1
|
Naiditch H, Betts MR, Larman HB, Levi M, Rosenberg AZ. Immunologic and inflammatory consequences of SARS-CoV-2 infection and its implications in renal disease. Front Immunol 2025; 15:1376654. [PMID: 40012912 PMCID: PMC11861071 DOI: 10.3389/fimmu.2024.1376654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 12/23/2024] [Indexed: 02/28/2025] Open
Abstract
The emergence of the COVID-19 pandemic made it critical to understand the immune and inflammatory responses to the SARS-CoV-2 virus. It became increasingly recognized that the immune response was a key mediator of illness severity and that its mechanisms needed to be better understood. Early infection of both tissue and immune cells, such as macrophages, leading to pyroptosis-mediated inflammasome production in an organ system critical for systemic oxygenation likely plays a central role in the morbidity wrought by SARS-CoV-2. Delayed transcription of Type I and Type III interferons by SARS-CoV-2 may lead to early disinhibition of viral replication. Cytokines such as interleukin-1 (IL-1), IL-6, IL-12, and tumor necrosis factor α (TNFα), some of which may be produced through mechanisms involving nuclear factor kappa B (NF-κB), likely contribute to the hyperinflammatory state in patients with severe COVID-19. Lymphopenia, more apparent among natural killer (NK) cells, CD8+ T-cells, and B-cells, can contribute to disease severity and may reflect direct cytopathic effects of SARS-CoV-2 or end-organ sequestration. Direct infection and immune activation of endothelial cells by SARS-CoV-2 may be a critical mechanism through which end-organ systems are impacted. In this context, endovascular neutrophil extracellular trap (NET) formation and microthrombi development can be seen in the lungs and other critical organs throughout the body, such as the heart, gut, and brain. The kidney may be among the most impacted extrapulmonary organ by SARS-CoV-2 infection owing to a high concentration of ACE2 and exposure to systemic SARS-CoV-2. In the kidney, acute tubular injury, early myofibroblast activation, and collapsing glomerulopathy in select populations likely account for COVID-19-related AKI and CKD development. The development of COVID-19-associated nephropathy (COVAN), in particular, may be mediated through IL-6 and signal transducer and activator of transcription 3 (STAT3) signaling, suggesting a direct connection between the COVID-19-related immune response and the development of chronic disease. Chronic manifestations of COVID-19 also include systemic conditions like Multisystem Inflammatory Syndrome in Children (MIS-C) and Adults (MIS-A) and post-acute sequelae of COVID-19 (PASC), which may reflect a spectrum of clinical presentations of persistent immune dysregulation. The lessons learned and those undergoing continued study likely have broad implications for understanding viral infections' immunologic and inflammatory consequences beyond coronaviruses.
Collapse
Affiliation(s)
- Hiam Naiditch
- Department of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Michael R. Betts
- Department of Microbiology and Institute of Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - H. Benjamin Larman
- Institute for Cell Engineering, Division of Immunology, Department of Pathology, Johns Hopkins University, Baltimore, MD, United States
| | - Moshe Levi
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, DC, United States
| | - Avi Z. Rosenberg
- Department of Pathology, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
2
|
Trimbake D, Singh D, K. YG, Babar P, S. VD, Tripathy AS. Durability of Functional SARS-CoV-2-Specific Immunological Memory and T Cell Response up to 8-9 Months Postrecovery From COVID-19. J Immunol Res 2025; 2025:9743866. [PMID: 39963186 PMCID: PMC11832264 DOI: 10.1155/jimr/9743866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 12/19/2024] [Indexed: 02/20/2025] Open
Abstract
Research on long-term follow-up in individuals who have recovered from coronavirus disease-19 (COVID-19) would yield insights regarding their immunity status and identify those who need booster vaccinations. This study evaluated the longevity of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-specific cellular and humoral memory responses, as well as T cell effector functionalities, at 1-2 months (n = 40), 8-9 months (n = 40), and 12 months/1 year (n = 27) following recovery from SARS-CoV-2 infection. CTL response by enzyme-linked immunospot (ELISPOT); levels of cytokine by Bio-Plex, natural killer (NK), CD4+ helper, and CD8+ cytotoxic T cell functionalities using flow cytometry; anti-SARS-CoV-2 IgG by ELISA; and levels of neutralizing antibodies (NAbs) by surrogate virus NAb assay were assessed. The levels of SARS-CoV-2-specific IgG and NAb at 1-2 and 8-9 months postrecovery were hand in hand and appeared declining. SARS-CoV-2-specific B, memory B and plasma cells, and T cells sustained up to 8-9 months. Increased expression of CD107a/IFN-γ by NK cells and cytotoxic T cells at 8-9 months could be indicative of SARS-CoV-2-specific effector functions. Recovered individuals with positive and negative IgG antibody status displayed T cell response up to 1 year and 8-9 months, respectively, emphasizing the durabilty of effector immunity up to 8-9 months regardless of IgG antibody status. Overall, the recovered individuals exhibited robust immunological memory, sustained T cell response with effector functionality against SARS-CoV-2 that persists for at least 8-9 months.
Collapse
Affiliation(s)
- Diptee Trimbake
- Department of Dengue and Chikungunya, Indian Council of Medical Research-National Institute of Virology, 20-A, Dr Ambedkar Road, Pune 411001, India
| | - Dharmendra Singh
- Department of Dengue and Chikungunya, Indian Council of Medical Research-National Institute of Virology, 20-A, Dr Ambedkar Road, Pune 411001, India
| | - Yogesh Gurav K.
- Department of Dengue and Chikungunya, Indian Council of Medical Research-National Institute of Virology, 20-A, Dr Ambedkar Road, Pune 411001, India
| | - Prasad Babar
- Department of Dengue and Chikungunya, Indian Council of Medical Research-National Institute of Virology, 20-A, Dr Ambedkar Road, Pune 411001, India
| | - Varsha Dange S.
- Department of Medicine, Pimpri Chinchwad Municipal Corporation, Pimpri, Pune 411018, Maharashtra, India
| | - Anuradha S. Tripathy
- Department of Dengue and Chikungunya, Indian Council of Medical Research-National Institute of Virology, 20-A, Dr Ambedkar Road, Pune 411001, India
| |
Collapse
|
3
|
Karaba AH, Xue J, Johnston TS, Traut CC, Dalrymple LS, Kossmann RJ, Blankson JN, Parikh CR, Ray SC. Longitudinal Characterization of SARS-CoV-2 Immunity in Hemodialysis Patients Post Omicron. Kidney Int Rep 2025; 10:406-415. [PMID: 39990910 PMCID: PMC11843127 DOI: 10.1016/j.ekir.2024.11.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 10/28/2024] [Accepted: 11/12/2024] [Indexed: 02/25/2025] Open
Abstract
Introduction Individuals receiving hemodialysis (HD) are at risk for severe COVID-19 and have attenuated responses to COVID-19 vaccines. Evolution of immunity and risk for subsequent infection with additional vaccinations and infections in this population is poorly understood. Methods An observational multicenter cohort of 55 patients receiving HD in community HD centers, majority (85%) with at least 2 doses of COVID-19 vaccine (56% female, age [median; interquartile range, IQR] of 67, [58.0-74.0] years), was followed-up with for 50 weeks between December, 2021 and April, 2023 and collected blood samples at enrollment, 8 weeks, and 24 weeks thereafter. Anti-SARS-CoV-2 IgG and ACE2 inhibition (surrogate neutralization) against ancestral, Delta, and Omicron subvariants was measured. T-cell responses to Spike and Mucleocapsid proteins were measured via enzyme-linked immunosorbent spot. Changes in antibody and T cell responses were assessed by paired Wilcoxon rank-sum testing and Fisher exact testing. Antibody responses were compared to thrice vaccinated healthy controls (HCs) as a benchmark for what optimal responses could have been in the early Omicron period. Results Neutralization did not increase over time, and HD participants had lower neutralization than HCs. Only 56% of HD participants had a positive T cell response to spike after the BA.1/2 wave. Antibody and cellular responses were concordant in only 34.5% at final visit. Antibody responses trended higher among those with prior COVID-19, but spike-specific T cell responses did not vary. Conclusions Original vaccine formulations and previous infection are insufficient to induce reliable SARS-CoV-2 responses in individuals on HD, suggesting that updated annual COVID-19 vaccines and transmission-based precautions remain critical in this population.
Collapse
Affiliation(s)
- Andrew H. Karaba
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jiashu Xue
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Trevor S. Johnston
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Caroline C. Traut
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | - Robert J. Kossmann
- Global Medical Office, Fresenius Medical Care, Waltham, Massachusetts, USA
| | - Joel N. Blankson
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Chirag R. Parikh
- Department of Medicine, Division of Nephrology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Stuart C. Ray
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
4
|
Boongird S, Srithongkul T, Sethakarun S, Bruminhent J, Kiertiburanakul S, Nongnuch A, Kitiyakara C, Sritippayawan S. Tixagevimab-cilgavimab for preventing breakthrough COVID-19 in dialysis patients: a prospective study. Clin Kidney J 2024; 17:sfae309. [PMID: 39539359 PMCID: PMC11558061 DOI: 10.1093/ckj/sfae309] [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: 06/26/2024] [Indexed: 11/16/2024] Open
Abstract
Background The effectiveness of tixagevimab-cilgavimab as pre-exposure prophylaxis (PrEP) against breakthrough coronavirus disease 2019 (COVID-19) in dialysis patients remains uncertain due to limited data. Methods In this multicenter prospective study, we enrolled vaccinated dialysis patients and divided them into two groups: a tixagevimab-cilgavimab group (received a 150 mg/150 mg intramuscular dose of tixagevimab-cilgavimab) and a control group (age-matched patients not receiving tixagevimab-cilgavimab). The primary outcome was the breakthrough COVID-19 rate at 6 months, whereas secondary outcomes included COVID-19-related hospitalization, intensive care unit admission, endotracheal intubation and mortality. The safety of tixagevimab-cilgavimab was assessed. Results Two hundred participants were enrolled, with equal numbers in each group (n = 100 each). Baseline characteristics were comparable between groups, except for a higher number of COVID-19 vaccine doses in the tixagevimab-cilgavimab group [median (IQR) 4 (3-5) vs. 3 (3-4); P = .01]. At 6 months, the breakthrough COVID-19 rates were comparable between the tixagevimab-cilgavimab (17%) and control (15%) groups (P = .66). However, the median (IQR) time to diagnosis of breakthrough infections tended to be longer in the tixagevimab-cilgavimab group [4.49 (2.81-4.98) vs 1.96 (1.65-2.91) months; P = .08]. Tixagevimab-cilgavimab significantly reduced COVID-19-related hospitalization rates (5.9% vs 40.0%; P = .02) among participants with breakthrough infections. All tixagevimab-cilgavimab-related adverse events were mild. Conclusion The use of tixagevimab-cilgavimab as PrEP in vaccinated dialysis patients during the Omicron surge did not prevent breakthrough infections but significantly reduced COVID-19-related hospitalizations. Further research should prioritize alternative strategies.
Collapse
Affiliation(s)
- Sarinya Boongird
- Division of Nephrology, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Thatsaphan Srithongkul
- Division of Nephrology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | | | - Jackrapong Bruminhent
- Division of Infectious Diseases, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Sasisopin Kiertiburanakul
- Division of Infectious Diseases, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Arkom Nongnuch
- Division of Nephrology, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Chagriya Kitiyakara
- Division of Nephrology, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Suchai Sritippayawan
- Division of Nephrology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
5
|
Bota SE, McArthur E, Naylor KL, Blake PG, Yau K, Hladunewich MA, Levin A, Oliver MJ. Long-Term Morbidity and Mortality of Coronavirus Disease 2019 in Patients Receiving Maintenance Dialysis: A Multicenter Population-Based Cohort Study. KIDNEY360 2024; 5:1116-1125. [PMID: 39151048 PMCID: PMC11371337 DOI: 10.34067/kid.0000000000000490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 06/04/2024] [Indexed: 08/18/2024]
Abstract
Key Points The rates of long-term mortality, reinfection, cardiovascular outcomes, and hospitalization were high among coronavirus disease 2019 (COVID-19) survivors on maintenance dialysis. Several risk factors, including intensive care unit admission related to COVID-19 and reinfection, were found to have a prolonged effect on survival. This study shows that the burden of COVID-19 remains high after the period of acute infection in the population receiving maintenance dialysis. Background Many questions remain about the population receiving maintenance dialysis who survived coronavirus disease 2019 (COVID-19). Previous literature has focused on outcomes associated with the initial severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, but it may underestimate the effect of disease. This study describes the long-term morbidity and mortality among patients receiving maintenance dialysis in Ontario, Canada, who survived SARS-CoV-2 infection and the risk factors associated with long-term mortality. Methods We conducted a population-based cohort study of patients receiving maintenance dialysis in Ontario, Canada, who tested positive for SARS-CoV-2 and survived 30 days between March 14, 2020, and December 1, 2021 (pre-Omicron), with follow-up until September 30, 2022. Our primary outcome was all-cause mortality while our secondary outcomes included reinfection, composite of cardiovascular (CV)–related death or hospitalization, all-cause hospitalization, and admission to long-term care or complex continuing care. We also examined risk factors associated with long-term mortality using multivariable Cox proportional hazards regression. Results We included 798 COVID-19 survivors receiving maintenance dialysis. After the first 30 days of infection, death occurred at a rate of 15.0 per 100 person-years (95% confidence interval [CI], 12.9 to 17.5) over a median follow-up of 1.4 years (interquartile range, 1.1–1.7) with a nadir of death at approximately 0.5 years. Reinfection, composite CV death or hospitalization, and all-cause hospitalization occurred at a rate (95% CI) of 15.9 (13.6 to 18.5), 17.4 (14.9 to 20.4), and 73.1 (66.6 to 80.2) per 100 person-years, respectively. In addition to traditional predictors of mortality, intensive care unit admission for COVID-19 had a prolonged effect on survival (adjusted hazard ratio, 2.6; 95% CI, 1.6 to 4.3). Reinfection with SARS-CoV-2 among 30-day survivors increased all-cause mortality (adjusted hazard ratio, 2.2; 95% CI, 1.4 to 3.3). Conclusions The burden of COVID-19 persists beyond the period of acute infection in the population receiving maintenance dialysis in Ontario with high rates of death, reinfection, all-cause hospitalization, and CV disease among COVID-19 survivors.
Collapse
Affiliation(s)
- Sarah E. Bota
- ICES, Toronto, Ontario, Canada
- Lawson Health Research Institute, London Health Sciences Centre, London, Ontario, Canada
| | - Eric McArthur
- ICES, Toronto, Ontario, Canada
- Lawson Health Research Institute, London Health Sciences Centre, London, Ontario, Canada
| | - Kyla L. Naylor
- ICES, Toronto, Ontario, Canada
- Lawson Health Research Institute, London Health Sciences Centre, London, Ontario, Canada
- Department of Epidemiology and Biostatistics, Western University, London, Ontario, Canada
| | - Peter G. Blake
- Lawson Health Research Institute, London Health Sciences Centre, London, Ontario, Canada
- Ontario Health, Toronto, Ontario, Canada
- Division of Nephrology, London Health Sciences Centre, London, Ontario, Canada
| | - Kevin Yau
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Michelle A. Hladunewich
- Ontario Health, Toronto, Ontario, Canada
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
- Division of Nephrology, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Adeera Levin
- Division of Nephrology, University of British Columbia, Vancouver, British Columbia, Canada
- BC Provincial Renal Agency, Vancouver, British Columbia, Canada
| | - Matthew J. Oliver
- Division of Nephrology, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| |
Collapse
|
6
|
Sanhueza ME, San Martín P, Brantes L, Caro S, Carrasco G, Machuca E. Efficacy of vaccination against the SARS-CoV-2 virus in patients with chronic kidney disease on hemodialysis. Hum Vaccin Immunother 2023; 19:2173904. [PMID: 36785953 PMCID: PMC10012891 DOI: 10.1080/21645515.2023.2173904] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/15/2023] Open
Abstract
SARS-CoV-2 has had a great impact on world health, patients on hemodialysis have a higher rate of infection and death due to COVID-19. Vaccination is important to control infection and improve the prognosis of infected patients. To describe the efficacy of vaccination against SARS-CoV-2 in Chilean patients on hemodialysis during the year 2021. Retrospective observational study. A total of 9,712 clinical records were reviewed. Data were presented as summary measures. Fisher's exact test, Mann-Whitney U test, and multivariate logistic regression were used for the analysis. Risk and survival analysis were calculated, considering a statistical significance of less than 0.05. The average age of the patients attended was 61.5 ± 14.6 years. Average time on dialysis 67.6 months and 35.0% diabetic. 93.2% of patients were vaccinated against SARS-CoV-2, 70.7% of them received booster doses. The risk of infection was higher for those who received one or no dose, compared to those who received booster doses against SARS-CoV-2: OR = 252.46 [165.13; 401.57]. Of the infected patients, 15.7% died from COVID-19. The risk of death was higher in unvaccinated or single-dose patients compared to those vaccinated with two doses: OR = 2.64 [2.23; 3.12]. Patients with two doses and a booster had a longer survival compared to those who received one or no dose of vaccination against SARS-CoV-2 (p < .05). The vaccination in Chile, which started in February 2021, has demonstrated that booster doses against SARS-CoV-2 significantly reduced the risk of infection, hospitalization, and death due to COVID-19 in patients on hemodialysis.
Collapse
Affiliation(s)
- María E Sanhueza
- National Medical Directorate, NephroCare-Chile,Santiago, Chile.,Nephrology DepartmentClinical Hospital, University of Chile, Santiago, Chile
| | | | - Loreto Brantes
- National Medical Directorate, NephroCare-Chile,Santiago, Chile
| | - Sylvia Caro
- National Medical Directorate, NephroCare-Chile,Santiago, Chile
| | | | - Eduardo Machuca
- National Medical Directorate, NephroCare-Chile,Santiago, Chile
| |
Collapse
|
7
|
Hsu CM, Weiner DE, Manley HJ, Li NC, Miskulin D, Harford A, Sanders R, Ladik V, Frament J, Argyropoulos C, Abreo K, Chin A, Gladish R, Salman L, Johnson D, Lacson EK. Serial SARS-CoV-2 Antibody Titers in Vaccinated Dialysis Patients: Prevalence of Unrecognized Infection and Duration of Seroresponse. Kidney Med 2023; 5:100718. [PMID: 37786901 PMCID: PMC10542005 DOI: 10.1016/j.xkme.2023.100718] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/04/2023] Open
Abstract
Rationale & Objective Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections are likely underdiagnosed, but the degree of underdiagnosis among patients receiving maintenance dialysis is unknown. The durability of the immune response after the third vaccine dose in this population also remains uncertain. This descriptive study tracked antibody levels to (1) assess the rate of undiagnosed infections and (2) characterize seroresponse durability after the third dose. Study Design Retrospective observational study. Setting & Participants SARS-CoV-2-vaccinated patients receiving maintenance dialysis through a national dialysis provider. Immunoglobulin G spike antibodies [anti-spike immunoglobulin (Ig) G] titers were assessed monthly after vaccination. Exposures Two and 3 doses of SARS-CoV-2 vaccine. Outcomes Undiagnosed and diagnosed SARS-CoV-2 infections; anti-spike IgG titers over time. Analytical Approach Undiagnosed SARS-CoV-2 infections were identified as an increase in anti-spike IgG titer of ≥100 BAU/mL, not associated with receipt of vaccine or diagnosed SARS-CoV-2 infection (by polymerase chain reaction test or antigen test). In descriptive analyses, anti-spike IgG titers were followed over time. Results Among 2,703 patients without previous coronavirus disease 2019 (COVID-19) who received an initial 2-dose vaccine series, 271 had diagnosed SARS-CoV-2 infections (3.4 per 10,000 patient-days) and 129 had undiagnosed SARS-CoV-2 infections (1.6 per 10,000 patient-days). Among 1,894 patients without previous COVID-19 who received a third vaccine dose, 316 had diagnosed SARS-CoV-2 infections (7.0 per 10,000 patient-days) and 173 had undiagnosed SARS-CoV-2 infections (3.8 per 10,000 patient-days). In both cohorts, anti-spike IgG levels declined over time. Of the initial 2-dose cohort, 66% had a titer of ≥500 BAU/mL in the first month, with 24% maintaining a titer of ≥500 BAU/mL at 6 months. Of the third dose cohort, 95% had a titer of ≥500 BAU/mL in the first month after the third dose, with 77% maintaining a titer of ≥500 BAU/mL at 6 months. Limitations The assays used had upper limits. Conclusions Among patients receiving maintenance dialysis, about 1 in every 3 SARS-CoV-2 infections was undiagnosed. Given this population's vulnerability to COVID-19, ongoing infection control measures are needed. A 3-dose primary mRNA vaccine series optimizes seroresponse rate and durability. Plain-Language Summary Patients receiving maintenance dialysis have been particularly vulnerable to COVID-19. Using serially measured antibodies, we found that a substantial proportion (about one-third) of SARS-CoV-2 infections among this population had been missed, both among those who had completed a 2-dose vaccine series and among those who had received a third vaccine dose. Such missed infections likely had only mild or minimal symptoms, but this failure to recognize all infections is concerning. Furthermore, vaccines have been effective among patients receiving dialysis, but our study additionally shows that the immune response wanes over time, even after a third dose. There is therefore a role for ongoing vigilance against this highly transmissible infection.
