n=6), treated with RTX therapy (n=13), or treated with csDMARDs/bDMARDs (n=31) were included. At BL, anti-S-AB titres in PID and csDMARD/bDMARD-treated patients were low (although significantly higher than RTX patients); measures of B-cell-mediated response increased significantly after booster vaccination. In the RTX cohort, low BL anti-S-AB and sVNT values did not improve after booster vaccination, but patients had significantly elevated IGRA responses post booster vaccination compared with the other groups. csDMARD/bDMARD-treated patients showed the highest BL values in all three assays with greater increases in all parameters after booster vaccination compared with patients with PID. CONCLUSION Patients with IMID on therapeutic B-cell depletion have low anti-S-AB and sVNT values before and after booster vaccination but show significantly higher levels of IGRA compared with other immunosuppressed patients, suggesting an underlying mechanism attempting to compensate compromised humoral immunity by upregulating T-cell responsiveness. PID appears to have a stronger impact on antiviral immune response than csDMARD/bDMARD treatment.
Collapse
Affiliation(s)
- Michaela Koehm
- Department of Rheumatology, Goethe University, Frankfurt am Main, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Frankfurt am Main, Germany
- Fraunhofer Cluster of Excellence Immune-mediated Diseases CIMD, Frankfurt am Main, Germany
| | - Maximilian Klippstein
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Frankfurt am Main, Germany
- Fraunhofer Cluster of Excellence Immune-mediated Diseases CIMD, Frankfurt am Main, Germany
| | - Stephanie Dauth
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Frankfurt am Main, Germany
- Fraunhofer Cluster of Excellence Immune-mediated Diseases CIMD, Frankfurt am Main, Germany
| | - Konstantin Hallmann
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Frankfurt am Main, Germany
- Fraunhofer Cluster of Excellence Immune-mediated Diseases CIMD, Frankfurt am Main, Germany
| | - Niko Kohmer
- Institute for Medical Virology, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Harald Burkhardt
- Department of Rheumatology, Goethe University, Frankfurt am Main, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Frankfurt am Main, Germany
- Fraunhofer Cluster of Excellence Immune-mediated Diseases CIMD, Frankfurt am Main, Germany
| | - Sandra Ciesek
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Frankfurt am Main, Germany
- Institute for Medical Virology, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt am Main, Germany
- Virology, German Centre for Infection Research, Frankfurt am Main, Germany
| | - Gerd Geisslinger
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Frankfurt am Main, Germany
- Fraunhofer Cluster of Excellence Immune-mediated Diseases CIMD, Frankfurt am Main, Germany
- Pharmazentrum Frankfurt/ZAFES, Institute of Clinical Pharmacology, Goethe University, Frankfurt am Main, Germany
| | - Holger F Rabenau
- Institute for Medical Virology, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Frank Behrens
- Department of Rheumatology, Goethe University, Frankfurt am Main, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Frankfurt am Main, Germany
- Fraunhofer Cluster of Excellence Immune-mediated Diseases CIMD, Frankfurt am Main, Germany
| |
Collapse
|
17
|
Loke J, Upasani V, Gaskell C, Fox S, Fletcher R, Thomas C, Hopkins L, Kumari A, Tang T, Yafai E, Boucher R, Homer V, Toth A, Chan YLT, Randall K, Rider T, O'Nions J, Drew V, Pillai A, Dungarwalla M, Murray D, Khan A, Wandroo F, Moore S, Krishnamurthy P, Huang YWJ, Knapper S, Byrne J, Zhao R, Craddock C, Parry H, Moss P, Stanworth SJ, Lowe DM. Defective T-cell response to COVID-19 vaccination in acute myeloid leukaemia and myelodysplastic syndromes. Br J Haematol 2023; 202:498-503. [PMID: 37303189 DOI: 10.1111/bjh.18894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 05/18/2023] [Accepted: 05/19/2023] [Indexed: 06/13/2023]
Abstract
Limited data exist on COVID-19 vaccination efficacy in patients with acute myeloid leukemia and myelodysplasia with excess blasts (AML/MDS-EB2). We report results from a prospective study, PACE (Patients with AML and COVID-19 Epidemiology). 93 patients provided samples post-vaccine 2 or 3 (PV2, PV3). Antibodies against SARS-COV-2 spike antigen were detectable in all samples. Neutralization of the omicron variant was poorer than ancestral variants but improved PV3. In contrast, adequate T-cell reactivity to SARS-COV-2 spike protein was seen in only 16/47 (34%) patients PV2 and 23/52 (44%) PV3. Using regression models, disease response (not in CR/Cri), and increasing age predicted poor T cell response.
Collapse
Affiliation(s)
- Justin Loke
- Centre for Clinical Haematology, Queen Elizabeth Hospital, Birmingham, UK
- CRUK Clinical Trials Unit, University of Birmingham, Birmingham, UK
- Dana Farber Cancer Institute, Boston, Massachusetts, USA
| | | | | | - Sonia Fox
- CRUK Clinical Trials Unit, University of Birmingham, Birmingham, UK
| | - Rachel Fletcher
- CRUK Clinical Trials Unit, University of Birmingham, Birmingham, UK
| | - Catherine Thomas
- CRUK Clinical Trials Unit, University of Birmingham, Birmingham, UK
| | - Louise Hopkins
- CRUK Clinical Trials Unit, University of Birmingham, Birmingham, UK
| | - Anita Kumari
- CRUK Clinical Trials Unit, University of Birmingham, Birmingham, UK
| | - Tina Tang
- CRUK Clinical Trials Unit, University of Birmingham, Birmingham, UK
| | - Emily Yafai
- CRUK Clinical Trials Unit, University of Birmingham, Birmingham, UK
| | - Rebecca Boucher
- CRUK Clinical Trials Unit, University of Birmingham, Birmingham, UK
| | - Victoria Homer
- CRUK Clinical Trials Unit, University of Birmingham, Birmingham, UK
| | - Arpad Toth
- Clatterbridge Cancer Hospital, Liverpool, UK
| | | | - Katie Randall
- South Warwickshire University NHS Foundation Trust, Warwick, UK
| | - Tom Rider
- Brighton and Sussex University Hospitals NHS Trust, Brighton, UK
| | | | | | | | | | | | | | - Farooq Wandroo
- Sandwell and West Birmingham Hospitals NHS Trust, West Bromwich, UK
| | - Sally Moore
- Royal United Hospital Bath NHS Foundation Trust, Bath, UK
| | | | | | | | - Jenny Byrne
- Nottingham University Hospitals Trust, Nottingham, UK
| | | | - Charles Craddock
- Centre for Clinical Haematology, Queen Elizabeth Hospital, Birmingham, UK
- CRUK Clinical Trials Unit, University of Birmingham, Birmingham, UK
| | - Helen Parry
- Centre for Clinical Haematology, Queen Elizabeth Hospital, Birmingham, UK
- University of Birmingham, Birmingham, UK
| | - Paul Moss
- Centre for Clinical Haematology, Queen Elizabeth Hospital, Birmingham, UK
- University of Birmingham, Birmingham, UK
| | - Simon J Stanworth
- Oxford University Hospitals, Oxford, UK
- University of Oxford, Oxford, UK
- NHS Blood and Transplant, Oxford, UK
| | - David M Lowe
- University College London, London, UK
- Royal Free London NHS Foundation Trust, London, UK
| |
Collapse
|
18
|
Steiner S, Schwarz T, Corman VM, Jeworowski LM, Bauer S, Drosten C, Scheibenbogen C, Hanitsch LG. Impaired B Cell Recall Memory and Reduced Antibody Avidity but Robust T Cell Response in CVID Patients After COVID-19 Vaccination. J Clin Immunol 2023; 43:869-881. [PMID: 36932291 PMCID: PMC10023009 DOI: 10.1007/s10875-023-01468-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 03/05/2023] [Indexed: 03/19/2023]
Abstract
PURPOSE Humoral and cellular immune responses were described after COVID-19 vaccination in patients with common variable immunodeficiency disorder (CVID). This study aimed to investigate SARS-CoV-2-specific antibody quality and memory function of B cell immunity as well as T cell responses after COVID-19 vaccination in seroresponding and non-responding CVID patients. METHODS We evaluated antibody avidity and applied a memory B cell ELSPOT assay for functional B cell recall memory response to SARS-CoV-2 after COVID-19 vaccination in CVID seroresponders. We comparatively analyzed SARS-CoV-2 spike reactive polyfunctional T cell response and reactive peripheral follicular T helper cells (pTFH) by flow cytometry in seroresponding and non-seroresponding CVID patients. All CVID patients had previously failed to mount a humoral response to pneumococcal conjugate vaccine. RESULTS SARS-CoV-2 spike antibody avidity of seroresponding CVID patients was significantly lower than in healthy controls. Only 30% of seroresponding CVID patients showed a minimal memory B cell recall response in ELISPOT assay. One hundred percent of CVID seroresponders and 83% of non-seroresponders had a detectable polyfunctional T cell response. Induction of antigen-specific CD4+CD154+CD137+CXCR5+ pTFH cells by the COVID-19 vaccine was higher in CVID seroresponder than in non-seroresponder. Levels of pTFH did not correlate with antibody response or avidity. CONCLUSION Reduced avidity and significantly impaired recall memory formation after COVID-19 vaccination in seroresponding CVID patients stress the importance of a more differentiated analysis of humoral immune response in CVID patients. Our observations challenge the clinical implications that follow the binary categorization into seroresponder and non-seroresponder.
Collapse
Affiliation(s)
- Sophie Steiner
- Institute of Medical Immunology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität Zu Berlin, Campus Virchow, Augustenburger Platz 1/Südstraße 2, 13353, Berlin, Germany
- Berlin Institute of Health, Berlin, Germany
| | - Tatjana Schwarz
- Berlin Institute of Health, Berlin, Germany
- Institute of Virology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, German Centre for Infection Research (DZIF), Associated Partner, Charitéplatz 1, 13353, Berlin, Germany
| | - Victor M Corman
- Berlin Institute of Health, Berlin, Germany
- Institute of Virology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, German Centre for Infection Research (DZIF), Associated Partner, Charitéplatz 1, 13353, Berlin, Germany
- Labor Berlin-Charité Vivantes GmbH, Berlin, Germany
| | - Lara M Jeworowski
- Berlin Institute of Health, Berlin, Germany
- Institute of Virology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, German Centre for Infection Research (DZIF), Associated Partner, Charitéplatz 1, 13353, Berlin, Germany
| | - Sandra Bauer
- Institute of Medical Immunology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität Zu Berlin, Campus Virchow, Augustenburger Platz 1/Südstraße 2, 13353, Berlin, Germany
- Berlin Institute of Health, Berlin, Germany
| | - Christian Drosten
- Berlin Institute of Health, Berlin, Germany
- Institute of Virology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, German Centre for Infection Research (DZIF), Associated Partner, Charitéplatz 1, 13353, Berlin, Germany
| | - Carmen Scheibenbogen
- Institute of Medical Immunology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität Zu Berlin, Campus Virchow, Augustenburger Platz 1/Südstraße 2, 13353, Berlin, Germany
- Berlin Institute of Health, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Charitéplatz 1, Berlin, Germany
| | - Leif G Hanitsch
- Institute of Medical Immunology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität Zu Berlin, Campus Virchow, Augustenburger Platz 1/Südstraße 2, 13353, Berlin, Germany.
