Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Front Immunol ; 15: 1296273, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38455062

RESUMEN

Background: SARS-CoV-2 vaccination in rheumatoid arthritis (RA) patients treated with B cell-depleting drugs induced limited seroconversion but robust cellular response. We aimed to document specific T and B cell immunity in response to vaccine booster doses and breakthrough infection (BTI). Methods: We included 76 RA patients treated with rituximab who received up to four SARS-CoV-2 vaccine doses or three doses plus BTI, in addition to vaccinated healthy donors (HD) and control patients treated with tumor necrosis factor inhibitor (TNFi). We quantified anti-SARS-CoV-2 receptor-binding domain (RBD) Spike IgG, anti-nucleocapsid (NC) IgG, 92 circulating inflammatory proteins, Spike-binding B cells, and Spike-specific T cells along with comprehensive high-dimensional phenotyping and functional assays. Findings: The time since the last rituximab infusion, persistent inflammation, and age were associated with the anti-SARS-CoV-2 RBD IgG seroconversion. The vaccine-elicited serological response was accompanied by an incomplete induction of peripheral Spike-specific memory B cells but occurred independently of T cell responses. Vaccine- and BTI-elicited cellular immunity was similar between RA and HD ex vivo in terms of frequency or phenotype of Spike-specific cytotoxic T cells and in vitro in terms of the functionality and differentiation profile of Spike-specific T cells. Interpretation: SARS-CoV-2 vaccination in RA can induce persistent effector T-cell responses that are reactivated by BTI. Paused rituximab medication allowed serological responses after a booster dose (D4), especially in RA with lower inflammation, enabling efficient humoral and cellular immunity after BTI, and contributed overall to the development of potential durable immunity.


Asunto(s)
Artritis Reumatoide , COVID-19 , Humanos , Rituximab/uso terapéutico , Vacunas contra la COVID-19 , SARS-CoV-2 , Infección Irruptiva , Estudios Prospectivos , Artritis Reumatoide/tratamiento farmacológico , Vacunación , Inflamación , Anticuerpos Antivirales , Inmunoglobulina G
2.
EBioMedicine ; 97: 104833, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37844534

RESUMEN

BACKGROUND: Kidney transplant recipients (KTR) are at high risk for severe COVID-19 and have demonstrated poor response to vaccination, making it unclear whether successive vaccinations offer immunity and protection. METHODS: We conducted a serologically guided interventional study where KTR patients that failed to seroconvert were revaccinated and also monitored seroconversion of KTR following the Norwegian vaccination program. We analysed IgG anti-RBD Spike responses from dose 2 (n = 432) up to after the 6th (n = 37) mRNA vaccine dose. The frequency and phenotype of Spike-specific T and B cell responses were assessed in the interventional cohort after 3-4 vaccine doses (n = 30). Additionally, we evaluated the Specific T and B cell response to breakthrough infection (n = 32), measured inflammatory cytokines and broadly cross-neutralizing antibodies, and defined the incidence of COVID-19-related hospitalizations and deaths. The Norwegian KTR cohort has a male dominance (2323 males, 1297 females), PBMC were collected from 114 male and 78 female donors. FINDINGS: After vaccine dose 3, most KTR developed Spike-specific T cell responses but had significantly reduced Spike-binding B cells and few memory cells. The B cell response included a cross-reactive subset that could bind Omicron VOC, which expanded after breakthrough infection (BTI) and gave rise to a memory IgG+ B cell response. After BTI, KTR had increased Spike-specific T cells, emergent non-Spike T and B cell responses, and a systemic inflammatory signature. Late seroconversion occurred after doses 5-6, but 38% (14/37) of KTR had no detectable immunity even after multiple vaccine doses. INTERPRETATION: Boosting vaccination can induce Spike-specific immunity that may expand in breakthrough infections highlighting the benefit of vaccination to protect this vulnerable population. FUNDING: CEPI and internal funds.


