1
|
Kobashi Y, Kawamura T, Shimazu Y, Kaneko Y, Nishikawa Y, Sugiyama A, Tani Y, Nakayama A, Yoshida M, Zho T, Yamamoto C, Saito H, Takita M, Wakui M, Kodama T, Tsubokura M. Kinetics of humoral and cellular immune responses 5 months post-COVID-19 booster dose by immune response groups at the peak immunity phase: An observational historical cohort study using the Fukushima vaccination community survey. Vaccine X 2024; 20:100553. [PMID: 39309610 PMCID: PMC11416657 DOI: 10.1016/j.jvacx.2024.100553] [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: 05/24/2024] [Revised: 09/03/2024] [Accepted: 09/05/2024] [Indexed: 09/25/2024] Open
Abstract
Background Understanding the waning of immunity after booster vaccinations is important to identify which immune-low populations should be prioritized. Methods We investigated longitudinal cellular and humoral immunity after the third vaccine dose in both high- and low-cellular and humoral immunity groups at the peak immunity phase after the booster vaccination in a large community-based cohort. Blood samples were collected from 1045 participants at peak (T1: median 54 days post-third dose) and decay (T2: median 145 days post-third dose) phases to assess IgG(S), neutralizing activity, and ELISpot responses. Participants were categorized into high/low ELISpot/IgG(S) groups at T1. Cellular and humoral responses were tracked for approximately five months after the third vaccination. Results In total, 983 participants were included in the cohort. IgG(S) geometric mean fold change between timepoints revealed greater waning in the >79 years age group (T2/T1 fold change: 0.27) and higher IgG(S) fold change in the low-ELISpot group at T1 (T2/T1 fold change: 0.32-0.33) than in the other groups, although ELISpot geometric mean remained stable. Conclusions Antibody level of those who did not respond well to third dose vaccination waned rapidly than those who responded well. Evidence-based vaccine strategies are essential in preventing potential health issues caused by vaccines, including side-effects.
Collapse
Affiliation(s)
- Yurie Kobashi
- Department of Radiation Health Management, Fukushima Medical University School of Medicine, Fukushima City, Fukushima, Japan
- Department of General Internal Medicine, Hirata Central Hospital, Hirata, Ishikawa District, Fukushima, Japan
| | - Takeshi Kawamura
- Isotope Science Center, The University of Tokyo, Tokyo, Japan
- Laboratory for Systems Biology and Medicine, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Yuzo Shimazu
- Department of Radiation Health Management, Fukushima Medical University School of Medicine, Fukushima City, Fukushima, Japan
| | - Yudai Kaneko
- Laboratory for Systems Biology and Medicine, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
- Medical & Biological Laboratories Co., Ltd, Tokyo, Japan
| | - Yoshitaka Nishikawa
- Department of General Internal Medicine, Hirata Central Hospital, Hirata, Ishikawa District, Fukushima, Japan
| | - Akira Sugiyama
- Isotope Science Center, The University of Tokyo, Tokyo, Japan
| | - Yuta Tani
- Medical Governance Research Institute, Minato-ku, Tokyo, Japan
| | - Aya Nakayama
- Isotope Science Center, The University of Tokyo, Tokyo, Japan
| | - Makoto Yoshida
- Medical Governance Research Institute, Minato-ku, Tokyo, Japan
| | - Tianchen Zho
- Department of Radiation Health Management, Fukushima Medical University School of Medicine, Fukushima City, Fukushima, Japan
| | - Chika Yamamoto
- Department of Radiation Health Management, Fukushima Medical University School of Medicine, Fukushima City, Fukushima, Japan
| | - Hiroaki Saito
- Department of Radiation Health Management, Fukushima Medical University School of Medicine, Fukushima City, Fukushima, Japan
| | - Morihito Takita
- Department of Radiation Health Management, Fukushima Medical University School of Medicine, Fukushima City, Fukushima, Japan
| | - Masatoshi Wakui
- Department of Laboratory Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Tatsuhiko Kodama
- Laboratory for Systems Biology and Medicine, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Masaharu Tsubokura
- Department of Radiation Health Management, Fukushima Medical University School of Medicine, Fukushima City, Fukushima, Japan
- Department of General Internal Medicine, Hirata Central Hospital, Hirata, Ishikawa District, Fukushima, Japan
- General Incorporated Association for Comprehensive Disaster Health Management Research Institute, Japan
| |
Collapse
|
2
|
Griffin DWJ, Dymock M, Wong G, Morrissey CO, Lewin SR, Cheng AC, Howard K, Marsh JA, Subbarao K, Hagenauer M, Roney J, Cunningham A, Snelling T, McMahon JH. Bringing optimised COVID-19 vaccine schedules to immunocompromised populations (BOOST-IC): study protocol for an adaptive randomised controlled clinical trial. Trials 2024; 25:485. [PMID: 39020446 PMCID: PMC11253462 DOI: 10.1186/s13063-024-08315-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 07/03/2024] [Indexed: 07/19/2024] Open
Abstract
BACKGROUND Immunocompromised hosts (ICH) experience more breakthrough infections and worse clinical outcomes following infection with COVID-19 than immunocompetent people. Prophylactic monoclonal antibody therapies can be challenging to access, and escape variants emerge rapidly. Immunity conferred through vaccination remains a central prevention strategy for COVID-19. COVID-19 vaccines do not elicit optimal immunity in ICH but boosting, through additional doses of vaccine improves humoral and cellular immune responses. This trial aims to assess the immunogenicity and safety of different COVID-19 vaccine booster strategies against SARS-CoV-2 for ICH in Australia. METHODS Bringing optimised COVID-19 vaccine schedules to immunocompromised populations (BOOST-IC) is an adaptive randomised trial of one or two additional doses of COVID-19 vaccines 3 months apart in people living with HIV, solid organ transplant (SOT) recipients, or those who have haematological malignancies (chronic lymphocytic leukaemia, non-Hodgkin lymphoma or multiple myeloma). Key eligibility criteria include having received 3 to 7 doses of Australian Therapeutic Goods Administration (TGA)-approved COVID-19 vaccines at least 3 months earlier, and having not received SARS-CoV-2-specific monoclonal antibodies in the 3 months prior to receiving the study vaccine. The primary outcome is the geometric mean concentration of anti-spike SARS-CoV-2 immunoglobulin G (IgG) 28 days after the final dose of the study vaccine. Key secondary outcomes include anti-spike SARS-CoV-2 IgG titres and the proportion of people seroconverting 6 and 12 months after study vaccines, local and systemic reactions in the 7 days after vaccination, adverse events of special interest, COVID-19 infection, mortality and quality of life. DISCUSSION This study will enhance the understanding of COVID-19 vaccine responses in ICH, and enable the development of safe, and optimised vaccine schedules in people with HIV, SOT, or haematological malignancy. TRIAL REGISTRATION ClinicalTrials.gov NCT05556720. Registered on 23rd August 2022.