Collapse
Affiliation(s)
| | | | | | | | - Dana Miskulin
- Division of Nephrology, Tufts Medical Center, Boston, MA
| | - Antonia Harford
- Division of Nephrology, University of New Mexico, Albuquerque, NM
| | | | | | | | | | - Kenneth Abreo
- Division of Nephrology, Louisiana State University Health Sciences Center, Shreveport, LA
| | - Andrew Chin
- Division of Nephrology, University of California, Davis, Sacramento, CA
| | | | - Loay Salman
- Division of Nephrology, Albany Medical College, Albany, NY
| | | | - Eduardo K. Lacson
- Division of Nephrology, Tufts Medical Center, Boston, MA
- Dialysis Clinic Inc, Nashville, TN
| |
Collapse
|
8
|
Amellal H, Assaid N, Akarid K, Maaroufi A, Ezzikouri S, Sarih M. Mix-and-match COVID-19 vaccines trigger high antibody response after the third dose vaccine in Moroccan health care workers. Vaccine X 2023; 14:100288. [PMID: 37008956 PMCID: PMC10039700 DOI: 10.1016/j.jvacx.2023.100288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 03/22/2023] [Accepted: 03/23/2023] [Indexed: 03/29/2023] Open
Abstract
Recent studies have shown that in individuals who have received two doses of COVID-19 vaccine, the level of IgG antibodies decreased over time. In addition, the resurgence of the epidemic due to variants has led the authorities in several countries, including Morocco, to extend the third dose to the entire adult population. In this study, we included 43 healthcare workers (HCWs) who were vaccinated with three doses. They were vaccinated with ChAdOx1 nCoV-19 for the first two doses and with BNT 162b2 or BBIBP-CorV vaccine for the third dose. Humoral response was assessed on the day of injection of the third dose of vaccine and one month after the third dose by measuring anti-receptor-binding domain (RBD) IgG levels. Seven months after the second dose, the median titer of anti-RBD IgG was higher in the group with a history of SARS-CoV-2 infection than in the group with no history of infection (1038 AU/mL vs. 76.05 AU/mL, respectively, p = 0.003). One month after the third dose, a significant increase in median level of anti-RBD in both groups was observed: from 76.05 AU/mL to 6127 AU/mL in the group with no history of infection and from 1038 AU/mL to 14,412 AU/mL in the group with history of infection. Notably, the BNT 162b2 vaccine elicits a high titer of anti-RBD antibody compared to the BBIBP-CorV vaccine. Median antibody titers were 21,991 AU/mL and 3640 AU/mL for BNT 162b2 and BBIBP-CorV vaccines, respectively (p = 0.0002). 23% of HCWs were infected with SARS-CoV-2 within the first two months after the third dose injection. However, all these patients developed mild symptoms and tested negative by RT-qPCR between 10 and 15 days after the onset of symptoms. Our findings support that the third dose of COVID-19 vaccine significantly improves the humoral response and protects against the severe disease.
Collapse
Affiliation(s)
- Houda Amellal
- Service de Parasitologie et des Maladies Vectorielles, Institut Pasteur du Maroc, Place Louis Pasteur, 20360 Casablanca, Morocco
- Health and Environment Laboratory, Biochemistry, Biotechnology and Immunophysiopathology Research Team, Aïn Chock Faculty of Sciences, Hassan II University of Casablanca, Casablanca, Morocco
| | - Najlaa Assaid
- Service de Parasitologie et des Maladies Vectorielles, Institut Pasteur du Maroc, Place Louis Pasteur, 20360 Casablanca, Morocco
| | - Khadija Akarid
- Health and Environment Laboratory, Biochemistry, Biotechnology and Immunophysiopathology Research Team, Aïn Chock Faculty of Sciences, Hassan II University of Casablanca, Casablanca, Morocco
| | - Abderrahmane Maaroufi
- Service de Parasitologie et des Maladies Vectorielles, Institut Pasteur du Maroc, Place Louis Pasteur, 20360 Casablanca, Morocco
| | - Sayeh Ezzikouri
- Virology Unit, Viral Hepatitis Laboratory, Institut Pasteur du Maroc, Casablanca 20360, Morocco
| | - M'hammed Sarih
- Service de Parasitologie et des Maladies Vectorielles, Institut Pasteur du Maroc, Place Louis Pasteur, 20360 Casablanca, Morocco
| |
Collapse
|
9
|
Miao J, Olson E, Houlihan S, Kattah A, Dillon J, Zoghby Z. Effects of SARS-CoV-2 vaccination on the severity of COVID-19 infection in patients on chronic dialysis. J Nephrol 2023; 36:1321-1328. [PMID: 37017924 PMCID: PMC10074356 DOI: 10.1007/s40620-023-01617-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 02/23/2023] [Indexed: 04/06/2023]
Abstract
BACKGROUND COVID-19 is associated with increased morbidity and mortality in patients with end-stage kidney disease on dialysis. Efficacy of SARS-CoV-2 vaccination to prevent severe COVID-19 disease in end-stage kidney disease patients remains limited. We compared the incidence of COVID-19-related hospitalization and death in dialysis patients based on SARS-CoV-2 vaccine status. METHODS Retrospective study of adults on chronic dialysis within Mayo Clinic Dialysis System in the Midwest (USA) between April 1st, 2020 and October 31st, 2022, who had a laboratory test positive for SARS-CoV-2 by PCR. Incidence of both COVID-19-related hospitalization and death were compared between vaccinated and unvaccinated patients. RESULTS SARS-CoV-2 infection was identified in 309 patients, including 183 vaccinated and 126 unvaccinated. The incidence of death (11.1% vs 3.8%, p = 0.02) and hospitalization (55.6% vs 23.5%, p < 0.001) was significantly higher in unvaccinated compared to vaccinated patients. Age at infection, sex, Charlson comorbidity index, dialysis modality, and hospital stays did not differ between the two groups. The incidence of hospitalization was significantly higher in partially vaccinated (63.6% vs 20.9%, p = 0.004) and unboosted (32% vs 16.4%, p = 0.04) patients compared to fully vaccinated and boosted, respectively. Among the 21 patients who died in the whole cohort, 47.6% (n = 10) died during the pre-vaccine period. The composite risk of death or hospitalization was lower among vaccinated patients after adjusting for age, sex and Charlson comorbidity index (OR 0.24, 95% CI 0.15-0.40). CONCLUSIONS This study supports the use of SARS-CoV-2 vaccination to improve COVID-19 outcomes in patients on chronic dialysis.
Collapse
Affiliation(s)
- Jing Miao
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Elsa Olson
- Home Dialysis Program and Vascular Access Clinic, Medical Nephrology, Department of Nursing, Mayo Clinic, Rochester, MN, USA
| | - Sally Houlihan
- Division of Nephrology and Hypertension, Department of Nursing, Mayo Clinic, Rochester, MN, USA
| | - Andrea Kattah
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - John Dillon
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Ziad Zoghby
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
| |
Collapse
|
10
|
Hiremath S, Blake PG, Yeung A, McGuinty M, Thomas D, Ip J, Brown PA, Pandes M, Burke A, Sohail QZ, To K, Blackwell L, Oliver M, Jain AK, Chagla Z, Cooper R. Early Experience with Modified Dose Nirmatrelvir/Ritonavir in Dialysis Patients with Coronavirus Disease 2019. Clin J Am Soc Nephrol 2023; 18:485-490. [PMID: 36723285 PMCID: PMC10103226 DOI: 10.2215/cjn.0000000000000107] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 01/18/2023] [Indexed: 02/02/2023]
Abstract
BACKGROUND Nirmatrelvir/ritonavir was approved for use in high-risk outpatients with coronavirus disease 2019 (COVID-19). However, patients with severe CKD were excluded from the phase 3 trial, and the drug is not recommended for those with GFR <30 ml/min per 1.73 m 2 . On the basis of available pharmacological data, we developed a modified low-dose regimen of nirmatrelvir/ritonavir 300/100 mg on day 1, followed by 150/100 mg daily from day 2 to 5. In this study, we report our experience with this modified dose regimen in dialysis patients in the Canadian province of Ontario. METHODS We included dialysis patients who developed COVID-19 and were treated with the modified dose nirmatrelvir/ritonavir regimen during a 60-day period between April 1 and May 31, 2022. Details of nirmatrelvir/ritonavir use and outcomes were captured manually, and demographic data were obtained from a provincial database. Data are presented with descriptive statistics. The principal outcomes we describe are 30-day hospitalization, 30-day mortality, and required medication changes with the modified dose regimen. RESULTS A total of 134 dialysis patients with COVID-19 received nirmatrelvir/ritonavir during the period of study. Fifty-six percent were men, and the mean age was 64 years. Most common symptoms were cough and/or sore throat (60%). Medication interactions were common with calcium channel blockers, statins being the most frequent. Most patients (128, 96%) were able to complete the course of nirmatrelvir/ritonavir, and none of the patients who received nirmatrelvir/ritonavir died of COVID-19 in the 30 days of follow-up. CONCLUSIONS A modified dose of nirmatrelvir/ritonavir use was found to be safe and well tolerated, with no serious adverse events being observed in a small sample of maintenance dialysis patients.
Collapse
Affiliation(s)
- Swapnil Hiremath
- Division of Nephrology, Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Peter G. Blake
- Division of Nephrology, Department of Medicine, Western University, London, Ontario, Canada
- Ontario Renal Network, Ontario Health, Toronto, Ontario, Canada
| | - Angie Yeung
- Ontario Renal Network, Ontario Health, Toronto, Ontario, Canada
| | - Michaeline McGuinty
- Division of Infectious Diseases, Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Doneal Thomas
- Ontario Renal Network, Ontario Health, Toronto, Ontario, Canada
| | - Jane Ip
- Ontario Renal Network, Ontario Health, Toronto, Ontario, Canada
| | - Pierre Antoine Brown
- Division of Nephrology, Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Michael Pandes
- Division of Nephrology, Department of Medicine, Mackenzie Health, Richmond Hill, Ontario, Canada
| | - Andrew Burke
- Grand River Hospital, Kitchener-Waterloo, Ontario, Canada
| | - Qazi Zain Sohail
- Division of Nephrology, Department of Medicine, Western University, London, Ontario, Canada
| | - Karen To
- Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Lindsay Blackwell
- Department of Pharmacy, London Health Sciences Centre, London, Ontario, Canada
| | - Matthew Oliver
- Division of Nephrology, Department of Medicine, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Arsh K. Jain
- Division of Nephrology, Department of Medicine, Western University, London, Ontario, Canada
| | - Zain Chagla
- Division of Infectious Disease, Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Rebecca Cooper
- Ontario Renal Network, Ontario Health, Toronto, Ontario, Canada
- Trillium Gift of Life Network, Ontario Health, Toronto, Ontario, Canada
| |
Collapse
|
11
|
Hsu CM, Weiner DE, Manley HJ, Miskulin D, Ladik V, Frament J, Argyropoulos C, Abreo K, Chin A, Gladish R, Salman L, Johnson D, Lacson EK. Serial SARS-CoV-2 antibody titers in vaccinated dialysis patients: prevalence of unrecognized infection and duration of seroresponse. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.03.16.23287322. [PMID: 36993760 PMCID: PMC10055593 DOI: 10.1101/2023.03.16.23287322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Rationale & Objective SARS-CoV-2 infections are likely underdiagnosed, but the degree of underdiagnosis among maintenance dialysis patients is unknown. Durability of the immune response after third vaccine doses in this population also remains uncertain. This study tracked antibody levels to 1) assess the rate of undiagnosed infections and 2) characterize seroresponse durability after third doses. Study Design Retrospective observational study. Setting & Participants SARS-CoV-2 vaccinated patients receiving maintenance dialysis through a national dialysis provider. Immunoglobulin G spike antibodies (anti-spike IgG) titers were assessed monthly following vaccination. Exposures Two and three doses of SARS-CoV-2 vaccine. Outcomes Undiagnosed and diagnosed SARS-CoV-2 infections; anti-spike IgG titers over time. Analytical Approach "Undiagnosed" SARS-CoV-2 infections were identified as an increase in anti-spike IgG titer of ≥ 100 BAU/mL, not associated with receipt of vaccine or "diagnosed" SARS-CoV-2 infection (by PCR or antigen test). In descriptive analyses, anti-spike IgG titers were followed over time. Results Among 2660 patients without prior COVID-19 who received an initial two-dose vaccine series, 371 (76%) SARS-CoV-2 infections were diagnosed and 115 (24%) were undiagnosed. Among 1717 patients without prior COVID-19 who received a third vaccine dose, 155 (80%) SARS-CoV-2 infections were diagnosed and 39 (20%) were undiagnosed. In both cohorts, anti-spike IgG levels declined over time. Of the initial two-dose cohort, 66% had a titer ≥ 500 BAU/mL in the first month, with 23% maintaining a titer ≥ 500 BAU/mL at six months. Of the third dose cohort, 95% had a titer ≥ 500 BAU/mL in the first month after the third dose, with 76% maintaining a titer ≥ 500 BAU/mL at six months. Limitations Assays used had upper limits. Conclusions Among maintenance dialysis patients, 20-24% of SARS-CoV-2 infections were undiagnosed. Given this population's vulnerability to COVID-19, ongoing infection control measures are needed. A three-dose primary mRNA vaccine series optimizes seroresponse rate and durability.
Collapse
|
12
|
Etemadi J, Motavalli R, Mirghaffari SA, Soltani-Zangbar MS, Hajivalili M, Ahmadian Heris J, Niknafs B, Zununi S, Sadeghi M, Rasi Hashemi S, Tayebi Khosroshahi H, Yousefi M. Potent SARS-CoV2-specific T-cell response in asymptomatic hemodialysis patients with hidden COVID-19 infection history. J Clin Lab Anal 2023; 37:e24863. [PMID: 36941528 PMCID: PMC10098065 DOI: 10.1002/jcla.24863] [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: 08/29/2022] [Revised: 01/24/2023] [Accepted: 03/04/2023] [Indexed: 03/23/2023] Open
Abstract
BACKGROUND COVID-19-related immune responses in patients with end-stage renal disease (ESRD) are characterized in detail by the humoral response, but their cellular immunity has not been clarified. Here, we evaluated virus-specific T cells in parallel with serology-related tests. METHODS In this study, 104 ESRD patients at the hemodialysis ward of Imam Reza hospital at Tabriz (Iran) were enrolled. After blood sampling, SARS-CoV2-specific humoral and cellular immune responses were evaluated by SARS-CoV2-specific IgM/IgG ELISA and peptide/MHCI-Tetramers flow cytometry, respectively. RESULTS Our results showed that 14 (13.5%) and 45 (43.3%) patients had specific SARS-CoV2 IgM and IgG in their sera, respectively. Immunophenotyping for SARS-CoV2-specific CD8+ T lymphocytes revealed that 68 (65.4%) patients had these types of cells. Among SARS-CoV2-specific CD8+ T lymphocytes positive subjects, 13 and 43 individuals had positive results for specific SARS-CoV2 IgM and IgG existence, respectively. Also, there was a relationship between specific SARS-CoV2 IgM (p = 0.031) and IgG (p < 0.0001) existence and having SARS-CoV2-specific TCD8+ lymphocytes in the studied population. CONCLUSION Despite not having clinical symptoms, a high rate of SARS-CoV2-specific T-cell response in asymptomatic ESRD patients may reveal a high burden of asymptomatic COVID-19 infection in these patients.
Collapse
Affiliation(s)
- Jalal Etemadi
- Kidney Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Roza Motavalli
- Department of Molecular Medicine, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Mohammad Sadegh Soltani-Zangbar
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Javad Ahmadian Heris
- Department of Allergy and Clinical Immunology, Pediatric Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Bahram Niknafs
- Kidney Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sepideh Zununi
- Kidney Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammadreza Sadeghi
- Department of Molecular Medicine, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | | | - Mehdi Yousefi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
13
|
Balčiuvienė V, Burčiuvienė A, Haarhaus M, Uogintaitė J, Janavičienė A, Santockienė L, Mitrikevičienė J, Aleknienė L, Keinaitė D. Waning Humoral Response 6 Month after Double Vaccination with the mRNA-BNT162b2 Vaccine in Hemodialysis Patients. Acta Med Litu 2023; 30:26-38. [PMID: 37575375 PMCID: PMC10417014 DOI: 10.15388/amed.2023.30.1.3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/10/2022] [Accepted: 12/21/2022] [Indexed: 01/26/2023] Open
Abstract
Introduction Although most hemodialysis patients (HDP) exhibit an initial seroresponse to vaccination against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), studies have shown this response to be lower compared to healthy subjects. This fact raised concerns regarding the durability of the immune response and effective protection against severe Coronavirus disease 2019 (COVID-19) in this vulnerable population. The aim of our study was to evaluate the change in antibody levels over time in HDP population. Materials and Methods We performed a prospective multicenter study, evaluating antibody response among HDP at 2 and at 6 months after complete two-dose vaccination course with the mRNA-BNT162b2 (Pfizer-BioNTech) vaccine. The study was performed in 14 hemodialysis units of a private dialysis provider in Lithuania. The serum samples of 189 HDP were tested for SARS-CoV-2 IgG against the Spike glycoprotein. Results 189 HDP participated in the study. Patients were 64.3±15.7 years of age, 116 (61.4%) were males and 73 (38.6%) were females. Among them, 183 (96.8%) were seropositive for anti-S IgG at 2 months after the second immunization dose. Six months after the second dose only 145 (76.7%) of study participants had positive anti-S IgG titers. The median level of anti-S IgG titers after 2 months was 383.1 BAU/mL (166.2-995.6) and after 6 months this level significantly decreased to 51.4 BAU/mL (22.0-104.0) (p<0.001). Seroresponses at both time points inversely correlated with increasing patient's age. Risk factor for absent response after 2 months included oncologic disease. Systemic autoimmune disease and a history of myocardial infarction increased risk to be seronegative 6 months after the second vaccine dose. Conclusions The majority of hemodialysis patients seroresponded after BNT162b2/Pfizer vaccination, but vaccine-induced humoral immunity wanes over time.
Collapse
Affiliation(s)
- Vilma Balčiuvienė
- Diaverum dialysis unit, Diaverum Lithuania, Josvainių 36, LT-57275 Kėdainiai, Lithuania
| | - Asta Burčiuvienė
- Diaverum dialysis unit, Diaverum Lithuania, Savanorių 68, LT-44147 Kaunas, Lithuania
| | | | | | - Asta Janavičienė
- Diaverum dialysis unit, Diaverum Lithuania, Žeimių 19, LT-55134 Jonava, Lithuania
| | - Lina Santockienė
- Diaverum dialysis unit, Diaverum Lithuania, Beržyno 27, LT-56172 Kaišiadorys, Lithuania
| | | | - Loreta Aleknienė
- Diaverum dialysis unit, Diaverum Lithuania, Savanorių 68, LT-44147 Kaunas, Lithuania
| | - Danutė Keinaitė
- Diaverum Lithuania, Mindaugo 23, LT-3214, Vilnius, Lithuania
| |
Collapse
|
14
|
Juric I, Katalinic L, Furic-Cunko V, Basic-Jukic N. Kidney Transplantation in Patients With the History of SARS-CoV-2 Infection. Transplant Proc 2022; 54:2673-2676. [PMID: 36184341 PMCID: PMC9444488 DOI: 10.1016/j.transproceed.2022.08.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Accepted: 08/26/2022] [Indexed: 01/09/2023]
Abstract
BACKGROUND The aim of this study is to present the experience and results of kidney transplantation in patients with the history of SARS-CoV-2 infection. METHODS We retrospectively analyzed waitlisted patients who had a history of SARS-CoV-2 infection and offered a kidney transplant between March 2020 and December 2021. RESULTS Of the 97 waitlisted potential kidney transplant recipients who were offered a kidney, 13 (13.4%) had a history of SARS-CoV-2 infection. All patients were tested negative for SARS-CoV-2 at the time of the kidney offer. Successful transplantation was performed in 9 patients (5 male; average age was 40.8 years), with the average time between SARS-CoV-2 infection and transplantation of 8 months. Four of 13 patients with a history of SARS-CoV-2 infection were finally not transplanted, with 2 patients not eligible for transplantation due to significant post-COVID findings in routine pretransplant chest CT scans, and 2 patients were not transplanted because of poor donor organ quality. CONCLUSIONS Kidney transplantation after SARS-CoV-2 infection is possible in a setting of full recovery from acute infection, negative PCR test, and no pneumonic infiltrates on chest CT scan. A growing number of waitlisted patients with a history of SARS-CoV-2 infection imposes the need for decision-making tools and guidelines for risk/benefit assessment in these patients.
Collapse
Affiliation(s)
- Ivana Juric
- Department of Nephrology, Arterial Hypertension, Dialysis and Transplantation, University Hospital Centre Zagreb, Zagreb, Croatia..
| | - Lea Katalinic
- Department of Nephrology, Arterial Hypertension, Dialysis and Transplantation, University Hospital Centre Zagreb, Zagreb, Croatia
| | - Vesna Furic-Cunko
- Department of Nephrology, Arterial Hypertension, Dialysis and Transplantation, University Hospital Centre Zagreb, Zagreb, Croatia
| | - Nikolina Basic-Jukic
- Department of Nephrology, Arterial Hypertension, Dialysis and Transplantation, University Hospital Centre Zagreb, Zagreb, Croatia
| |
Collapse
|
15
|
Chen Q, Zhu K, Liu X, Zhuang C, Huang X, Huang Y, Yao X, Quan J, Lin H, Huang S, Su Y, Wu T, Zhang J, Xia N. The Protection of Naturally Acquired Antibodies Against Subsequent SARS-CoV-2 Infection: A Systematic Review and Meta-Analysis. Emerg Microbes Infect 2022; 11:793-803. [PMID: 35195494 PMCID: PMC8920404 DOI: 10.1080/22221751.2022.2046446] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 02/21/2022] [Indexed: 01/18/2023]
Abstract
The specific antibodies induced by SARS-CoV-2 infection may provide protection against a subsequent infection. However, the efficacy and duration of protection provided by naturally acquired immunity against subsequent SARS-CoV-2 infection remain controversial. We systematically searched for the literature describing COVID-19 reinfection published before 07 February 2022. The outcomes were the pooled incidence rate ratio (IRR) for estimating the risk of subsequent infection. The Newcastle-Ottawa Scale (NOS) was used to assess the quality of the included studies. Statistical analyses were conducted using the R programming language 4.0.2. We identified 19 eligible studies including more than 3.5 million individuals without the history of COVID-19 vaccination. The efficacy of naturally acquired antibodies against reinfection was estimated at 84% (pooled IRR = 0.16, 95% CI: 0.14-0.18), with higher efficacy against symptomatic COVID-19 cases (pooled IRR = 0.09, 95% CI = 0.07-0.12) than asymptomatic infection (pooled IRR = 0.28, 95% CI = 0.14-0.54). In the subgroup analyses, the pooled IRRs of COVID-19 infection in health care workers (HCWs) and the general population were 0.22 (95% CI = 0.16-0.31) and 0.14 (95% CI = 0.12-0.17), respectively, with a significant difference (P = 0.02), and those in older (over 60 years) and younger (under 60 years) populations were 0.26 (95% CI = 0.15-0.48) and 0.16 (95% CI = 0.14-0.19), respectively. The risk of subsequent infection in the seropositive population appeared to increase slowly over time. In conclusion, naturally acquired antibodies against SARS-CoV-2 can significantly reduce the risk of subsequent infection, with a protection efficacy of 84%.Registration number: CRD42021286222.