- Berlin Institute of Health, Berlin, Germany.
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Charitéplatz 1, Berlin, Germany.
| |
Collapse
|
19
|
Pac M, Casanova JL, Tuzankina I, Maródi L. Editorial: Advances in primary immunodeficiencies (inborn errors of immunity) in Central-Eastern Europe, volume II. Front Immunol 2023; 14:1221137. [PMID: 37383230 PMCID: PMC10294667 DOI: 10.3389/fimmu.2023.1221137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 05/23/2023] [Indexed: 06/30/2023] Open
Affiliation(s)
- Malgorzata Pac
- Department of Immunology, The Children’s Memorial Health Institute, Warsaw, Poland
| | - Jean-Laurent Casanova
- St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University, New York, NY, United States
- Howard Hughes Medical Institute, New York, NY, United States
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Necker Hospital for Sick Children and Institut National de la Sante et de la Recherche Medicale (INSERM), Paris, France
- University Paris Cité, Imagine Institute, Paris, France
| | - Irina Tuzankina
- Institute of Immunology and Physiology of Ural Branch of the Russian Academy of Sciences, Yekaterinburg, Russia
| | - László Maródi
- St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University, New York, NY, United States
- Department of Dermatology, Primary Immunodeficiency Clinical Unit and Laboratory, Semmelweis University, Budapest, Hungary
| |
Collapse
|
20
|
Cao C, Li S, Zhou G, Xu C, Chen X, Qiu H, Li X, Liu Y, Cao H, Bi C. Global trends in COVID-19 Alzheimer's related research: a bibliometric analysis. Front Neurol 2023; 14:1193768. [PMID: 37342784 PMCID: PMC10278887 DOI: 10.3389/fneur.2023.1193768] [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: 03/31/2023] [Accepted: 05/09/2023] [Indexed: 06/23/2023] Open
Abstract
Background The COVID-19 pandemic has significantly impacted public health, putting people with Alzheimer's disease at significant risk. This study used bibliometric analysis method to conduct in-depth research on the relationship between COVID-19 and Alzheimer's disease, as well as to predict its development trends. Methods The Web of Science Core Collection was searched for relevant literature on Alzheimer's and Coronavirus-19 during 2019-2023. We used a search query string in our advanced search. Using Microsoft Excel 2021 and VOSviewer software, a statistical analysis of primary high-yield authors, research institutions, countries, and journals was performed. Knowledge networks, collaboration maps, hotspots, and regional trends were analyzed using VOSviewer and CiteSpace. Results During 2020-2023, 866 academic studies were published in international journals. United States, Italy, and the United Kingdom rank top three in the survey; in terms of productivity, the top three schools were Harvard Medical School, the University of Padua, and the University of Oxford; Bonanni, Laura, from Gabriele d'Annunzio University (Italy), Tedeschi, Gioacchino from the University of Campania Luigi Vanvitelli (Italy), Vanacore, Nicola from Natl Ctr Dis Prevent and Health Promot (Italy), Reddy, P. Hemachandra from Texas Tech University (USA), and El Haj, Mohamad from University of Nantes (France) were the authors who published the most articles; The Journal of Alzheimer's Disease is the journals with the most published articles; "COVID-19," "Alzheimer's disease," "neurodegenerative diseases," "cognitive impairment," "neuroinflammation," "quality of life," and "neurological complications" have been the focus of attention in the last 3 years. Conclusion The disease caused by the COVID-19 virus infection related to Alzheimer's disease has attracted significant attention worldwide. The major hot topics in 2020 were: "Alzheimer' disease," COVID-19," risk factors," care," and "Parkinson's disease." During the 2 years 2021 and 2022, researchers were also interested in "neurodegenerative diseases," "cognitive impairment," and "quality of life," which require further investigation.
Collapse
Affiliation(s)
- Chenjun Cao
- Department of Psychiatry, School of Clinical Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Department of Psychiatry, Hunan Brain Hospital (The Second People's Hospital of Hunan Province), Changsha, Hunan, China
| | - Sixin Li
- Department of Psychiatry, School of Clinical Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Department of Psychiatry, Hunan Brain Hospital (The Second People's Hospital of Hunan Province), Changsha, Hunan, China
| | - Gaoya Zhou
- Department of Neurology, School of Clinical Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Department of Neurology, Hunan Brain Hospital (The Second People's Hospital of Hunan Province), Changsha, Hunan, China
| | - Caijuan Xu
- Department of Psychiatry, School of Clinical Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Department of Psychiatry, Hunan Brain Hospital (The Second People's Hospital of Hunan Province), Changsha, Hunan, China
| | - Xi Chen
- Department of Psychiatry, School of Clinical Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Department of Psychiatry, Hunan Brain Hospital (The Second People's Hospital of Hunan Province), Changsha, Hunan, China
| | - Huiwen Qiu
- Department of Psychiatry, School of Clinical Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Department of Psychiatry, Hunan Brain Hospital (The Second People's Hospital of Hunan Province), Changsha, Hunan, China
| | - Xinyu Li
- Department of Psychiatry, School of Clinical Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Department of Psychiatry, Hunan Brain Hospital (The Second People's Hospital of Hunan Province), Changsha, Hunan, China
| | - Ying Liu
- Department of Psychiatry, School of Clinical Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Department of Psychiatry, Hunan Brain Hospital (The Second People's Hospital of Hunan Province), Changsha, Hunan, China
| | - Hui Cao
- Department of Psychiatry, School of Clinical Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Department of Psychiatry, Hunan Brain Hospital (The Second People's Hospital of Hunan Province), Changsha, Hunan, China
| | - Changlong Bi
- Department of Neurosurgery, Xiangya Hospital, Center South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
21
|
Faustini SE, Hall A, Brown S, Roberts S, Hill H, Stamataki Z, Jenner MW, Owen RG, Pratt G, Cook G, Richter A, Drayson MT, Kaiser MF, Heaney JLJ. Immune responses to COVID-19 booster vaccinations in intensively anti-CD38 antibody treated patients with ultra-high-risk multiple myeloma: results from the Myeloma UK (MUK) nine OPTIMUM trial. Br J Haematol 2023; 201:845-850. [PMID: 36895158 DOI: 10.1111/bjh.18714] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 02/10/2023] [Accepted: 02/14/2023] [Indexed: 03/11/2023]
Abstract
Multiple myeloma (MM) and anti-MM therapy cause profound immunosuppression, leaving patients vulnerable to coronavirus disease 2019 (COVID-19) and other infections. We investigated anti-severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) antibodies longitudinally in ultra-high-risk patients with MM receiving risk-adapted, intensive anti-CD38 combined therapy in the Myeloma UK (MUK) nine trial. Despite continuous intensive therapy, seroconversion was achieved in all patients, but required a greater number of vaccinations compared to healthy individuals, highlighting the importance of booster vaccinations in this population. Reassuringly, high antibody cross-reactivity was found with current variants of concern, prior to Omicron subvariant adapted boostering. Multiple booster vaccine doses can provide effective protection from COVID-19, even with intensive anti-CD38 therapy for high-risk MM.
Collapse
Affiliation(s)
- Sian E Faustini
- Clinical Immunology Service, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Andrew Hall
- Clinical Trials Research Unit, Leeds Institute of Clinical Trials Research, University of Leeds, Leeds, UK
| | - Sarah Brown
- Clinical Trials Research Unit, Leeds Institute of Clinical Trials Research, University of Leeds, Leeds, UK
| | - Sadie Roberts
- Clinical Trials Research Unit, Leeds Institute of Clinical Trials Research, University of Leeds, Leeds, UK
| | - Harriet Hill
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Zania Stamataki
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Matthew W Jenner
- Department of Haematology, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Roger G Owen
- The Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Guy Pratt
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Gordon Cook
- Clinical Trials Research Unit, Leeds Institute of Clinical Trials Research, University of Leeds, Leeds, UK
- The Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Alex Richter
- Clinical Immunology Service, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Mark T Drayson
- Clinical Immunology Service, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Martin F Kaiser
- The Royal Marsden Hospital NHS Foundation Trust, London, UK
- The Institute of Cancer Research, London, UK
| | - Jennifer L J Heaney
- Clinical Immunology Service, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| |
Collapse
|
22
|
van Leeuwen LPM, Grobben M, GeurtsvanKessel CH, Ellerbroek PM, de Bree GJ, Potjewijd J, Rutgers A, Jolink H, van de Veerdonk FL, van Gils MJ, de Vries RD, Dalm VASH. Immune Responses 6 Months After mRNA-1273 COVID-19 Vaccination and the Effect of a Third Vaccination in Patients with Inborn Errors of Immunity. J Clin Immunol 2023:10.1007/s10875-023-01514-7. [PMID: 37231290 DOI: 10.1007/s10875-023-01514-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 05/08/2023] [Indexed: 05/27/2023]
Abstract
PURPOSE Patients with inborn errors of immunity (IEI) are at increased risk of severe coronavirus disease-2019 (COVID-19). Effective long-term protection against COVID-19 is therefore of great importance in these patients, but little is known about the decay of the immune response after primary vaccination. We studied the immune responses 6 months after two mRNA-1273 COVID-19 vaccines in 473 IEI patients and subsequently the response to a third mRNA COVID-19 vaccine in 50 patients with common variable immunodeficiency (CVID). METHODS In a prospective multicenter study, 473 IEI patients (including X-linked agammaglobulinemia (XLA) (N = 18), combined immunodeficiency (CID) (N = 22), CVID (N = 203), isolated or undefined antibody deficiencies (N = 204), and phagocyte defects (N = 16)), and 179 controls were included and followed up to 6 months after two doses of the mRNA-1273 COVID-19 vaccine. Additionally, samples were collected from 50 CVID patients who received a third vaccine 6 months after primary vaccination through the national vaccination program. SARS-CoV-2-specific IgG titers, neutralizing antibodies, and T cell responses were assessed. RESULTS At 6 months after vaccination, the geometric mean antibody titers (GMT) declined in both IEI patients and healthy controls, when compared to GMT 28 days after vaccination. The trajectory of this decline did not differ between controls and most IEI cohorts; however, antibody titers in CID, CVID, and isolated antibody deficiency patients more often dropped to below the responder cut-off compared to controls. Specific T cell responses were still detectable in 77% of controls and 68% of IEI patients at 6 months post vaccination. A third mRNA vaccine resulted in an antibody response in only two out of 30 CVID patients that did not seroconvert after two mRNA vaccines. CONCLUSION A similar decline in IgG titers and T cell responses was observed in patients with IEI when compared to healthy controls 6 months after mRNA-1273 COVID-19 vaccination. The limited beneficial benefit of a third mRNA COVID-19 vaccine in previous non-responder CVID patients implicates that other protective strategies are needed for these vulnerable patients.