Collapse
Affiliation(s)
- David W J Griffin
- Department of Infectious Diseases, Alfred Hospital and School of Translational Medicine, Monash University, Melbourne, Australia.
| | - Michael Dymock
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, Perth, Australia
| | - Germaine Wong
- Centre for Kidney Research, The Children's Hospital at Westmead, Westmead, NSW, Australia
- Department of Renal Medicine, Westmead Hospital, Westmead, NSW, Australia
- Sydney School of Public Health, Faculty of Medicine & Health, University of Sydney, Sydney, NSW, Australia
| | - C Orla Morrissey
- Department of Infectious Diseases, Alfred Hospital and School of Translational Medicine, Monash University, Melbourne, Australia
| | - Sharon R Lewin
- Department of Infectious Diseases, Alfred Hospital and School of Translational Medicine, Monash University, Melbourne, Australia
- Department of Infectious Diseases, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
- Victorian Infectious Diseases Service, Royal Melbourne Hospital, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Allen C Cheng
- Department of Infectious Diseases, Monash Medical Centre, Melbourne, Australia
- Monash University School of Clinical Sciences at Monash Health, Clayton, Australia
| | - Kirsten Howard
- Sydney School of Public Health, Faculty of Medicine & Health, University of Sydney, Sydney, NSW, Australia
- Menzies Centre for Health Policy and Economics, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Julie A Marsh
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, Perth, Australia
- Centre for Child Health Research, School of Medicine, The University of Western Australia, Perth, Australia
| | - Kanta Subbarao
- WHO Collaborating Centre for Reference and Research On Influenza at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Michelle Hagenauer
- Department of Infectious Diseases, Alfred Hospital and School of Translational Medicine, Monash University, Melbourne, Australia
| | - Janine Roney
- Department of Infectious Diseases, Alfred Hospital and School of Translational Medicine, Monash University, Melbourne, Australia
| | - Anthony Cunningham
- Westmead Institute for Medical Research, University of Sydney, Sydney, NSW, Australia
| | - Tom Snelling
- Sydney School of Public Health, Faculty of Medicine & Health, University of Sydney, Sydney, NSW, Australia
| | - James H McMahon
- Department of Infectious Diseases, Alfred Hospital and School of Translational Medicine, Monash University, Melbourne, Australia
- Department of Infectious Diseases, Monash Medical Centre, Melbourne, Australia
| |
Collapse
|
3
|
Naylor KL, Knoll GA, Treleaven D, Kang Y, Garg AX, Stirling K, Kim SJ. Comparison of COVID-19 Hospitalization and Death Between Solid Organ Transplant Recipients and the General Population in Canada, 2020-2022. Transplant Direct 2024; 10:e1670. [PMID: 38953040 PMCID: PMC11216672 DOI: 10.1097/txd.0000000000001670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 05/11/2024] [Indexed: 07/03/2024] Open
Abstract
Background Solid organ transplant recipients have a high risk of severe outcomes from SARS-CoV-2 infection. A comprehensive understanding of the impact of the COVID-19 pandemic across multiple waves in the solid organ transplant population and how this compares to the general population is limited. We conducted a population-based cohort study using linked administrative healthcare databases from Ontario, Canada to answer this question. Methods We included 15 306 solid organ transplant recipients and 12 160 904 individuals from the general population. Our primary outcome was the rate (per 100 person-years) of severe COVID-19 (ie, hospitalization or death with a positive SARS-CoV-2 test) occurring between January 25, 2020, and November 30, 2022. Results Compared with the general population, solid organ transplant recipients had almost a 6 times higher rate of severe COVID-19 (20.39 versus 3.44 per 100 person-years), with almost 5.5 times as high a rate of death alone (4.19 versus 0.77 per 100 person-years). Transplant recipients with severe COVID-19 were substantially younger (60.1 versus 66.5 y) and had more comorbidities. The rate of severe COVID-19 declined over time in the solid organ transplant population, with an incidence rate of 41.25 per 100 person-years in the first wave (January 25, 2020, to August 31, 2020) and 18.41 in the seventh wave (June 19, 2022, to November 30, 2022, Omicron era). Conclusions Solid organ transplant recipients remain at high risk of severe outcomes when they are infected with SARS-CoV-2. Resources and strategies to mitigate the impact of SARS-CoV-2 exposure are needed in this vulnerable patient population.
Collapse
Affiliation(s)
- Kyla L. Naylor
- ICES, ON, Canada
- Department of Epidemiology & Biostatistics, Western University, London, ON, Canada
- Lawson Health Research Institute, London Health Sciences Centre, London, ON, Canada
| | - Gregory A. Knoll
- Department of Medicine (Nephrology), University of Ottawa and the Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | | | - Yuguang Kang
- ICES, ON, Canada
- Lawson Health Research Institute, London Health Sciences Centre, London, ON, Canada
| | - Amit X. Garg
- ICES, ON, Canada
- Department of Epidemiology & Biostatistics, Western University, London, ON, Canada
- Lawson Health Research Institute, London Health Sciences Centre, London, ON, Canada
| | - Kathryn Stirling
- Lawson Health Research Institute, London Health Sciences Centre, London, ON, Canada
| | - S. Joseph Kim
- ICES, ON, Canada
- Division of Nephrology, University Health Network, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
4
|
Li Y, Han M, Li X. Clinical and prognostic implications of hyaluronic acid in patients with COVID-19 reinfection and first infection. Front Microbiol 2024; 15:1406581. [PMID: 38881657 PMCID: PMC11178136 DOI: 10.3389/fmicb.2024.1406581] [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: 03/25/2024] [Accepted: 05/20/2024] [Indexed: 06/18/2024] Open
Abstract
Objective Previous research has shown that human identical sequences of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) promote coronavirus disease 2019 (COVID-19) progression by upregulating hyaluronic acid (HA). However, the association of HA with mortality and long COVID in SARS-CoV-2 reinfection and first infection is unclear. Methods Patients with COVID-19 at Beijing Ditan Hospital from September 2023 to November 2023 were consecutively enrolled. SARS-CoV-2 reinfections were matched 1:2 with first infections using a nearest neighbor propensity score matching algorithm. We compared the hospital outcomes between patients with COVID-19 reinfection and first infection. The association between HA levels and mortality and long COVID in the matched cohort was analyzed. Results The reinfection rate among COVID-19 hospitalized patients was 25.4% (62 cases). After propensity score matching, we found that reinfection was associated with a better clinical course and prognosis, including lower levels of C-reactive protein and erythrocyte sedimentation rate, fewer cases of bilateral lung infiltration and respiratory failure, and shorter viral clearance time and duration of symptoms (p < 0.05). HA levels were significantly higher in patients with primary infection [128.0 (90.5, 185.0) vs. 94.5 (62.0, 167.3), p = 0.008], those with prolonged viral clearance time [90.5 (61.5, 130.8) vs. 130.0 (95.0, 188.0), p < 0.001], and deceased patients [105.5 (76.8, 164.5) vs. 188.0 (118.0, 208.0), p = 0.002]. Further analysis showed that HA was an independent predictor of death (AUC: 0.789), and the risk of death increased by 4.435 times (OR = 5.435, 95% CI = 1.205-24.510, p = 0.028) in patients with high HA levels. We found that patients with HA levels above 116 ng/mL had an increased risk of death. However, the incidence of long COVID was similar in the different HA level groups (p > 0.05). Conclusion Serum HA may serve as a novel biomarker for predicting COVID-19 mortality in patients with SARS-CoV-2 reinfection and first infection. However, HA levels may not be associated with long COVID.