Collapse
Affiliation(s)
- Qi Chen
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Strait Collaborative Innovation Center of Biomedicine and Pharmaceutics, School of Public Health, Xiamen University, Xiamen City, Fujian Province, People’s Republic of China
| | - Kongxin Zhu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Strait Collaborative Innovation Center of Biomedicine and Pharmaceutics, School of Public Health, Xiamen University, Xiamen City, Fujian Province, People’s Republic of China
| | - Xiaohui Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Strait Collaborative Innovation Center of Biomedicine and Pharmaceutics, School of Public Health, Xiamen University, Xiamen City, Fujian Province, People’s Republic of China
| | - Chunlan Zhuang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Strait Collaborative Innovation Center of Biomedicine and Pharmaceutics, School of Public Health, Xiamen University, Xiamen City, Fujian Province, People’s Republic of China
| | - Xingcheng Huang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Strait Collaborative Innovation Center of Biomedicine and Pharmaceutics, School of Public Health, Xiamen University, Xiamen City, Fujian Province, People’s Republic of China
| | - Yue Huang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Strait Collaborative Innovation Center of Biomedicine and Pharmaceutics, School of Public Health, Xiamen University, Xiamen City, Fujian Province, People’s Republic of China
| | - Xingmei Yao
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Strait Collaborative Innovation Center of Biomedicine and Pharmaceutics, School of Public Health, Xiamen University, Xiamen City, Fujian Province, People’s Republic of China
| | - Jiali Quan
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Strait Collaborative Innovation Center of Biomedicine and Pharmaceutics, School of Public Health, Xiamen University, Xiamen City, Fujian Province, People’s Republic of China
| | - Hongyan Lin
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Strait Collaborative Innovation Center of Biomedicine and Pharmaceutics, School of Public Health, Xiamen University, Xiamen City, Fujian Province, People’s Republic of China
| | - Shoujie Huang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Strait Collaborative Innovation Center of Biomedicine and Pharmaceutics, School of Public Health, Xiamen University, Xiamen City, Fujian Province, People’s Republic of China
| | - Yingying Su
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Strait Collaborative Innovation Center of Biomedicine and Pharmaceutics, School of Public Health, Xiamen University, Xiamen City, Fujian Province, People’s Republic of China
| | - Ting Wu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Strait Collaborative Innovation Center of Biomedicine and Pharmaceutics, School of Public Health, Xiamen University, Xiamen City, Fujian Province, People’s Republic of China
| | - Jun Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Strait Collaborative Innovation Center of Biomedicine and Pharmaceutics, School of Public Health, Xiamen University, Xiamen City, Fujian Province, People’s Republic of China
| | - Ningshao Xia
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Strait Collaborative Innovation Center of Biomedicine and Pharmaceutics, School of Public Health, Xiamen University, Xiamen City, Fujian Province, People’s Republic of China
- The Research Unit of Frontier Technology of Structural Vaccinology of Chinese Academy of Medical Sciences, Xiamen City, Fujian Province, People's Republic of China
| |
Collapse
|
16
|
Bernardo AP, Carioni P, Stuard S, Kotanko P, Usvyat LA, Kovarova V, Arkossy O, Bellocchio F, Tupputi A, Gervasoni F, Winter A, Zhang Y, Zhang H, Ponce P, Neri L. Effectiveness of COVID-19 vaccines in a large European hemodialysis cohort. FRONTIERS IN NEPHROLOGY 2022; 2:1037754. [PMID: 37675035 PMCID: PMC10479614 DOI: 10.3389/fneph.2022.1037754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 10/17/2022] [Indexed: 09/08/2023]
Abstract
Background Hemodialysis patients have high-risk of severe SARS-CoV-2 infection but were unrepresented in randomized controlled trials evaluating the safety and efficacy of COVID-19 vaccines. We estimated the real-world effectiveness of COVID-19 vaccines in a large international cohort of hemodialysis patients. Methods In this historical, 1:1 matched cohort study, we included adult hemodialysis patients receiving treatment from December 1, 2020, to May 31, 2021. For each vaccinated patient, an unvaccinated control was selected among patients registered in the same country and attending a dialysis session around the first vaccination date. Matching was based on demographics, clinical characteristics, past COVID-19 infections and a risk score representing the local background risk of infection at vaccination dates. We estimated the effectiveness of mRNA and viral-carrier COVID-19 vaccines in preventing infection and mortality rates from a time-dependent Cox regression stratified by country. Results In the effectiveness analysis concerning mRNA vaccines, we observed 850 SARS-CoV-2 infections and 201 COVID-19 related deaths among the 28110 patients during a mean follow up of 44 ± 40 days. In the effectiveness analysis concerning viral-carrier vaccines, we observed 297 SARS-CoV-2 infections and 64 COVID-19 related deaths among 12888 patients during a mean follow up of 48 ± 32 days. We observed 18.5/100-patient-year and 8.5/100-patient-year fewer infections and 5.4/100-patient-year and 5.2/100-patient-year fewer COVID-19 related deaths among patients vaccinated with mRNA and viral-carrier vaccines respectively, compared to matched unvaccinated controls. Estimated vaccine effectiveness at days 15, 30, 60 and 90 after the first dose of a mRNA vaccine was: for infection, 41.3%, 54.5%, 72.6% and 83.5% and, for death, 33.1%, 55.4%, 80.1% and 91.2%. Estimated vaccine effectiveness after the first dose of a viral-carrier vaccine was: for infection, 38.3% without increasing over time and, for death, 56.6%, 75.3%, 92.0% and 97.4%. Conclusion In this large, real-world cohort of hemodialyzed patients, mRNA and viral-carrier COVID-19 vaccines were associated with reduced COVID-19 related mortality. Additionally, we observed a strong reduction of SARS-CoV-2 infection in hemodialysis patients receiving mRNA vaccines.
Collapse
Affiliation(s)
- Ana Paula Bernardo
- Fresenius Medical Care Portugal / Nephrocare Portugal, Vila Nova de Gaia, Portugal
- Unit for Multidisciplinary Research in Biomedicine (UMIB), Institute of Biomedical Sciences Abel Salazar (ICBAS), Porto University, Oporto, Portugal
| | - Paola Carioni
- Fresenius Medical Care Italia SpA, Palazzo Pignano, Italy
| | - Stefano Stuard
- Fresenius Medical Care Deutschland GmbH, Bad Homburg, Germany
| | - Peter Kotanko
- Renal Research Institute, New York, NY, United States
- Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | | | | | - Otto Arkossy
- Fresenius Medical Care Deutschland GmbH, Bad Homburg, Germany
| | | | | | | | - Anke Winter
- Fresenius Medical Care Deutschland GmbH, Bad Homburg, Germany
| | - Yan Zhang
- Fresenius Medical Care Deutschland GmbH, Bad Homburg, Germany
| | - Hanjie Zhang
- Renal Research Institute, New York, NY, United States
| | - Pedro Ponce
- Fresenius Medical Care Portugal / Nephrocare Portugal, Lisboa, Portugal
| | - Luca Neri
- Fresenius Medical Care Italia SpA, Palazzo Pignano, Italy
| |
Collapse
|
17
|
Falahi S, Sayyadi H, Kenarkoohi A. Immunogenicity of COVID-19 mRNA vaccines in hemodialysis patients: Systematic review and meta-analysis. Health Sci Rep 2022; 5:e874. [PMID: 36210877 PMCID: PMC9528953 DOI: 10.1002/hsr2.874] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 09/21/2022] [Accepted: 09/22/2022] [Indexed: 11/05/2022] Open
Abstract
Background and Aims Vaccine response is a concern in hemodialysis patients. Given that hemodialysis patients were not included in clinical trials, we aimed to synthesize the available evidence on the immunogenicity of coronavirus disease 2019 (COVID-19) mRNA vaccines in hemodialysis patients. Methods We searched Scopus, PubMed, Sciencedirect, and finally google scholar databases for studies on COVID-19 mRNA-vaccines immunogenicity in hemodialysis patients up to December 1, 2021. Eligible articles measured antibodies against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike or Receptor-Binding Domain Antibody (S/RBD) postimmunization with COVID-19 mRNA vaccines. The immunogenicity of the vaccine was evaluated using seroconversion rates measured between 21 and 30 days after the first immunization and between 14 and 36 days post the second dose. We included studies including participants without a history of COVID-19 before vaccination. Healthy controls or health-care workers served as the control groups. After selecting eligible articles, the data were finally extracted from included articles. We used a random effects model to estimate the pooled seroconversion rate after COVID-19 mRNA vaccine administration. We assessed the heterogeneity between studies with the I 2 statistical index. Result We selected 39 eligible citations comprising 806 cases and 336 controls for the first dose and 6314 cases and 927 controls for the second dose for statistical analysis. After the first dose of mRNA vaccines, the seroconversion rate was 36% (95% confidence interval [CI]: 0.24-0.47) and 68% (95% CI: 0.45-0.91) in hemodialysis patients and the control group, respectively. While seroconversion rate after the second dose of mRNA vaccines was 86% (95% CI: 0.81-0.91) and 100% (95% CI: 1.00-1.00) in hemodialysis patients and the control group, respectively. Conclusion Although the immune response of hemodialysis patients to the second dose of the SARS-CoV-2 mRNA vaccine is very promising, the seroconversion rate of dialysis patients is lower than healthy controls. Periodically assessment of antibody levels of hemodialysis patients at short intervals is recommended.
Collapse
Affiliation(s)
- Shahab Falahi
- Zoonotic Diseases Research CenterIlam University of Medical SciencesIlamIran
| | - Hojjat Sayyadi
- Department of Biostatistics, Faculty of HealthIlam University of Medical SciencesIlamIran
| | - Azra Kenarkoohi
- Zoonotic Diseases Research CenterIlam University of Medical SciencesIlamIran
- Department of Microbiology, Faculty of MedicineIlam University of Medical SciencesIlamIran
| |
Collapse
|
18
|
Ponticelli C, Campise M. COVID-19 Vaccination in Kidney Transplant Candidates and Recipients. Vaccines (Basel) 2022; 10:vaccines10111808. [PMID: 36366317 PMCID: PMC9692413 DOI: 10.3390/vaccines10111808] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 10/14/2022] [Accepted: 10/20/2022] [Indexed: 12/03/2022] Open
Abstract
Kidney transplant candidates and kidney transplant recipients (KTRs) are at particular risk of severe complications of COVID-19 disease. In Western countries, mortality in affected hospitalized KTRs ranges between 19% and 50%. COVID-19 vaccination remains the most important measure to prevent the severity of infection in candidates and recipients of kidney transplant. However, the uraemic condition may affect the vaccine-induced immunity in patients with advanced chronic kidney disease (CKD) and in KTRs. Retention of uraemic toxins, dysbiosis, dysmetabolism, and dialysis can diminish the normal response to vaccination, leading to dysfunction of inflammatory and immune cells. In KTRs the efficacy of vaccines may be reduced by the immunosuppressive medications, and more than half of kidney transplant recipients are unable to build an immune response even after four administrations of anti-COVID-19 vaccines. The lack of antibody response leaves these patients at high risk for SARS-CoV-2 infection and severe COVID-19 disease. The aim of the present review is to focus on the main reasons for the impaired immunological response among candidates and kidney transplant recipients and to highlight some of the present options available to solve the problem.
Collapse
Affiliation(s)
| | - Mariarosaria Campise
- Department of Nephrology, Dialysis and Kidney Transplantation, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
- Correspondence:
| |
Collapse
|
19
|
Flacco ME, Acuti Martellucci C, Baccolini V, De Vito C, Renzi E, Villari P, Manzoli L. Risk of reinfection and disease after SARS-CoV-2 primary infection: Meta-analysis. Eur J Clin Invest 2022; 52:e13845. [PMID: 35904405 PMCID: PMC9353414 DOI: 10.1111/eci.13845] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 07/09/2022] [Accepted: 07/27/2022] [Indexed: 11/29/2022]
Abstract
INTRODUCTION A precise estimate of the frequency and severity of SARS-CoV-2 reinfections would be critical to optimize restriction and vaccination policies for the hundreds of millions previously infected subjects. We performed a meta-analysis to evaluate the risk of reinfection and COVID-19 following primary infection. METHODS We searched MedLine, Scopus and preprint repositories for cohort studies evaluating the onset of new infections among baseline SARS-CoV-2-positive subjects. Random-effect meta-analyses of proportions were stratified by gender, exposure risk, vaccination status, viral strain, time between episodes, and reinfection definition. RESULTS Ninety-one studies, enrolling 15,034,624 subjects, were included. Overall, 158,478 reinfections were recorded, corresponding to a pooled rate of 0.97% (95% CI: 0.71%-1.27%), with no substantial differences by definition criteria, exposure risk or gender. Reinfection rates were still 0.66% after ≥12 months from first infection, and the risk was substantially lower among vaccinated subjects (0.32% vs. 0.74% for unvaccinated individuals). During the first 3 months of Omicron wave, the reinfection rates reached 3.31%. Overall rates of severe/lethal COVID-19 were very low (2-7 per 10,000 subjects according to definition criteria) and were not affected by strain predominance. CONCLUSIONS A strong natural immunity follows the primary infection and may last for more than one year, suggesting that the risk and health care needs of recovered subjects might be limited. Although the reinfection rates considerably increased during the Omicron wave, the risk of a secondary severe or lethal disease remained very low. The risk-benefit profile of multiple vaccine doses for this subset of population needs to be carefully evaluated.
Collapse
Affiliation(s)
- Maria Elena Flacco
- Department of Environmental and Preventive Sciences, University of Ferrara, Ferrara, Italy
| | | | - Valentina Baccolini
- Department of Public Health and Infectious Diseases, Sapienza University, Rome, Italy
| | - Corrado De Vito
- Department of Public Health and Infectious Diseases, Sapienza University, Rome, Italy
| | - Erika Renzi
- Department of Public Health and Infectious Diseases, Sapienza University, Rome, Italy
| | - Paolo Villari
- Department of Public Health and Infectious Diseases, Sapienza University, Rome, Italy
| | - Lamberto Manzoli
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| |
Collapse
|
20
|
Martin P, Gleeson S, Clarke CL, Thomson T, Edwards H, Spensley K, Mortimer P, McIntyre S, Cox A, Pickard G, Lightstone L, Thomas D, McAdoo SP, Kelleher P, Prendecki M, Willicombe M. Comparison of immunogenicity and clinical effectiveness between BNT162b2 and ChAdOx1 SARS-CoV-2 vaccines in people with end-stage kidney disease receiving haemodialysis: A prospective, observational cohort study. THE LANCET REGIONAL HEALTH. EUROPE 2022; 21:100478. [PMID: 36105885 PMCID: PMC9463038 DOI: 10.1016/j.lanepe.2022.100478] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Background People with end-stage kidney disease, including people on haemodialysis, are susceptible to greater COVID-19 related morbidity and mortality. This study compares the immunogenicity and clinical effectiveness of BNT162B2 versus ChAdOx1 in haemodialysis patients. Methods In this observational cohort study, 1021 patients were followed-up from time of vaccination until December 2021. All patients underwent weekly RT-PCR screening. Patients were assessed for nucleocapsid(anti-NP) and spike(anti-S) antibodies at timepoints after second(V2) and third(V3) vaccinations. 191 patients were investigated for T-cell responses. Vaccine effectiveness (VE) for prevention of infection, hospitalisation and mortality was evaluated using the formula VE=(1-adjustedHR)x100. Findings 45.7% (467/1021) had evidence of prior infection. There was no difference in the proportion of infection-naïve patients who seroconverted by vaccine type, but median anti-S antibody titres were higher post-BNT162b2 compared with ChAdOx1; 462(152-1171) and 78(20-213) BAU/ml respectively, p<0.001. Concomitant immunosuppressant use was a risk factor for non-response, OR 0.12[95% CI 0.05-0.25] p<0.001. Post-V3 (all BNT162b2), median anti-S antibody titres remained higher in those receiving BNT162b2 versus ChAdOx1 as primary doses; 2756(187-1246) and 1250(439-2635) BAU/ml respectively, p=0.003.Anti-S antibodies waned over time. Hierarchical levels of anti-S post-V2 predicted risk of infection; patients with no/low anti-S being at highest risk. VE for preventing infection, hospitalisation and death was 53% (95% CI 6-75), 77% (95% CI 30-92) and 93% (95% CI 59-99) respectively, with no difference seen by vaccine type. Interpretation Serum anti-S concentrations predict risk of breakthrough infection. Anti-S responses vary dependent upon clinical features, infection history and vaccine type. Monitoring of serological responses may enable individualised approaches to vaccine boosters in at risk populations. Funding National Institute for Health Research (NIHR) Biomedical Research Centre based at Imperial College Healthcare NHS Trust and Imperial College London.
Collapse
Affiliation(s)
- Paul Martin
- Imperial College Renal and Transplant Centre, Imperial College Healthcare NHS Trust, Hammersmith Hospital, London W12 0HS, United Kingdom
| | - Sarah Gleeson
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, Hammersmith campus, Du Cane Road, London W12 0NN, United Kingdom
| | - Candice L. Clarke
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, Hammersmith campus, Du Cane Road, London W12 0NN, United Kingdom
| | - Tina Thomson
- Imperial College Renal and Transplant Centre, Imperial College Healthcare NHS Trust, Hammersmith Hospital, London W12 0HS, United Kingdom
| | - Helena Edwards
- Imperial College Renal and Transplant Centre, Imperial College Healthcare NHS Trust, Hammersmith Hospital, London W12 0HS, United Kingdom
| | - Katrina Spensley
- Imperial College Renal and Transplant Centre, Imperial College Healthcare NHS Trust, Hammersmith Hospital, London W12 0HS, United Kingdom
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, Hammersmith campus, Du Cane Road, London W12 0NN, United Kingdom
| | - Paige Mortimer
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, Hammersmith campus, Du Cane Road, London W12 0NN, United Kingdom
| | - Stacey McIntyre
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, Hammersmith campus, Du Cane Road, London W12 0NN, United Kingdom
| | - Alison Cox
- Department of Infection and Immunity Sciences, Northwest London Pathology NHS trust, Charing Cross Hospital, Fulham Palace Road, London W6 6RF, United Kingdom
| | - Graham Pickard
- Department of Infection and Immunity Sciences, Northwest London Pathology NHS trust, Charing Cross Hospital, Fulham Palace Road, London W6 6RF, United Kingdom
| | - Liz Lightstone
- Imperial College Renal and Transplant Centre, Imperial College Healthcare NHS Trust, Hammersmith Hospital, London W12 0HS, United Kingdom
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, Hammersmith campus, Du Cane Road, London W12 0NN, United Kingdom
| | - David Thomas
- Imperial College Renal and Transplant Centre, Imperial College Healthcare NHS Trust, Hammersmith Hospital, London W12 0HS, United Kingdom
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, Hammersmith campus, Du Cane Road, London W12 0NN, United Kingdom
| | - Stephen P. McAdoo
- Imperial College Renal and Transplant Centre, Imperial College Healthcare NHS Trust, Hammersmith Hospital, London W12 0HS, United Kingdom
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, Hammersmith campus, Du Cane Road, London W12 0NN, United Kingdom
| | - Peter Kelleher
- Department of Infection and Immunity Sciences, Northwest London Pathology NHS trust, Charing Cross Hospital, Fulham Palace Road, London W6 6RF, United Kingdom
- Department of Infectious Diseases, Imperial College London, Chelsea and Westminster Campus, Fulham Road, London SW10 9NH, United Kingdom
| | - Maria Prendecki
- Imperial College Renal and Transplant Centre, Imperial College Healthcare NHS Trust, Hammersmith Hospital, London W12 0HS, United Kingdom
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, Hammersmith campus, Du Cane Road, London W12 0NN, United Kingdom
| | - Michelle Willicombe
- Imperial College Renal and Transplant Centre, Imperial College Healthcare NHS Trust, Hammersmith Hospital, London W12 0HS, United Kingdom
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, Hammersmith campus, Du Cane Road, London W12 0NN, United Kingdom
| |
Collapse
|
21
|
Torres R, Toro L, Sanhueza ME, Lorca E, Ortiz M, Pefaur J, Clavero R, Machuca E, Gonzalez F, Herrera P, Mocarquer A, Frias A, Roessler E, Muñoz C, Nuñez M, Aravena C, Quintana E, Lemus J, Lillo M, Reynolds E, Morales A, Pais E, Fiabane A, Parra-Lucares A, Garrido C, Mendez-Valdes G, Villa E, Mansilla R, Sotomayor G, Gonzalez M, Miranda C, Briones E, Gomez E, Mezzano S, Bernales W, Rocca X, Espinoza O, Zuñiga E, Aragon H, Badilla M, Valenzuela M, Escobar L, Zamora D, Flores I, Tapia B, Borquez T, Herrera P. Clinical Efficacy of SARS-CoV-2 Vaccination in Hemodialysis Patients. Kidney Int Rep 2022; 7:2176-2185. [PMID: 35874643 PMCID: PMC9287586 DOI: 10.1016/j.ekir.2022.07.007] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 07/01/2022] [Accepted: 07/05/2022] [Indexed: 12/31/2022] Open
Abstract
INTRODUCTION The COVID-19 pandemic is a global public health problem. Patients with end-stage renal disease on hemodialysis are at a higher risk of infection and mortality than the general population. Worldwide, a vaccination campaign has been developed that has been shown to reduce severe infections and deaths in the general population. However, there are currently limited data on the clinical efficacy of vaccinations in the hemodialysis population. METHODS A national multicenter observational cohort was performed in Chile to evaluate the clinical efficacy of anti-SARS-CoV-2 vaccination in end-stage renal disease patients on chronic hemodialysis from February 2021 to August 2021. In addition, the BNT162b2 (Pfizer-BioNTech) and CoronaVac (Sinovac) vaccines were evaluated. The efficacy of vaccination in preventing SARS-CoV-2 infection, hospitalizations, and deaths associated with COVID-19 was determined. RESULTS A total of 12,301 patients were evaluated; 10,615 (86.3%) received a complete vaccination (2 doses), 490 (4.0%) received incomplete vaccination, and 1196 (9.7%) were not vaccinated. During follow-up, 1362 (11.0%) patients developed COVID-19, and 150 died (case fatality rate: 11.0%). The efficacy of the complete vaccination in preventing infection was 18.1% (95% confidence interval [CI]:11.8-23.8%), and prevention of death was 66.0% (95% CI:60.6-70.7%). When comparing both vaccines, BNT162b2 and CoronaVac were effective in reducing infection and deaths associated with COVID-19. Nevertheless, the BNT162b2 vaccine had higher efficacy in preventing infection (42.6% vs. 15.0%) and deaths (90.4% vs. 64.8%) compared to CoronaVac. CONCLUSION The results of our study suggest that vaccination against SARS-CoV-2 in patients on chronic hemodialysis was effective in preventing infection and death associated with COVID-19.