Asunto(s)
COVID-19 , Trasplante de Riñón , Humanos , Femenino , Masculino , Vacunas contra la COVID-19 , SARS-CoV-2 , COVID-19/prevención & control , Trasplante de Riñón/efectos adversos , Leucocitos Mononucleares , Infección Irruptiva , Inmunoglobulina G , Anticuerpos Antivirales , Receptores de Trasplantes , Vacunación
3.
Tidsskr Nor Laegeforen ; 143(11)2023 08 15.
Artículo en Inglés, Noruego | MEDLINE | ID: mdl-37589359

RESUMEN

BACKGROUND: In Norway, treatment with COVID-19 convalescent plasma has been given through the NORPLASMA project. The treatment was initially offered to critically ill patients after an individual assessment, but from December 2020, the indication was limited to critically ill, immunocompromised patients. In this article we describe clinical characteristics, comorbidity and mortality in patients who received convalescent plasma in these two periods. MATERIAL AND METHOD: From 22 April 2020 to 30 March 2022, a total of 79 patients were included in the observational studies NORPLASMA MONITOR and the Norwegian SARS-CoV-2 study. The patients had received a total of 193 units of convalescent plasma at 15 Norwegian hospitals/nursing homes; 62 in South-Eastern Norway Regional Health Authority, 8 in Western Norway Regional Health Authority and 9 in Central Norway Regional Health Authority. Information on immune status, comorbidity and course of infection was retrieved from the patient records after informed written consent was obtained. RESULTS: Of 79 patients with a median age of 65 years (interquartile range 51-⁠73) who were treated with convalescent plasma, 31 (39 %) died during hospitalisation. A total of 59 patients were immunocompromised, and of these, 20 died in hospital compared to 11 of 20 who were assumed to be immunocompetent. Median number of comorbidities was 2 (interquartile range 1-4). The patients received a median of two plasma units (min.-max. 1-21). Two of the patients developed mild allergic skin reactions. INTERPRETATION: Convalescent plasma was well tolerated by patients with COVID-19. Immunocompromised patients may have benefitted from the treatment, with lower mortality than for those assumed to be immunocompetent.


Asunto(s)
COVID-19 , Dermatitis Atópica , Anciano , Humanos , COVID-19/terapia , Sueroterapia para COVID-19 , Enfermedad Crítica/terapia , SARS-CoV-2 , Persona de Mediana Edad
4.
Tidsskr Nor Laegeforen ; 143(11)2023 08 15.
Artículo en Inglés, Noruego | MEDLINE | ID: mdl-37589362

RESUMEN

BACKGROUND: At the start of the pandemic, the Norwegian Directorate of Health and Norwegian blood banks initiated the production of COVID-19 convalescent plasma within the framework of clinical studies. In this article we describe the blood donors who participated. MATERIAL AND METHOD: Blood donors who had recovered from COVID-19 were recruited to donate single donor plasma for the purpose of patient treatment. Data on the course of infection, leukocyte antibodies and antibody level against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) per plasma unit were registered after informed consent was obtained. We calculated a disease score defined as the total number of self-reported symptoms/findings and hospitalisation where relevant (score 0-⁠11). RESULTS: A total of 1644 plasma units were collected from 266 plasma donors at 12 blood banks. Median disease score was 5 (interquartile range 3-⁠6), and 15 donors had recovered from pneumonia and/or been hospitalised. A total of 599/1644 plasma units from 106/266 donors met our requirement for SARS-CoV-2 antibody content (> 60 % inhibition of virus binding to angiotensin-converting enzyme 2 (ACE2)) or positive virus neutralisation test. The antibody level in donors waned over time following infection, and showed no clear correlation with disease score. INTERPRETATION: The number of symptoms and findings in blood donors could not predict antibody response at individual level, and antibody testing was crucial for the production of effective convalescent plasma.