Collapse
Affiliation(s)
- Yanyan Li
- Center of Integrative Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Ming Han
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Infectious Diseases, Beijing, China
| | - Xin Li
- Center of Integrative Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
5
|
Rabezanahary H, Gilbert C, Santerre K, Scarrone M, Gilbert M, Thériault M, Brousseau N, Masson JF, Pelletier JN, Boudreau D, Trottier S, Baz M. Live virus neutralizing antibodies against pre and post Omicron strains in food and retail workers in Québec, Canada. Heliyon 2024; 10:e31026. [PMID: 38826717 PMCID: PMC11141348 DOI: 10.1016/j.heliyon.2024.e31026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 05/08/2024] [Accepted: 05/09/2024] [Indexed: 06/04/2024] Open
Abstract
Background Measuring the ability of SARS-CoV-2 antibodies to neutralize live viruses remains an effective approach to quantify the level of protection of individuals. We assessed the neutralization activity against the ancestral SARS-CoV-2, Delta, Omicron BA.1, BA.2, BA.2.12.1, BA.4 and BA.5 strains, in 280 vaccinated restaurant/bar, grocery and hardware store workers in Québec, Canada. Methods Participants were recruited during the emergence of Omicron BA.1 variant. The neutralizing activity of participant sera was assessed by microneutralization assay. Results Serum neutralizing antibody (NtAb) titers of all participants against the ancestral SARS-CoV-2 strain were comparable with those against Delta variant (ranges of titers 10-2032 and 10-2560, respectively), however, their response was significantly reduced against Omicron BA.1, BA2, BA.2.12.1, BA.4 and BA.5 (10-1016, 10-1016, 10-320, 10-80 and 10-254, respectively). Individuals who received 2 doses of vaccine had significantly reduced NtAb titers against all SARS-CoV-2 strains compared to those infected and then vaccinated (≥1 dose), vaccinated (≥2 doses) and then infected, or those who received 3 doses of vaccine. Participants vaccinated with 2 or 3 doses of vaccine and then infected had the highest NtAb titers against all SARS-CoV-2 strains tested. Conclusion We assessed for the first time the NtAb response in food and retail workers. We found that vaccination prior to the emergence of Omicron BA.1 was associated with higher neutralizing activity against pre-Omicron variants, suggesting the importance of updating vaccines to increase antibody response against new SARS-CoV-2 variants. Vaccination followed by infection was associated with higher neutralizing activity against all SARS-CoV-2 strains tested.
Collapse
Affiliation(s)
- Henintsoa Rabezanahary
- Division of Infectious and Immune Diseases, CHU de Québec Research Center, QC, Quebec, Canada
- Department of Microbiology, Infectious Disease and Immunology, Faculty of Medicine, Université Laval, QC, Quebec, Canada
| | - Caroline Gilbert
- Division of Infectious and Immune Diseases, CHU de Québec Research Center, QC, Quebec, Canada
- Department of Microbiology, Infectious Disease and Immunology, Faculty of Medicine, Université Laval, QC, Quebec, Canada
| | - Kim Santerre
- Division of Infectious and Immune Diseases, CHU de Québec Research Center, QC, Quebec, Canada
- Department of Microbiology, Infectious Disease and Immunology, Faculty of Medicine, Université Laval, QC, Quebec, Canada
| | - Martina Scarrone
- Division of Infectious and Immune Diseases, CHU de Québec Research Center, QC, Quebec, Canada
- Department of Microbiology, Infectious Disease and Immunology, Faculty of Medicine, Université Laval, QC, Quebec, Canada
| | - Megan Gilbert
- Division of Infectious and Immune Diseases, CHU de Québec Research Center, QC, Quebec, Canada
- Department of Microbiology, Infectious Disease and Immunology, Faculty of Medicine, Université Laval, QC, Quebec, Canada
| | - Mathieu Thériault
- Division of Infectious and Immune Diseases, CHU de Québec Research Center, QC, Quebec, Canada
- Department of Microbiology, Infectious Disease and Immunology, Faculty of Medicine, Université Laval, QC, Quebec, Canada
| | - Nicholas Brousseau
- Division of Infectious and Immune Diseases, CHU de Québec Research Center, QC, Quebec, Canada
- Biological Risks Department, Institut National de Santé Publique du Québec, Québec, QC, G1V 5B3, Canada
| | - Jean-François Masson
- Department of Chemistry, Quebec Center for Advanced Materials, Regroupement québécois sur les Matériaux de Pointe, and Centre Interdisciplinaire de Recherche sur le Cerveau et l'apprentissage, Université de Montréal, Montréal, Canada
| | - Joelle N. Pelletier
- Department of Chemistry, Department of Biochemistry, Université de Montréal, Montréal, QC H2V 0B3, Canada
- PROTEO-The Québec Network for Research on Protein Function, Engineering, and Applications, Québec, Canada
| | - Denis Boudreau
- Département de Chimie et Centre d'Optique, Photonique et laser (COPL), Université Laval, Québec, Canada
| | - Sylvie Trottier
- Division of Infectious and Immune Diseases, CHU de Québec Research Center, QC, Quebec, Canada
- Department of Microbiology, Infectious Disease and Immunology, Faculty of Medicine, Université Laval, QC, Quebec, Canada
| | - Mariana Baz
- Division of Infectious and Immune Diseases, CHU de Québec Research Center, QC, Quebec, Canada
- Department of Microbiology, Infectious Disease and Immunology, Faculty of Medicine, Université Laval, QC, Quebec, Canada
| |
Collapse
|
6
|
Boan P. Prior Infection Reduces Omicron COVID-19 Severity in Solid Organ Transplant Recipients. Transplantation 2024; 108:1070-1071. [PMID: 38291591 DOI: 10.1097/tp.0000000000004905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Affiliation(s)
- Peter Boan
- Department of Infectious Diseases, Fiona Stanley Hospital, Murdoch, WA, Australia
- Department of Microbiology, PathWest Laboratory Medicine WA, Fiona Stanley Hospital, Murdoch, WA, Australia
| |
Collapse
|
7
|
Solera JT, Árbol BG, Mittal A, Hall VG, Marinelli T, Bahinskaya I, Humar A, Kumar D. COVID-19 Reinfection Has Better Outcomes Than the First Infection in Solid Organ Transplant Recipients. Transplantation 2024; 108:1249-1256. [PMID: 38291585 DOI: 10.1097/tp.0000000000004902] [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: 02/01/2024]
Abstract
BACKGROUND Solid organ transplant recipients face an increased risk of severe coronavirus disease 2019 (COVID-19) and are vulnerable to repeat severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections. In nonimmunocompromised individuals, SARS-CoV-2 reinfections are milder likely because of cross-protective immunity. We sought to determine whether SARS-CoV-2 reinfection exhibits milder manifestations than primary infection in transplant recipients. METHODS Using a large, prospective cohort of adult transplant patients with COVID-19, we identified patients with SARS-CoV-2 reinfections. We performed a 1:1 nearest neighbor propensity score matching to control potential confounders, including the COVID-19 variant. We compared outcomes including oxygen requirement, hospitalization, and intensive care unit admission within 30 d after diagnosis between patients with reinfection and those with the first episode of COVID-19. RESULTS Between 2020 and 2023, 103 reinfections were identified in a cohort of 1869 transplant recipients infected with SARS-CoV-2 (incidence of 2.7% per year). These included 50 kidney (48.5%), 27 lung (26.2%), 7 heart (6.8%), 6 liver (5.8%), and 13 multiorgan (12.6%) transplants. The median age was 54.5 y (interquartile range [IQR], 40.5-65.5) and the median time from transplant to first infection was 6.6 y (IQR, 2.8-11.2). The time between the primary COVID-19 and reinfection was 326 d (IQR, 226-434). Three doses or more of SARS-CoV-2 vaccine are received by 87.4% of patients. After propensity score matching, reinfections were associated with significantly lower hospitalization (5.8% versus 19.4%; risk ratio, 0.3; 95% CI, 0.12-0.71) and oxygen requirement (3.9% versus 13.6%; risk ratio, 0.29; 95% CI, 0.10-0.84). In a within-patient analysis only in the reinfection group, the second infection was milder than the first (3.9% required oxygen versus 19.4%, P < 0.0001), and severe first COVID-19 was the only predictor of severe reinfection. CONCLUSIONS Transplant recipients with COVID-19 reinfection present better outcomes than those with the first infection, providing clinical evidence for the development of cross-protective immunity.