Collapse
Affiliation(s)
- Rubén Torres
- Fuerza de Trabajo anti COVID-19, Sociedad Chilena de Nefrología, Chile
- Division of Nephrology, Department of Medicine, Hospital Clínico Universidad de Chile, Santiago, Chile
| | - Luis Toro
- Fuerza de Trabajo anti COVID-19, Sociedad Chilena de Nefrología, Chile
- Division of Nephrology, Department of Medicine, Hospital Clínico Universidad de Chile, Santiago, Chile
- Centro de Investigación Clínica Avanzada, Hospital Clínico Universidad de Chile, Santiago, Chile
| | - María Eugenia Sanhueza
- Fuerza de Trabajo anti COVID-19, Sociedad Chilena de Nefrología, Chile
- Division of Nephrology, Department of Medicine, Hospital Clínico Universidad de Chile, Santiago, Chile
| | - Eduardo Lorca
- Fuerza de Trabajo anti COVID-19, Sociedad Chilena de Nefrología, Chile
- Division of Nephrology, Hospital del Salvador, Santiago, Chile
| | - Mireya Ortiz
- Fuerza de Trabajo anti COVID-19, Sociedad Chilena de Nefrología, Chile
- Department of Nephrology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Jacqueline Pefaur
- Fuerza de Trabajo anti COVID-19, Sociedad Chilena de Nefrología, Chile
- Division of Nephrology, Hospital Barros Luco Trudeau, Santiago, Chile
| | - Rene Clavero
- Fuerza de Trabajo anti COVID-19, Sociedad Chilena de Nefrología, Chile
- Division of Nephrology, Hospital Gustavo Fricke, Valparaiso, Chile
| | - Eduardo Machuca
- Fuerza de Trabajo anti COVID-19, Sociedad Chilena de Nefrología, Chile
- Fresenius Medical Care Chile, Chile
| | | | - Patricia Herrera
- Fuerza de Trabajo anti COVID-19, Sociedad Chilena de Nefrología, Chile
- Division of Nephrology, Hospital del Salvador, Santiago, Chile
| | - Alfredo Mocarquer
- Fuerza de Trabajo anti COVID-19, Sociedad Chilena de Nefrología, Chile
- Dialisis Gran Avenida, Santiago, Chile
| | - Alondra Frias
- Fuerza de Trabajo anti COVID-19, Sociedad Chilena de Nefrología, Chile
- Division of Nephrology, Hospital Regional de Talca, Talca, Chile
| | - Eric Roessler
- Fuerza de Trabajo anti COVID-19, Sociedad Chilena de Nefrología, Chile
- Department of Nephrology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Carolina Muñoz
- Fuerza de Trabajo anti COVID-19, Sociedad Chilena de Nefrología, Chile
- Division of Nephrology, Hospital Puerto Montt, Puerto Montt, Chile
| | - Miguel Nuñez
- Fuerza de Trabajo anti COVID-19, Sociedad Chilena de Nefrología, Chile
- Division of Nephrology, Hospital Puerto Montt, Puerto Montt, Chile
| | - Cesar Aravena
- Fuerza de Trabajo anti COVID-19, Sociedad Chilena de Nefrología, Chile
- Dialisis Municipal La Granja, Santiago, Chile
| | - Enrique Quintana
- Fuerza de Trabajo anti COVID-19, Sociedad Chilena de Nefrología, Chile
- Division of Nephrology, Hospital Padre Hurtado, Santiago, Chile
| | - Juan Lemus
- Fuerza de Trabajo anti COVID-19, Sociedad Chilena de Nefrología, Chile
- Dialysis Center, Santiago, Chile
| | - Mario Lillo
- Fuerza de Trabajo anti COVID-19, Sociedad Chilena de Nefrología, Chile
- Centro Médico de Diálisis, Santiago, Chile
| | - Enrique Reynolds
- Fuerza de Trabajo anti COVID-19, Sociedad Chilena de Nefrología, Chile
- Division of Nephrology, Hospital del Salvador, Santiago, Chile
| | - Alvaro Morales
- Fuerza de Trabajo anti COVID-19, Sociedad Chilena de Nefrología, Chile
- Division of Nephrology, Hospital del Salvador, Santiago, Chile
| | - Edgard Pais
- Fuerza de Trabajo anti COVID-19, Sociedad Chilena de Nefrología, Chile
- Division of Nephrology, Complejo Asistencial Sótero del Río, Santiago, Chile
| | - Andrea Fiabane
- Fuerza de Trabajo anti COVID-19, Sociedad Chilena de Nefrología, Chile
- Division of Nephrology, Hospital Barros Luco Trudeau, Santiago, Chile
| | - Alfredo Parra-Lucares
- Division of Critical Care Medicine, Department of Medicine, Hospital Clínico Universidad de Chile, Santiago, Chile
| | - Cristian Garrido
- Department of Radiology, Hospital Clinico Universidad de Chile, Santiago, Chile
| | | | - Eduardo Villa
- Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Rodrigo Mansilla
- Fuerza de Trabajo anti COVID-19, Sociedad Chilena de Nefrología, Chile
- Division of Nephrology, Hospital Clínico de Magallanes, Punta Arenas, Chile
| | - Germana Sotomayor
- Fuerza de Trabajo anti COVID-19, Sociedad Chilena de Nefrología, Chile
| | - Marcela Gonzalez
- Fuerza de Trabajo anti COVID-19, Sociedad Chilena de Nefrología, Chile
- Division of Nephrology, Hospital Militar de Santiago, Santiago, Chile
| | - Cecilia Miranda
- Fuerza de Trabajo anti COVID-19, Sociedad Chilena de Nefrología, Chile
- CID Servicio Integral de Salud, Santiago, Chile
| | - Eduardo Briones
- Fuerza de Trabajo anti COVID-19, Sociedad Chilena de Nefrología, Chile
- Division of Nephrology, Clinica Indisa, Santiago, Chile
| | - Esteban Gomez
- Fuerza de Trabajo anti COVID-19, Sociedad Chilena de Nefrología, Chile
- Division of Nephrology, Hospital Clinico San Borja Arriaran, Santiago, Chile
| | - Sergio Mezzano
- Fuerza de Trabajo anti COVID-19, Sociedad Chilena de Nefrología, Chile
- Faculty of Medicine, Universidad Austral de Chile, Valdivia, Santiago, Chile
| | - Waldo Bernales
- Fuerza de Trabajo anti COVID-19, Sociedad Chilena de Nefrología, Chile
- Division of Nephrology, Hospital Regional de Coyhaique, Coyhaique, Chile
| | - Ximena Rocca
- Fuerza de Trabajo anti COVID-19, Sociedad Chilena de Nefrología, Chile
- Division of Nephrology, Hospital del Salvador, Santiago, Chile
| | - Oscar Espinoza
- Fuerza de Trabajo anti COVID-19, Sociedad Chilena de Nefrología, Chile
- Division of Nephrology, Hospital del Salvador, Santiago, Chile
| | - Eric Zuñiga
- Fuerza de Trabajo anti COVID-19, Sociedad Chilena de Nefrología, Chile
- Division of Nephrology, Hospital Clinico Universidad de Antofagasta, Antofagasta, Chile
| | - Henry Aragon
- Fuerza de Trabajo anti COVID-19, Sociedad Chilena de Nefrología, Chile
- Division of Nephrology, Hospital San José, Santiago, Chile
| | - Marta Badilla
- Fuerza de Trabajo anti COVID-19, Sociedad Chilena de Nefrología, Chile
- Division of Nephrology, Hospital de Puerto Natales, Puerto Natales, Chile
| | - Marcela Valenzuela
- Fuerza de Trabajo anti COVID-19, Sociedad Chilena de Nefrología, Chile
- Division of Nephrology, Complejo Asistencial Sótero del Río, Santiago, Chile
| | - Luis Escobar
- Fuerza de Trabajo anti COVID-19, Sociedad Chilena de Nefrología, Chile
- Division of Nephrology, Hospital San José, Santiago, Chile
| | - Daniela Zamora
- Fuerza de Trabajo anti COVID-19, Sociedad Chilena de Nefrología, Chile
- Division of Nephrology, Hospital Barros Luco Trudeau, Santiago, Chile
| | - Ivan Flores
- Fuerza de Trabajo anti COVID-19, Sociedad Chilena de Nefrología, Chile
- Division of Nephrology, Hospital San Juan de Dios de Curicó, Curicó, Chile
| | - Beatriz Tapia
- Fuerza de Trabajo anti COVID-19, Sociedad Chilena de Nefrología, Chile
- Division of Nephrology, Hospital Gustavo Fricke, Valparaiso, Chile
| | - Tamara Borquez
- Fuerza de Trabajo anti COVID-19, Sociedad Chilena de Nefrología, Chile
- Division of Nephrology, Hospital Clínico Regional Dr. Guillermo Grant Benavente, Concepción, Chile
| | - Patricio Herrera
- Fuerza de Trabajo anti COVID-19, Sociedad Chilena de Nefrología, Chile
- Servicio de Salud Chiloé, X Región, Chile
| |
Collapse
|
22
|
Moore LR, Al-Jaddou N, Wodeyar H, Sharma A, Schulz M, Rao A, Abraham K. SARS-CoV-2 in dialysis patients and the impact of vaccination. BMC Nephrol 2022; 23:317. [PMID: 36131241 PMCID: PMC9490718 DOI: 10.1186/s12882-022-02940-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 06/30/2022] [Indexed: 11/30/2022] Open
Abstract
Background In centre haemodialysis (ICHD) patients have been identified as high risk of contracting Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) infection due to frequent healthcare contact and poor innate and adaptive immunity. Our ICHD patients were offered immunisation from January 2021. We aimed to assess outcomes following SARS-CoV-2 infection and report on the effect of vaccination in our ICHD patients. Methods Demographics, SARS-CoV-2 status, hospitalisation, mortality and vaccination status were analysed. From 11th March 2020 to 31st March 2021, 662 ICHD patients were included in the study and these patients were then followed up until 31st August 2021. Results SARS-CoV-2 infection occurred in 28.4% with 51.1% of them requiring hospitalisation in contrast to community infection rates of 13.9% and hospitalisation of 9.0%. 28-day mortality was 19.2% in comparison to 1.9% of the community. Mortality increased to 34.0% over the study period. Mortality over the study period was 1.8 times in infected patients (HR 1.81 (1.32–2.49) P < 0.001) despite adjustment for age, gender and ethnicity. 91.3% of ICHD patients have now received both doses of SARS-CoV-2 vaccinations. Conclusions ICHD patients are at increased risk of acquiring SARS-CoV-2, with increased rates of hospitalisation and mortality. The increased mortality extends well beyond the 28 days post-infection and persists in those who have recovered. Peaks and troughs in infection rates mirrored community trends. Preliminary data indicates that the SARS-CoV-2 vaccination provides protection to ICHD patients, with ICHD case rates now comparable to that of the local population.
Collapse
Affiliation(s)
- Louise Rachel Moore
- Liverpool University Hospitals NHS FT, Liverpool, UK. .,Nephrology Department, Liverpool University Hospitals NHS Foundation Trust, Liverpool, UK.
| | | | | | | | | | - Anirudh Rao
- Liverpool University Hospitals NHS FT, Liverpool, UK
| | | |
Collapse
|
23
|
Glowinska I, Labij-Reduta B, Juzwiuk J, Lukaszewicz M, Pietruczuk A, Poplawska A, Daniluk-Jamro A, Kakareko K, Rydzewska-Rosolowska A, Naumnik B, Koc-Zorawska E, Zorawski M, Hryszko T. Factors Influencing Longevity of Humoral Response to SARS-CoV-2 Vaccination in Patients with End Stage Kidney Disease Receiving Renal Replacement Therapy. J Clin Med 2022; 11:jcm11174984. [PMID: 36078913 PMCID: PMC9457318 DOI: 10.3390/jcm11174984] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 08/02/2022] [Accepted: 08/23/2022] [Indexed: 01/14/2023] Open
Abstract
COVID-19 has severely affected the population of patients with end stage renal disease. Current data have proved a two-dose vaccination schedule against SARS-CoV-2 to be effective among dialyzed patients. There are limited data on the longevity and modulating factors of humoral response after vaccination. We performed a prospective longitudinal cohort study to determine longevity of the humoral response after SARS-CoV-2 vaccine. The study included 191 adult patients on hemodialysis and peritoneal dialysis. All participants had been vaccinated with three doses, either with BNT162b2 (Pfizer-BioNTech) (n = 109) or mRNA-1273 (Moderna) (n = 82). Anti-spike protein receptor-binding domain antibodies (anti-S IgG) were assessed using SARS-CoV-2 (RBD) IgG ELISA EIA-6150 IVD assay at baseline, on the 21st day and 43rd day, before a booster dose and two weeks thereafter. We found that before vaccination, 37.7% of the cohort had anti-S IgG titres concordant with seroconversion. After two-dose vaccination, seroconversion occurred in 97% of patients. The booster dose evoked a ~12-fold increase in antibody level. Obesity increased more than two-fold the odds for a decrease in anti-S IgG. Previous COVID-19 infection enhanced longevity of the humoral response following vaccination. In patients with previous COVID-19 infection, the BNT162b2 vaccine was associated with a higher odds of anti-S IgG waning compared to the mRNA-1273 vaccine. In conclusion, we report that obesity predisposes patients to protective antibody waning, hybrid immunity enhances odds for higher anti-S IgG concentrations and vaccine efficacy may be influenced by previous SARS-CoV-2 infection. The results might provide a rationale for vaccination protocol design.
Collapse
Affiliation(s)
- Irena Glowinska
- 2nd Department of Nephrology and Hypertension with Dialysis Unit, Medical University of Bialystok, ul. M. Sklodowskiej-Curie 24A, 15-276 Bialystok, Poland
- Correspondence:
| | - Barbara Labij-Reduta
- 1st Department of Nephrology and Transplantation with Dialysis Unit, Medical University of Bialystok, ul. Zurawia 14, 15-540 Bialystok, Poland
| | - Jerzy Juzwiuk
- Dialysis Unit, SPZOZ in Hajnowka, ul. A. Dowgirda 9, 17-200 Hajnowka, Poland
| | - Magdalena Lukaszewicz
- Nephrology and Dialysis Unit, Regional Specialist Hospital in Suwalki, ul. Szpitalna 60, 16-400 Suwalki, Poland
| | - Adam Pietruczuk
- Dialysis Unit, SPZOZ in Sokolka, ul. W. Sikorskiego 40, 16-100 Sokolka, Poland
| | - Agata Poplawska
- Nephrology and Dialysis Unit, Regional Specialist Hospital in Lomza, Al. J. Piłsudskiego 11, 18-404 Lomza, Poland
| | | | - Katarzyna Kakareko
- 2nd Department of Nephrology and Hypertension with Dialysis Unit, Medical University of Bialystok, ul. M. Sklodowskiej-Curie 24A, 15-276 Bialystok, Poland
| | - Alicja Rydzewska-Rosolowska
- 2nd Department of Nephrology and Hypertension with Dialysis Unit, Medical University of Bialystok, ul. M. Sklodowskiej-Curie 24A, 15-276 Bialystok, Poland
| | - Beata Naumnik
- 1st Department of Nephrology and Transplantation with Dialysis Unit, Medical University of Bialystok, ul. Zurawia 14, 15-540 Bialystok, Poland
| | - Ewa Koc-Zorawska
- 2nd Department of Nephrology and Hypertension with Dialysis Unit, Medical University of Bialystok, ul. M. Sklodowskiej-Curie 24A, 15-276 Bialystok, Poland
| | - Marcin Zorawski
- Department of Clinical Medicine, Medical University of Bialystok, 15-254 Bialystok, Poland
| | - Tomasz Hryszko
- 2nd Department of Nephrology and Hypertension with Dialysis Unit, Medical University of Bialystok, ul. M. Sklodowskiej-Curie 24A, 15-276 Bialystok, Poland
| |
Collapse
|
24
|
Cantarelli C, Angeletti A, Perin L, Russo LS, Sabiu G, Podestà MA, Cravedi P. Immune responses to SARS-CoV-2 in dialysis and kidney transplantation. Clin Kidney J 2022; 15:1816-1828. [PMID: 36147709 PMCID: PMC9384565 DOI: 10.1093/ckj/sfac174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Indexed: 11/15/2022] Open
Abstract
Despite progressive improvements in the management of patients with coronavirus disease 2019 (COVID-19), individuals with end-stage kidney disease (ESKD) are still at high risk of infection-related complications. Although the risk of infection in these patients is comparable to that of the general population, their lower rate of response to vaccination is a matter of concern. When prevention strategies fail, infection is often severe. Comorbidities affecting patients on maintenance dialysis and kidney transplant recipients clearly account for the increased risk of severe COVID-19, while the role of uremia and chronic immunosuppression is less clear. Immune monitoring studies have identified differences in the innate and adaptive immune response against the virus that could contribute to the increased disease severity. In particular, individuals on dialysis show signs of T cell exhaustion that may impair antiviral response. Similar to kidney transplant recipients, antibody production in these patients occurs, but with delayed kinetics compared with the general population, leaving them more exposed to viral expansion during the early phases of infection. Overall, unique features of the immune response during COVID-19 in individuals with ESKD may occur with severe comorbidities affecting these individuals in explaining their poor outcomes.
Collapse
Affiliation(s)
- Chiara Cantarelli
- UO Nefrologia, Azienda Ospedaliero-Universitaria di Parma , Parma , Italy
| | - Andrea Angeletti
- Division of Nephrology, Dialysis, Transplantation, IRCCS Istituto Giannina Gaslini
| | - Laura Perin
- GOFARR Laboratory for Organ Regenerative Research and Cell Therapeutics in Urology, Saban Research Institute, Division of Urology, Children's Hospital Los Angeles , Los Angeles, CA , USA ; , Los Angeles, CA
- Department of Urology, Keck School of Medicine, University of Southern California , Los Angeles, CA , USA ; , Los Angeles, CA
| | - Luis Sanchez Russo
- Department of Medicine, Icahn School of Medicine at Mount Sinai , New York, NY
| | - Gianmarco Sabiu
- Nephrology and Dialysis Unit, ASST Fatebenefratelli Sacco, Università degli Studi di Milano , Italy
| | - Manuel Alfredo Podestà
- Nephrology Unit, Department of Health Sciences, Università degli Studi di Milano , Italy
| | - Paolo Cravedi
- Department of Medicine, Icahn School of Medicine at Mount Sinai , New York, NY
| |
Collapse
|
25
|
Presence of specific SARS-COV2 antibodies in hemodialysis patients and their caregivers after the first wave of COVID-19. Sci Rep 2022; 12:11972. [PMID: 35831388 PMCID: PMC9279503 DOI: 10.1038/s41598-022-15913-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 06/30/2022] [Indexed: 12/02/2022] Open
Abstract
Hemodialysis (HD) patients are at risk for severe COVID-19 and cannot comply with social distancing. SARS-COV2 seroprevalence in French patients and caregivers after the first wave of COVID-19 is unknown. SeroCOVIDial is a prospective study conducted between June and December 2020. SARS-COV2 seroprevalence was evaluated by a rapid serological test (BIOSYNEX) in HD patients and caregivers, and the presence or not of anti-SARS-COV2 neutralizing or non-neutralizing antibodies in patients was also determined by ELISA and seroneutralization. In June 2020, 451 HD patients and 238 caregivers were included. Overall SARS-COV2 seroprevalence was 8.4% (patients) and 6.7% (caregivers), and was 87.1% (patients) and 90.0% (caregivers) in participants with a previously documented SARS-COV2 infection. Overall seroprevalence reached 13.8% (patients) and 12.6% (caregivers) following the second epidemic wave. During the follow-up, 38 (8.4%) patients died (9 of COVID-19). Among the 44 (10.6%) patients who became infected, only two were seropositive at M0. The levels of anti-SARS-COV2 antibodies decreased over time in patients and caregivers. The BIOSYNEX test showed 82.9% sensitivity and 97.7% specificity. Prevalence of anti-SARS-COV2 antibodies was low in HD patients and caregivers after the first epidemic wave but rose after the second wave. A rapid serological test showed good performances and could be useful for future monitoring of anti-SARS-COV2 antibodies.