Asunto(s)
Donantes de Sangre , COVID-19 , Humanos , COVID-19/terapia , SARS-CoV-2 , Sueroterapia para COVID-19 , Anticuerpos Antivirales
5.
Front Immunol ; 14: 1210899, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37503339

RESUMEN

Poor overall survival of hematopoietic stem cell transplantation (HSCT) recipients who developed COVID-19 underlies the importance of SARS-CoV-2 vaccination. Previous studies of vaccine efficacy have reported weak humoral responses but conflicting results on T cell immunity. Here, we have examined the relationship between humoral and T cell response in 48 HSCT recipients who received two doses of Moderna's mRNA-1273 or Pfizer/BioNTech's BNT162b2 vaccines. Nearly all HSCT patients had robust T cell immunity regardless of protective humoral responses, with 18/48 (37%, IQR 8.679-5601 BAU/mL) displaying protective IgG anti-receptor binding domain (RBD) levels (>2000 BAU/mL). Flow cytometry analysis of activation induced markers (AIMs) revealed that 90% and 74% of HSCT patients showed reactivity towards immunodominant spike peptides in CD8+ and CD4+ T cells, respectively. The response rate increased to 90% for CD4+ T cells as well when we challenged the cells with a complete set of overlapping peptides spanning the entire spike protein. T cell response was detectable as early as 3 months after transplant, but only CD4+ T cell reactivity correlated with IgG anti-RBD level and time after transplantation. Boosting increased seroconversion rate, while only one patient developed COVID-19 requiring hospitalization. Our data suggest that HSCT recipients with poor serological responses were protected from severe COVID-19 by vaccine-induced T cell responses.


Asunto(s)
Vacunas contra la COVID-19 , COVID-19 , Trasplante de Células Madre Hematopoyéticas , Humanos , Vacuna BNT162 , Linfocitos T CD4-Positivos , Linfocitos T CD8-positivos , Estudios de Cohortes , Vacunas contra la COVID-19/inmunología , Inmunoglobulina G , Estudios Prospectivos , SARS-CoV-2
8.
J Neurol Neurosurg Psychiatry ; 94(1): 19-22, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-34670844

RESUMEN

INTRODUCTION: The effect of disease-modifying therapies (DMT) on vaccine responses is largely unknown. Understanding the development of protective immunity is of paramount importance to fight the COVID-19 pandemic. OBJECTIVE: To characterise humoral immunity after mRNA-COVID-19 vaccination of people with multiple sclerosis (pwMS). METHODS: All pwMS in Norway fully vaccinated against SARS-CoV-2 were invited to a national screening study. Humoral immunity was assessed by measuring anti-SARS-CoV-2 SPIKE RBD IgG response 3-12 weeks after full vaccination, and compared with healthy subjects. RESULTS: 528 pwMS and 627 healthy subjects were included. Reduced humoral immunity (anti-SARS-CoV-2 IgG <70 arbitrary units) was present in 82% and 80% of all pwMS treated with fingolimod and rituximab, respectively, while patients treated with other DMT showed similar rates as healthy subjects and untreated pwMS. We found a significant correlation between time since the last rituximab dose and the development of humoral immunity. Revaccination in two seronegative patients induced a weak antibody response. CONCLUSIONS: Patients treated with fingolimod or rituximab should be informed about the risk of reduced humoral immunity and vaccinations should be timed carefully in rituximab patients. Our results identify the need for studies regarding the durability of vaccine responses, the role of cellular immunity and revaccinations.