Collapse
Affiliation(s)
- Javier T Solera
- Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada
| | | | | | | | | | | | | | | |
Collapse
|
8
|
Mikhailov M, Budde K, Halleck F, Eleftheriadis G, Naik MG, Schrezenmeier E, Bachmann F, Choi M, Duettmann W, von Hoerschelmann E, Koch N, Liefeldt L, Lücht C, Straub-Hohenbleicher H, Waiser J, Weber U, Zukunft B, Osmanodja B. COVID-19 Outcomes in Kidney Transplant Recipients in a German Transplant Center. J Clin Med 2023; 12:6103. [PMID: 37763043 PMCID: PMC10531713 DOI: 10.3390/jcm12186103] [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: 08/10/2023] [Revised: 09/02/2023] [Accepted: 09/06/2023] [Indexed: 09/29/2023] Open
Abstract
Kidney transplant recipients (KTRs) show higher morbidity and mortality from COVID-19 than the general population and have an impaired response to vaccination. We analyzed COVID-19 incidence and clinical outcomes in a single-center cohort of approximately 2500 KTRs. Between 1 February 2020 and 1 July 2022, 578 KTRs were infected with SARS-CoV-2, with 25 (4%) recurrent infections. In total, 208 KTRs (36%) were hospitalized, and 39 (7%) died. Among vaccinated patients, infection with the Omicron variant had a mortality of 2%. Unvaccinated patients infected with the Omicron variant showed mortality (9% vs. 11%) and morbidity (hospitalization 52% vs. 54%, ICU admission 12% vs. 18%) comparable to the pre-Omicron era. Multivariable analysis revealed that being unvaccinated (OR = 2.15, 95% CI [1.38, 3.35]), infection in the pre-Omicron era (OR = 3.06, 95% CI [1.92, 4.87]), and higher patient age (OR = 1.04, 95% CI [1.03, 1.06]) are independent risk factors for COVID-19 hospitalization, whereas a steroid-free immunosuppressive regimen was found to reduce the risk of COVID-19 hospitalization (OR = 0.51, 95% CI [0.33, 0.79]). This suggests that both virological changes in the Omicron variant and vaccination reduce the risk for morbidity and mortality from COVID-19 in KTRs. Our data extend the knowledge from the general population to KTRs and provide important insights into outcomes during the Omicron era.
Collapse
Affiliation(s)
- Michael Mikhailov
- Department of Nephrology and Medical Intensive Care, Charité–Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany; (K.B.); (F.H.); (G.E.); (M.G.N.); (E.S.); (F.B.); (M.C.); (W.D.); (E.v.H.); (N.K.); (L.L.); (C.L.); (H.S.-H.); (J.W.); (U.W.); (B.Z.); (B.O.)
- Clinic for Anaesthesiology and Intensive Care Medicine, Charité–Universitätsmedizin Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, 12200 Berlin, Germany
| | - Klemens Budde
- Department of Nephrology and Medical Intensive Care, Charité–Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany; (K.B.); (F.H.); (G.E.); (M.G.N.); (E.S.); (F.B.); (M.C.); (W.D.); (E.v.H.); (N.K.); (L.L.); (C.L.); (H.S.-H.); (J.W.); (U.W.); (B.Z.); (B.O.)
| | - Fabian Halleck
- Department of Nephrology and Medical Intensive Care, Charité–Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany; (K.B.); (F.H.); (G.E.); (M.G.N.); (E.S.); (F.B.); (M.C.); (W.D.); (E.v.H.); (N.K.); (L.L.); (C.L.); (H.S.-H.); (J.W.); (U.W.); (B.Z.); (B.O.)
| | - Georgios Eleftheriadis
- Department of Nephrology and Medical Intensive Care, Charité–Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany; (K.B.); (F.H.); (G.E.); (M.G.N.); (E.S.); (F.B.); (M.C.); (W.D.); (E.v.H.); (N.K.); (L.L.); (C.L.); (H.S.-H.); (J.W.); (U.W.); (B.Z.); (B.O.)
| | - Marcel G. Naik
- Department of Nephrology and Medical Intensive Care, Charité–Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany; (K.B.); (F.H.); (G.E.); (M.G.N.); (E.S.); (F.B.); (M.C.); (W.D.); (E.v.H.); (N.K.); (L.L.); (C.L.); (H.S.-H.); (J.W.); (U.W.); (B.Z.); (B.O.)
| | - Eva Schrezenmeier
- Department of Nephrology and Medical Intensive Care, Charité–Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany; (K.B.); (F.H.); (G.E.); (M.G.N.); (E.S.); (F.B.); (M.C.); (W.D.); (E.v.H.); (N.K.); (L.L.); (C.L.); (H.S.-H.); (J.W.); (U.W.); (B.Z.); (B.O.)
| | - Friederike Bachmann
- Department of Nephrology and Medical Intensive Care, Charité–Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany; (K.B.); (F.H.); (G.E.); (M.G.N.); (E.S.); (F.B.); (M.C.); (W.D.); (E.v.H.); (N.K.); (L.L.); (C.L.); (H.S.-H.); (J.W.); (U.W.); (B.Z.); (B.O.)
| | - Mira Choi
- Department of Nephrology and Medical Intensive Care, Charité–Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany; (K.B.); (F.H.); (G.E.); (M.G.N.); (E.S.); (F.B.); (M.C.); (W.D.); (E.v.H.); (N.K.); (L.L.); (C.L.); (H.S.-H.); (J.W.); (U.W.); (B.Z.); (B.O.)
| | - Wiebke Duettmann
- Department of Nephrology and Medical Intensive Care, Charité–Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany; (K.B.); (F.H.); (G.E.); (M.G.N.); (E.S.); (F.B.); (M.C.); (W.D.); (E.v.H.); (N.K.); (L.L.); (C.L.); (H.S.-H.); (J.W.); (U.W.); (B.Z.); (B.O.)
| | - Ellen von Hoerschelmann
- Department of Nephrology and Medical Intensive Care, Charité–Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany; (K.B.); (F.H.); (G.E.); (M.G.N.); (E.S.); (F.B.); (M.C.); (W.D.); (E.v.H.); (N.K.); (L.L.); (C.L.); (H.S.-H.); (J.W.); (U.W.); (B.Z.); (B.O.)