Collapse
|
26
|
Prasithsirikul W, Nopsopon T, Phutrakool P, Suwanwattana P, Kantagowit P, Pongpirul W, Jongkaewwattana A, Pongpirul K. ChAdOx1 nCoV-19 Immunogenicity and Immunological Response Following COVID-19 Infection in Patients Receiving Maintenance Hemodialysis. Vaccines (Basel) 2022; 10:vaccines10060959. [PMID: 35746567 PMCID: PMC9230560 DOI: 10.3390/vaccines10060959] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 06/11/2022] [Accepted: 06/14/2022] [Indexed: 12/17/2022] Open
Abstract
Patients with end-stage renal disease (ESRD) receiving hemodialysis (HD) were found to have a decreased immune response following mRNA COVID-19 immunization. ChAdOx1 nCoV-19 was a promising COVID-19 vaccine that performed well in the general population, but the evidence on immunogenicity in ESRD with HD patients was limited. Moreover, the immunological response to COVID-19 infection was inconclusive in patients with ESRD and HD. The aim of this study was to investigate the immunogenicity of ChAdOx1 nCoV-19 vaccination and the immunological response after COVID-19 infection in ESRD patients with HD. The blood samples were obtained at baseline, 1-month, and 3-month follow-up after each shot or recovery. All participants were measured for anti-spike IgG by the ELISA method, using Euroimmun. This study found a significant increase in anti-spike IgG after 1 month of two-shot ChAdOx1 nCoV-19 vaccination, followed by a significant decrease after 3 months. On the other hand, the anti-spike IgG was maintained in the post-recovery group. There was no significant difference in the change of anti-spike IgG between the one-shot ChAdOx1 nCoV-19-vaccinated and post-recovery groups for both 1-month and 3-month follow-ups. The seroconversion rate for the vaccinated group was 60.32% at 1 month after one-shot vaccination and slightly dropped to 58.73% at the 3-month follow-up, then was 92.06% at 1 month after two-shot vaccination and reduced to 82.26% at the 3-month follow-up. For the recovered group, the seroconversion rate was 95.65% at 1 month post-recovery and 92.50% at 3-month follow-up. This study demonstrated the immunogenicity of two-dose ChAdOx1 nCoV-19 in ESRD patients with HD for humoral immunity. After COVID-19 infection, the humoral immune response was strong and could be maintained for at least three months.
Collapse
Affiliation(s)
- Wisit Prasithsirikul
- Bamrasnaradura Infectious Diseases Institute, Nonthaburi 11000, Thailand; (W.P.); (P.S.); (W.P.)
| | - Tanawin Nopsopon
- Department of Preventive and Social Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (P.P.); (P.K.)
- School of Global Health, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
- Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA 02215, USA
- Correspondence: (T.N.); (K.P.)
| | - Phanupong Phutrakool
- Department of Preventive and Social Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (P.P.); (P.K.)
- School of Global Health, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Pawita Suwanwattana
- Bamrasnaradura Infectious Diseases Institute, Nonthaburi 11000, Thailand; (W.P.); (P.S.); (W.P.)
| | - Piyawat Kantagowit
- Department of Preventive and Social Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (P.P.); (P.K.)
- School of Global Health, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Wannarat Pongpirul
- Bamrasnaradura Infectious Diseases Institute, Nonthaburi 11000, Thailand; (W.P.); (P.S.); (W.P.)
| | - Anan Jongkaewwattana
- National Center of Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathum Thani 12120, Thailand;
| | - Krit Pongpirul
- Department of Preventive and Social Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (P.P.); (P.K.)
- School of Global Health, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
- Bumrungrad International Hospital, Bangkok 10110, Thailand
- Correspondence: (T.N.); (K.P.)
| |
Collapse
|
27
|
Tillmann FP, Figiel L, Ricken J, Still H, Korte C, Plaßmann G, Harth A, Jörres A, von Landenberg P. Effect of Third and Fourth mRNA-Based Booster Vaccinations on SARS-CoV-2 Neutralizing Antibody Titer Formation, Risk Factors for Non-Response, and Outcome after SARS-CoV-2 Omicron Breakthrough Infections in Patients on Chronic Hemodialysis: A Prospective Multicenter Cohort Study. J Clin Med 2022; 11:jcm11113187. [PMID: 35683580 PMCID: PMC9181211 DOI: 10.3390/jcm11113187] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 05/09/2022] [Accepted: 05/16/2022] [Indexed: 02/06/2023] Open
Abstract
The aim of this study is to determine the effect of repeated vaccinations on neutralizing SARS-CoV-2 IgG antibody titers, evaluate risk factors for immunological non-response, and to report breakthrough infections in chronic hemodialysis patients. Methods: A prospective, multi-center cohort study in 163 chronic hemodialysis patients was conducted. Antibody titers were measured three months after second, third, and fourth (10 pts) booster vaccinations. SARS-CoV-2 neutralizing antibody titers in BAU/mL and % inhibition were divided into three categories (<216, 216–433, >433 and <33, 33–66, and >66%). Somers’s test, paired t-test, and univariable and multivariable logistic regression analysis were applied to evaluate differences in antibody levels and search for risk factors for vaccination failure defined as neutralizing titers <50% and/or need for repeated booster vaccinations. Furthermore, we report on a case series to describe characteristics of patients after four vaccinations (n = 10) and breakthrough infections (n = 20). Results: Third dose boosters resulted in higher proportions of patients with neutralizing antibody levels >66% as compared to after the second dose (64.7% after second dose vs. 88.9% after third dose, p = 0.003), as well as in a respective increase in neutralizing titer levels in % from 68 ± 33% to 89 ± 24 (p < 0.001). The proportion of patients with IgG-titers below 216 BAU/mL decreased from 38.6 to 10.5% (p ≤ 0.001). Age (p = 0.004, OR 1.066, 95% CI 1.020–1.114) and presence of immunosuppressive medications (p = 0.002, OR 8.267, 95% CI 2.206–30.975) were identified as major risk factors for vaccination failure. Repeated booster vaccinations ≥4 times were effective in 8 out of 10 former low-responders (80%) without any side effects or safety concerns. Breakthrough infections showed a clinically mild course but were associated with prolonged viral shedding on PCR-testing ranging 7–29 (mean 13) days. Conclusions: Third and fourth mRNA-based booster vaccinations resulted in higher and longer lasting SARS-CoV-2 antibody levels as compared to after two dosages. The presence of immunosuppressive medication and repeat vaccinations are major potentially modifiable measures to increase antibody levels in non-or low-responders. Breakthrough infections with SARS-CoV-2 Omicron were associated with prolonged viral shedding but clinically mild disease courses.
Collapse
Affiliation(s)
- Frank-Peter Tillmann
- Department of Medicine I—Nephrology, Transplantation & Medical Intensive Care, Medical Center Cologne-Merheim, University Witten/Herdecke, Ostmerheimer Str. 200, D-51109 Cologne, Germany; (A.H.); (A.J.)
- Nephrologisches Zentrum Ibbenbüren, Gravenhorsterstr. 1, D-49477 Ibbenbüren, Germany;
- Correspondence:
| | - Lars Figiel
- Nephrologisches Zentrum Emsdetten, Nordwalderstr. 48-50, D-48282 Emsdetten, Germany; (L.F.); (C.K.)
| | - Johannes Ricken
- Nephrologisches Zentrum Rheine, Neuenkirchenerstr. 104, D-48431 Rheine, Germany; (J.R.); (G.P.)
| | - Hermann Still
- Nephrologisches Zentrum Ibbenbüren, Gravenhorsterstr. 1, D-49477 Ibbenbüren, Germany;
| | - Christoph Korte
- Nephrologisches Zentrum Emsdetten, Nordwalderstr. 48-50, D-48282 Emsdetten, Germany; (L.F.); (C.K.)
| | - Grete Plaßmann
- Nephrologisches Zentrum Rheine, Neuenkirchenerstr. 104, D-48431 Rheine, Germany; (J.R.); (G.P.)
| | - Ana Harth
- Department of Medicine I—Nephrology, Transplantation & Medical Intensive Care, Medical Center Cologne-Merheim, University Witten/Herdecke, Ostmerheimer Str. 200, D-51109 Cologne, Germany; (A.H.); (A.J.)
| | - Achim Jörres
- Department of Medicine I—Nephrology, Transplantation & Medical Intensive Care, Medical Center Cologne-Merheim, University Witten/Herdecke, Ostmerheimer Str. 200, D-51109 Cologne, Germany; (A.H.); (A.J.)
| | | |
Collapse
|
28
|
Ashby DR, Caplin B, Corbett RW, Asgari E, Kumar N, Sarnowski A, Hull R, Makanjuola D, Cole N, Chen J, Nyberg S, McCafferty K, Zaman F, Cairns H, Sharpe C, Bramham K, Motallebzadeh R, Anwari KJ, Salama AD, Banerjee D. Severity of COVID-19 after Vaccination among Hemodialysis Patients: An Observational Cohort Study. Clin J Am Soc Nephrol 2022; 17:843-850. [PMID: 35649718 PMCID: PMC9269655 DOI: 10.2215/cjn.16621221] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 03/22/2022] [Indexed: 12/18/2022]
Abstract
BACKGROUND AND OBJECTIVES Patients receiving hemodialysis are at high risk from coronavirus disease 2019 (COVID-19) and demonstrate impaired immune responses to vaccines. There have been several descriptions of their immunologic responses to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccination, but few studies have described the clinical efficacy of vaccination in patients on hemodialysis. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS In a multicenter observational study of the London hemodialysis population undergoing surveillance PCR testing during the period of vaccine rollout with BNT162b2 and AZD1222, all of those positive for SARS-CoV-2 were identified. Clinical outcomes were analyzed according to predictor variables, including vaccination status, using a mixed effects logistic regression model. Risk of infection was analyzed in a subgroup of the base population using a Cox proportional hazards model with vaccination status as a time-varying covariate. RESULTS SARS-CoV-2 infection was identified in 1323 patients of different ethnicities (Asian/other, 30%; Black, 38%; and White, 32%), including 1047 (79%) unvaccinated, 86 (7%) after first-dose vaccination, and 190 (14%) after second-dose vaccination. The majority of patients had a mild course; however, 515 (39%) were hospitalized, and 172 (13%) died. Older age, diabetes, and immune suppression were associated with greater illness severity. In regression models adjusted for age, comorbidity, and time period, prior two-dose vaccination was associated with a 75% (95% confidence interval, 56 to 86) lower risk of admission and 88% (95% confidence interval, 70 to 95) fewer deaths compared with unvaccinated patients. No loss of protection was seen in patients over 65 years or with increasing time since vaccination, and no difference was seen between vaccine types. CONCLUSIONS These data demonstrate a substantially lower risk of severe COVID-19 after vaccination in patients on dialysis who become infected with SARS-CoV-2.
Collapse
Affiliation(s)
- Damien R Ashby
- Renal and Transplant Centre, Hammersmith Hospital, Imperial College Healthcare NHS Trust, London, United Kingdom .,Department of Immunology and Inflammation, Imperial College London, London, United Kingdom
| | - Ben Caplin
- Department of Renal Medicine, University College London, London, United Kingdom
| | - Richard W Corbett
- Renal and Transplant Centre, Hammersmith Hospital, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Elham Asgari
- Kidney Services, Guy's and St. Thomas' NHS Foundation Trust, London, United Kingdom
| | - Nicola Kumar
- Kidney Services, Guy's and St. Thomas' NHS Foundation Trust, London, United Kingdom
| | - Alexander Sarnowski
- Renal and Transplantation, St. George's University Hospitals NHS Foundation Trust, London, United Kingdom
| | - Richard Hull
- Renal and Transplantation, St. George's University Hospitals NHS Foundation Trust, London, United Kingdom
| | - David Makanjuola
- South West Thames Renal and Transplantation Unit, Epsom and St. Helier University Hospitals NHS Trust, London, United Kingdom
| | - Nicholas Cole
- South West Thames Renal and Transplantation Unit, Epsom and St. Helier University Hospitals NHS Trust, London, United Kingdom
| | - Jian Chen
- Renal Service, Barts Health NHS Trust, London, United Kingdom
| | - Sofia Nyberg
- Renal Service, Barts Health NHS Trust, London, United Kingdom
| | | | - Faryal Zaman
- Department of Renal Medicine, King's College Hospital NHS Foundation Trust, London, United Kingdom
| | - Hugh Cairns
- Department of Renal Medicine, King's College Hospital NHS Foundation Trust, London, United Kingdom
| | - Claire Sharpe
- Department of Renal Medicine, King's College Hospital NHS Foundation Trust, London, United Kingdom
| | - Kate Bramham
- Department of Renal Medicine, King's College Hospital NHS Foundation Trust, London, United Kingdom
| | - Reza Motallebzadeh
- Renal Medicine, Royal Free London NHS Foundation Trust, London, United Kingdom
| | - Kashif Jamil Anwari
- Renal Medicine, Royal Free London NHS Foundation Trust, London, United Kingdom
| | - Alan D Salama
- Department of Renal Medicine, University College London, London, United Kingdom
| | | | | |
Collapse
|
29
|
El Karoui K, De Vriese AS. COVID-19 in dialysis: clinical impact, immune response, prevention, and treatment. Kidney Int 2022; 101:883-894. [PMID: 35176326 PMCID: PMC8842412 DOI: 10.1016/j.kint.2022.01.022] [Citation(s) in RCA: 89] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 01/01/2022] [Accepted: 01/20/2022] [Indexed: 01/07/2023]
Abstract
The COVID-19 pandemic has profound adverse effects on the population on dialysis. Patients requiring dialysis are at an increased risk of SARS-CoV-2 infection and mortality, and many have experienced psychological distress as well as delayed or suboptimal care. COVID-19 survivors have prolonged viral shedding, but generally develop a robust and long-lasting humoral immune response that correlates with initial disease severity. However, protection against reinfection is incomplete. A growing body of evidence reveals delayed and blunted immune responses to SARS-CoV-2 vaccination. Administration of a third dose within 1 to 2 months of prime-boost vaccination significantly increases antibody levels, in particular in patients with poor initial responses. Patients on dialysis have inferior immune responses to adenoviral vector vaccines than to mRNA vaccines. The immunogenicity of the mRNA-1273 vaccine is markedly better than that of the BNT162b2 vaccine, most likely by virtue of its higher mRNA content. Despite suboptimal immune responses in patients on dialysis, preliminary data suggest that vaccination partially protects against infection and severe disease requiring hospitalization. However, progressive waning of immunity and emergence of SARS-CoV-2 variants with a high potential of immune escape call for a booster dose in all patients on dialysis 4 to 6 months after prime-boost vaccination. Patients with persistent poor vaccine responses may be candidates for primary prophylaxis strategies. In the absence of specific data in patients on dialysis, therapeutic strategies in the event of established COVID-19 must be extrapolated from evidence obtained in the population not on dialysis. Neutralizing monoclonal antibodies may be an attractive option after a high-risk exposure or during the early course of infection.
Collapse
Affiliation(s)
- Khalil El Karoui
- Department of Nephrology and Transplantation, Hôpitaux Universitaires Henri Mondor, Fédération Hospitalo-Universitaire TRUE, Université Paris Est, Créteil, France
| | - An S De Vriese
- Division of Nephrology and Infectious Diseases, AZ Sint-Jan Brugge-Oostende AV, Brugge, Belgium; Department of Internal Medicine, Ghent University, Ghent, Belgium.
| |
Collapse
|
30
|
Shankar S, Beckett J, Tipton T, Ogbe A, Kasanyinga M, Dold C, Lumley S, Dengu F, Rompianesi G, Elgilani F, Longet S, Deeks A, Payne RP, Duncan CJ, Richter A, de Silva TI, Turtle L, Bull K, Barnardo M, Friend PJ, Dunachie SJ, Hester J, Issa F, Barnes E, Carroll MW, Klenerman P. SARS-CoV-2-Specific T Cell Responses Are Not Associated with Protection against Reinfection in Hemodialysis Patients. J Am Soc Nephrol 2022; 33:883-887. [PMID: 35361708 PMCID: PMC9063898 DOI: 10.1681/asn.2021121587] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Patients on hemodialysis (HD) are vulnerable to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and mount poor neutralizing antibody responses after two-dose vaccination. Although serological responses have been associated with reduced rates of reinfection, the relationship between cellular immunogenicity and protection has not been established. We report, for the first time, high incidence of reinfection in patients on HD who are vaccine naive (25%), which identifies that T cell responses do not predict protection against reinfection. Instead, patients on HD who went on to become reinfected had mounted highly variable and sometimes robust proliferative T cell responses to a broad array of SARS-CoV-2 peptide pools during the primary infection. The understanding that SARS-CoV-2–specific T cell responses are not predictive of protection against future infection will be a critical issue when measuring clinical efficacy of vaccination in these vulnerable cohorts, particularly when facing rapidly emerging variants of concern.
Collapse
Affiliation(s)
- Sushma Shankar
- Transplantation Research and Immunology Group, Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
- Oxford Transplant Centre, Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
- Oxford University Hospitals National Health Service (NHS) Foundation Trust, Oxford, United Kingdom
| | - Joseph Beckett
- Transplantation Research and Immunology Group, Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
| | - Tom Tipton
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Ane Ogbe
- Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, United Kingdom
| | - Mwila Kasanyinga
- Oxford Vaccine Group, University of Oxford, Oxford, United Kingdom
| | - Christina Dold
- Oxford Vaccine Group, University of Oxford, Oxford, United Kingdom
| | - Sheila Lumley
- Oxford University Hospitals National Health Service (NHS) Foundation Trust, Oxford, United Kingdom
- Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Fungai Dengu
- Oxford Transplant Centre, Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
- Oxford University Hospitals National Health Service (NHS) Foundation Trust, Oxford, United Kingdom
| | - Gianluca Rompianesi
- Oxford Transplant Centre, Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
- Oxford University Hospitals National Health Service (NHS) Foundation Trust, Oxford, United Kingdom
| | - Faysal Elgilani
- Oxford Transplant Centre, Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
- Oxford University Hospitals National Health Service (NHS) Foundation Trust, Oxford, United Kingdom
| | - Stephanie Longet
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Alexandra Deeks
- Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, United Kingdom
| | - Rebecca P. Payne
- Translational and Clinical Research Institute Immunity and Inflammation Theme, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Christopher J.A. Duncan
- Translational and Clinical Research Institute Immunity and Inflammation Theme, Newcastle University, Newcastle upon Tyne, United Kingdom
- Department of Infection and Tropical Medicine, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Alex Richter
- Institute of Cancer and Genomic Science, College of Medical and Dental Science, University of Birmingham, Birmingham, United Kingdom
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - Thushan I. de Silva
- Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, United Kingdom
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, United Kingdom
| | - Lance Turtle
- National Institute for Health Research Health Protection Research Unit in Emerging and Zoonotic Infections, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
- Liverpool University Hospitals NHS Trust, Liverpool, United Kingdom
| | - Katherine Bull
- Oxford University Hospitals National Health Service (NHS) Foundation Trust, Oxford, United Kingdom
- Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Martin Barnardo
- Oxford Transplant Centre, Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
- Oxford University Hospitals National Health Service (NHS) Foundation Trust, Oxford, United Kingdom
| | - Peter J. Friend
- Oxford Transplant Centre, Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
- Oxford University Hospitals National Health Service (NHS) Foundation Trust, Oxford, United Kingdom
| | - Susanna J. Dunachie
- Oxford University Hospitals National Health Service (NHS) Foundation Trust, Oxford, United Kingdom
- Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, United Kingdom
- Oxford Centre for Global Health Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
| | - Joanna Hester
- Transplantation Research and Immunology Group, Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
| | - Fadi Issa
- Transplantation Research and Immunology Group, Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
| | - Eleanor Barnes
- Oxford University Hospitals National Health Service (NHS) Foundation Trust, Oxford, United Kingdom
- Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, United Kingdom
| | - Miles W. Carroll
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Paul Klenerman
- Oxford University Hospitals National Health Service (NHS) Foundation Trust, Oxford, United Kingdom
- Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
31
|
Sanders JSF, Bemelman FJ, Messchendorp AL, Baan CC, van Baarle D, van Binnendijk R, Diavatopoulos DA, Frölke SC, Geers D, GeurtsvanKessel CH, den Hartog G, van der Heiden M, Imhof C, Kho MM, Koopmans MP, Malahe SRK, Mattheussens WB, van der Molen R, van Mourik D, Remmerswaal EB, Rots N, Vart P, de Vries RD, Gansevoort RT, Hilbrands LB, Reinders ME, RECOVAC Collaborators *. The RECOVAC Immune-response Study: The Immunogenicity, Tolerability, and Safety of COVID-19 Vaccination in Patients With Chronic Kidney Disease, on Dialysis, or Living With a Kidney Transplant. Transplantation 2022; 106:821-834. [PMID: 34753894 PMCID: PMC8942603 DOI: 10.1097/tp.0000000000003983] [Citation(s) in RCA: 130] [Impact Index Per Article: 43.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 09/22/2021] [Accepted: 09/26/2021] [Indexed: 11/29/2022]
Abstract
BACKGROUND In kidney patients COVID-19 is associated with severely increased morbidity and mortality. A comprehensive comparison of the immunogenicity, tolerability, and safety of COVID-19 vaccination in different cohorts of kidney patients and a control cohort is lacking. METHODS This investigator driven, prospective, controlled multicenter study included 162 participants with chronic kidney disease (CKD) stages G4/5 (eGFR < 30 mL/min/1.73m2), 159 participants on dialysis, 288 kidney transplant recipients, and 191 controls. Participants received 2 doses of the mRNA-1273 COVID-19 vaccine (Moderna). The primary endpoint was seroconversion. RESULTS Transplant recipients had a significantly lower seroconversion rate when compared with controls (56.9% versus 100%, P < 0.001), with especially mycophenolic acid, but also, higher age, lower lymphocyte concentration, lower eGFR, and shorter time after transplantation being associated with nonresponder state. Transplant recipients also showed significantly lower titers of neutralizing antibodies and T-cell responses when compared with controls. Although a high seroconversion rate was observed for participants with CKD G4/5 (100%) and on dialysis (99.4%), mean antibody concentrations in the CKD G4/5 cohort and dialysis cohort were lower than in controls (2405 [interquartile interval 1287-4524] and 1650 [698-3024] versus 3186 [1896-4911] BAU/mL, P = 0.06 and P < 0.001, respectively). Dialysis patients and especially kidney transplant recipients experienced less systemic vaccination related adverse events. No specific safety issues were noted. CONCLUSIONS The immune response following vaccination in patients with CKD G4/5 and on dialysis is almost comparable to controls. In contrast, kidney transplant recipients have a poor response. In this latter, patient group development of alternative vaccination strategies are warranted.