Asunto(s)
COVID-19 , Esclerosis Múltiple , Humanos , Inmunización Secundaria , Inmunidad Humoral , Rituximab/uso terapéutico , Esclerosis Múltiple/tratamiento farmacológico , Clorhidrato de Fingolimod/uso terapéutico , Vacunas contra la COVID-19/uso terapéutico , Pandemias , SARS-CoV-2 , COVID-19/prevención & control , Vacunación , Anticuerpos Antivirales , Inmunoglobulina G , ARN Mensajero
9.
NPJ Vaccines ; 7(1): 174, 2022 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-36585405

RESUMEN

Diagnostic assays currently used to monitor the efficacy of COVID-19 vaccines measure levels of antibodies to the receptor-binding domain of ancestral SARS-CoV-2 (RBDwt). However, the predictive value for protection against new variants of concern (VOCs) has not been firmly established. Here, we used bead-based arrays and flow cytometry to measure binding of antibodies to spike proteins and receptor-binding domains (RBDs) from VOCs in 12,000 serum samples. Effects of sera on RBD-ACE2 interactions were measured as a proxy for neutralizing antibodies. The samples were obtained from healthy individuals or patients on immunosuppressive therapy who had received two to four doses of COVID-19 vaccines and from COVID-19 convalescents. The results show that anti-RBDwt titers correlate with the levels of binding- and neutralizing antibodies against the Alpha, Beta, Gamma, Delta, Epsilon and Omicron variants. The benefit of multiplexed analysis lies in the ability to measure a wide range of anti-RBD titers using a single dilution of serum for each assay. The reactivity patterns also yield an internal reference for neutralizing activity and binding antibody units per milliliter (BAU/ml). Results obtained with sera from vaccinated healthy individuals and patients confirmed and extended results from previous studies on time-dependent waning of antibody levels and effects of immunosuppressive agents. We conclude that anti-RBDwt titers correlate with levels of neutralizing antibodies against VOCs and propose that our method may be implemented to enhance the precision and throughput of immunomonitoring.

10.
RMD Open ; 8(2)2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36328399

RESUMEN

OBJECTIVES: Humoral vaccine responses to SARS-CoV-2 vaccines are impaired and short lasting in patients with immune-mediated inflammatory diseases (IMID) following two vaccine doses. To protect these vulnerable patients against severe COVID-19 disease, a three-dose primary vaccination strategy has been implemented in many countries. The aim of this study was to evaluate humoral response and safety of primary vaccination with three doses in patients with IMID. METHODS: Patients with IMID on immunosuppressive therapy and healthy controls receiving three-dose and two-dose primary SARS-CoV-2 vaccination, respectively, were included in this prospective observational cohort study. Anti-Spike antibodies were assessed 2-4 weeks, and 12 weeks following each dose. The main outcome was anti-Spike antibody levels 2-4 weeks following three doses in patients with IMID and two doses in controls. Additional outcomes were the antibody decline rate and adverse events. RESULTS: 1100 patients and 303 controls were included. Following three-dose vaccination, patients achieved median (IQR) antibody levels of 5720 BAU/mL (2138-8732) compared with 4495 (1591-6639) in controls receiving two doses, p=0.27. Anti-Spike antibody levels increased with median 1932 BAU/mL (IQR 150-4978) after the third dose. The interval between the vaccine doses and vaccination with mRNA-1273 or a combination of vaccines were associated with antibody levels following the third dose. Antibody levels had a slower decline-rate following the third than the second vaccine dose, p<0.001. Adverse events were reported by 464 (47%) patients and by 196 (78%) controls. Disease flares were reported by 70 (7%) patients. CONCLUSIONS: This study shows that additional vaccine doses to patients with IMID contribute to strong and sustained immune-responses comparable to healthy persons vaccinated twice, and supports repeated vaccination of patients with IMID. TRIAL REGISTRATION NUMBER: NCT04798625.