Collapse
Affiliation(s)
- Leanne P M van Leeuwen
- Department of Viroscience, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands
- Travel Clinic, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Marloes Grobben
- Department of Medical Microbiology and Infection Prevention, Amsterdam Institute for Infection and Immunity, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Corine H GeurtsvanKessel
- Department of Viroscience, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands
| | | | - Godelieve J de Bree
- Department of Infectious Diseases, Amsterdam UMC, Amsterdam, The Netherlands
| | - Judith Potjewijd
- Department of Internal Medicine, Division Nephrology and Clinical Immunology, Maastricht UMC, Maastricht, The Netherlands
| | - Abraham Rutgers
- Department of Rheumatology and Clinical Immunology, UMC Groningen, Groningen, The Netherlands
| | - Hetty Jolink
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Frank L van de Veerdonk
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Marit J van Gils
- Department of Medical Microbiology and Infection Prevention, Amsterdam Institute for Infection and Immunity, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Rory D de Vries
- Department of Viroscience, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Virgil A S H Dalm
- Department of Internal Medicine, Division of Allergy & Clinical Immunology, Erasmus MC University Medical Center Rotterdam, Doctor Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands.
- Department of Immunology, Erasmus MC University Medical Center Rotterdam, Doctor Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands.
| |
Collapse
|
23
|
Lucane Z, Slisere B, Gersone G, Papirte S, Gailite L, Tretjakovs P, Kurjane N. Cytokine Response Following SARS-CoV-2 Antigen Stimulation in Patients with Predominantly Antibody Deficiencies. Viruses 2023; 15:v15051146. [PMID: 37243231 DOI: 10.3390/v15051146] [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: 03/29/2023] [Revised: 05/02/2023] [Accepted: 05/09/2023] [Indexed: 05/28/2023] Open
Abstract
Predominantly antibody deficiencies (PADs) are inborn disorders characterized by immune dysregulation and increased susceptibility to infections. Response to vaccination, including severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), may be impaired in these patients, and studies on responsiveness correlates, including cytokine signatures to antigen stimulation, are sparse. In this study, we aimed to describe the spike-specific cytokine response following whole-blood stimulation with SARS-CoV-2 spike peptides in patients with PAD (n = 16 with common variable immunodeficiency and n = 15 with selective IgA deficiency) and its relationship with the occurrence of coronavirus disease 2019 (COVID-19) during up to 10-month follow-up period. Spike-induced antibody and cytokine production was measured using ELISA (anti-spike IgG, IFN-γ) and xMAP technology (interleukin-1β (IL-1β), IL-4, IL-6, IL-10, IL-15, IL-17A, IL-21, TNF-α, TGF-β1). No difference was found in the production of cytokines between patients with PAD and controls. Anti-spike IgG and cytokine levels did not predict contraction of COVID-19. The only cytokine that distinguished between vaccinated and naturally infected unvaccinated PAD patients was IFN-γ (median 0.64 (IQR = 1.08) in vaccinated vs. 0.10 (IQR = 0.28) in unvaccinated). This study describes the spike-specific cytokine response to SARS-CoV-2 antigens, which is not predictive of contracting COVID-19 during the follow-up.
Collapse
Affiliation(s)
- Zane Lucane
- Department of Biology and Microbiology, Riga Stradins University, LV-1007 Riga, Latvia
| | - Baiba Slisere
- The Joint Laboratory, Pauls Stradins Clinical University Hospital, LV-1002 Riga, Latvia
- Department of Internal Diseases, Riga Stradins University, LV-1007 Riga, Latvia
| | - Gita Gersone
- Department of Human Physiology and Biochemistry, Riga Stradins University, LV-1007 Riga, Latvia
| | - Sindija Papirte
- Faculty of Medicine, Riga Stradins University, LV-1007 Riga, Latvia
| | - Linda Gailite
- Scientific Laboratory of Molecular Genetics, Riga Stradins University, LV-1007 Riga, Latvia
| | - Peteris Tretjakovs
- Department of Human Physiology and Biochemistry, Riga Stradins University, LV-1007 Riga, Latvia
| | - Natalja Kurjane
- Department of Biology and Microbiology, Riga Stradins University, LV-1007 Riga, Latvia
- Outpatient Clinic, Pauls Stradins Clinical University Hospital, LV-1002 Riga, Latvia
- Outpatient Clinic, Children's Clinical University Hospital, LV-1004 Riga, Latvia
| |
Collapse
|
24
|
Tani Y, Takita M, Kobashi Y, Wakui M, Zhao T, Yamamoto C, Saito H, Kawashima M, Sugiura S, Nishikawa Y, Omata F, Shimazu Y, Kawamura T, Sugiyama A, Nakayama A, Kaneko Y, Kodama T, Kami M, Tsubokura M. Varying Cellular Immune Response against SARS-CoV-2 after the Booster Vaccination: A Cohort Study from Fukushima Vaccination Community Survey, Japan. Vaccines (Basel) 2023; 11:vaccines11050920. [PMID: 37243024 DOI: 10.3390/vaccines11050920] [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: 04/03/2023] [Revised: 04/24/2023] [Accepted: 04/27/2023] [Indexed: 05/28/2023] Open
Abstract
Booster vaccination reduces the incidence of severe cases and mortality related to COVID-19, with cellular immunity playing an important role. However, little is known about the proportion of the population that has achieved cellular immunity after booster vaccination. Thus, we conducted a Fukushima cohort database and assessed humoral and cellular immunity in 2526 residents and healthcare workers in Fukushima Prefecture in Japan through continuous blood collection every 3 months from September 2021. We identified the proportion of people with induced cellular immunity after booster vaccination using the T-SPOT.COVID test, and analyzed their background characteristics. Among 1089 participants, 64.3% (700/1089) had reactive cellular immunity after booster vaccination. Multivariable analysis revealed the following independent predictors of reactive cellular immunity: age < 40 years (adjusted odds ratio: 1.81; 95% confidence interval: 1.19-2.75; p-value: 0.005) and adverse reactions after vaccination (1.92, 1.19-3.09, 0.007). Notably, despite IgG(S) and neutralizing antibody titers of ≥500 AU/mL, 33.9% (349/1031) and 33.5% (341/1017) of participants, respectively, did not have reactive cellular immunity. In summary, this is the first study to evaluate cellular immunity at the population level after booster vaccination using the T-SPOT.COVID test, albeit with several limitations. Future studies will need to evaluate previously infected subjects and their T-cell subsets.
Collapse
Affiliation(s)
- Yuta Tani
- Medical Governance Research Institute, Tokyo 108-0074, Japan
| | - Morihito Takita
- Medical Governance Research Institute, Tokyo 108-0074, Japan
- Department of Radiation Health Management, Fukushima Medical University, Fukushima 960-1295, Japan
| | - Yurie Kobashi
- Department of Radiation Health Management, Fukushima Medical University, Fukushima 960-1295, Japan
- Department of General Internal Medicine, Hirata Central Hospital, Fukushima 963-8202, Japan
| | - Masatoshi Wakui
- Department of Laboratory Medicine, Keio University School of Medicine, Tokyo 160-0016, Japan
| | - Tianchen Zhao
- Department of Radiation Health Management, Fukushima Medical University, Fukushima 960-1295, Japan
| | - Chika Yamamoto
- Department of Radiation Health Management, Fukushima Medical University, Fukushima 960-1295, Japan
| | - Hiroaki Saito
- Department of Radiation Health Management, Fukushima Medical University, Fukushima 960-1295, Japan
- Department of Internal Medicine, Soma Central Hospital, Fukushima 976-0016, Japan
| | - Moe Kawashima
- Department of Radiation Health Management, Fukushima Medical University, Fukushima 960-1295, Japan
| | - Sota Sugiura
- Medical Governance Research Institute, Tokyo 108-0074, Japan
| | - Yoshitaka Nishikawa
- Department of General Internal Medicine, Hirata Central Hospital, Fukushima 963-8202, Japan
| | - Fumiya Omata
- Department of General Internal Medicine, Hirata Central Hospital, Fukushima 963-8202, Japan
| | - Yuzo Shimazu
- Department of General Internal Medicine, Hirata Central Hospital, Fukushima 963-8202, Japan
| | - Takeshi Kawamura
- Proteomics Laboratory, Isotope Science Center, The University of Tokyo, Tokyo 113-0032, Japan
- Laboratory for Systems Biology and Medicine, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo 153-8904, Japan
| | - Akira Sugiyama
- Proteomics Laboratory, Isotope Science Center, The University of Tokyo, Tokyo 113-0032, Japan
| | - Aya Nakayama
- Proteomics Laboratory, Isotope Science Center, The University of Tokyo, Tokyo 113-0032, Japan
| | - Yudai Kaneko
- Laboratory for Systems Biology and Medicine, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo 153-8904, Japan
- Medical and Biological Laboratories Co., Ltd., Tokyo 105-0012, Japan
| | - Tetsuhiko Kodama
- Laboratory for Systems Biology and Medicine, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo 153-8904, Japan
| | - Masahiro Kami
- Medical Governance Research Institute, Tokyo 108-0074, Japan
| | - Masaharu Tsubokura
- Department of Radiation Health Management, Fukushima Medical University, Fukushima 960-1295, Japan
- Department of General Internal Medicine, Hirata Central Hospital, Fukushima 963-8202, Japan
- Department of Internal Medicine, Soma Central Hospital, Fukushima 976-0016, Japan
| |
Collapse
|
25
|
Tangye SG. Impact of SARS-CoV-2 infection and COVID-19 on patients with inborn errors of immunity. J Allergy Clin Immunol 2023; 151:818-831. [PMID: 36522221 PMCID: PMC9746792 DOI: 10.1016/j.jaci.2022.11.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 11/02/2022] [Accepted: 11/04/2022] [Indexed: 12/15/2022]
Abstract
Since the arrival of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in December 2019, its characterization as a novel human pathogen, and the resulting coronavirus disease 2019 (COVID-19) pandemic, over 6.5 million people have died worldwide-a stark and sobering reminder of the fundamental and nonredundant roles of the innate and adaptive immune systems in host defense against emerging pathogens. Inborn errors of immunity (IEI) are caused by germline variants, typically in single genes. IEI are characterized by defects in development and/or function of cells involved in immunity and host defense, rendering individuals highly susceptible to severe, recurrent, and sometimes fatal infections, as well as immune dysregulatory conditions such as autoinflammation, autoimmunity, and allergy. The study of IEI has revealed key insights into the molecular and cellular requirements for immune-mediated protection against infectious diseases. Indeed, this has been exemplified by assessing the impact of SARS-CoV-2 infection in individuals with previously diagnosed IEI, as well as analyzing rare cases of severe COVID-19 in otherwise healthy individuals. This approach has defined fundamental aspects of mechanisms of disease pathogenesis, immunopathology in the context of infection with a novel pathogen, and therapeutic options to mitigate severe disease. This review summarizes these findings and illustrates how the study of these rare experiments of nature can inform key features of human immunology, which can then be leveraged to improve therapies for treating emerging and established infectious diseases.