| | - Nadine Koch
- Department of Nephrology and Medical Intensive Care, Charité–Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany; (K.B.); (F.H.); (G.E.); (M.G.N.); (E.S.); (F.B.); (M.C.); (W.D.); (E.v.H.); (N.K.); (L.L.); (C.L.); (H.S.-H.); (J.W.); (U.W.); (B.Z.); (B.O.)
| | - Lutz Liefeldt
- Department of Nephrology and Medical Intensive Care, Charité–Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany; (K.B.); (F.H.); (G.E.); (M.G.N.); (E.S.); (F.B.); (M.C.); (W.D.); (E.v.H.); (N.K.); (L.L.); (C.L.); (H.S.-H.); (J.W.); (U.W.); (B.Z.); (B.O.)
| | - Christian Lücht
- Department of Nephrology and Medical Intensive Care, Charité–Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany; (K.B.); (F.H.); (G.E.); (M.G.N.); (E.S.); (F.B.); (M.C.); (W.D.); (E.v.H.); (N.K.); (L.L.); (C.L.); (H.S.-H.); (J.W.); (U.W.); (B.Z.); (B.O.)
| | - Henriette Straub-Hohenbleicher
- Department of Nephrology and Medical Intensive Care, Charité–Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany; (K.B.); (F.H.); (G.E.); (M.G.N.); (E.S.); (F.B.); (M.C.); (W.D.); (E.v.H.); (N.K.); (L.L.); (C.L.); (H.S.-H.); (J.W.); (U.W.); (B.Z.); (B.O.)
| | - Johannes Waiser
- Department of Nephrology and Medical Intensive Care, Charité–Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany; (K.B.); (F.H.); (G.E.); (M.G.N.); (E.S.); (F.B.); (M.C.); (W.D.); (E.v.H.); (N.K.); (L.L.); (C.L.); (H.S.-H.); (J.W.); (U.W.); (B.Z.); (B.O.)
| | - Ulrike Weber
- Department of Nephrology and Medical Intensive Care, Charité–Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany; (K.B.); (F.H.); (G.E.); (M.G.N.); (E.S.); (F.B.); (M.C.); (W.D.); (E.v.H.); (N.K.); (L.L.); (C.L.); (H.S.-H.); (J.W.); (U.W.); (B.Z.); (B.O.)
| | - Bianca Zukunft
- Department of Nephrology and Medical Intensive Care, Charité–Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany; (K.B.); (F.H.); (G.E.); (M.G.N.); (E.S.); (F.B.); (M.C.); (W.D.); (E.v.H.); (N.K.); (L.L.); (C.L.); (H.S.-H.); (J.W.); (U.W.); (B.Z.); (B.O.)
| | - Bilgin Osmanodja
- Department of Nephrology and Medical Intensive Care, Charité–Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany; (K.B.); (F.H.); (G.E.); (M.G.N.); (E.S.); (F.B.); (M.C.); (W.D.); (E.v.H.); (N.K.); (L.L.); (C.L.); (H.S.-H.); (J.W.); (U.W.); (B.Z.); (B.O.)
| |
Collapse
|
9
|
Liew MY, Mathews JI, Li A, Singh R, Jaramillo SA, Weiss ZF, Bowman K, Ankomah PO, Ghantous F, Lewis GD, Neuringer I, Bitar N, Lipiner T, Dighe AS, Kotton CN, Seaman MS, Lemieux JE, Goldberg MB. Delayed and Attenuated Antibody Responses to Coronavirus Disease 2019 Vaccination With Poor Cross-Variant Neutralization in Solid-Organ Transplant Recipients-A Prospective Longitudinal Study. Open Forum Infect Dis 2023; 10:ofad369. [PMID: 37577118 PMCID: PMC10414143 DOI: 10.1093/ofid/ofad369] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 07/12/2023] [Indexed: 08/15/2023] Open
Abstract
Background Therapeutically immunosuppressed transplant recipients exhibit attenuated responses to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccines. To elucidate the kinetics and variant cross-protection of vaccine-induced antibodies in this population, we conducted a prospective longitudinal study in heart and lung transplant recipients receiving the SARS-CoV-2 messenger RNA (mRNA) 3-dose vaccination series. Methods We measured longitudinal serum antibody and neutralization responses against the ancestral and major variants of SARS-CoV-2 in SARS-CoV-2-uninfected lung (n = 18) and heart (n = 17) transplant recipients, non-lung-transplanted patients with cystic fibrosis (n = 7), and healthy controls (n = 12) before, during, and after the primary mRNA vaccination series. Results Among healthy controls, strong anti-spike responses arose immediately following vaccination and displayed cross-neutralization against all variants. In contrast, among transplant recipients, after the first 2 vaccine doses, increases in antibody concentrations occurred gradually, and cross-neutralization was completely absent against the Omicron B.1.1.529 variant. However, most (73%) of the transplant recipients had a significant response to the third vaccine dose, reaching levels comparable to those of healthy controls, with improved but attenuated neutralization of immune evasive variants, particularly Beta, Gamma, and Omicron. Responses in non-lung-transplanted patients with cystic fibrosis paralleled those in healthy controls. Conclusions In this prospective, longitudinal analysis of variant-specific antibody responses, lung and heart transplant recipients display delayed and defective responses to the first 2 SARS-CoV-2 vaccine doses but significantly augmented responses to a third dose. Gaps in antibody-mediated immunity among transplant recipients are compounded by decreased neutralization against Omicron variants, leaving many patients with substantially weakened immunity against currently circulating variants.
Collapse
Affiliation(s)
- May Y Liew
- Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Josh I Mathews
- Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Amy Li
- Department of Pediatrics, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Rohan Singh
- Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Salvador A Jaramillo
- Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Zoe F Weiss
- Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Kathryn Bowman
- Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Pierre O Ankomah
- Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Fadi Ghantous
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Gregory D Lewis
- Heart Transplant Program, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Isabel Neuringer
- Pulmonary and Critical Care, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Natasha Bitar
- Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Taryn Lipiner
- Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Anand S Dighe
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Camille N Kotton
- Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Michael S Seaman
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Jacob E Lemieux
- Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
- Infectious Disease and Microbiome Program, The Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, Massachusetts, USA
- Department of Microbiology, Harvard Medical School, Boston, Massachusetts, USA
| | - Marcia B Goldberg
- Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
- Infectious Disease and Microbiome Program, The Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, Massachusetts, USA
- Department of Microbiology, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
10
|
Müller TR, Sekine T, Trubach D, Niessl J, Chen P, Bergman P, Blennow O, Hansson L, Mielke S, Nowak P, Vesterbacka J, Akber M, Olofsson A, Amaya Hernandez SP, Gao Y, Cai C, Söderdahl G, Smith CIE, Österborg A, Loré K, Sällberg Chen M, Ljungman P, Ljunggren HG, Karlsson AC, Saini SK, Aleman S, Buggert M. Additive effects of booster mRNA vaccination and SARS-CoV-2 Omicron infection on T cell immunity across immunocompromised states. Sci Transl Med 2023; 15:eadg9452. [PMID: 37437015 PMCID: PMC7615622 DOI: 10.1126/scitranslmed.adg9452] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 06/23/2023] [Indexed: 07/14/2023]
Abstract
Suboptimal immunity to SARS-CoV-2 mRNA vaccination has frequently been observed in individuals with various immunodeficiencies. Given the increased antibody evasion properties of emerging SARS-CoV-2 subvariants, it is necessary to assess whether other components of adaptive immunity generate resilient and protective responses against infection. We assessed T cell responses in 279 individuals, covering five different immunodeficiencies and healthy controls, before and after booster mRNA vaccination, as well as after Omicron infection in a subset of patients. We observed robust and persistent Omicron-reactive T cell responses that increased markedly upon booster vaccination and correlated directly with antibody titers across all patient groups. Poor vaccination responsiveness in immunocompromised or elderly individuals was effectively counteracted by the administration of additional vaccine doses. Functionally, Omicron-reactive T cell responses exhibited a pronounced cytotoxic profile and signs of longevity, characterized by CD45RA+ effector memory subpopulations with stem cell-like properties and increased proliferative capacity. Regardless of underlying immunodeficiency, booster-vaccinated and Omicron-infected individuals appeared protected against severe disease and exhibited enhanced and diversified T cell responses against conserved and Omicron-specific epitopes. Our findings indicate that T cells retain the ability to generate highly functional responses against newly emerging variants, even after repeated antigen exposure and a robust immunological imprint from ancestral SARS-CoV-2 mRNA vaccination.