Asunto(s)
Vacunas contra la COVID-19 , COVID-19 , Humanos , COVID-19/prevención & control , Vacunas contra la COVID-19/efectos adversos , Terapia de Inmunosupresión , Estudios Prospectivos , SARS-CoV-2 , Vacunación , Vacunas Virales/efectos adversos
11.
BMC Med ; 20(1): 378, 2022 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-36199139

RESUMEN

BACKGROUND: The durability of vaccine-induced humoral immunity against SARS-CoV-2 in patients with immune-mediated inflammatory diseases (IMIDs) on immunosuppressive therapy is not known. The aim of this study was to compare the persistence of anti-Spike antibodies following two-dose SARS-CoV-2 vaccination between IMID patients and healthy controls and to identify factors associated with antibody decline. METHODS: IMID patients on immunosuppressive medication enrolled in the prospective observational Nor-vaC study were included. Participants received two-dose SARS-CoV-2 vaccination. Serum collected at two time points following vaccination (first assessment within 6-48 days, second within 49-123 days) were analyzed for antibodies binding the receptor-binding domain (RBD) of the SARS-CoV-2 Spike protein. Multivariable regression models estimated percent reduction in anti-RBD over 30 days and factors associated with reduction. RESULTS: A total of 1108 patients (403 rheumatoid arthritis, 195 psoriatic arthritis, 195 spondyloarthritis, 124 ulcerative colitis, 191 Crohn's disease) and 134 controls provided blood samples within the defined intervals (median 19 days [IQR 15-24] and 97 days [87-105] after second vaccine dose). Antibody levels were lower in patients compared to controls at both time points, with median anti-RBD 2806 BAU/ml [IQR 1018-6068] in patients and 6187 BAU/ml [4105-7496] in controls (p<0.001) at first assessment, and 608 BAU/ml [IQR 58-1053] in patients and 1520 BAU/ml [979-3766] in controls (p<0.001) at second assessment. At second assessment, low anti-RBD antibody levels (defined as <200 BAU/ml) were found in 449 (41%) patients, and 6 (5%) controls (p<0.001). The change was - 83% in patients and - 66% in controls (p<0.001). Patients had a greater estimated 30 days percent reduction in anti-RBD levels compared to controls - 4.9 (95% CI - 7.4 to - 2.4), (p<0.05). Among therapies, mono- or combination treatment with tumor necrosis factor inhibitors was associated with the greatest decline. CONCLUSIONS: Within 4 months after vaccination, antibody levels declined considerably in both IMID patients and controls. Patients had lower initial antibody levels and a more pronounced decline compared to healthy controls and were therefore more likely to decline to low antibody levels. These results support that IMID patients need additional vaccine doses at an earlier stage than healthy individuals.


Asunto(s)
COVID-19 , Vacunas , Anticuerpos Antivirales , COVID-19/prevención & control , Vacunas contra la COVID-19 , Humanos , Terapia de Inmunosupresión , Estudios Prospectivos , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus , Inhibidores del Factor de Necrosis Tumoral , Vacunación
12.
Front Immunol ; 13: 964525, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36159859

RESUMEN

Background: Results showing that sera from double vaccinated individuals have minimal neutralizing activity against Omicron have been interpreted as indicating the need for a third vaccine dose for protection. However, there is little information about early immune responses to Omicron infection in double vaccinated individuals. Methods: We measured inflammatory mediators, antibodies to the SARS-CoV-2 spike and nucleocapsid proteins, and spike peptide-induced release of interferon gamma in whole blood in 51 double-vaccinated individuals infected with Omicron, in 14 infected with Delta, and in 18 healthy controls. The median time points for the first and second samples were 7 and 14 days after symptom onset, respectively. Findings: Infection with Omicron or Delta led to a rapid and similar increase in antibodies to the receptor-binding domain (RBD) of Omicron protein and spike peptide-induced interferon gamma in whole blood. Both the Omicron- and the Delta-infected patients had a mild and transient increase in inflammatory parameters. Interpretation: The results suggest that two vaccine doses are sufficient to mount a rapid and potent immune response upon infection in healthy individuals of with the Omicron variant. Funding: The study was funded by the Oslo University Hospital, and by grants from The Coalition for Epidemic Preparedness Innovations, Research Council of Norway (no 312780, 324272), South-Eastern Norway Regional Health Authority (no 2019067, 2021071, 10357, 2021047, 33612, 2021087, 2017092), EU Horizon 2020 grant no 848099, a philantropic donation from Vivaldi Invest A/S, and The European Virus Archive Global.