Collapse
Affiliation(s)
- Stuart G Tangye
- Garvan Institute of Medical Research, Darlinghurst, Darlinghurst, Australia; St Vincent's Clinical School, University of New South Wales Sydney, Randwick, Randwick, Australia; Clinical Immunogenomics Research Consortium of Australasia (CIRCA).
| |
Collapse
|
26
|
Mohamed KM, Guevara-Hoyer K, García CJ, Bravo LG, Jiménez-Huete A, de la Peña AR, Valeros BM, Velázquez CC, López EC, Cabello N, Estrada V, Corbí ÁL, Fernández-Arquero M, Ocaña A, Delgado-Iribarren A, Martínez-Novillo M, Bolaños E, Anguita E, Peña A, Benavente C, Benítez Fuentes JD, Pérez Segura P, Sánchez-Ramón S. Specific Cellular and Humoral Immune Responses to the Neoantigen RBD of SARS-CoV-2 in Patients with Primary and Secondary Immunodeficiency and Healthy Donors. Biomedicines 2023; 11:biomedicines11041042. [PMID: 37189660 DOI: 10.3390/biomedicines11041042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/14/2023] [Accepted: 03/24/2023] [Indexed: 03/30/2023] Open
Abstract
Patients with antibody deficiency disorders, such as primary immunodeficiency (PID) or secondary immunodeficiency (SID) to B-cell lymphoproliferative disorder (B-CLPD), are two groups vulnerable to developing the severe or chronic form of coronavirus disease caused by SARS-CoV-2 (COVID-19). The data on adaptive immune responses against SARS-CoV-2 are well described in healthy donors, but still limited in patients with antibody deficiency of a different cause. Herein, we analyzed spike-specific IFN-γ and anti-spike IgG antibody responses at 3 to 6 months after exposure to SARS-CoV-2 derived from vaccination and/or infection in two cohorts of immunodeficient patients (PID vs. SID) compared to healthy controls (HCs). Pre-vaccine anti-SARS-CoV-2 cellular responses before vaccine administration were measured in 10 PID patients. Baseline cellular responses were detectable in 4 out of 10 PID patients who had COVID-19 prior to vaccination, perceiving an increase in cellular responses after two-dose vaccination (p < 0.001). Adequate specific cellular responses were observed in 18 out of 20 (90%) PID patients, in 14 out of 20 (70%) SID patients and in 74 out of 81 (96%) HCs after vaccination (and natural infection in some cases). Specific IFN-γ response was significantly higher in HC with respect to PID (1908.5 mUI/mL vs. 1694.1 mUI/mL; p = 0.005). Whereas all SID and HC patients mounted a specific humoral immune response, only 80% of PID patients showed positive anti-SARS-CoV-2 IgG. The titer of anti-SARS-CoV-2 IgG was significantly lower in SID compared with HC patients (p = 0.040), without significant differences between PID and HC patients (p = 0.123) and between PID and SID patients (p =0.683). High proportions of PID and SID patients showed adequate specific cellular responses to receptor binding domain (RBD) neoantigen, with a divergence between the two arms of the adaptive immune response in PID and SID patients. We also focused on the correlation of protection of positive SARS-CoV-2 cellular response to omicron exposure: 27 out of 81 (33.3%) HCs referred COVID-19 detected by PCR or antigen test, 24 with a mild course, 1 with moderate symptoms and the remaining 2 with bilateral pneumonia that were treated in an outpatient basis. Our results might support the relevance of these immunological studies to determine the correlation of protection with severe disease and for deciding the need for additional boosters on a personalized basis. Follow-up studies are required to evaluate the duration and variability in the immune response to COVID-19 vaccination or infection.
Collapse
Affiliation(s)
- Kauzar Mohamed Mohamed
- Department of Immunology, Laboratory Medicine Institute (IML) and Fundación para la Investigación Biomédica del Hospital Clínico San Carlos (IdISSC), Hospital Clínico San Carlos, Calle Profesor Martín Lagos SN, 28040 Madrid, Spain
| | - Kissy Guevara-Hoyer
- Department of Immunology, Laboratory Medicine Institute (IML) and Fundación para la Investigación Biomédica del Hospital Clínico San Carlos (IdISSC), Hospital Clínico San Carlos, Calle Profesor Martín Lagos SN, 28040 Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Complutense University, 28040 Madrid, Spain
| | - Carlos Jiménez García
- Department of Immunology, Laboratory Medicine Institute (IML) and Fundación para la Investigación Biomédica del Hospital Clínico San Carlos (IdISSC), Hospital Clínico San Carlos, Calle Profesor Martín Lagos SN, 28040 Madrid, Spain
| | - Laura García Bravo
- Department of Immunology, Laboratory Medicine Institute (IML) and Fundación para la Investigación Biomédica del Hospital Clínico San Carlos (IdISSC), Hospital Clínico San Carlos, Calle Profesor Martín Lagos SN, 28040 Madrid, Spain
| | | | - Antonia Rodríguez de la Peña
- Department of Immunology, Laboratory Medicine Institute (IML) and Fundación para la Investigación Biomédica del Hospital Clínico San Carlos (IdISSC), Hospital Clínico San Carlos, Calle Profesor Martín Lagos SN, 28040 Madrid, Spain
| | - Beatriz Mediero Valeros
- Department of Immunology, Laboratory Medicine Institute (IML) and Fundación para la Investigación Biomédica del Hospital Clínico San Carlos (IdISSC), Hospital Clínico San Carlos, Calle Profesor Martín Lagos SN, 28040 Madrid, Spain
| | - Cristina Cañizares Velázquez
- Department of Immunology, Laboratory Medicine Institute (IML) and Fundación para la Investigación Biomédica del Hospital Clínico San Carlos (IdISSC), Hospital Clínico San Carlos, Calle Profesor Martín Lagos SN, 28040 Madrid, Spain
| | - Esther Culebras López
- Department of Microbiology, IML and IdISSC, Hospital Clínico San Carlos, 28040 Madrid, Spain
| | - Noemí Cabello
- Unit of Infectious Diseases, Department of Internal Medicine, Hospital Clínico San Carlos, Calle Profesor Martín Lagos SN, 28040 Madrid, Spain
| | - Vicente Estrada
- Unit of Infectious Diseases, Department of Internal Medicine, Hospital Clínico San Carlos, Calle Profesor Martín Lagos SN, 28040 Madrid, Spain
| | - Ángel L Corbí
- Centro de Investigaciones Biológicas (CSIC), C./Ramiro de Maeztu, 9, 28040 Madrid, Spain
| | - Miguel Fernández-Arquero
- Department of Immunology, Laboratory Medicine Institute (IML) and Fundación para la Investigación Biomédica del Hospital Clínico San Carlos (IdISSC), Hospital Clínico San Carlos, Calle Profesor Martín Lagos SN, 28040 Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Complutense University, 28040 Madrid, Spain
| | - Alberto Ocaña
- Department of Microbiology, IML and IdISSC, Hospital Clínico San Carlos, 28040 Madrid, Spain
| | | | - Mercedes Martínez-Novillo
- Clinical Analysis Department, Laboratory Medicine Institute (IML) and Fundación para la Investigación Biomédica del Hospital Clínico San Carlos (IdISSC), Hospital Clínico San Carlos, Calle Profesor Martín Lagos SN, 28040 Madrid, Spain
| | - Estefanía Bolaños
- Department of Hematology, Hospital Clínico San Carlos, IML, IdISSC, Calle Profesor Martín Lagos SN, 28040 Madrid, Spain
| | - Eduardo Anguita
- Department of Hematology, Hospital Clínico San Carlos, IML, IdISSC, Calle Profesor Martín Lagos SN, 28040 Madrid, Spain
| | - Ascensión Peña
- Department of Hematology, Hospital Clínico San Carlos, IML, IdISSC, Calle Profesor Martín Lagos SN, 28040 Madrid, Spain
| | - Celina Benavente
- Department of Hematology, Hospital Clínico San Carlos, IML, IdISSC, Calle Profesor Martín Lagos SN, 28040 Madrid, Spain
| | - Javier David Benítez Fuentes
- Department of Medical Oncology, Hospital Clínico San Carlos, Calle Profesor Martín Lagos SN, 28040 Madrid, Spain
| | - Pedro Pérez Segura
- Department of Medical Oncology, Hospital Clínico San Carlos, Calle Profesor Martín Lagos SN, 28040 Madrid, Spain
| | - Silvia Sánchez-Ramón
- Department of Immunology, Laboratory Medicine Institute (IML) and Fundación para la Investigación Biomédica del Hospital Clínico San Carlos (IdISSC), Hospital Clínico San Carlos, Calle Profesor Martín Lagos SN, 28040 Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Complutense University, 28040 Madrid, Spain
- Department of Clinical Immunology, Hospital Universitario Clínico San Carlos and IdISSC, Calle Profesor Martín Lagos SN, 28040 Madrid, Spain
| |
Collapse
|
27
|
Immunological Findings in a Group of Individuals Who Were Poor or Non-Responders to Standard Two-Dose SARS-CoV-2 Vaccines. Vaccines (Basel) 2023; 11:vaccines11020461. [PMID: 36851338 PMCID: PMC9963224 DOI: 10.3390/vaccines11020461] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 02/08/2023] [Accepted: 02/14/2023] [Indexed: 02/19/2023] Open
Abstract
Coronavirus disease (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has been declared a pandemic. However, data on the poor or non-responders to SARS-CoV-2 vaccines in the general population are limited. The objective of this study was to comprehensively compare the immunological characteristics of poor or non-responders to SARS-CoV-2 vaccines in the 18-59-year group with those in the ≥60-year group using internationally recognized cut-off values. The main outcome was effective seroconversion characterized by an anti-SARS-CoV-2 spike IgG level of at least a four-fold increase from baseline. Profiling of naïve immune cells was analyzed prior to vaccination to demonstrate baseline immunity. The outcomes of effective seroconversion in patients aged 18-59 years with those in patients aged ≥60 years were compared. The quantitative level of anti-spike IgG was significantly lower in individuals aged ≥60 and men aged 18-59 years. There were 7.5% of poor or non-responders among the 18-59 years and 11.7% of poor or non-responders in the ≥60 years using a four-fold increase parameter. There were 37.0-58.1% with low lymphocyte count (<1000/mm3), 33.3-45.2% with low CD4 cell counts (<500/mm3), and 74.1-96.8% with low B cell counts (<100/mm3) in the non-seroconversion group. An individual with an anti-SARS-CoV-2 spike IgG titer below 50 BAU/mL might be considered a poor or non-responder between 14 and 90 days after the last vaccine dose. Booster vaccination or additional protective measures should be recommended to poor or non-responders as soon as possible to reduce disease severity and mortality.