Collapse
Affiliation(s)
- Jan-Stephan F. Sanders
- Division of Nephrology, Department of Internal Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Frederike J. Bemelman
- Renal Transplant Unit, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - A. Lianne Messchendorp
- Division of Nephrology, Department of Internal Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Carla C. Baan
- Department of Internal Medicine, Nephrology, and Transplantation, Erasmus MC Transplant Institute, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Debbie van Baarle
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, Groningen, The Netherlands
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - Rob van Binnendijk
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - Dimitri A. Diavatopoulos
- Radboud Center for Infectious Diseases, Radboud University Medical Center Nijmegen, Nijmegen, The Netherlands
- Radboud Institute for Molecular Life Sciences, Department of Laboratory Medicine, Laboratory of Medical Immunology, Radboud University Medical Center Nijmegen, Nijmegen, The Netherlands
| | - Sophie C. Frölke
- Renal Transplant Unit, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Daryl Geers
- Department of Viroscience, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | - Gerco den Hartog
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - Marieke van der Heiden
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, Groningen, The Netherlands
| | - Celine Imhof
- Division of Nephrology, Department of Internal Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, Groningen, The Netherlands
| | - Marcia M.L. Kho
- Department of Internal Medicine, Nephrology, and Transplantation, Erasmus MC Transplant Institute, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | | | - Wouter B. Mattheussens
- Department of Nephrology, Radboud University Medical Center, Radboud Institute for Health Sciences, Nijmegen, The Netherlands
| | - Renate van der Molen
- Radboud Institute for Molecular Life Sciences, Department of Laboratory Medicine, Laboratory of Medical Immunology, Radboud University Medical Center Nijmegen, Nijmegen, The Netherlands
| | - Djenolan van Mourik
- Department of Viroscience, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Ester B.M. Remmerswaal
- Department of Experimental Immunology, Amsterdam Infection and Immunity Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Nynke Rots
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - Priya Vart
- Division of Nephrology, Department of Internal Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Rory D. de Vries
- Department of Viroscience, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Ron T. Gansevoort
- Division of Nephrology, Department of Internal Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Luuk B. Hilbrands
- Department of Nephrology, Radboud University Medical Center, Radboud Institute for Health Sciences, Nijmegen, The Netherlands
| | - Marlies E.J. Reinders
- Department of Internal Medicine, Nephrology, and Transplantation, Erasmus MC Transplant Institute, Erasmus Medical Center, Rotterdam, The Netherlands
| | - RECOVAC Collaborators*
- Division of Nephrology, Department of Internal Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Renal Transplant Unit, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Department of Internal Medicine, Nephrology, and Transplantation, Erasmus MC Transplant Institute, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, Groningen, The Netherlands
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
- Radboud Center for Infectious Diseases, Radboud University Medical Center Nijmegen, Nijmegen, The Netherlands
- Radboud Institute for Molecular Life Sciences, Department of Laboratory Medicine, Laboratory of Medical Immunology, Radboud University Medical Center Nijmegen, Nijmegen, The Netherlands
- Department of Viroscience, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Nephrology, Radboud University Medical Center, Radboud Institute for Health Sciences, Nijmegen, The Netherlands
- Department of Experimental Immunology, Amsterdam Infection and Immunity Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
32
|
A case of COVID-19 reinfection in a hemodialysis patient: the role of antibody in SARS-CoV-2 infection. CEN Case Rep 2022; 11:422-427. [PMID: 35266095 PMCID: PMC8906520 DOI: 10.1007/s13730-022-00697-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 02/24/2022] [Indexed: 11/07/2022] Open
Abstract
Hemodialysis patients are vulnerable to severe and lethal COVID-19, and their protective immunity against COVID-19 is not yet fully understood. Therefore, we report a case of COVID-19 reinfection in a hemodialysis patient 81 days after the first episode and discuss the role of antibodies in SARS-CoV-2 infection. A hemodialysis patient developed asymptomatic COVID-19 due to an outbreak in a hospital on October 29th, 2020. As he was hospitalized and did not develop any symptoms, he was discharged on November 9th. On January 18th, he presented with symptomatic COVID-19 due to close household contact. Then, he developed respiratory failure and was transferred to National Center for Global Health and Medicine if he would need intensive care. He recovered with oxygen inhalation, favipiravir, and steroid treatment, and was discharged on February 12th. To evaluate anti-SARS-CoV-2 antibodies during two hospital stays, we measured immunoglobulin (Ig) G specific for S1 subunit of Spike (S) protein of SARS-CoV-2 (IgG-S1) , IgG specific for the full-length S protein (anti-Spike IgG) and neutralizing antibodies. No seroconversion occurred 5 days after initial infection, the seroconversion of IgG-S1 was observed 10 days after the second infection. Similar to IgG-S1 antibody titer results, anti-Spike IgG and neutralizing antibodies increased from 12 days after the second infection. In conclusion, we experienced a case of COVID-19 reinfection in a hemodialysis patient 81 days after the first episode and showed the kinetics and role of antibodies in SARS-CoV-2 infection. Further studies are needed to understand SARS-CoV-2 reinfection risk in hemodialysis patients and its clinical significance.
Collapse
|
33
|
Raja N, Rajagopalan A, Arunachalam J, Prasath A, Durai R, Rajendran M. Humoral response to viral vector COVID-19 vaccine in hemodialysis patients. Kidney Res Clin Pract 2022; 41:342-350. [PMID: 35286797 PMCID: PMC9184837 DOI: 10.23876/j.krcp.21.184] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 10/31/2021] [Indexed: 11/04/2022] Open
Affiliation(s)
- Niranjan Raja
- Department of Nephrology, Madurai Medical College, Madurai, India
- Correspondence: Niranjan Raja Department of Nephrology, Madurai Medical College, Panagal road, Alwarpuram, Madurai, Tamil Nadu 625020, India. E-mail:
| | - Arul Rajagopalan
- Department of Nephrology, Madurai Medical College, Madurai, India
| | | | - Arun Prasath
- Department of Nephrology, Madurai Medical College, Madurai, India
| | - Rakesh Durai
- Department of Nephrology, Madurai Medical College, Madurai, India
| | | |
Collapse
|
34
|
Crespo M, Barrilado-Jackson A, Padilla E, Eguía J, Echeverria-Esnal D, Cao H, Faura A, Folgueiras M, Solà-Porta E, Pascual S, Barbosa F, Hurtado S, Ribera L, Río-No L, Pérez-Sáez MJ, Redondo-Pachón D, Pascual J. Negative immune responses to two-dose mRNA COVID-19 vaccines in renal allograft recipients assessed with simple antibody and interferon gamma release assay cellular monitoring. Am J Transplant 2022; 22:786-800. [PMID: 34551187 PMCID: PMC8653097 DOI: 10.1111/ajt.16854] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 09/13/2021] [Accepted: 09/16/2021] [Indexed: 01/25/2023]
Abstract
Studies are urgently needed to characterize immunogenicity, efficacy, and safety of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) mRNA vaccines in kidney transplant (KT) recipients, excluded from major clinical trials. Complex ELISPOT and other cellular response techniques have been applied, but simpler tools are needed. An easy-to-use real-world monitoring of SARS-CoV-2 IgG antibodies against the Spike protein and QuantiFERON® SARS-CoV-2 IFNγ release assay (IGRA) were performed at baseline and 28 days after the second dose in KT recipients and controls (dialysis patients and healthy ones). All healthy controls and >95% dialysis controls became positive for anti-S IgG antibodies, while only 63.3% of KT patients seroconverted with a very low antibody level. A positive IGRA was documented in 96.9% of controls, 89.3% peritoneal dialysis, 77.6% hemodialysis, 61.3% of KT patients transplanted more than 1 year ago and only 36% of those transplanted within the previous 12 months. Overall, 100% of healthy controls, 95.4% of dialysis patients and 78.8% KT recipients developed any immune response (humoral and/or cellular) against SARS-CoV-2. KT patients showed low rates of immune responses to mRNA Coronavirus infectious disease 2019 vaccines, especially those with recent transplantations. Simple humoral and cellular monitoring is advisable, so that repeated doses may be scheduled according to the results.
Collapse
Affiliation(s)
- Marta Crespo
- Department of Nephrology, Hospital del Mar, Barcelona, Spain,Correspondence Marta Crespo, Department of Nephrology, Hospital del Mar, Barcelona, Spain.
| | | | | | - Jorge Eguía
- Laboratori de Referència de Catalunya SA, Barcelona, Spain
| | - Daniel Echeverria-Esnal
- Pharmacy Department, Hospital del Mar, Barcelona, Spain,Infectious Pathology and Antimicrobials Research Group (IPAR), Institut Hospital del Mar for Medical Research, Barcelona, Spain
| | - Higini Cao
- Department of Nephrology, Hospital del Mar, Barcelona, Spain
| | - Anna Faura
- Department of Nephrology, Hospital del Mar, Barcelona, Spain
| | | | | | - Sergi Pascual
- Department of Nephrology, Hospital del Mar, Barcelona, Spain
| | | | - Sara Hurtado
- Department of Nephrology, Fresenius Medical Care Services, Barcelona, Spain
| | - Laura Ribera
- Department of Nephrology, Fresenius Medical Care Services, Barcelona, Spain
| | - Laura Río-No
- Pharmacy Department, Hospital del Mar, Barcelona, Spain
| | | | | | - Julio Pascual
- Department of Nephrology, Hospital del Mar, Barcelona, Spain,Department of Nephrology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | | |
Collapse
|
35
|
Pan D, Kim JW, Nazareth J, Assadi S, Bellass A, Leach J, Brosnan JG, Ahmed A, Starcevic F, Sze S, Martin CA, Williams CM, Barer MR, Sahota A, Patel P, Tattersall A, Cooper A, Pareek M, Haldar P. Quantification and prognostic significance of interferon-γ secreting SARS-CoV-2 responsive T cells in hospitalized patients with acute COVID-19. J Infect 2022; 84:418-467. [PMID: 34801527 PMCID: PMC8603398 DOI: 10.1016/j.jinf.2021.11.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 11/16/2021] [Indexed: 11/23/2022]
Affiliation(s)
- Daniel Pan
- Department of Respiratory Sciences, University of Leicester, United Kingdom; Department of Infectious Diseases and HIV Medicine, University Hospitals of Leicester NHS Trust, United Kingdom.
| | - Jee Whang Kim
- Department of Respiratory Sciences, University of Leicester, United Kingdom; Respiratory Biomedical Research Center, University of Leicester, United Kingdom
| | - Joshua Nazareth
- Department of Respiratory Sciences, University of Leicester, United Kingdom; Department of Infectious Diseases and HIV Medicine, University Hospitals of Leicester NHS Trust, United Kingdom
| | - Sara Assadi
- Department of Infectious Diseases and HIV Medicine, University Hospitals of Leicester NHS Trust, United Kingdom
| | - Adam Bellass
- Department of Infectious Diseases and HIV Medicine, University Hospitals of Leicester NHS Trust, United Kingdom
| | - Jack Leach
- University of Leicester Medical School, United Kingdom
| | | | - Adam Ahmed
- University of Leicester Medical School, United Kingdom
| | - Fleur Starcevic
- Department of Infectious Diseases and HIV Medicine, University Hospitals of Leicester NHS Trust, United Kingdom
| | - Shirley Sze
- Department of Cardiovascular Sciences, University of Leicester, United Kingdom
| | - Christopher A Martin
- Department of Respiratory Sciences, University of Leicester, United Kingdom; Department of Infectious Diseases and HIV Medicine, University Hospitals of Leicester NHS Trust, United Kingdom
| | - Caroline M Williams
- Department of Respiratory Sciences, University of Leicester, United Kingdom; Department of Microbiology, University Hospitals of Leicester NHS Trust, United Kingdom
| | - Michael R Barer
- Department of Respiratory Sciences, University of Leicester, United Kingdom; Department of Microbiology, University Hospitals of Leicester NHS Trust, United Kingdom
| | - Amandip Sahota
- Department of Infectious Diseases and HIV Medicine, University Hospitals of Leicester NHS Trust, United Kingdom
| | - Prashanth Patel
- Department of Chemical and Metabolic Diseases, University Hospitals of Leicester NHS Trust, United Kingdom
| | | | - Andrea Cooper
- Department of Respiratory Sciences, University of Leicester, United Kingdom; Trudeau Institute, Saranac Lake, New York, USA
| | - Manish Pareek
- Department of Respiratory Sciences, University of Leicester, United Kingdom; Department of Infectious Diseases and HIV Medicine, University Hospitals of Leicester NHS Trust, United Kingdom
| | - Pranabashis Haldar
- Department of Respiratory Sciences, University of Leicester, United Kingdom; Respiratory Biomedical Research Center, University of Leicester, United Kingdom
| |
Collapse
|
36
|
Parshina E, Zulkarnaev A, Tolkach A, Ivanov A, Kislyy P, Gaipov A. Patients receiving hemodialysis do not lose SARS-CoV-2 antibodies more rapidly than non-renal controls: a prospective cohort study. Ren Fail 2022; 44:392-398. [PMID: 35220855 PMCID: PMC8890585 DOI: 10.1080/0886022x.2022.2042310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
Background Method Results Conclusion
Collapse
Affiliation(s)
- Ekaterina Parshina
- Department of Nephrology and Dialysis, Saint-Petersburg State University Hospital, Saint-Petersburg, Russia
| | - Alexey Zulkarnaev
- Surgical Department of Transplantology and Dialysis, Moscow Regional Research and Clinical Institute ("MONIKI"), Moscow, Russia
| | - Alexey Tolkach
- Department of Nephrology and Dialysis, Saint-Petersburg State University Hospital, Saint-Petersburg, Russia
| | - Andrey Ivanov
- Human Genetics Department, Saint Petersburg State University Hospital, Saint-Petersburg, Russian Federation
| | - Pavel Kislyy
- Department of Nephrology and Dialysis, Saint-Petersburg State University Hospital, Saint-Petersburg, Russia
| | - Abduzhappar Gaipov
- Department of Medicine, Nazarbayev University School of Medicine, Nur-Sultan, Kazakhstan
| |
Collapse
|
37
|
Jahn M, Korth J, Dorsch O, Anastasiou OE, Krawczyk A, Brochhagen L, van de Sand L, Sorge-Hädicke B, Tyczynski B, Witzke O, Dittmer U, Dolff S, Wilde B, Kribben A. Decline of Humoral Responses 6 Months after Vaccination with BNT162b2 (Pfizer–BioNTech) in Patients on Hemodialysis. Vaccines (Basel) 2022; 10:vaccines10020327. [PMID: 35214785 PMCID: PMC8878048 DOI: 10.3390/vaccines10020327] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 02/04/2022] [Accepted: 02/15/2022] [Indexed: 02/04/2023] Open
Abstract
This study analyzed binding and neutralizing antibody titers up to 6 months after standard vaccination with BNT162b2 (two doses of 30 µg each) in SARS-CoV-2 naïve patients (n = 59) on hemodialysis. Humoral vaccine responses were measured before and 6, 12, and 24 weeks after the first vaccination. A chemiluminescent immunoassay (CLIA) was used to quantify SARS-CoV-2 IgG against the spike glycoprotein. SARS-CoV-2 neutralizing activity was tested against the wild-type virus. A multivariable binary regression model was used to identify risk factors for the absence of humoral immune responses at 6 months. At week 6, vaccine-specific seroconversion was detected in 96.6% of all patients with median anti-SARS-CoV-2 IgGs of 918 BAU/mL. At weeks 12 and 24, seroconversion rates decreased to 91.5% and 79.7%, and corresponding median binding antibody titers declined to 298 BAU/mL and 89 BAU/mL, respectively. Neutralizing antibodies showed a decay from 79.6% at week 6 to 32.8% at week 24. The risk factor with the strongest association for vanishing immune responses was low serum albumin (p = 0.018). Regarding vaccine-specific humoral responses 6 months after the standard BNT162b2 vaccination schedule, SARS-CoV-2 naïve patients receiving hemodialysis must be considered at risk of becoming infected with SARS-CoV-2 and being infectious.
Collapse
Affiliation(s)
- Michael Jahn
- Department of Nephrology, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany; (J.K.); (B.T.); (B.W.); (A.K.)
- Correspondence:
| | - Johannes Korth
- Department of Nephrology, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany; (J.K.); (B.T.); (B.W.); (A.K.)
| | - Oliver Dorsch
- KfH Kuratorium für Dialyse und Nierentransplantation e.V, Friesener Straße 37a, 96317 Kronach, Germany;
| | - Olympia Evdoxia Anastasiou
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Virchowstr. 179, 45147 Essen, Germany; (O.E.A.); (U.D.)
| | - Adalbert Krawczyk
- Department of Infectious Diseases, West German Centre of Infectious Diseases, University Hospital Essen, University of Duisburg-Essen, Hufelandstr. 55, 45147 Essen, Germany; (A.K.); (L.B.); (L.v.d.S.); (O.W.); (S.D.)
| | - Leonie Brochhagen
- Department of Infectious Diseases, West German Centre of Infectious Diseases, University Hospital Essen, University of Duisburg-Essen, Hufelandstr. 55, 45147 Essen, Germany; (A.K.); (L.B.); (L.v.d.S.); (O.W.); (S.D.)
| | - Lukas van de Sand
- Department of Infectious Diseases, West German Centre of Infectious Diseases, University Hospital Essen, University of Duisburg-Essen, Hufelandstr. 55, 45147 Essen, Germany; (A.K.); (L.B.); (L.v.d.S.); (O.W.); (S.D.)
| | - Burkhard Sorge-Hädicke
- KfH Kuratorium für Dialyse und Nierentransplantation e.V, Alfried-Krupp-Str. 43, 45131 Essen, Germany;
| | - Bartosz Tyczynski
- Department of Nephrology, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany; (J.K.); (B.T.); (B.W.); (A.K.)
| | - Oliver Witzke
- Department of Infectious Diseases, West German Centre of Infectious Diseases, University Hospital Essen, University of Duisburg-Essen, Hufelandstr. 55, 45147 Essen, Germany; (A.K.); (L.B.); (L.v.d.S.); (O.W.); (S.D.)
| | - Ulf Dittmer
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Virchowstr. 179, 45147 Essen, Germany; (O.E.A.); (U.D.)
| | - Sebastian Dolff
- Department of Infectious Diseases, West German Centre of Infectious Diseases, University Hospital Essen, University of Duisburg-Essen, Hufelandstr. 55, 45147 Essen, Germany; (A.K.); (L.B.); (L.v.d.S.); (O.W.); (S.D.)
| | - Benjamin Wilde
- Department of Nephrology, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany; (J.K.); (B.T.); (B.W.); (A.K.)
| | - Andreas Kribben
- Department of Nephrology, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany; (J.K.); (B.T.); (B.W.); (A.K.)
| |
Collapse
|
38
|
Bassi J, Giannini O, Silacci-Fregni C, Pertusini L, Hitz P, Terrot T, Franzosi Y, Muoio F, Saliba C, Meury M, Dellota EA, Dillen JR, Hernandez P, Czudnochowski N, Cameroni E, Beria N, Ventresca M, Badellino A, Lavorato-Hadjeres S, Lecchi E, Bonora T, Mattiolo M, Trinci G, Garzoni D, Bonforte G, Forni-Ogna V, Giunzioni D, Berwert L, Gupta RK, Ferrari P, Ceschi A, Cippà P, Corti D, Lanzavecchia A, Piccoli L. Poor neutralization and rapid decay of antibodies to SARS-CoV-2 variants in vaccinated dialysis patients. PLoS One 2022; 17:e0263328. [PMID: 35143540 PMCID: PMC8830698 DOI: 10.1371/journal.pone.0263328] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 01/14/2022] [Indexed: 12/11/2022] Open
Abstract
Patients on dialysis are at risk of severe course of SARS-CoV-2 infection. Understanding the neutralizing activity and coverage of SARS-CoV-2 variants of vaccine-elicited antibodies is required to guide prophylactic and therapeutic COVID-19 interventions in this frail population. By analyzing plasma samples from 130 hemodialysis and 13 peritoneal dialysis patients after two doses of BNT162b2 or mRNA-1273 vaccines, we found that 35% of the patients had low-level or undetectable IgG antibodies to SARS-CoV-2 Spike (S). Neutralizing antibodies against the vaccine-matched SARS-CoV-2 and Delta variant were low or undetectable in 49% and 77% of patients, respectively, and were further reduced against other emerging variants. The fraction of non-responding patients was higher in SARS-CoV-2-naïve hemodialysis patients immunized with BNT162b2 (66%) than those immunized with mRNA-1273 (23%). The reduced neutralizing activity correlated with low antibody avidity. Patients followed up to 7 months after vaccination showed a rapid decay of the antibody response with an average 21- and 10-fold reduction of neutralizing antibodies to vaccine-matched SARS-CoV-2 and Delta variant, which increased the fraction of non-responders to 84% and 90%, respectively. These data indicate that dialysis patients should be prioritized for additional vaccination boosts. Nevertheless, their antibody response to SARS-CoV-2 must be continuously monitored to adopt the best prophylactic and therapeutic strategy.