Collapse
Affiliation(s)
- Thomas R. Müller
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Takuya Sekine
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Darya Trubach
- Department of Health Technology, Section of Experimental and Translational Immunology, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Julia Niessl
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Puran Chen
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Peter Bergman
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
- Department of Laboratory Medicine, Clinical Immunology, Karolinska Institutet, Stockholm, Sweden
- Department of Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Ola Blennow
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
- Department of Transplantation, Karolinska University Hospital, Stockholm, Sweden
- Department of Medicine Huddinge, Infectious Diseases, Karolinska Institutet, Stockholm, Sweden
| | - Lotta Hansson
- Department of Hematology, Karolinska University Hospital, Stockholm, Sweden
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Stephan Mielke
- Department of Laboratory Medicine, Biomolecular and Cellular Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska Comprehensive Cancer Center, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Piotr Nowak
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
- Department of Medicine Huddinge, Infectious Diseases, Karolinska Institutet, Stockholm, Sweden
- Laboratory for Molecular Infection Medicine Sweden MIMS, Umeå University, Sweden
| | - Jan Vesterbacka
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
- Department of Medicine Huddinge, Infectious Diseases, Karolinska Institutet, Stockholm, Sweden
| | - Mira Akber
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Anna Olofsson
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Susana Patricia Amaya Hernandez
- Department of Health Technology, Section of Experimental and Translational Immunology, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Yu Gao
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Curtis Cai
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Gunnar Söderdahl
- Department of Transplantation, Karolinska University Hospital, Stockholm, Sweden
- Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - C. I. Edvard Smith
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
- Department of Laboratory Medicine, Biomolecular and Cellular Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska Comprehensive Cancer Center, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Anders Österborg
- Department of Hematology, Karolinska University Hospital, Stockholm, Sweden
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Karin Loré
- Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | | | - Per Ljungman
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska Comprehensive Cancer Center, Karolinska University Hospital Huddinge, Stockholm, Sweden
- Department of Medicine Huddinge, Hematology, Karolinska Institutet, Stockholm
| | - Hans-Gustaf Ljunggren
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Annika C. Karlsson
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
- Karolinska University Laboratory, Clinical Microbiology, Karolinska University Hospital, Stockholm, Sweden
| | - Sunil Kumar Saini
- Department of Health Technology, Section of Experimental and Translational Immunology, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Soo Aleman
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
- Department of Medicine Huddinge, Infectious Diseases, Karolinska Institutet, Stockholm, Sweden
| | - Marcus Buggert
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
11
|
Meredith RT, Bermingham MD, Bentley K, Agah S, Aboagye-Odei A, Yarham RAR, Mills H, Shaikh M, Hoye N, Stanton RJ, Chadwick DR, Oliver MA. Differential cellular and humoral immune responses in immunocompromised individuals following multiple SARS-CoV-2 vaccinations. Front Cell Infect Microbiol 2023; 13:1207313. [PMID: 37424787 PMCID: PMC10327606 DOI: 10.3389/fcimb.2023.1207313] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 06/06/2023] [Indexed: 07/11/2023] Open
Abstract
Introduction The heterogeneity of the immunocompromised population means some individuals may exhibit variable, weak or reduced vaccine-induced immune responses, leaving them poorly protected from COVID-19 disease despite receiving multiple SARS-CoV-2 vaccinations. There is conflicting data on the immunogenicity elicited by multiple vaccinations in immunocompromised groups. The aim of this study was to measure both humoral and cellular vaccine-induced immunity in several immunocompromised cohorts and to compare them to immunocompetent controls. Methods Cytokine release in peptide-stimulated whole blood, and neutralising antibody and baseline SARS-CoV-2 spike-specific IgG levels in plasma were measured in rheumatology patients (n=29), renal transplant recipients (n=46), people living with HIV (PLWH) (n=27) and immunocompetent participants (n=64) post third or fourth vaccination from just one blood sample. Cytokines were measured by ELISA and multiplex array. Neutralising antibody levels in plasma were determined by a 50% neutralising antibody titre assay and SARS-CoV-2 spike specific IgG levels were quantified by ELISA. Results In infection negative donors, IFN-γ, IL-2 and neutralising antibody levels were significantly reduced in rheumatology patients (p=0.0014, p=0.0415, p=0.0319, respectively) and renal transplant recipients (p<0.0001, p=0.0005, p<0.0001, respectively) compared to immunocompetent controls, with IgG antibody responses similarly affected. Conversely, cellular and humoral immune responses were not impaired in PLWH, or between individuals from all groups with previous SARS-CoV-2 infections. Discussion These results suggest that specific subgroups within immunocompromised cohorts could benefit from distinct, personalised immunisation or treatment strategies. Identification of vaccine non-responders could be critical to protect those most at risk.
Collapse
Affiliation(s)
| | | | - Kirsten Bentley
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Sayeh Agah
- InBio, Charlottesville, VA, United States
| | - Abigail Aboagye-Odei
- Department of Infectious Diseases, South Tees Hospitals National Health Service (NHS) Foundation Trust, Middlesbrough, England, United Kingdom
| | | | | | - Muddassir Shaikh
- Department of Kidney Services, South Tees Hospitals National Health Service (NHS) Foundation Trust, Middlesbrough, England, United Kingdom
| | - Neil Hoye
- Department of Rheumatology, South Tees Hospitals National Health Service (NHS) Foundation Trust, Middlesbrough, England, United Kingdom
| | - Richard J. Stanton
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - David R. Chadwick
- Department of Infectious Diseases, South Tees Hospitals National Health Service (NHS) Foundation Trust, Middlesbrough, England, United Kingdom
| | | |
Collapse
|
12
|
Lee B, Nanishi E, Levy O, Dowling DJ. Precision Vaccinology Approaches for the Development of Adjuvanted Vaccines Targeted to Distinct Vulnerable Populations. Pharmaceutics 2023; 15:1766. [PMID: 37376214 PMCID: PMC10305121 DOI: 10.3390/pharmaceutics15061766] [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: 05/04/2023] [Revised: 06/11/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
Infection persists as one of the leading global causes of morbidity and mortality, with particular burden at the extremes of age and in populations who are immunocompromised or suffer chronic co-morbid diseases. By focusing discovery and innovation efforts to better understand the phenotypic and mechanistic differences in the immune systems of diverse vulnerable populations, emerging research in precision vaccine discovery and development has explored how to optimize immunizations across the lifespan. Here, we focus on two key elements of precision vaccinology, as applied to epidemic/pandemic response and preparedness, including (a) selecting robust combinations of adjuvants and antigens, and (b) coupling these platforms with appropriate formulation systems. In this context, several considerations exist, including the intended goals of immunization (e.g., achieving immunogenicity versus lessening transmission), reducing the likelihood of adverse reactogenicity, and optimizing the route of administration. Each of these considerations is accompanied by several key challenges. On-going innovation in precision vaccinology will expand and target the arsenal of vaccine components for protection of vulnerable populations.