Collapse
|
28
|
Milito C, Firinu D, Bez P, Villa A, Punziano A, Lagnese G, Costanzo G, van Leeuwen LPM, Piazza B, Deiana CM, d’Ippolito G, Del Giacco SR, Rattazzi M, Spadaro G, Quinti I, Scarpa R, Dalm VASH, Cinetto F. A beacon in the dark: COVID-19 course in CVID patients from two European countries: Different approaches, similar outcomes. Front Immunol 2023; 14:1093385. [PMID: 36845159 PMCID: PMC9944020 DOI: 10.3389/fimmu.2023.1093385] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 01/09/2023] [Indexed: 02/10/2023] Open
Abstract
Background CVID patients present an increased risk of prolonged SARS-CoV-2 infection and re-infection and a higher COVID-19-related morbidity and mortality compared to the general population. Since 2021, different therapeutic and prophylactic strategies have been employed in vulnerable groups (vaccination, SARS-CoV-2 monoclonal antibodies and antivirals). The impact of treatments over the last 2 years has not been explored in international studies considering the emergence of viral variants and different management between countries. Methods A multicenter retrospective/prospective real-life study comparing the prevalence and outcomes of SARS-CoV-2 infection between a CVID cohort from four Italian Centers (IT-C) and one cohort from the Netherlands (NL-C), recruiting 773 patients. Results 329 of 773 CVID patients were found positive for SARS-CoV-2 infection between March 1st, 2020 and September 1st 2022. The proportion of CVID patients infected was comparable in both national sub-cohorts. During all waves, chronic lung disease, "complicated" phenotype, chronic immunosuppressive treatment and cardiovascular comorbidities impacted on hospitalization, whereas risk factors for mortality were older age, chronic lung disease, and bacterial superinfections. IT-C patients were significantly more often treated, both with antivirals and mAbs, than NL-C patients. Outpatient treatment, available only in Italy, started from the Delta wave. Despite this, no significant difference was found for COVID-19 severity between the two cohorts. However, pooling together specific SARS-CoV-2 outpatient treatments (mAbs and antivirals), we found a significant effect on the risk of hospitalization starting from Delta wave. Vaccination with ≥ 3 doses shortened RT-PCR positivity, with an additional effect only in patients receiving antivirals. Conclusions The two sub-cohorts had similar COVID-19 outcomes despite different treatment approaches. This points out that specific treatment should now be reserved for selected subgroups of CVID patients, based on pre-existing conditions.
Collapse
Affiliation(s)
- Cinzia Milito
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Davide Firinu
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Patrick Bez
- Rare Diseases Referral Center, Internal Medicine 1, Ca’ Foncello Hospital, AULSS2 Marca Trevigiana, Department of Medicine (DIMED), University of Padova, Padova, Italy
| | - Annalisa Villa
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Alessandra Punziano
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Gianluca Lagnese
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Giulia Costanzo
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Leanne P. M. van Leeuwen
- Department of Viroscience, Travel Clinic, Erasmus University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Beatrice Piazza
- Rare Diseases Referral Center, Internal Medicine 1, Ca’ Foncello Hospital, AULSS2 Marca Trevigiana, Department of Medicine (DIMED), University of Padova, Padova, Italy
| | - Carla Maria Deiana
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | | | | | - Marcello Rattazzi
- Rare Diseases Referral Center, Internal Medicine 1, Ca’ Foncello Hospital, AULSS2 Marca Trevigiana, Department of Medicine (DIMED), University of Padova, Padova, Italy
| | - Giuseppe Spadaro
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Isabella Quinti
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Riccardo Scarpa
- Rare Diseases Referral Center, Internal Medicine 1, Ca’ Foncello Hospital, AULSS2 Marca Trevigiana, Department of Medicine (DIMED), University of Padova, Padova, Italy,*Correspondence: Riccardo Scarpa,
| | - Virgil A. S. H. Dalm
- Department of Internal Medicine, Division of Allergy and Clinical Immunology, Department of Immunology, Erasmus University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Francesco Cinetto
- Rare Diseases Referral Center, Internal Medicine 1, Ca’ Foncello Hospital, AULSS2 Marca Trevigiana, Department of Medicine (DIMED), University of Padova, Padova, Italy
| |
Collapse
|
29
|
Long-Term Immunological Memory of SARS-CoV-2 Is Present in Patients with Primary Antibody Deficiencies for up to a Year after Vaccination. Vaccines (Basel) 2023; 11:vaccines11020354. [PMID: 36851231 PMCID: PMC9959530 DOI: 10.3390/vaccines11020354] [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: 01/02/2023] [Revised: 01/30/2023] [Accepted: 02/01/2023] [Indexed: 02/05/2023] Open
Abstract
Some studies have found increased coronavirus disease-19 (COVID-19)-related morbidity and mortality in patients with primary antibody deficiencies. Immunization against COVID-19 may, therefore, be particularly important in these patients. However, the durability of the immune response remains unclear in such patients. In this study, we evaluated the cellular and humoral response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) antigens in a cross-sectional study of 32 patients with primary antibody deficiency (n = 17 with common variable immunodeficiency (CVID) and n = 15 with selective IgA deficiency) and 15 healthy controls. Serological and cellular responses were determined using enzyme-linked immunosorbent assay and interferon-gamma release assays. The subsets of B and T lymphocytes were measured using flow cytometry. Of the 32 patients, 28 had completed the vaccination regimen with a median time after vaccination of 173 days (IQR = 142): 27 patients showed a positive spike-peptide-specific antibody response, and 26 patients showed a positive spike-peptide-specific T-cell response. The median level of antibody response in CVID patients (5.47 ratio (IQR = 4.08)) was lower compared to healthy controls (9.43 ratio (IQR = 2.13)). No difference in anti-spike T-cell response was found between the groups. The results of this study indicate that markers of the sustained SARS-CoV-2 spike-specific immune response are detectable several months after vaccination in patients with primary antibody deficiencies comparable to controls.
Collapse
|
30
|
Milito C, Cinetto F, Garzi G, Palladino A, Puca M, Brambilla E, De Vitis C, Costanzo G, Scarpa R, Punziano A, Lagnese G, Del Giacco S, Spadaro G, Quinti I, Firinu D. Safety of mRNA COVID-19 Vaccines in Patients with Inborn Errors of Immunity: an Italian Multicentric Study. J Clin Immunol 2023; 43:299-307. [PMID: 36374363 PMCID: PMC9662105 DOI: 10.1007/s10875-022-01402-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 10/31/2022] [Indexed: 11/16/2022]
Abstract
PURPOSE Little is known about vaccine safety in inborn errors of immunity (IEI) patients during the current vaccination campaign for COVID-19. To better investigate the reactogenicity and adverse event profile after two, three, and four doses of mRNA vaccines, we conducted an observational, multicentric study on 342 PID patients from four Italian Referral Centres. METHODS We conducted a survey on self-reported adverse reactions in IEI patients who received mRNA vaccine by administering a questionnaire after each dose. RESULTS Over the whole study period, none of the patients needed hospitalization or had hypersensitivity reactions, including anaphylaxis and delayed injection site reaction. After two vaccination doses, 35.4% of patients showed only local reactogenicity-related symptoms (RrS), 44.4% reported both systemic and local RrS, and 5% reported only systemic RrS. In more than 60% of cases, local or systemic RrS were mild. After the first and second booster doses, patients showed fewer adverse events (AEs) than after the first vaccination course. Patients aged 50 years and older reported adverse events and RrS less frequently. Among AEs requiring treatment, one common variable immune deficiency patient affected by T cell large granular lymphocytic leukemia developed neutropenia and one patient had Bell's paralysis perhaps during herpes zoster reactivation. CONCLUSION Although our follow-up period is relatively short, the safety data we reported are reassuring. This data would help to contrast the vaccine hesitancy often manifested by patients with IEI and to better inform their healthcare providers.
Collapse
Affiliation(s)
- Cinzia Milito
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Francesco Cinetto
- Department of Medicine-DIMED, University of Padova, Padua, Italy
- Rare Diseases Referral Center, Internal Medicine I, Ca' Foncello Hospital, AULSS2 Marca Trevigiana, Treviso, Italy
| | - Giulia Garzi
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Andrea Palladino
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Marco Puca
- Department of Medical Sciences and Public Health, University of Cagliari, Azienda Ospedaliero Universitaria, SS 554-Bivio Sestu, 09042, Monserrato, CA, Italy
| | - Elena Brambilla
- Department of Medicine-DIMED, University of Padova, Padua, Italy
- Rare Diseases Referral Center, Internal Medicine I, Ca' Foncello Hospital, AULSS2 Marca Trevigiana, Treviso, Italy
| | - Camilla De Vitis
- Department of Medicine-DIMED, University of Padova, Padua, Italy
- Rare Diseases Referral Center, Internal Medicine I, Ca' Foncello Hospital, AULSS2 Marca Trevigiana, Treviso, Italy
| | - Giulia Costanzo
- Department of Medical Sciences and Public Health, University of Cagliari, Azienda Ospedaliero Universitaria, SS 554-Bivio Sestu, 09042, Monserrato, CA, Italy
| | - Riccardo Scarpa
- Department of Medicine-DIMED, University of Padova, Padua, Italy
- Rare Diseases Referral Center, Internal Medicine I, Ca' Foncello Hospital, AULSS2 Marca Trevigiana, Treviso, Italy
| | - Alessandra Punziano
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Gianluca Lagnese
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Stefano Del Giacco
- Department of Medical Sciences and Public Health, University of Cagliari, Azienda Ospedaliero Universitaria, SS 554-Bivio Sestu, 09042, Monserrato, CA, Italy
| | - Giuseppe Spadaro
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Isabella Quinti
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Davide Firinu
- Department of Medical Sciences and Public Health, University of Cagliari, Azienda Ospedaliero Universitaria, SS 554-Bivio Sestu, 09042, Monserrato, CA, Italy.