Asunto(s)
COVID-19 , Vacunas Virales , Anticuerpos Antivirales , COVID-19/prevención & control , Humanos , Mediadores de Inflamación , Interferón gamma , Proteínas de la Nucleocápside , SARS-CoV-2
13.
Nat Commun ; 13(1): 4165, 2022 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-35851055

RESUMEN

The SARS-CoV-2 Omicron variant has more than 15 mutations in the receptor binding domain of the Spike protein enabling increased transmissibility and viral escape from antibodies in vaccinated individuals. It is unclear how vaccine immunity protects against Omicron infection. Here we show that vaccinated participants at a super-spreader event have robust recall response of humoral and pre-existing cellular immunity induced by the vaccines, and an emergent de novo T cell response to non-Spike antigens. Individuals with Omicron SARS-CoV-2 breakthrough infections have significantly increased activated SARS-CoV-2 wild type Spike-specific cytotoxic T cells, activated follicular helper (TFH) cells, functional T cell responses, boosted humoral responses, and rapid release of Spike and RBD-specific IgG+ B cell plasmablasts and memory B cells into circulation. Omicron breakthrough infection affords significantly increased de novo memory T cell responses to non-Spike viral antigens. Concerted T and B cell responses may provide durable and broad immunity.


Asunto(s)
COVID-19 , Vacunas Virales , Adulto , Anticuerpos Antivirales , Humanos , Inmunidad , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus , Proteínas del Envoltorio Viral
14.
Tidsskr Nor Laegeforen ; 142(9)2022 06 14.
Artículo en Inglés, Noruego | MEDLINE | ID: mdl-35699539

RESUMEN

The collection and use of convalescent plasma to treat COVID-19 has taught us important lessons about the organisation, testing and selection of blood donors and patients. This is knowledge that can be used in the next pandemic.


Asunto(s)
Donantes de Sangre , COVID-19 , Anticuerpos Antivirales , COVID-19/terapia , Humanos , Inmunización Pasiva , SARS-CoV-2 , Sueroterapia para COVID-19
15.
Influenza Other Respir Viruses ; 16(6): 1004-1013, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35770841

RESUMEN

BACKGROUND: One year into the COVID-19 pandemic, the cumulative number of confirmed COVID-19 cases in Norway was still low. In January 2021, when the Norwegian COVID-19 vaccination campaign started, the national seroprevalence estimate of SARS-CoV-2 antibodies was 3.2%. We have conducted a nationwide cross-sectional study in August 2021 to investigate the overall prevalence of SARS-CoV-2 antibodies in Norway after 8 months of COVID-19 mass vaccination and a third wave of SARS-CoV-2 infection. METHODS: Residual sera were collected from laboratories across Norway in August 2021. In IgG antibodies against the spike protein, the spike receptor binding domain (RBD) and the nucleocapsid protein of SARS-CoV-2 were measured by a bead-based flow cytometric assay. RESULTS: In total, 1926 residual sera were collected from individuals aged 0-98 years; 55.1% were from women. The overall national estimated seroprevalence from vaccination and/or infection was 62.6% (credible interval [CrI] 60.1%-65.2%) based on having antibodies against both spike and RBD. Estimated seroprevalence increased with age. Among all samples, 11.7% had antibodies against nucleocapsid. For unvaccinated children <12 years, the seroprevalence estimate due to SARS-CoV-2 infection was 12.5% (95% CrI 9.3%-16.1%). Of seropositive samples from the unvaccinated children, 31.9% lacked anti-nucleocapsid antibodies. CONCLUSIONS: The high overall SARS-CoV-2 seroprevalence estimates are in line with Norwegian registry data. Vaccination, not infection, contributed the most to the high seroprevalence in August 2021. Lack of antibodies against nucleocapsid should not automatically be interpreted as absence of previous infection as this could lead to underestimation of COVID-19 cases in seroprevalence studies.