Collapse
Affiliation(s)
- Jessica Bassi
- Humabs BioMed SA, A Subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | - Olivier Giannini
- Faculty of Biomedical Sciences, Università della Svizzera italiana, Lugano, Switzerland
- Department of Medicine, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | | | - Laura Pertusini
- Division of Nephrology, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Paolo Hitz
- Faculty of Biomedical Sciences, Università della Svizzera italiana, Lugano, Switzerland
| | - Tatiana Terrot
- Clinical Trial Unit, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Yves Franzosi
- Clinical Trial Unit, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Francesco Muoio
- Humabs BioMed SA, A Subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | - Christian Saliba
- Humabs BioMed SA, A Subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | - Marcel Meury
- Vir Biotechnology, San Francisco, California, United States of America
| | | | - Josh R. Dillen
- Vir Biotechnology, San Francisco, California, United States of America
| | - Patrick Hernandez
- Vir Biotechnology, San Francisco, California, United States of America
| | | | - Elisabetta Cameroni
- Humabs BioMed SA, A Subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | - Nicola Beria
- Division of Nephrology, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | | | - Alberto Badellino
- Division of Nephrology, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | | | - Elisabetta Lecchi
- Division of Nephrology, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Tecla Bonora
- Division of Nephrology, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Matteo Mattiolo
- Division of Nephrology, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Guido Trinci
- Division of Nephrology, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Daniela Garzoni
- Division of Nephrology, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Giuseppe Bonforte
- Division of Nephrology, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | | | - Davide Giunzioni
- Division of Nephrology, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Lorenzo Berwert
- Division of Nephrology, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Ravindra K. Gupta
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Cambridge, United Kingdom
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Paolo Ferrari
- Faculty of Biomedical Sciences, Università della Svizzera italiana, Lugano, Switzerland
- Division of Nephrology, Ente Ospedaliero Cantonale, Lugano, Switzerland
- Clinical School, University of New South Wales, Sydney, Australia
| | - Alessandro Ceschi
- Faculty of Biomedical Sciences, Università della Svizzera italiana, Lugano, Switzerland
- Clinical Trial Unit, Ente Ospedaliero Cantonale, Lugano, Switzerland
- Division of Clinical Pharmacology and Toxicology, Institute of Pharmacological Science of Southern Switzerland, Ente Ospedaliero Cantonale, Lugano, Switzerland
- Department of Clinical Pharmacology and Toxicology, University Hospital Zurich, Zurich, Switzerland
| | - Pietro Cippà
- Department of Medicine, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
- Division of Nephrology, Ente Ospedaliero Cantonale, Lugano, Switzerland
- Faculty of Medicine, University of Zurich, Zurich, Switzerland
| | - Davide Corti
- Humabs BioMed SA, A Subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | | | - Luca Piccoli
- Humabs BioMed SA, A Subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| |
Collapse
|
39
|
Al-Muhaiteeb A, AlSahow A, Al-Yousef A, AlHelal B, Alrajab H, Bahbahani Y, Dewidar N, Fanous GNM. Response to and outcomes of the Pfizer BNT162B2 vaccine in hemodialysis patients-A prospective observational study. Hemodial Int 2022; 26:216-222. [PMID: 34989097 PMCID: PMC9304137 DOI: 10.1111/hdi.13005] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 12/27/2021] [Accepted: 12/28/2021] [Indexed: 01/11/2023]
Abstract
Introduction COVID‐19 infection is associated with high mortality among hemodialysis patients. Standard vaccine response is generally lower among these patients. The adequate antibody titer response and the outcome of COVID‐19 vaccine responders versus non‐responders are unknown. Methods Hemodialysis patients on maintenance hemodialysis who have received two doses of Pfizer BNT162B2 vaccine were studied. Antibody response was tested after 14 days of the second dose. LIAISON SARS‐CoV2 S1/S2 IgG test by DiaSorin (Italy) was used to assess antibody response. Patients were followed between 3 and 7 months after vaccination for COVID‐19 infection, hospitalization and death related to COVID‐19. Findings A total of 138 patients received two doses of Pfizer BNT162B2 vaccine. One hundred and twenty‐seven patients had adequate response to the vaccine with IgG level ≥ 15 AU/ml versus 11 patients had poor response with IgG level ≤ 15 AU/ml. The response was 92% (127/138). Patient with history of prior COVID‐19 infection had higher antibody titer mean of 339 ± 113 versus 157 ± 140 for patient with no prior history of COVID‐19. Seven patients in both groups had COVID‐19 infection post vaccine. Among the responders, five patients had COIVD‐19 infection and two were hospitalized. These two patients had lower antibody titer of 23.9 and 75.2 AU/ml. In comparison, three patients who were not hospitalized had higher antibody titer 96.3, 118, and 319 AU/ml, respectively. In the non‐responders one patient was hospitalized and one death occurred with rate of infection of 18%. Discussion Seropositive patients with low antibody titer might be associated with worse outcome among responders. The ideal antibody titer level among dialysis patient is not known. Also, prior COVID‐19 infection is associated with higher response to vaccine with higher antibody titer. All non‐responders did not have prior COVID‐19 infection. More research is required to further evaluated protective antibody titer.
Collapse
Affiliation(s)
| | - Ali AlSahow
- Division of Nephrology, Jahra Hospital, Jahra City, Kuwait
| | - Anas Al-Yousef
- Division of Nephrology, Al Amiri Hospital, Kuwait City, Kuwait
| | | | - Heba Alrajab
- Division of Nephrology, Farwaniya Hospital, Sabah Al Nasser, Kuwait
| | | | - Noha Dewidar
- Division of Nephrology, Jahra Hospital, Jahra City, Kuwait
| | | |
Collapse
|
40
|
Chimon A, Ferrière E, Lammouchi MA, Jouan N, Michel PA, Saloum K, Morand-Joubert L, Schnuriger A, Leruez-Ville M, Fourgeaud J, Dahmane D, Bentaarit B, Guéry B, Fessi H, Kazdaghli H, Sounni F, Fearon T, Boudhabhay I, Pawlotsky JM, El Karoui K, Fourati S, Sakhi H. OUP accepted manuscript. Clin Kidney J 2022; 15:1785-1788. [PMID: 35999965 PMCID: PMC9383950 DOI: 10.1093/ckj/sfac137] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Indexed: 11/17/2022] Open
Affiliation(s)
| | | | | | | | | | - Kenda Saloum
- Virology Department, Trousseau Hospital, APHP, Sorbonne University, Paris, France
| | | | - Aurélie Schnuriger
- Virology Department, Trousseau Hospital, APHP, Sorbonne University, Paris, France
| | | | - Jacques Fourgeaud
- Virology Department, Necker Hospital, APHP, University of Paris, Paris, France
| | - Djamal Dahmane
- Nephrology Department, Mondor Hospital, APHP, University Paris Est, Créteil, France
| | - Boutheina Bentaarit
- Nephrology Department, Mondor Hospital, APHP, University Paris Est, Créteil, France
| | - Bruno Guéry
- Nephrology Department, Necker Hospital, APHP, University of Paris, Paris, France
| | - Hafedh Fessi
- Nephrology Department, Tenon Hospital, APHP, Sorbonne University, Paris, France
| | - Hajer Kazdaghli
- Nephrology Department, Tenon Hospital, APHP, Sorbonne University, Paris, France
| | - Farah Sounni
- Nephrology Department, Tenon Hospital, APHP, Sorbonne University, Paris, France
| | - Timothée Fearon
- Nephrology Department, Tenon Hospital, APHP, Sorbonne University, Paris, France
| | - Idris Boudhabhay
- Nephrology Department, Necker Hospital, APHP, University of Paris, Paris, France
| | | | | | | | | |
Collapse
|
41
|
Bassi J, Giannini O, Silacci-Fregni C, Pertusini L, Hitz P, Terrot T, Franzosi Y, Muoio F, Saliba C, Meury M, Dellota EA, Dillen JR, Hernandez P, Czudnochowski N, Cameroni E, Beria N, Ventresca M, Badellino A, Lavorato-Hadjeres S, Lecchi E, Bonora T, Mattiolo M, Trinci G, Garzoni D, Bonforte G, Forni-Ogna V, Giunzioni D, Berwert L, Gupta RK, Ferrari P, Ceschi A, Cippà P, Corti D, Lanzavecchia A, Piccoli L. Poor neutralization and rapid decay of antibodies to SARS-CoV-2 variants in vaccinated dialysis patients. PLoS One 2022. [PMID: 35143540 DOI: 10.1101/2021.10.05.21264054v2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/16/2023] Open
Abstract
Patients on dialysis are at risk of severe course of SARS-CoV-2 infection. Understanding the neutralizing activity and coverage of SARS-CoV-2 variants of vaccine-elicited antibodies is required to guide prophylactic and therapeutic COVID-19 interventions in this frail population. By analyzing plasma samples from 130 hemodialysis and 13 peritoneal dialysis patients after two doses of BNT162b2 or mRNA-1273 vaccines, we found that 35% of the patients had low-level or undetectable IgG antibodies to SARS-CoV-2 Spike (S). Neutralizing antibodies against the vaccine-matched SARS-CoV-2 and Delta variant were low or undetectable in 49% and 77% of patients, respectively, and were further reduced against other emerging variants. The fraction of non-responding patients was higher in SARS-CoV-2-naïve hemodialysis patients immunized with BNT162b2 (66%) than those immunized with mRNA-1273 (23%). The reduced neutralizing activity correlated with low antibody avidity. Patients followed up to 7 months after vaccination showed a rapid decay of the antibody response with an average 21- and 10-fold reduction of neutralizing antibodies to vaccine-matched SARS-CoV-2 and Delta variant, which increased the fraction of non-responders to 84% and 90%, respectively. These data indicate that dialysis patients should be prioritized for additional vaccination boosts. Nevertheless, their antibody response to SARS-CoV-2 must be continuously monitored to adopt the best prophylactic and therapeutic strategy.
Collapse
Affiliation(s)
- Jessica Bassi
- Humabs BioMed SA, A Subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | - Olivier Giannini
- Faculty of Biomedical Sciences, Università della Svizzera italiana, Lugano, Switzerland
- Department of Medicine, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | | | - Laura Pertusini
- Division of Nephrology, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Paolo Hitz
- Faculty of Biomedical Sciences, Università della Svizzera italiana, Lugano, Switzerland
| | - Tatiana Terrot
- Clinical Trial Unit, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Yves Franzosi
- Clinical Trial Unit, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Francesco Muoio
- Humabs BioMed SA, A Subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | - Christian Saliba
- Humabs BioMed SA, A Subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | - Marcel Meury
- Vir Biotechnology, San Francisco, California, United States of America
| | | | - Josh R Dillen
- Vir Biotechnology, San Francisco, California, United States of America
| | - Patrick Hernandez
- Vir Biotechnology, San Francisco, California, United States of America
| | | | - Elisabetta Cameroni
- Humabs BioMed SA, A Subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | - Nicola Beria
- Division of Nephrology, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | | | - Alberto Badellino
- Division of Nephrology, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | | | - Elisabetta Lecchi
- Division of Nephrology, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Tecla Bonora
- Division of Nephrology, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Matteo Mattiolo
- Division of Nephrology, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Guido Trinci
- Division of Nephrology, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Daniela Garzoni
- Division of Nephrology, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Giuseppe Bonforte
- Division of Nephrology, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | | | - Davide Giunzioni
- Division of Nephrology, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Lorenzo Berwert
- Division of Nephrology, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Ravindra K Gupta
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Cambridge, United Kingdom
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Paolo Ferrari
- Faculty of Biomedical Sciences, Università della Svizzera italiana, Lugano, Switzerland
- Division of Nephrology, Ente Ospedaliero Cantonale, Lugano, Switzerland
- Clinical School, University of New South Wales, Sydney, Australia
| | - Alessandro Ceschi
- Faculty of Biomedical Sciences, Università della Svizzera italiana, Lugano, Switzerland
- Clinical Trial Unit, Ente Ospedaliero Cantonale, Lugano, Switzerland
- Division of Clinical Pharmacology and Toxicology, Institute of Pharmacological Science of Southern Switzerland, Ente Ospedaliero Cantonale, Lugano, Switzerland
- Department of Clinical Pharmacology and Toxicology, University Hospital Zurich, Zurich, Switzerland
| | - Pietro Cippà
- Department of Medicine, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
- Division of Nephrology, Ente Ospedaliero Cantonale, Lugano, Switzerland
- Faculty of Medicine, University of Zurich, Zurich, Switzerland
| | - Davide Corti
- Humabs BioMed SA, A Subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | | | - Luca Piccoli
- Humabs BioMed SA, A Subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| |
Collapse
|
42
|
Pallett SJ, Jones R, Abdulaal A, Pallett MA, Rayment M, Patel A, Denny SJ, Mughal N, Khan M, Rosadas de Oliveira C, Pantelidis P, Randell P, Toumazou C, O'Shea MK, Tedder R, McClure MO, Davies GW, Moore LS. Variability in detection of SARS-CoV-2-specific antibody responses following mild infection: a prospective multicentre cross-sectional study, London, United Kingdom, 17 April to 17 July 2020. Euro Surveill 2022; 27:2002076. [PMID: 35086612 PMCID: PMC8796290 DOI: 10.2807/1560-7917.es.2022.27.4.2002076] [Citation(s) in RCA: 3] [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: 12/10/2020] [Accepted: 07/03/2021] [Indexed: 12/13/2022] Open
Abstract
IntroductionImmunoassays targeting different SARS-CoV-2-specific antibodies are employed for seroprevalence studies. The degree of variability between immunoassays targeting anti-nucleocapsid (anti-NP; the majority) vs the potentially neutralising anti-spike antibodies (including anti-receptor-binding domain; anti-RBD), particularly in mild or asymptomatic disease, remains unclear.AimsWe aimed to explore variability in anti-NP and anti-RBD antibody detectability following mild symptomatic or asymptomatic SARS-CoV-2 infection and analyse antibody response for correlation with symptomatology.MethodsA multicentre prospective cross-sectional study was undertaken (April-July 2020). Paired serum samples were tested for anti-NP and anti-RBD IgG antibodies and reactivity expressed as binding ratios (BR). Multivariate linear regression was performed analysing age, sex, time since onset, symptomatology, anti-NP and anti-RBD antibody BR.ResultsWe included 906 adults. Antibody results (793/906; 87.5%; 95% confidence interval: 85.2-89.6) and BR strongly correlated (ρ = 0.75). PCR-confirmed cases were more frequently identified by anti-RBD (129/130) than anti-NP (123/130). Anti-RBD testing identified 83 of 325 (25.5%) cases otherwise reported as negative for anti-NP. Anti-NP presence (+1.75/unit increase; p < 0.001), fever (≥ 38°C; +1.81; p < 0.001) or anosmia (+1.91; p < 0.001) were significantly associated with increased anti-RBD BR. Age (p = 0.85), sex (p = 0.28) and cough (p = 0.35) were not. When time since symptom onset was considered, we did not observe a significant change in anti-RBD BR (p = 0.95) but did note decreasing anti-NP BR (p < 0.001).ConclusionSARS-CoV-2 anti-RBD IgG showed significant correlation with anti-NP IgG for absolute seroconversion and BR. Higher BR were seen in symptomatic individuals, particularly those with fever. Inter-assay variability (12.5%) was evident and raises considerations for optimising seroprevalence testing strategies/studies.
Collapse
Affiliation(s)
- Scott Jc Pallett
- Centre of Defence Pathology, Royal Centre for Defence Medicine, Queen Elizabeth Hospital Birmingham, Birmingham, United Kingdom
- Chelsea and Westminster Hospital NHS Foundation Trust, London, United Kingdom
| | - Rachael Jones
- Chelsea and Westminster Hospital NHS Foundation Trust, London, United Kingdom
| | - Ahmed Abdulaal
- Chelsea and Westminster Hospital NHS Foundation Trust, London, United Kingdom
| | - Mitchell A Pallett
- Department of Infectious Disease, MRC Centre for Molecular Bacteriology and Infection, Imperial College London, United Kingdom
| | - Michael Rayment
- Chelsea and Westminster Hospital NHS Foundation Trust, London, United Kingdom
| | - Aatish Patel
- Chelsea and Westminster Hospital NHS Foundation Trust, London, United Kingdom
| | - Sarah J Denny
- Chelsea and Westminster Hospital NHS Foundation Trust, London, United Kingdom
| | - Nabeela Mughal
- Chelsea and Westminster Hospital NHS Foundation Trust, London, United Kingdom
- North West London Pathology, London, United Kingdom
| | - Maryam Khan
- Department of Infectious Disease, Faculty of Medicine, Imperial College London, St Mary's Campus, London, United Kingdom
| | - Carolina Rosadas de Oliveira
- Department of Infectious Disease, Faculty of Medicine, Imperial College London, St Mary's Campus, London, United Kingdom
| | | | - Paul Randell
- North West London Pathology, London, United Kingdom
| | - Christofer Toumazou
- Faculty of Engineering, Department of Electrical and Electronic Engineering, Imperial College London, South Kensington Campus, London, United Kingdom
| | - Matthew K O'Shea
- Centre of Defence Pathology, Royal Centre for Defence Medicine, Queen Elizabeth Hospital Birmingham, Birmingham, United Kingdom
- Institute of Immunology and Immunotherapy, College of Medical & Dental Sciences, University of Birmingham, Edgbaston, Birmingham, United Kingdom
| | - Richard Tedder
- Department of Infectious Disease, Faculty of Medicine, Imperial College London, St Mary's Campus, London, United Kingdom
| | - Myra O McClure
- Department of Infectious Disease, Faculty of Medicine, Imperial College London, St Mary's Campus, London, United Kingdom
| | - Gary W Davies
- Chelsea and Westminster Hospital NHS Foundation Trust, London, United Kingdom
| | - Luke Sp Moore
- Chelsea and Westminster Hospital NHS Foundation Trust, London, United Kingdom
- North West London Pathology, London, United Kingdom
- Imperial College London, NIHR Health Protection Research Unit in Healthcare Associated Infections and Antimicrobial Resistance, London, United Kingdom
| |
Collapse
|
43
|
Poulikakos D, Chinnadurai R, Mcgee Y, Gray S, Clough T, Clarke N, Murphy T, Wickens O, Mitchell C, Darby D, Paul J, Chadwick P, Sethi S, Sinha S, Kalra PA, Donne R. A Quality Improvement Project to Minimize COVID-19 Infections in Patients Receiving Haemodialysis and the Role of Routine Surveillance Using Nose and Throat Swabs for SARS-CoV-2 rRT-PCR and Serum Antibody Testing. Nephron Clin Pract 2021; 146:335-342. [PMID: 34937042 PMCID: PMC8805076 DOI: 10.1159/000520654] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 10/22/2021] [Indexed: 11/19/2022] Open
Abstract
Background Patients receiving in-centre haemodialysis (ICHD) are highly vulnerable to COVID-19. Objective We created a quality improvement (QI) project aimed to eliminate outbreaks of COVID-19 in haemodialysis units and evaluated the utility of surveillance rRT-PCR test and SARS-CoV-2 serum antibodies for prompt identification of patients infected with COVID-19. Methods A multifaceted QI programme including a bundle of infection prevention control (IPC) measures was implemented across 5 ICHD units following the first wave of the pandemic in June 2020. Primary outcomes evaluated before and after QI implementation were incidence of outbreaks and severe COVID-19 illness defined as COVID-19-related death or hospitalization. Secondary outcomes included the proportion of patients identified in the pre-symptomatic/asymptomatic phase on surveillance rRT-PCR screening and the incidence and longevity of SARS-CoV-2 antibody response. Results Following the implementation of the QI project, there were no further outbreaks. Pre- and post-implementation comparison showed a significant reduction in COVID-19-related mortality and hospitalization (26 vs. 13 events, respectively, p < 0.001). Surveillance rRT-PCR screening identified 39 asymptomatic or pre-symptomatic cases out of a total of 59 rRT-PCR-positive patients (39/59, 66%). SARS-CoV-2 antibody levels were detected in 72/74 (97%) rRT-PCR-positive patients. Amongst rRT-PCR-positive patients diagnosed before August 2020, 96% had detectable antibodies until January 2021 (days from the rRT-PCR test to last antibody testing, 245–280). Conclusions Systematic implementation of a bundle of IPC measures using QI methodology and surveillance rRT-PCR eliminated outbreaks in HD facilities. Most HD patients mount and sustain antibody response to COVID-19 for over 8 months.