| |
Collapse
|
31
|
Nadesalingam A, Cantoni D, Aguinam ET, Chan AC, Paloniemi M, Ohlendorf L, George C, Carnell G, Lyall J, Ferrari M, Temperton N, Wagner R, Castillo-Olivares J, Baxendale H, Heeney JL. Vaccination and protective immunity to SARS-CoV-2 omicron variants in people with immunodeficiencies. THE LANCET. MICROBE 2023; 4:e58-e59. [PMID: 36332646 PMCID: PMC9625114 DOI: 10.1016/s2666-5247(22)00297-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/16/2022] [Accepted: 10/03/2022] [Indexed: 11/07/2022]
Affiliation(s)
- Angalee Nadesalingam
- Laboratory of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Cambridge CB3 OES, UK
| | - Diego Cantoni
- Viral Pseudotype Unit, Medway School of Pharmacy, University of Kent, Medway, UK
| | - Ernest T Aguinam
- Laboratory of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Cambridge CB3 OES, UK
| | - Andrew Cy Chan
- Laboratory of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Cambridge CB3 OES, UK
| | - Minna Paloniemi
- Laboratory of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Cambridge CB3 OES, UK
| | - Luis Ohlendorf
- Laboratory of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Cambridge CB3 OES, UK
| | - Charlotte George
- Laboratory of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Cambridge CB3 OES, UK
| | - George Carnell
- Laboratory of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Cambridge CB3 OES, UK
| | - Jon Lyall
- Laboratory of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Cambridge CB3 OES, UK
| | - Matteo Ferrari
- Laboratory of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Cambridge CB3 OES, UK
| | - Nigel Temperton
- Viral Pseudotype Unit, Medway School of Pharmacy, University of Kent, Medway, UK
| | - Ralf Wagner
- Institute of Medical Microbiology and Hygiene, University of Regensburg, Regensburg, Germany
| | - Javier Castillo-Olivares
- Laboratory of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Cambridge CB3 OES, UK
| | - Helen Baxendale
- Clinical Immunology Department, Royal Papworth NHS Foundation Trust, Cambridge, UK
| | - Jonathan L Heeney
- Laboratory of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Cambridge CB3 OES, UK.
| |
Collapse
|
32
|
Erra L, Uriarte I, Colado A, Paolini MV, Seminario G, Fernández JB, Tau L, Bernatowiez J, Moreira I, Vishnopolska S, Rumbo M, Cassarino C, Vijoditz G, López AL, Curciarello R, Rodríguez D, Rizzo G, Ferreyra M, Ferreyra Mufarregue LR, Badano MN, Pérez Millán MI, Quiroga MF, Baré P, Ibañez I, Pozner R, Borge M, Docena G, Bezrodnik L, Almejun MB. COVID-19 Vaccination Responses with Different Vaccine Platforms in Patients with Inborn Errors of Immunity. J Clin Immunol 2023; 43:271-285. [PMID: 36251205 PMCID: PMC9574808 DOI: 10.1007/s10875-022-01382-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Accepted: 10/05/2022] [Indexed: 02/07/2023]
Abstract
Patients with inborn errors of immunity (IEI) in Argentina were encouraged to receive licensed Sputnik, AstraZeneca, Sinopharm, Moderna, and Pfizer vaccines, even though most of the data of humoral and cellular responses combination on available vaccines comes from trials conducted in healthy individuals. We aimed to evaluate the safety and immunogenicity of the different vaccines in IEI patients in Argentina. The study cohort included adults and pediatric IEI patients (n = 118) and age-matched healthy controls (HC) (n = 37). B cell response was evaluated by measuring IgG anti-spike/receptor binding domain (S/RBD) and anti-nucleocapsid(N) antibodies by ELISA. Neutralization antibodies were also assessed with an alpha-S protein-expressing pseudo-virus assay. The T cell response was analyzed by IFN-γ secretion on S- or N-stimulated PBMC by ELISPOT and the frequency of S-specific circulating T follicular-helper cells (TFH) was evaluated by flow cytometry.No moderate/severe vaccine-associated adverse events were observed. Anti-S/RBD titers showed significant differences in both pediatric and adult IEI patients versus the age-matched HC cohort (p < 0.05). Neutralizing antibodies were also significantly lower in the patient cohort than in age-matched HC (p < 0.01). Positive S-specific IFN-γ response was observed in 84.5% of IEI patients and 82.1% presented S-specific TFH cells. Moderna vaccines, which were mainly administered in the pediatric population, elicited a stronger humoral response in IEI patients, both in antibody titer and neutralization capacity, but the cellular immune response was similar between vaccine platforms. No difference in humoral response was observed between vaccinated patients with and without previous SARS-CoV-2 infection.In conclusion, COVID-19 vaccines showed safety in IEI patients and, although immunogenicity was lower than HC, they showed specific anti-S/RBD IgG, neutralizing antibody titers, and T cell-dependent cellular immunity with IFN-γ secreting cells. These findings may guide the recommendation for a vaccination with all the available vaccines in IEI patients to prevent COVID-19 disease.
Collapse
Affiliation(s)
- Lorenzo Erra
- Departamento de Fisiología, Biología Molecular y Celular, Instituto de Biociencias, Biotecnología y Biología Traslacional (IB3) e Instituto de Química Biológica (IQUIBICEN), FCEN, UBA, CONICET, Buenos Aires, CABA, Argentina
| | - Ignacio Uriarte
- Escuela Superior de Medicina, Universidad Nacional Mar del Plata-Hospital Interzonal Especializado Materno Infantil Don Vitorio Tetamanti, Mar del Plata, Buenos Aires, Argentina
| | - Ana Colado
- Instituto de Medicina Experimental (IMEX), CONICET-Academia Nacional de Medicina, Buenos Aires, CABA, Argentina
| | | | | | - Julieta Belén Fernández
- Departamento de Fisiología, Biología Molecular y Celular, Instituto de Biociencias, Biotecnología y Biología Traslacional (IB3) e Instituto de Química Biológica (IQUIBICEN), FCEN, UBA, CONICET, Buenos Aires, CABA, Argentina
| | - Lorena Tau
- Laboratorio de Salud Pública de La Facultad de Ciencias Exactas, Departamento de Ciencias Biológicas, Facultad de Ciencias Exactas, Instituto de Estudios Inmunológicos y Fisiopatológicos (IIFP), UNLP, CONICET, Asociado a CIC PBA, UNLP, La Plata, Argentina
| | - Juliana Bernatowiez
- Instituto de Medicina Experimental (IMEX), CONICET-Academia Nacional de Medicina, Buenos Aires, CABA, Argentina
| | - Ileana Moreira
- Centro de Inmunología Clínica, Buenos Aires, CABA, Argentina
| | - Sebastián Vishnopolska
- Departamento de Fisiología, Biología Molecular y Celular, Instituto de Biociencias, Biotecnología y Biología Traslacional (IB3) e Instituto de Química Biológica (IQUIBICEN), FCEN, UBA, CONICET, Buenos Aires, CABA, Argentina
| | - Martín Rumbo
- Laboratorio de Salud Pública de La Facultad de Ciencias Exactas, Departamento de Ciencias Biológicas, Facultad de Ciencias Exactas, Instituto de Estudios Inmunológicos y Fisiopatológicos (IIFP), UNLP, CONICET, Asociado a CIC PBA, UNLP, La Plata, Argentina
| | - Chiara Cassarino
- Instituto de Medicina Experimental (IMEX), CONICET-Academia Nacional de Medicina, Buenos Aires, CABA, Argentina
| | - Gustavo Vijoditz
- Hospital Nacional Profesor Alejandro Posadas, Buenos Aires, Argentina
| | - Ana Laura López
- Hospital General de Agudos C. G. Durand, Buenos Aires, CABA, Argentina
| | - Renata Curciarello
- Laboratorio de Salud Pública de La Facultad de Ciencias Exactas, Departamento de Ciencias Biológicas, Facultad de Ciencias Exactas, Instituto de Estudios Inmunológicos y Fisiopatológicos (IIFP), UNLP, CONICET, Asociado a CIC PBA, UNLP, La Plata, Argentina
| | - Diego Rodríguez
- Escuela Superior de Medicina, Universidad Nacional Mar del Plata-Hospital Interzonal Especializado Materno Infantil Don Vitorio Tetamanti, Mar del Plata, Buenos Aires, Argentina
| | - Gastón Rizzo
- Laboratorio de Salud Pública de La Facultad de Ciencias Exactas, Departamento de Ciencias Biológicas, Facultad de Ciencias Exactas, Instituto de Estudios Inmunológicos y Fisiopatológicos (IIFP), UNLP, CONICET, Asociado a CIC PBA, UNLP, La Plata, Argentina
| | - Malena Ferreyra
- Laboratorio de Salud Pública de La Facultad de Ciencias Exactas, Departamento de Ciencias Biológicas, Facultad de Ciencias Exactas, Instituto de Estudios Inmunológicos y Fisiopatológicos (IIFP), UNLP, CONICET, Asociado a CIC PBA, UNLP, La Plata, Argentina
| | | | - María Noel Badano
- Instituto de Medicina Experimental (IMEX), CONICET-Academia Nacional de Medicina, Buenos Aires, CABA, Argentina
| | - María Inés Pérez Millán
- Departamento de Fisiología, Biología Molecular y Celular, Instituto de Biociencias, Biotecnología y Biología Traslacional (IB3) e Instituto de Química Biológica (IQUIBICEN), FCEN, UBA, CONICET, Buenos Aires, CABA, Argentina
| | - María Florencia Quiroga
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), CONICET, Buenos Aires, CABA, Argentina
| | - Patricia Baré
- Instituto de Medicina Experimental (IMEX), CONICET-Academia Nacional de Medicina, Buenos Aires, CABA, Argentina
| | - Itatí Ibañez
- Instituto de Química Física de los Materiales, Medio Ambiente y Energía (INQUIMAE), CONICET, FCEN, UBA, Buenos Aires, CABA, Argentina
| | - Roberto Pozner
- Instituto de Medicina Experimental (IMEX), CONICET-Academia Nacional de Medicina, Buenos Aires, CABA, Argentina
| | - Mercedes Borge
- Instituto de Medicina Experimental (IMEX), CONICET-Academia Nacional de Medicina, Buenos Aires, CABA, Argentina
| | - Guillermo Docena
- Laboratorio de Salud Pública de La Facultad de Ciencias Exactas, Departamento de Ciencias Biológicas, Facultad de Ciencias Exactas, Instituto de Estudios Inmunológicos y Fisiopatológicos (IIFP), UNLP, CONICET, Asociado a CIC PBA, UNLP, La Plata, Argentina
| | | | - María Belén Almejun
- Departamento de Fisiología, Biología Molecular y Celular, Instituto de Biociencias, Biotecnología y Biología Traslacional (IB3) e Instituto de Química Biológica (IQUIBICEN), FCEN, UBA, CONICET, Buenos Aires, CABA, Argentina.