Asunto(s)
COVID-19 , SARS-CoV-2 , Anticuerpos Antivirales , COVID-19/epidemiología , Vacunas contra la COVID-19 , Niño , Estudios Transversales , Femenino , Humanos , Inmunoglobulina G , Proteínas de la Nucleocápside , Pandemias , Prevalencia , Estudios Seroepidemiológicos , Glicoproteína de la Espiga del Coronavirus
17.
Br J Haematol ; 197(6): 697-708, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35254660

RESUMEN

B-cell depletion induced by anti-cluster of differentiation 20 (CD20) monoclonal antibody (mAb) therapy of patients with lymphoma is expected to impair humoral responses to severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) vaccination, but effects on CD8 T-cell responses are unknown. Here, we investigated humoral and CD8 T-cell responses following two vaccinations in patients with lymphoma undergoing anti-CD20-mAb therapy as single agent or in combination with chemotherapy or other anti-neoplastic agents during the last 9 months prior to inclusion, and in healthy age-matched blood donors. Antibody measurements showed that seven of 110 patients had antibodies to the receptor-binding domain of the SARS-CoV-2 Spike protein 3-6 weeks after the second dose of vaccination. Peripheral blood CD8 T-cell responses against prevalent human leucocyte antigen (HLA) class I SARS-CoV-2 epitopes were determined by peptide-HLA multimer analysis. Strong CD8 T-cell responses were observed in samples from 20/29 patients (69%) and 12/16 (75%) controls, with similar median response magnitudes in the groups and some of the strongest responses observed in patients. We conclude that despite the absence of humoral immune responses in fully SARS-CoV-2-vaccinated, anti-CD20-treated patients with lymphoma, their CD8 T-cell responses reach similar frequencies and magnitudes as for controls. Patients with lymphoma on B-cell depleting therapies are thus likely to benefit from current coronavirus disease 2019 (COVID-19) vaccines, and development of vaccines aimed at eliciting T-cell responses to non-Spike epitopes might provide improved protection.


Asunto(s)
Linfocitos T CD8-positivos , Vacunas contra la COVID-19 , COVID-19 , Linfoma , Rituximab , Anticuerpos Antivirales , Linfocitos T CD8-positivos/inmunología , COVID-19/prevención & control , Vacunas contra la COVID-19/inmunología , Epítopos , Humanos , Linfoma/tratamiento farmacológico , Rituximab/uso terapéutico , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus , Vacunación
18.
Lancet Rheumatol ; 4(3): e177-e187, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-34977602