Collapse
Affiliation(s)
- Dimitrios Poulikakos
- Renal Department, Salford Royal NHS Foundation Trust, The Northern Care Alliance NHS Group, Manchester, United Kingdom
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Rajkumar Chinnadurai
- Renal Department, Salford Royal NHS Foundation Trust, The Northern Care Alliance NHS Group, Manchester, United Kingdom
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
- *Rajkumar Chinnadurai,
| | - Yvonne Mcgee
- Renal Department, Salford Royal NHS Foundation Trust, The Northern Care Alliance NHS Group, Manchester, United Kingdom
| | - Simon Gray
- Renal Department, Salford Royal NHS Foundation Trust, The Northern Care Alliance NHS Group, Manchester, United Kingdom
| | - Toni Clough
- Renal Department, Salford Royal NHS Foundation Trust, The Northern Care Alliance NHS Group, Manchester, United Kingdom
| | - Nicola Clarke
- Renal Department, Salford Royal NHS Foundation Trust, The Northern Care Alliance NHS Group, Manchester, United Kingdom
| | - Tracey Murphy
- Renal Department, Salford Royal NHS Foundation Trust, The Northern Care Alliance NHS Group, Manchester, United Kingdom
| | - Olivia Wickens
- Renal Department, Salford Royal NHS Foundation Trust, The Northern Care Alliance NHS Group, Manchester, United Kingdom
| | - Carol Mitchell
- Infection Control Department, Salford Royal NHS Foundation Trust, The Northern Care Alliance NHS Group, Manchester, United Kingdom
| | - Denise Darby
- Chemical Pathology Department, Salford Royal NHS Foundation Trust, The Northern Care Alliance NHS Group, Manchester, United Kingdom
| | - Joel Paul
- Virology Department, The Northern Care Alliance NHS Group, Manchester, United Kingdom
| | - Paul Chadwick
- Microbiology Department, Salford Royal NHS Foundation Trust, The Northern Care Alliance NHS Group, Manchester, United Kingdom
| | - Su Sethi
- Public Health England, North West Region Specialised Commissioning Team, NHS, London, United Kingdom
| | - Smeeta Sinha
- Renal Department, Salford Royal NHS Foundation Trust, The Northern Care Alliance NHS Group, Manchester, United Kingdom
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Philip A. Kalra
- Renal Department, Salford Royal NHS Foundation Trust, The Northern Care Alliance NHS Group, Manchester, United Kingdom
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Rosie Donne
- Renal Department, Salford Royal NHS Foundation Trust, The Northern Care Alliance NHS Group, Manchester, United Kingdom
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
44
|
Affeldt P, Koehler FC, Brensing KA, Adam V, Burian J, Butt L, Gies M, Grundmann F, Hinrichs S, Johannis W, Kalisch N, Meyer-Delpho M, Oehm S, Platen E, Schöler C, Heger E, Steger G, Stippel D, Ziegelhöfer A, Benzing T, Klein F, Kurschat C, Müller RU, Di Cristanziano V. Immune Responses to SARS-CoV-2 Infection and Vaccination in Dialysis Patients and Kidney Transplant Recipients. Microorganisms 2021; 10:4. [PMID: 35056453 PMCID: PMC8779774 DOI: 10.3390/microorganisms10010004] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 12/12/2021] [Accepted: 12/15/2021] [Indexed: 12/24/2022] Open
Abstract
Dialysis patients and kidney transplant (KTX) recipients suffer from an impaired immune system and show a decreased response to the severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2) vaccination. We performed a retrospective analysis of 1505 serological SARS-CoV-2 measurements obtained from 887 dialysis patients and 86 KTX recipients. The results were separated by patient subgroups (dialysis/KTX) as well as SARS-CoV-2 status. The latter criterion included SARS-CoV-2-naïve patients with or without COVID-19 vaccination and convalescent patients receiving a booster shot. Serologies of 27 vaccinated healthy individuals served as the reference group. Vaccine-induced cellular immune response was quantified by an interferon-γ release assay in 32 KTX recipients. We determined seroconversion rates of 92.6%, 93.4%, and 71.4% in dialysis patients vaccinated with either BNT162b2, mRNA-1273, or AZD1222, respectively. Vaccination-induced anti-SARS-CoV-2 antibody titers were lower in dialysis patients compared to healthy individuals, and vaccination with mRNA-1273 induced higher titers than BNT162b2. The initial seroconversion rate was 39.5% in KTX recipients vaccinated with BNT162b2. A linear regression model identified medication with mycophenolate-mofetil/mycophenolic acid as an independent risk factor for missing seroconversion. Within a cohort of 32 KTX recipients, cellular and humoral immune reactivity to SARS-CoV-2 was detectable in three patients only. Conclusively, vaccine-induced seroconversion rates were similar in dialysis patients compared to healthy individuals but were strongly impaired in KTX recipients. Anti-SARS-CoV-2 IgG titers elicited by double active immunization were significantly lower in both cohorts compared to healthy individuals, and immune responses to vaccination vanished quickly.
Collapse
Affiliation(s)
- Patrick Affeldt
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; (P.A.); (F.C.K.); (V.A.); (L.B.); (F.G.); (S.H.); (N.K.); (S.O.); (T.B.); (C.K.)
| | - Felix Carlo Koehler
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; (P.A.); (F.C.K.); (V.A.); (L.B.); (F.G.); (S.H.); (N.K.); (S.O.); (T.B.); (C.K.)
- CECAD, Faculty of Medicine and University Hospital of Cologne, University of Cologne, 50931 Cologne, Germany
| | | | - Vivien Adam
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; (P.A.); (F.C.K.); (V.A.); (L.B.); (F.G.); (S.H.); (N.K.); (S.O.); (T.B.); (C.K.)
| | - Julia Burian
- Nieren- und Dialysezentrum Rheinbach, 53359 Rheinbach, Germany;
| | - Linus Butt
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; (P.A.); (F.C.K.); (V.A.); (L.B.); (F.G.); (S.H.); (N.K.); (S.O.); (T.B.); (C.K.)
- CECAD, Faculty of Medicine and University Hospital of Cologne, University of Cologne, 50931 Cologne, Germany
| | - Martin Gies
- KfH-Nierenzentrum Köln-Longerich, 50737 Cologne, Germany; (M.G.); (C.S.)
| | - Franziska Grundmann
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; (P.A.); (F.C.K.); (V.A.); (L.B.); (F.G.); (S.H.); (N.K.); (S.O.); (T.B.); (C.K.)
| | - Steffen Hinrichs
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; (P.A.); (F.C.K.); (V.A.); (L.B.); (F.G.); (S.H.); (N.K.); (S.O.); (T.B.); (C.K.)
| | - Wibke Johannis
- Institute for Clinical Chemistry, Faculty of Medicine and University Hospital of Cologne, University of Cologne, 50937 Cologne, Germany;
| | - Nils Kalisch
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; (P.A.); (F.C.K.); (V.A.); (L.B.); (F.G.); (S.H.); (N.K.); (S.O.); (T.B.); (C.K.)
| | | | - Simon Oehm
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; (P.A.); (F.C.K.); (V.A.); (L.B.); (F.G.); (S.H.); (N.K.); (S.O.); (T.B.); (C.K.)
| | - Eva Platen
- Nierenzentrum Eifel, 53894 Mechernich, Germany;
| | - Claudia Schöler
- KfH-Nierenzentrum Köln-Longerich, 50737 Cologne, Germany; (M.G.); (C.S.)
| | - Eva Heger
- Institute of Virology, Faculty of Medicine and University Hospital of Cologne, University of Cologne, 50935 Cologne, Germany; (E.H.); (G.S.); (A.Z.); (F.K.)
| | - Gertrud Steger
- Institute of Virology, Faculty of Medicine and University Hospital of Cologne, University of Cologne, 50935 Cologne, Germany; (E.H.); (G.S.); (A.Z.); (F.K.)
| | - Dirk Stippel
- Department of General, Visceral, Cancer and Transplant Surgery, Faculty of Medicine, University Hospital Cologne, University of Cologne, 50937 Cologne, Germany;
| | - Aileen Ziegelhöfer
- Institute of Virology, Faculty of Medicine and University Hospital of Cologne, University of Cologne, 50935 Cologne, Germany; (E.H.); (G.S.); (A.Z.); (F.K.)
| | - Thomas Benzing
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; (P.A.); (F.C.K.); (V.A.); (L.B.); (F.G.); (S.H.); (N.K.); (S.O.); (T.B.); (C.K.)
- CECAD, Faculty of Medicine and University Hospital of Cologne, University of Cologne, 50931 Cologne, Germany
| | - Florian Klein
- Institute of Virology, Faculty of Medicine and University Hospital of Cologne, University of Cologne, 50935 Cologne, Germany; (E.H.); (G.S.); (A.Z.); (F.K.)
| | - Christine Kurschat
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; (P.A.); (F.C.K.); (V.A.); (L.B.); (F.G.); (S.H.); (N.K.); (S.O.); (T.B.); (C.K.)
- CECAD, Faculty of Medicine and University Hospital of Cologne, University of Cologne, 50931 Cologne, Germany
| | - Roman-Ulrich Müller
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; (P.A.); (F.C.K.); (V.A.); (L.B.); (F.G.); (S.H.); (N.K.); (S.O.); (T.B.); (C.K.)
- CECAD, Faculty of Medicine and University Hospital of Cologne, University of Cologne, 50931 Cologne, Germany
| | - Veronica Di Cristanziano
- Institute of Virology, Faculty of Medicine and University Hospital of Cologne, University of Cologne, 50935 Cologne, Germany; (E.H.); (G.S.); (A.Z.); (F.K.)
| |
Collapse
|
45
|
Malipiero G, Moratto A, Infantino M, D'Agaro P, Piscianz E, Manfredi M, Grossi V, Benvenuti E, Bulgaresi M, Benucci M, Villalta D. Assessment of humoral and cellular immunity induced by the BNT162b2 SARS-CoV-2 vaccine in healthcare workers, elderly people, and immunosuppressed patients with autoimmune disease. Immunol Res 2021; 69:576-583. [PMID: 34417958 PMCID: PMC8379062 DOI: 10.1007/s12026-021-09226-z] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 08/11/2021] [Indexed: 01/01/2023]
Abstract
The development of vaccines to prevent SARS-CoV-2 infection has mainly relied on the induction of neutralizing antibodies (nAbs) to the Spike protein of SARS-CoV-2, but there is growing evidence that T cell immune response can contribute to protection as well. In this study, an anti-receptor binding domain (RBD) antibody assay and an INFγ-release assay (IGRA) were used to detect humoral and cellular responses to the Pfizer-BioNTech BNT162b2 vaccine in three separate cohorts of COVID-19-naïve patients: 108 healthcare workers (HCWs), 15 elderly people, and 5 autoimmune patients treated with immunosuppressive agents. After the second dose of vaccine, the mean values of anti-RBD antibodies (Abs) and INFγ were 123.33 U/mL (range 27.55-464) and 1513 mIU/mL (range 145-2500) in HCWs and 210.7 U/mL (range 3-500) and 1167 mIU/mL (range 83-2500) in elderly people. No correlations between age and immune status were observed. On the contrary, a weak but significant positive correlation was found between INFγ and anti-RBD Abs values (rho = 0.354, p = 0.003). As to the autoimmune cohort, anti-RBD Abs were not detected in the two patients with absent peripheral CD19+B cells, despite high INFγ levels being observed in all 5 patients after vaccination. Even though the clinical relevance of T cell response has not yet been established as a correlate of vaccine-induced protection, IGRA testing has showed optimal sensitivity and specificity to define vaccine responders, even in patients lacking a cognate antibody response to the vaccine.
Collapse
Affiliation(s)
- Giacomo Malipiero
- Immunology and Allergy Unit, Ospedale S-Maria degli Angeli, Pordenone, Italy
| | - Anna Moratto
- Immunology and Allergy Unit, Ospedale S-Maria degli Angeli, Pordenone, Italy
| | - Maria Infantino
- Immunology and Allergy Laboratory Unit, San Giovanni Di Dio Hospital, Florence, Italy
| | - Pierlanfranco D'Agaro
- Laboratory for Hygiene and Public Health, University Hospital of Trieste, Trieste, Italy
| | - Elisa Piscianz
- Laboratory for Hygiene and Public Health, University Hospital of Trieste, Trieste, Italy
| | - Mariangela Manfredi
- Immunology and Allergy Laboratory Unit, San Giovanni Di Dio Hospital, Florence, Italy
| | - Valentina Grossi
- Immunology and Allergy Laboratory Unit, San Giovanni Di Dio Hospital, Florence, Italy
| | - Enrico Benvenuti
- Geriatric Unit Firenze-Empoli, Santa Maria Annunziata Hospital, Florence, Italy
| | - Matteo Bulgaresi
- Geriatric Unit Firenze-Empoli, Santa Maria Annunziata Hospital, Florence, Italy
| | - Maurizio Benucci
- Rheumatology Unit, San Giovanni Di Dio Hospital, Florence, Italy
| | - Danilo Villalta
- Immunology and Allergy Unit, Ospedale S-Maria degli Angeli, Pordenone, Italy.
| |
Collapse
|
46
|
Bertrand D, Hamzaoui M, Drouot L, Lamulle J, Hanoy M, Edet S, Laurent C, Lebourg L, Etienne I, Lemoine M, Le Roy F, Nezam D, Mauger E, Boyer O, Guerrot D, Candon S. SARS-CoV-2-specific Humoral and Cellular Immunities in Kidney Transplant Recipients and Dialyzed Patients Recovered From Severe and Nonsevere COVID-19. Transplant Direct 2021; 7:e792. [PMID: 34805494 PMCID: PMC8601300 DOI: 10.1097/txd.0000000000001230] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 07/16/2021] [Accepted: 07/17/2021] [Indexed: 12/23/2022] Open
Abstract
Kidney transplantation and dialysis are two major risk factors for severe forms of coronavirus disease 2019 (COVID-19). The dynamics of the immune response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in this population remain largely unknown. Methods We report here the analysis of anti-SARS-CoV-2 antibody- and T cell-mediated immune responses in 26 kidney transplant recipients (KTRs) and 11 dialyzed patients (DPs) who recovered from COVID-19. Results After a mean time of 83 ± 26 d post-symptom onset for KTRs and 97 ± 31 d for DPs, 20 KTRs (76.9%) and 10 DPs (90.9%) displayed anti-S1 immunoglobulin G SARS-CoV-2 antibodies (P = 0.34), at similar titers in both groups. SARS-CoV-2-specific interferon-γ-producing T cells were evidenced in 26 KTRs (100%) and 10 DPs (90.9%). Total numbers of SARS-CoV-2-reactive T cells were high and not statistically different between the 2 groups. No correlation between the severity of the disease and the number of reactive T cells was found in KTRs. In 5 KTRs, also evaluated 10 mo after COVID-19, weak or absent antibody response was observed, whereas specific memory T-cell response was detected in all cases. Conclusion T-cell response persisted up to 3 mo post-symptom onset, even in KTRs in whom full immunosuppressive regimen was reinstated at recovery, and seems to be present up to 10 mo after infection. Our findings have implications in the understanding of the natural course of the disease in transplant patients and DPs.
Collapse
Affiliation(s)
- Dominique Bertrand
- Department of Nephrology, Transplantation and Hemodialysis, Rouen University Hospital, Rouen, France
| | - Mouad Hamzaoui
- Department of Nephrology, Transplantation and Hemodialysis, Rouen University Hospital, Rouen, France
| | - Laurent Drouot
- Normandy University, UNIROUEN, INSERM U1234, Rouen, France
| | - Julie Lamulle
- Normandy University, UNIROUEN, INSERM U1234, Rouen, France
| | - Mélanie Hanoy
- Department of Nephrology, Transplantation and Hemodialysis, Rouen University Hospital, Rouen, France
| | - Stéphane Edet
- Department of Nephrology, Transplantation and Hemodialysis, Rouen University Hospital, Rouen, France.,ANIDER, Rouen, France
| | - Charlotte Laurent
- Department of Nephrology, Transplantation and Hemodialysis, Rouen University Hospital, Rouen, France
| | - Ludivine Lebourg
- Department of Nephrology, Transplantation and Hemodialysis, Rouen University Hospital, Rouen, France
| | - Isabelle Etienne
- Department of Nephrology, Transplantation and Hemodialysis, Rouen University Hospital, Rouen, France
| | - Mathilde Lemoine
- Department of Nephrology, Transplantation and Hemodialysis, Rouen University Hospital, Rouen, France
| | - Frank Le Roy
- Department of Nephrology, Transplantation and Hemodialysis, Rouen University Hospital, Rouen, France
| | - Dorian Nezam
- Department of Nephrology, Transplantation and Hemodialysis, Rouen University Hospital, Rouen, France
| | - Eleusis Mauger
- Department of Nephrology, Transplantation and Hemodialysis, Rouen University Hospital, Rouen, France
| | - Olivier Boyer
- Normandy University, UNIROUEN, INSERM U1234, Rouen, France.,Department of Immunology and Biotherapies, Rouen University Hospital, Rouen, France
| | - Dominique Guerrot
- Department of Nephrology, Transplantation and Hemodialysis, Rouen University Hospital, Rouen, France
| | - Sophie Candon
- Normandy University, UNIROUEN, INSERM U1234, Rouen, France.,Department of Immunology and Biotherapies, Rouen University Hospital, Rouen, France
| |
Collapse
|
47
|
Pallett SJC, Wake R, Youngs J, Pope C, Tan NK, Taylor J, Hawkins L, Witney AA, Laing KG, Monahan IM, Akay M, Cox A, Groppelli E, Kelleher P, Miller P, Bicanic T. Adjunctive viral cell culture supports treatment decision-making in patients with secondary humoral immunodeficiency and persistent SARS-CoV-2 infection. Br J Haematol 2021; 196:1170-1174. [PMID: 34766331 PMCID: PMC8652771 DOI: 10.1111/bjh.17957] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 10/30/2021] [Indexed: 12/26/2022]
Affiliation(s)
- Scott J C Pallett
- Infection Care Group, St George's University Hospitals NHS Foundation Trust, London, UK.,Centre of Defence Pathology, Royal Centre for Defence Medicine, Edgbaston, Birmingham, UK
| | - Rachel Wake
- Infection Care Group, St George's University Hospitals NHS Foundation Trust, London, UK.,Institute for Infection and Immunity, St George's University of London, Cranmer Terrace, London, UK
| | - Jonathan Youngs
- Infection Care Group, St George's University Hospitals NHS Foundation Trust, London, UK
| | - Cassie Pope
- Infection Care Group, St George's University Hospitals NHS Foundation Trust, London, UK.,Institute for Infection and Immunity, St George's University of London, Cranmer Terrace, London, UK
| | - Ngee Keong Tan
- South West London Pathology, Jenner Wing, St. George's Hospital, London, UK
| | - Joshua Taylor
- South West London Pathology, Jenner Wing, St. George's Hospital, London, UK
| | - Lois Hawkins
- Infection Care Group, St George's University Hospitals NHS Foundation Trust, London, UK
| | - Adam A Witney
- Institute for Infection and Immunity, St George's University of London, Cranmer Terrace, London, UK
| | - Kenneth G Laing
- Institute for Infection and Immunity, St George's University of London, Cranmer Terrace, London, UK
| | - Irene M Monahan
- Institute for Infection and Immunity, St George's University of London, Cranmer Terrace, London, UK
| | - Melek Akay
- Department of Haematology, St George's University Hospitals NHS Foundation Trust, London, UK
| | - Alison Cox
- Centre for Immunology and Vaccinology, Department of Medicine, Imperial College London, London, UK
| | - Elisabetta Groppelli
- Institute for Infection and Immunity, St George's University of London, Cranmer Terrace, London, UK
| | - Peter Kelleher
- Centre for Immunology and Vaccinology, Department of Medicine, Imperial College London, London, UK
| | - Paul Miller
- Department of Haematology, St George's University Hospitals NHS Foundation Trust, London, UK
| | - Tihana Bicanic
- Infection Care Group, St George's University Hospitals NHS Foundation Trust, London, UK.,Institute for Infection and Immunity, St George's University of London, Cranmer Terrace, London, UK
| |
Collapse
|
48
|
De Vriese AS, Van Praet J, Reynders M, Heylen L, Viaene L, Caluwé R, Schoutteten M, De Bacquer D. Longevity and correlation with disease severity of the humoral and cellular response to SARS-CoV-2 infection in haemodialysis patients. Clin Kidney J 2021; 14:2446-2448. [PMID: 34754442 PMCID: PMC8573012 DOI: 10.1093/ckj/sfab147] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 08/10/2021] [Indexed: 12/31/2022] Open
Affiliation(s)
- An S De Vriese
- Division of Nephrology and Infectious Diseases, AZ Sint-Jan Brugge-Oostende AV, Brugge, Belgium
| | - Jens Van Praet
- Division of Nephrology and Infectious Diseases, AZ Sint-Jan Brugge-Oostende AV, Brugge, Belgium
| | - Marijke Reynders
- Department of Internal Medicine, Ghent University, Ghent, Belgium
| | - Line Heylen
- Division of Nephrology, Ziekenhuis Oost-Limburg, Genk, Belgium
| | | | - Rogier Caluwé
- Division of Nephrology, OLV Hospital, Aalst, Belgium
| | | | - Dirk De Bacquer
- Department of Public Health and Primary Care, Ghent University, Ghent, Belgium
| |
Collapse
|
49
|
Kamińska D, Augustyniak-Bartosik H, Kościelska-Kasprzak K, Żabińska M, Bartoszek D, Poznański P, Kuriata-Kordek M, Kusztal M, Mazanowska O, Krajewska M. Comparing Humoral and Cellular Adaptive Immunity during Convalescent Phase of COVID-19 in Hemodialysis Patients and Kidney Transplant Recipients. J Clin Med 2021; 10:jcm10214833. [PMID: 34768356 PMCID: PMC8585082 DOI: 10.3390/jcm10214833] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 10/12/2021] [Accepted: 10/19/2021] [Indexed: 12/13/2022] Open
Abstract
Background. It is still unclear whether COVID-19 convalescent kidney transplant recipients (KTR) and hemodialysis (HD) patients can develop anti-SARS-CoV-2 adaptive immunity. The aim was to characterize and compare the immune response to the virus in HD patients and KTR. Methods. The study included 26 HD patients and 54 KTR—both convalescent (14 HD, 25 KTR) and unexposed. The immune response was assessed by determining the anti-SARS-CoV-2 antibodies in serum and specific T cell response via the interferon-gamma release assay (IGRA). Moreover, blood-morphology-derived parameters, immune cell phenotypes, and acute phase reactants were evaluated. Results. KRT and HD convalescents presented similar serum levels of anti-SARS-CoV-2 IgG and IgA. A negative correlation occurred between IgG and time after the infection was observed. There was a strong relationship between the prevalence of anti-SARS-CoV-2 cellular and humoral responses in both groups. Convalescent IGRA response was significantly higher in HD patients compared to KTR. Conclusions. HD patients and KTR develop humoral and cellular responses after COVID-19. The antibodies levels are similar in both groups of patients. SARS-CoV-2-reactive T cell response is stronger in HD patients compared to KTR. The SARS-CoV-2-specific IgG level decreases with time while IgA and a cellular response are maintained. IGRA proved to be a valuable test for the assessment of specific cellular immunity in immunocompromised HD patients and KTR.
Collapse
|
50
|
|