- Pabellón II, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Intendente Güiraldes 2160-Ciudad Universitaria-CABA C1428EG, Buenos Aires, Argentina.
| |
Collapse
|
33
|
Murray CE, O’Brien C, Alamin S, Phelan SH, Argue R, Kiersey R, Gardiner M, Naughton A, Keogh E, Holmes P, Naughton S, Scanlon A, Sloan A, McCrea P, Sui J, Dunne J, Conlon N. Cellular and humoral immunogenicity of the COVID-19 vaccine and COVID-19 disease severity in individuals with immunodeficiency. Front Immunol 2023; 14:1131604. [PMID: 37033955 PMCID: PMC10080028 DOI: 10.3389/fimmu.2023.1131604] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Accepted: 03/06/2023] [Indexed: 04/11/2023] Open
Abstract
Background A well-coordinated adaptive immune response is crucial for limiting COVID-19 disease. Some individuals with immunodeficiency are at a high risk of developing severe COVID-19. Therefore, the development of standardized methods for measuring different arms of the vaccine response in the setting of immunodeficiency is of particular interest. In this study, we compared the vaccine response of individuals living with immunodeficiency with healthy controls in terms of interferon gamma (IFN-γ) production and spike protein-specific antibody level post primary COVID-19 vaccination and booster vaccines. Additionally, the disease severity of those individuals who contracted COVID-19 was assessed. Methods Whole blood was stimulated overnight from 71 participants and 99 healthy controls. Commercially available PepTivator® peptide pool and trimeric spike protein stimulation were used. ELISA was used to analyze IFN-γ levels. The total SARS-CoV-2 spike protein antibody titre was measured using a Roche Elecsys® S total antibody assay. Patient characteristics, COVID-19 infection status and IDDA 2.1 'Kaleidoscope' scores were recorded. Vaccine responses were scored from zero to three. Results 99% of healthy controls, 89% of individuals with IEI and 76% with secondary immunodeficiency (SID) had an IFN-γ level above the validated reference range after peptide mix stimulation following primary vaccination. There was an increase in IFN-γ levels in patients with inborn errors of immunity (IEI) following the booster vaccine (p = 0.0156). 100% of healthy controls, 70% of individuals living with IEI and 64% of individuals living with SID had detectable spike protein-specific antibody levels following the primary vaccination. 55% of immunodeficiency patients who had mild COVID-19 and 10% with moderate/severe COVID-19 had detectable antibody and IFN-γ levels post vaccine. The mean pre-infection IDDA 2.1 scores were higher in individuals who developed moderate/severe COVID-19 (25.2 compared to 9.41). Conclusions Covid whole-blood IGRA is a highly accurate, straightforward and robust assay and can be easily adapted to measure cellular response to COVID-19. A complete evaluation of the vaccine response may be particularly important for individuals living with immunodeficiency. A clinical immunodeficiency score and a validated vaccine response score may be valuable tools in estimating COVID-19 disease risk and identifying individuals living with immunodeficiency who may benefit from enhanced vaccination schedules.
Collapse
Affiliation(s)
- C. E. Murray
- Department of Immunology, St. James’s Hospital, Dublin, Ireland
- *Correspondence: C. E. Murray,
| | - C. O’Brien
- Department of Immunology, St. James’s Hospital, Dublin, Ireland
| | - S. Alamin
- Department of Immunology, St. James’s Hospital, Dublin, Ireland
| | - S. H. Phelan
- Department of Immunology, St. James’s Hospital, Dublin, Ireland
| | - R. Argue
- Department of Immunology, St. James’s Hospital, Dublin, Ireland
- Wellcome Trust Clinical Research Facility, St. James's Hospital, Dublin, Ireland
| | - R. Kiersey
- Department of Immunology, St. James’s Hospital, Dublin, Ireland
| | - M. Gardiner
- Department of Immunology, St. James’s Hospital, Dublin, Ireland
| | - A. Naughton
- Department of Immunology, St. James’s Hospital, Dublin, Ireland
| | - E. Keogh
- Department of Biochemistry, St. James’s Hospital, Dublin, Ireland
| | - P. Holmes
- Department of Biochemistry, St. James’s Hospital, Dublin, Ireland
| | - S. Naughton
- Department of Biochemistry, St. James’s Hospital, Dublin, Ireland
| | - A. Scanlon
- Department of Biochemistry, St. James’s Hospital, Dublin, Ireland
| | - A. Sloan
- Department of Immunology, St. James’s Hospital, Dublin, Ireland
| | - P. McCrea
- Department of Immunology, St. James’s Hospital, Dublin, Ireland
| | - J. Sui
- Department of Immunology, St. James’s Hospital, Dublin, Ireland
- STTAR Bioresource, St. James’s Hospital, Dublin, Ireland
| | - J. Dunne
- Department of Immunology, St. James’s Hospital, Dublin, Ireland
| | - N. Conlon
- Department of Immunology, St. James’s Hospital, Dublin, Ireland
- Wellcome Trust Clinical Research Facility, St. James's Hospital, Dublin, Ireland
- STTAR Bioresource, St. James’s Hospital, Dublin, Ireland
- School of Medicine, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
34
|
Mustafa SS, Rider NL, Jolles S. Immunosuppression in Patients With Primary Immunodeficiency-Walking the Line. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2022; 10:3088-3096. [PMID: 36049628 DOI: 10.1016/j.jaip.2022.08.025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 08/22/2022] [Accepted: 08/22/2022] [Indexed: 12/14/2022]
Abstract
Individuals with primary immunodeficiency (PIDD) experience not only infectious complications but also immune dysregulation leading to autoimmunity, inflammation, and lymphoproliferative manifestations. Management of these complications often requires treatment with additional immunosuppressive medications, which pose an additional risk of infectious complications. Immunosuppression in individuals with PIDD therefore requires careful assessment and consideration of risks and benefits. Medications should be closely monitored, and strategies for risk mitigation of adverse events considered, such as exposure reduction, appropriate vaccination, use of antibiotics/antivirals, and optimization of immunoglobulin replacement therapy. In a subset of individuals who are not tolerating immune modulation or experiencing disease progression despite appropriate interventions, hematopoietic stem-cell transplantation is a management option.
Collapse
Affiliation(s)
- S Shahzad Mustafa
- Rochester Regional Health, Division of Allergy, Immunology, and Rheumatology, University of Rochester School of Medicine and Dentistry, Rochester, NY; Liberty University College of Osteopathic Medicine and the Liberty Mountain, Chair, Division of Clinical Informatics; Associate Professor of Pediatrics, Allergy-Immunology Medical Group, Rochester, NY.
| | - Nicholas L Rider
- Liberty University College of Osteopathic Medicine and the Liberty Mountain Medical Group, Lynchburg, Va
| | - Stephen Jolles
- Immunodeficiency Centre for Wales, University Hospital of Wales, Cardiff, United Kingdom
| |
Collapse
|
35
|
Bracke C, Miranda C, González S, Casas I, Cardona PJ, Benitez RM, Sopena N, Reynaga EA, Massanella M, Clotet B, Carrillo J, Mateu L, Pedro-Botet ML. Correlation between Clinical and Immunological Variables and Humoral Response to SARS-CoV-2 Vaccination in Adult Patients with Antibody Deficiency Disorders. Pathogens 2022; 11:1364. [PMID: 36422615 PMCID: PMC9696841 DOI: 10.3390/pathogens11111364] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 11/03/2022] [Accepted: 11/13/2022] [Indexed: 08/05/2023] Open
Abstract
BACKGROUND Prophylactic vaccination has proven to be the most effective strategy to fight the COVID-19 pandemic. METHODS This was a prospective observational cohort study involving 30 predominantly antibody deficiency disorders (ADD)-afflicted adult patients on immunoglobulin replacement therapy vaccinated with three doses of the mRNA-1273 COVID-19 vaccine, and 10 healthy controls. Anti-RBD IgG antibodies were determined in plasma samples collected just before the first dose of mRNA-based COVID-19 vaccine and on weeks 4, 8, 24, and 28 following the first vaccination. Patients were categorized based on the levels of anti-RBD antibodies determined on w8 as non-, low-, and responders. Chi-square and Kruskal-Wallis tests were used to see if any variables correlated with humoral response levels. Any adverse effects of the mRNA-based vaccine were also noted. RESULTS The COVID-19 vaccine was safe and well-tolerated. The humoral response elicited at w8 after vaccination depended on the type of ADD, the type of immunoglobulin deficiency, the presence of granulomatous lymphocytic interstitial lung disease, recent use of immunosuppressive drugs, and the switched memory B cells counts. The third vaccine dose boosted humoral response in previous responders to second dose but seldom in non-responders. CONCLUSIONS The humoral response of patients with predominant ADD depends mostly on the type of immunodeficiency and on the frequency of B and T cell populations.