RESUMEN

BACKGROUND: In rituximab-treated patients with rheumatoid arthritis, humoral and cellular immune responses after two or three doses of SARS-CoV-2 vaccines are not well characterised. We aimed to address this knowledge gap. METHODS: This prospective, cohort study (Nor-vaC) was done at two hospitals in Norway. For this sub-study, we enrolled patients with rheumatoid arthritis on rituximab treatment and healthy controls who received SARS-CoV-2 vaccines according to the Norwegian national vaccination programme. Patients with insufficient serological responses to two doses (antibody to the receptor-binding domain [RBD] of the SARS-CoV-2 spike protein concentration <100 arbitrary units [AU]/mL) were allotted a third vaccine dose. Antibodies to the RBD of the SARS-CoV-2 spike protein were measured in serum 2-4 weeks after the second and third doses. Vaccine-elicited T-cell responses were assessed in vitro using blood samples taken before and 7-10 days after the second dose and 3 weeks after the third dose from a subset of patients by stimulating cryopreserved peripheral blood mononuclear cells with spike protein peptides. The main outcomes were the proportions of participants with serological responses (anti-RBD antibody concentrations of ≥70 AU/mL) and T-cell responses to spike peptides following two and three doses of SARS-CoV-2 vaccines. The study is registered at ClinicalTrials.gov, NCT04798625, and is ongoing. FINDINGS: Between Feb 9, 2021, and May 27, 2021, 90 patients were enrolled, 87 of whom donated serum and were included in our analyses (69 [79·3%] women and 18 [20·7%] men). 1114 healthy controls were included (854 [76·7%] women and 260 [23·3%] men). 49 patients were allotted a third vaccine dose. 19 (21·8%) of 87 patients, compared with 1096 (98·4%) of 1114 healthy controls, had a serological response after two doses (p<0·0001). Time since last rituximab infusion (median 267 days [IQR 222-324] in responders vs 107 days [80-152] in non-responders) and vaccine type (mRNA-1273 vs BNT162b2) were significantly associated with serological response (adjusting for age and sex). After two doses, 10 (53%) of 19 patients had CD4+ T-cell responses and 14 (74%) had CD8+ T-cell responses. A third vaccine dose induced serological responses in eight (16·3%) of 49 patients, but induced CD4+ and CD8+ T-cell responses in all patients assessed (n=12), including responses to the SARS-CoV-2 delta variant (B.1.617.2). Adverse events were reported in 32 (48%) of 67 patients and in 191 (78%) of 244 healthy controls after two doses, with the frequency not increasing after the third dose. There were no serious adverse events or deaths. INTERPRETATION: This study provides important insight into the divergent humoral and cellular responses to two and three doses of SARS-CoV-2 vaccines in rituximab-treated patients with rheumatoid arthritis. A third vaccine dose given 6-9 months after a rituximab infusion might not induce a serological response, but could be considered to boost the cellular immune response. FUNDING: The Coalition for Epidemic Preparedness Innovations, Research Council of Norway Covid, the KG Jebsen Foundation, Oslo University Hospital, the University of Oslo, the South-Eastern Norway Regional Health Authority, Dr Trygve Gythfeldt og frues forskningsfond, the Karin Fossum Foundation, and the Research Foundation at Diakonhjemmet Hospital.

20.
Influenza Other Respir Viruses ; 16(2): 204-212, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34751488

RESUMEN

BACKGROUND: Infection with the novel coronavirus SARS-CoV-2 induces antibodies that can be used as a proxy for COVID-19. We present a repeated nationwide cross-sectional study assessing the seroprevalence of SARS-CoV-2, the infection fatality rate (IFR), and infection hospitalization rate (IHR) during the first year of the pandemic in Norway. METHODS: Residual serum samples were solicited in April/May 2020 (Round 1), in July/August 2020 (Round 2) and in January 2021 (Round 3). Antibodies against SARS-CoV-2 were measured using a flow cytometer-based assay. Aggregate data on confirmed cases, COVID-19-associated deaths and hospitalizations were obtained from the Emergency preparedness registry for COVID-19 (Beredt C19), and the seroprevalence estimates were used to estimate IFR and IHR. RESULTS: Antibodies against SARS-CoV-2 were measured in 4840 samples. The estimated seroprevalence increased from 0.8% (95% credible interval [CrI] 0.4%-1.3%) after the first wave of the pandemic (Rounds 1 and 2 combined) to 3.2% (95% CrI 2.3%-4.2%) (Round 3). The IFR and IHR were higher in the first wave than in the second wave and increased with age. The IFR was 0.2% (95% CrI 0.1%-0.3%), and IHR was 0.9% (95% CrI 0.6%-1.5%) for the second wave. CONCLUSIONS: The seroprevalence estimates show a cumulative increase of SARS-CoV-2 infections over time in the Norwegian population and suggest some under-recording of confirmed cases. The IFR and IHR were low, corresponding to the relatively low number of COVID-19-associated deaths and hospitalizations in Norway. Most of the Norwegian population was still susceptible to SARS-CoV-2 infection after the first year of the pandemic.


Asunto(s)
COVID-19 , Anticuerpos Antivirales , Estudios Transversales , Humanos , Noruega/epidemiología , Pandemias , SARS-CoV-2 , Estudios Seroepidemiológicos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...