Collapse
Affiliation(s)
- Branden Lee
- Precision Vaccines Program, Boston Children’s Hospital, Boston, MA 02115, USA; (B.L.); (E.N.); (O.L.)
| | - Etsuro Nanishi
- Precision Vaccines Program, Boston Children’s Hospital, Boston, MA 02115, USA; (B.L.); (E.N.); (O.L.)
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Ofer Levy
- Precision Vaccines Program, Boston Children’s Hospital, Boston, MA 02115, USA; (B.L.); (E.N.); (O.L.)
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - David J. Dowling
- Precision Vaccines Program, Boston Children’s Hospital, Boston, MA 02115, USA; (B.L.); (E.N.); (O.L.)
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
13
|
Werbel WA, Karaba AH, Chiang TPY, Massie AB, Brown DM, Watson N, Chahoud M, Thompson EA, Johnson AC, Avery RK, Cochran WV, Warren D, Liang T, Fribourg M, Huerta C, Samaha H, Klein SL, Bettinotti MP, Clarke WA, Sitaras I, Rouphael N, Cox AL, Bailey JR, Pekosz A, Tobian AAR, Durand CM, Bridges ND, Larsen CP, Heeger PS, Segev DL. Persistent SARS-CoV-2-specific immune defects in kidney transplant recipients following third mRNA vaccine dose. Am J Transplant 2023; 23:744-758. [PMID: 36966905 PMCID: PMC10037915 DOI: 10.1016/j.ajt.2023.03.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/17/2023] [Accepted: 03/17/2023] [Indexed: 03/26/2023]
Abstract
Kidney transplant recipients (KTRs) show poorer response to SARS-CoV-2 mRNA vaccination, yet response patterns and mechanistic drivers following third doses are ill-defined. We administered third monovalent mRNA vaccines to n = 81 KTRs with negative or low-titer anti-receptor binding domain (RBD) antibody (n = 39 anti-RBDNEG; n = 42 anti-RBDLO), compared with healthy controls (HCs, n = 19), measuring anti-RBD, Omicron neutralization, spike-specific CD8+%, and SARS-CoV-2-reactive T cell receptor (TCR) repertoires. By day 30, 44% anti-RBDNEG remained seronegative; 5% KTRs developed BA.5 neutralization (vs 68% HCs, P < .001). Day 30 spike-specific CD8+% was negative in 91% KTRs (vs 20% HCs; P = .07), without correlation to anti-RBD (rs = 0.17). Day 30 SARS-CoV-2-reactive TCR repertoires were detected in 52% KTRs vs 74% HCs (P = .11). Spike-specific CD4+ TCR expansion was similar between KTRs and HCs, yet KTR CD8+ TCR depth was 7.6-fold lower (P = .001). Global negative response was seen in 7% KTRs, associated with high-dose MMF (P = .037); 44% showed global positive response. Of the KTRs, 16% experienced breakthrough infections, with 2 hospitalizations; prebreakthrough variant neutralization was poor. Absent neutralizing and CD8+ responses in KTRs indicate vulnerability to COVID-19 despite 3-dose mRNA vaccination. Lack of neutralization despite CD4+ expansion suggests B cell dysfunction and/or ineffective T cell help. Development of more effective KTR vaccine strategies is critical. (NCT04969263).
Collapse
Affiliation(s)
- William A Werbel
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
| | - Andrew H Karaba
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Teresa Po-Yu Chiang
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Allan B Massie
- Department of Surgery, New York University Grossman School of Medicine, New York, New York, USA; Department of Population Health, New York University Grossman School of Medicine, New York, New York, USA
| | - Diane M Brown
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Natasha Watson
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Maggie Chahoud
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Elizabeth A Thompson
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | - Robin K Avery
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Willa V Cochran
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Daniel Warren
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Tao Liang
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Miguel Fribourg
- Translational Transplant Research Center, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | | | - Hady Samaha
- Department of Medicine, Emory University, Atlanta, Georgia, USA
| | - Sabra L Klein
- Department of Medicine, Emory University, Atlanta, Georgia, USA
| | - Maria P Bettinotti
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - William A Clarke
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Ioannis Sitaras
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Nadine Rouphael
- Department of Medicine, Emory University, Atlanta, Georgia, USA
| | - Andrea L Cox
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Department of Medicine, Emory University, Atlanta, Georgia, USA; Bloomberg Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Justin R Bailey
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Andrew Pekosz
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Aaron A R Tobian
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Christine M Durand
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Nancy D Bridges
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | | | - Peter S Heeger
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Dorry L Segev
- Department of Surgery, New York University Grossman School of Medicine, New York, New York, USA; Department of Population Health, New York University Grossman School of Medicine, New York, New York, USA
| |
Collapse
|
14
|
Ferreira VH, Hu Q, Kurtesi A, Solera JT, Ierullo M, Gingras AC, Kumar D, Humar A. Impact of Omicron BA.1 infection on BA.4/5 immunity in transplant recipients. Am J Transplant 2023; 23:278-283. [PMID: 36744606 PMCID: PMC9835003 DOI: 10.1016/j.ajt.2022.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/10/2022] [Accepted: 10/20/2022] [Indexed: 01/13/2023]
Abstract
Mutations in the spike protein of SARS-CoV-2 have allowed Omicron subvariants to escape neutralizing antibodies. The degree to which this occurs in transplant recipients is poorly understood. We measured BA.4/5 cross-neutralizing responses in 75 mostly vaccinated transplant recipients who recovered from BA.1 infection. Sera were collected at 1 and 6 months post-BA.1 infection, and a lentivirus pseudovirus neutralization assay was performed using spike constructs corresponding to BA.1 and BA.4/5. Uninfected immunized transplant recipients and health care worker controls were used for comparison. Following BA.1 infection, the proportion of transplant recipients with neutralizing antibody responses was 88.0% (66/75) against BA.1 and 69.3% (52/75) against BA.4/5 (P = .005). The neutralization level against BA.4/5 was approximately 17-fold lower than that against BA.1 (IQR 10.6- to 45.1-fold lower, P < .0001). BA.4/5 responses declined over time and by ≥0.5 log10 (approximately 3-fold) in almost half of the patients by 6 months. BA.4/5-neutralizing antibody titers in transplant recipients with breakthrough BA.1 infection were similar to those in immunized health care workers but significantly lower than those in uninfected triple-vaccinated transplant recipients. These results provide evidence that transplant recipients are at ongoing risk for BA.4/5 infection despite vaccination and prior Omicron strain infection, and additional mitigation strategies may be required to prevent severe disease in this cohort.