Collapse
Affiliation(s)
- Carmen Bracke
- Department of Infectious Diseases, Germans Trias i Pujol Hospital, 08916 Badalona, Spain
| | - Cristina Miranda
- Fight AIDS and Infectious Diseases Foundation, 08916 Badalona, Spain
| | - Sandra González
- Fight AIDS and Infectious Diseases Foundation, 08916 Badalona, Spain
| | - Irma Casas
- Department of Preventive Medicine, Germans Trias i Pujol Hospital, 08916 Badalona, Spain
| | - Pere Joan Cardona
- Microbiology Department, Germans Trias i Pujol Hospital, 08916 Badalona, Spain
- Department of Genetics and Microbiology, Autonomous University of Barcelona, 08193 Cerdanyola, Spain
- Respiratory Disease Networking Biomedical Research Center (CIBERes), Carlos III Health Institute, 28029 Madrid, Spain
| | - Rosa Maria Benitez
- Department of Infectious Diseases, Germans Trias i Pujol Hospital, 08916 Badalona, Spain
| | - Nieves Sopena
- Department of Infectious Diseases, Germans Trias i Pujol Hospital, 08916 Badalona, Spain
- Department of Genetics and Microbiology, Autonomous University of Barcelona, 08193 Cerdanyola, Spain
- Respiratory Disease Networking Biomedical Research Center (CIBERes), Carlos III Health Institute, 28029 Madrid, Spain
| | | | - Marta Massanella
- IrsiCaixa AIDS Research Institute, Germans Trias i Pujol Hospital, 08916 Badalona, Spain
- Infectious Disease Networking Biomedical Research Center (CIBERINFEC), Carlos III Health Institute, 28029 Madrid, Spain
| | - Bonaventura Clotet
- Department of Infectious Diseases, Germans Trias i Pujol Hospital, 08916 Badalona, Spain
- Department of Genetics and Microbiology, Autonomous University of Barcelona, 08193 Cerdanyola, Spain
- IrsiCaixa AIDS Research Institute, Germans Trias i Pujol Hospital, 08916 Badalona, Spain
- Infectious Disease Networking Biomedical Research Center (CIBERINFEC), Carlos III Health Institute, 28029 Madrid, Spain
| | - Jorge Carrillo
- IrsiCaixa AIDS Research Institute, Germans Trias i Pujol Hospital, 08916 Badalona, Spain
- Infectious Disease Networking Biomedical Research Center (CIBERINFEC), Carlos III Health Institute, 28029 Madrid, Spain
| | - Lourdes Mateu
- Department of Infectious Diseases, Germans Trias i Pujol Hospital, 08916 Badalona, Spain
- Department of Genetics and Microbiology, Autonomous University of Barcelona, 08193 Cerdanyola, Spain
- Respiratory Disease Networking Biomedical Research Center (CIBERes), Carlos III Health Institute, 28029 Madrid, Spain
| | - Maria Luisa Pedro-Botet
- Department of Infectious Diseases, Germans Trias i Pujol Hospital, 08916 Badalona, Spain
- Department of Genetics and Microbiology, Autonomous University of Barcelona, 08193 Cerdanyola, Spain
- Respiratory Disease Networking Biomedical Research Center (CIBERes), Carlos III Health Institute, 28029 Madrid, Spain
| |
Collapse
|
36
|
Göschl L, Mrak D, Grabmeier-Pfistershammer K, Stiasny K, Haslacher H, Schneider L, Deimel T, Kartnig F, Tobudic S, Aletaha D, Burgmann H, Bonelli M, Pickl WF, Förster-Waldl E, Scheinecker C, Vossen MG. Reactogenicity and immunogenicity of the second COVID-19 vaccination in patients with inborn errors of immunity or mannan-binding lectin deficiency. Front Immunol 2022; 13:974987. [PMID: 36189225 PMCID: PMC9515892 DOI: 10.3389/fimmu.2022.974987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 08/16/2022] [Indexed: 11/29/2022] Open
Abstract
Background Patients with inborn errors of immunity (IEI) are at increased risk for severe courses of SARS-CoV-2 infection. COVID-19 vaccination provides effective protection in healthy individuals. However, it remains unclear whether vaccination is efficient and safe in patients with constitutional dysfunctions of the immune system. Thus, we analyzed the humoral response, adverse reactions and assessed the disease activity of the underlying disease after COVID-19 vaccination in a cohort of patients suffering from IEIs or mannan-binding lectin deficiency (MBLdef). Methods Vaccination response was assessed after basic immunization using the Elecsys anti-SARS-CoV-2 S immunoassay and via Vero E6 cell based assay to detect neutralization capabilities. Phenotyping of lymphocytes was performed by flow cytometry. Patient charts were reviewed for disease activity, autoimmune phenomena as well as immunization status and reactogenicity of the vaccination. Activity of the underlying disease was assessed using a patient global numeric rating scale (NRS). Results Our cohort included 11 individuals with common variable immunodeficiency (CVID), one patient with warts hypogammaglobulinemia immunodeficiency myelokathexis (WHIM) syndrome, two patients with X-linked agammaglobulinemia (XLA), one patient with Muckle Wells syndrome, two patients with cryopyrin-associated periodic syndrome, one patient with Interferon-gamma (IFN-gamma) receptor defect, one patient with selective deficiency in pneumococcal antibody response combined with a low MBL level and seven patients with severe MBL deficiency. COVID-19 vaccination was generally well tolerated with little to no triggering of autoimmune phenomena. 20 out of 26 patients developed an adequate humoral vaccine response. 9 out of 11 patients developed a T cell response comparable to healthy control subjects. Tested immunoglobulin replacement therapy (IgRT) preparations contained Anti-SARS-CoV-2 S antibodies implicating additional protection through IgRT. Summary In summary the data support the efficacy and safety of a COVID-19 vaccination in patients with IEIs/MBLdef. We recommend evaluation of the humoral immune response and testing for virus neutralization after vaccination in this cohort.
Collapse
Affiliation(s)
- Lisa Göschl
- Division of Rheumatology, University Clinics of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Daniel Mrak
- Division of Rheumatology, University Clinics of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | | | - Karin Stiasny
- Center for Virology, Medical University of Vienna, Vienna, Austria
| | - Helmuth Haslacher
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Lisa Schneider
- Division of Infectious Diseases and Tropical Medicine, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Thomas Deimel
- Division of Rheumatology, University Clinics of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Felix Kartnig
- Division of Rheumatology, University Clinics of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Selma Tobudic
- Division of Infectious Diseases and Tropical Medicine, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Daniel Aletaha
- Division of Rheumatology, University Clinics of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Heinz Burgmann
- Division of Infectious Diseases and Tropical Medicine, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Michael Bonelli
- Division of Rheumatology, University Clinics of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Winfried F. Pickl
- Institute of Immunology, Centre for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Elisabeth Förster-Waldl
- Division of Neonatology, Pediatric Intensive Care and Neuropediatrics with Centre for Congenital Immunodeficiencies & Jeffrey Modell Center Vienna, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Clemens Scheinecker
- Division of Rheumatology, University Clinics of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Matthias Gerhard Vossen
- Division of Infectious Diseases and Tropical Medicine, Department of Medicine I, Medical University of Vienna, Vienna, Austria
- *Correspondence: Matthias Gerhard Vossen,
| |
Collapse
|
37
|
Shields AM, Faustini SE, Hill HJ, Al-Taei S, Tanner C, Ashford F, Workman S, Moreira F, Verma N, Wagg H, Heritage G, Campton N, Stamataki Z, Drayson MT, Klenerman P, Thaventhiran JED, Elkhalifa S, Goddard S, Johnston S, Huissoon A, Bethune C, Elcombe S, Lowe DM, Patel SY, Savic S, Richter AG, Burns SO. Increased Seroprevalence and Improved Antibody Responses Following Third Primary SARS-CoV-2 Immunisation: An Update From the COV-AD Study. Front Immunol 2022; 13:912571. [PMID: 35720400 PMCID: PMC9201027 DOI: 10.3389/fimmu.2022.912571] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 05/06/2022] [Indexed: 11/29/2022] Open
Abstract
Background Patients with primary and secondary antibody deficiency are vulnerable to COVID-19 and demonstrate diminished responses following two-dose SARS-CoV-2 vaccine schedules. Third primary vaccinations have been deployed to enhance their humoral and cellular immunity. Objectives To determine the immunogenicity of the third primary SARS-CoV-2 immunisation in a heterogeneous cohort of patients with antibody deficiency. Methods Participants enrolled in the COV-AD study were sampled before and after their third vaccine dose. Serological and cellular responses were determined using ELISA, live-virus neutralisation and ELISPOT assays. Results Following a two-dose schedule, 100% of healthy controls mounted a serological response to SARS-CoV-2 vaccination, however, 38.6% of individuals with antibody deficiency remained seronegative. A third primary SARS-CoV-2 vaccine significantly increased anti-spike glycoprotein antibody seroprevalence from 61.4% to 76.0%, the magnitude of the antibody response, its neutralising capacity and induced seroconversion in individuals who were seronegative after two vaccine doses. Vaccine-induced serological responses were broadly cross-reactive against the SARS-CoV-2 B.1.1.529 variant of concern, however, seroprevalence and antibody levels remained significantly lower than healthy controls. No differences in serological responses were observed between individuals who received AstraZeneca ChAdOx1 nCoV-19 and Pfizer BioNTech 162b2 during their initial two-dose vaccine schedule. SARS-CoV-2 infection-naive participants who had received a heterologous vaccine as a third dose were significantly more likely to have a detectable T cell response following their third vaccine dose (61.5% vs 11.1%). Conclusion These data support the widespread use of third primary immunisations to enhance humoral immunity against SARS-CoV-2 in individuals with antibody deficiency.
Collapse
Affiliation(s)
- Adrian M. Shields
- Clinical Immunology Service, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
- Department of Clinical Immunology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - Sian E. Faustini
- Clinical Immunology Service, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Harriet J. Hill
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Saly Al-Taei
- Clinical Immunology Service, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Chloe Tanner
- Clinical Immunology Service, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Fiona Ashford
- Clinical Immunology Service, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Sarita Workman
- Department of Immunology, Royal Free London NHS Foundation Trust, London, United Kingdom
| | - Fernando Moreira
- Department of Immunology, Royal Free London NHS Foundation Trust, London, United Kingdom
| | - Nisha Verma
- Department of Immunology, Royal Free London NHS Foundation Trust, London, United Kingdom
| | - Hollie Wagg
- Institute of Translational Medicine, University of Birmingham, Birmingham, United Kingdom
| | - Gail Heritage
- Institute of Translational Medicine, University of Birmingham, Birmingham, United Kingdom
| | - Naomi Campton
- Institute of Translational Medicine, University of Birmingham, Birmingham, United Kingdom
| | - Zania Stamataki
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Mark T. Drayson
- Clinical Immunology Service, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Paul Klenerman
- Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | | | - Shuayb Elkhalifa
- Department of Immunology, Salford Royal NHS Foundation Trust, Salford, United Kingdom
| | - Sarah Goddard
- Department of Clinical Immunology, University Hospitals North Midlands, Stoke-on-Trent, United Kingdom
| | - Sarah Johnston
- Department of Clinical Immunology, North Bristol NHS Trust, Bristol, United Kingdom
| | - Aarnoud Huissoon
- Department of Clinical Immunology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - Claire Bethune
- Department of Allergy and Clinical Immunology, University Hospitals Plymouth NHS Trust, Plymouth, United Kingdom
| | - Suzanne Elcombe
- Department of Allergy and Clinical Immunology, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle, United Kingdom
| | - David M. Lowe
- Department of Immunology, Royal Free London NHS Foundation Trust, London, United Kingdom
- Institute of Immunity and Transplantation, University College London, London, United Kingdom
| | - Smita Y. Patel
- Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- National Institute for Health and Care Research (NIHR) Biomedical Research Centre (BRC) Oxford Biomedical Centre, University of Oxford, Oxford, United Kingdom
| | - Sinisa Savic
- Department of Allergy and Clinical Immunology, Leeds Teaching Hospitals NHS Trust, Leeds, United Kingdom
| | - Alex G. Richter
- Clinical Immunology Service, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
- Department of Clinical Immunology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - Siobhan O. Burns
- Department of Immunology, Royal Free London NHS Foundation Trust, London, United Kingdom
- Institute of Immunity and Transplantation, University College London, London, United Kingdom
| |
Collapse
|