Collapse
Affiliation(s)
- Victor H. Ferreira
- University Health Network, Ajmera Transplant Centre, Toronto, Ontario, Canada
| | - Queenie Hu
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Sinai Health, Toronto, Ontario, Canada
| | - Alexandra Kurtesi
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Sinai Health, Toronto, Ontario, Canada
| | - Javier T. Solera
- University Health Network, Ajmera Transplant Centre, Toronto, Ontario, Canada
| | - Matthew Ierullo
- University Health Network, Ajmera Transplant Centre, Toronto, Ontario, Canada
| | - Anne-Claude Gingras
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Sinai Health, Toronto, Ontario, Canada,Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Deepali Kumar
- University Health Network, Ajmera Transplant Centre, Toronto, Ontario, Canada
| | - Atul Humar
- University Health Network, Ajmera Transplant Centre, Toronto, Ontario, Canada.
| |
Collapse
|
15
|
Moioffer SJ, Berton RR, McGonagill PW, Jensen IJ, Griffith TS, Badovinac VP. Inefficient Recovery of Repeatedly Stimulated Memory CD8 T Cells after Polymicrobial Sepsis Induction Leads to Changes in Memory CD8 T Cell Pool Composition. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:168-179. [PMID: 36480268 PMCID: PMC9840817 DOI: 10.4049/jimmunol.2200676] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 11/09/2022] [Indexed: 01/03/2023]
Abstract
Long-lasting sepsis-induced immunoparalysis has been principally studied in primary (1°) memory CD8 T cells; however, the impact of sepsis on memory CD8 T cells with a history of repeated cognate Ag encounters is largely unknown but important in understanding the role of sepsis in shaping the pre-existing memory CD8 T cell compartment. Higher-order memory CD8 T cells are crucial in providing immunity against common pathogens that reinfect the host or are generated by repeated vaccination. In this study, we analyzed peripheral blood from septic patients and show that memory CD8 T cells with defined Ag specificity for recurring CMV infection proliferate less than bulk populations of central memory CD8 T cells. Using TCR-transgenic T cells to generate 1° and higher-order (quaternary [4°]) memory T cells within the same host, we demonstrate that the susceptibility and loss of both memory subsets are similar after sepsis induction, and sepsis diminished Ag-dependent and -independent (bystander) functions of these memory subsets equally. Both the 1° and 4° memory T cell populations proliferated in a sepsis-induced lymphopenic environment; however, due to the intrinsic differences in baseline proliferative capacity, expression of receptors (e.g., CD127/CD122), and responsiveness to homeostatic cytokines, 1° memory T cells become overrepresented over time in sepsis survivors. Finally, IL-7/anti-IL-7 mAb complex treatment early after sepsis induction preferentially rescued the proliferation and accumulation of 1° memory T cells, whereas recovery of 4° memory T cells was less pronounced. Thus, inefficient recovery of repeatedly stimulated memory cells after polymicrobial sepsis induction leads to changes in memory T cell pool composition, a notion with important implications in devising strategies to recover the number and function of pre-existing memory CD8 T cells in sepsis survivors.
Collapse
Affiliation(s)
| | - Roger R. Berton
- Department of Pathology, University of Iowa, Iowa City, IA;,Interdisciplinary Program in Immunology, University of Iowa, Iowa City, IA
| | | | - Isaac J. Jensen
- Columbia University Irving Medical Center, University of Minnesota, Minneapolis, MN
| | - Thomas S. Griffith
- Department of Urology, University of Minnesota, Minneapolis, MN,,Minneapolis Veterans Affairs Health Care System, Minneapolis, MN
| | - Vladimir P. Badovinac
- Department of Pathology, University of Iowa, Iowa City, IA;,Interdisciplinary Program in Immunology, University of Iowa, Iowa City, IA
| |
Collapse
|
16
|
Utilization of the Abbott SARS-CoV-2 IgG II Quant Assay To Identify High-Titer Anti-SARS-CoV-2 Neutralizing Plasma against Wild-Type and Variant SARS-CoV-2 Viruses. Microbiol Spectr 2022; 10:e0281122. [PMID: 36125288 PMCID: PMC9602363 DOI: 10.1128/spectrum.02811-22] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
There is evidence that COVID-19 convalescent plasma may improve outcomes of patients with impaired immune systems; however, more clinical trials are required. Although we have previously used a 50% plaque reduction/neutralization titer (PRNT50) assay to qualify convalescent plasma for clinical trials and virus-like particle (VLP) assays to validate PRNT50 methodologies, these approaches are time-consuming and expensive. Here, we characterized the ability of the Abbott severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) IgG II Quant assay to identify high- and low-titer plasma for wild-type and variant (Alpha, Beta, Gamma, and Delta) SARS-CoV-2 characterized by both VLP assays and PRNT50. Plasma specimens previously tested in wild-type, Alpha, Beta, Gamma, and Delta VLP neutralization assays were selected based on availability. Selected specimens were evaluated by the Abbott SARS-CoV-2 IgG II Quant assay [Abbott anti-Spike (S); Abbott, Chicago, IL], and values in units per milliliter were converted to binding antibody units (BAU) per milliliter. Sixty-three specimens were available for analysis. Abbott SARS-CoV-2 IgG II Quant assay values in BAU per milliliter were significantly different between high- and low-titer specimens for wild-type (Mann-Whitney U = 42, P < 0.0001), Alpha (Mann-Whitney U = 38, P < 0.0001), Beta (Mann-Whitney U = 29, P < 0.0001), Gamma (Mann-Whitney U = 0, P < 0.0001), and Delta (Mann-Whitney U = 42, P < 0.0001). A conservative approach using the highest 95% confidence interval (CI) values from wild-type and variant of concern (VOC) SARS-CoV-2 experiments would identify a potential Abbott SARS-CoV-2 IgG II Quant assay cutoff of ≥7.1 × 103 BAU/mL. IMPORTANCE The United States Food and Drug Administration (FDA) issued an Emergency Use Authorization (EUA) for the use of COVID-19 convalescent plasma (CCP) to treat hospitalized patients with COVID-19 in August 2020. However, by 4 February 2021, the FDA had revised the convalescent plasma EUA. This revision limited the authorization for high-titer COVID-19 convalescent plasma and restricted patient groups to hospitalized patients with COVID-19 early in their disease course or hospitalized patients with impaired humoral immunity. Traditionally our group utilized 50% plaque reduction/neutralization titer (PRNT50) assays to qualify CCP in Canada. Since that time, the Abbott SARS-CoV-2 IgG II Quant assay (Abbott, Chicago IL) was developed for the qualitative and quantitative determination of IgG against the SARS-CoV-2. Here, we characterized the ability of the Abbott SARS-CoV-2 IgG II Quant assay to identify high- and low-titer plasma for wild-type and variant (Alpha, Beta, Gamma, and Delta) SARS-CoV-2.
Collapse
|