Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Annals of the Rheumatic Diseases ; 82(Suppl 1):867-868, 2023.
Article in English | ProQuest Central | ID: covidwho-20233202

ABSTRACT

BackgroundPrevious studies have shown that using a finger prick as the primary method for blood withdrawal is an efficient way to collect blood samples remotely, and data on blood levels from a finger prick are directly comparable to that obtained by a venepuncture. During the COVID-19 pandemic, we therefore complemented our large digital research platform with serum collection via a home finger prick testing in order to collect samples without the need of visits to a hospital. This repeatedly enabled us to rapidly answer new and relevant clinical research questions about COVID-19, thereby showing the potential of the finger prick for research purposes. However, the use of finger pricks in a research or clinical practice setting is still uncommon, and not yet tested on a large scale. In addition, there is limited data on peoples' willingness and ability to successfully use the finger prick at home, especially in patients with inflammatory rheumatic diseases (iRD) who may have impaired hand function.ObjectivesTo investigate the feasibility of finger prick testing in combination with a digital research platform by evaluating the success rate and patients' perspective towards the use of the finger prick.MethodsData were collected from an ongoing prospective cohort study including patients with iRD from the Amsterdam Rheumatology & immunology Center and healthy controls. Serum samples were collected up to eight times during follow-up via blood withdrawal by venepuncture at the local research institute or via a finger prick that could be performed at home. For the latter option, participants were instructed to collect three drops of blood, which would yield approximately 40-80 µL of serum after clotting. All study participants were questioned about their preference for a particular sampling method for individual healthcare and for scientific research. Participants who received a finger prick test before June 26, 2021, were asked to complete a digital evaluation questionnaire of the finger prick after their attempt. The finger prick was defined as failed when less than 10 µL of serum could be recovered from the collection device, or if no sample was returned to the laboratory and participants indicated in the questionnaires that they did not succeed in collecting the required amount of serum.ResultsA total of 3080 patients with iRD and 1102 healthy controls were included in the study. Of these, 2135 (69%) patients and 899 (82%) controls attempted to execute at least one finger prick, and 1439 (67%) patients and 712 (21%) controls executed multiple finger pricks. The first finger prick was successfully done by 92% (CI 90 – 93) of iRD patients, 94% (CI 92 – 95) of healthy controls, 93% (CI: 92 – 94) of all participants aged 70 years or younger, and 89% (CI 86 – 92) of all participants aged above 70 years (Table 1). Sex did not impact these success rates. Repeated failure occurred in 11 of 1439 (0.8%) patients and 4 of 712 (0.6%) controls. The two most common reasons for perceived failure of the finger prick were related to insufficient blood yield when applying the finger prick. Finally, both patients and controls were less willing to perform a finger prick for individual healthcare compared to scientific research;31% of patients and 61% of controls were willing to perform a finger prick for scientific research compared to 19% of patients and 39% of controls for healthcare. The most important reason for this was lower confidence in the execution and laboratory measurements when blood was drawn via a venepuncture compared to a finger prick.ConclusionIn this study, we demonstrated that the vast majority of participants, among which elderly and patients of whom hand function may be impaired by an underlying rheumatic disease, were able to successfully draw the required amount of blood for serological analyses. This shows that the finger prick testing is suitable for a high-throughput implementation to monitor patients remotely, which will likely contribute to improving the efficiency and cost-effectiveness of b th healthcare and scientific research.REFERENCES:NIL.Acknowledgements:NIL.Disclosure of InterestsNone Declared.

2.
Scand J Rheumatol ; : 1-4, 2022 Jul 14.
Article in English | MEDLINE | ID: covidwho-2236565

ABSTRACT

OBJECTIVE: To evaluate the relationship between reported coronavirus disease 2019 (COVID-19)-like symptoms and the presence of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) antibodies in patients with an immune-mediated inflammatory disorder or post-solid organ transplantation (IMIDT) with and without immunosuppressive medication (imed) and controls. METHOD: The IENIMINI cohort was a prospective cohort study set up in the Netherlands in March 2020, with 2 monthly (paper) or weekly (online) questionnaires about COVID-19-like symptoms. Participants from this cohort who reported these symptoms between March 2020 and November 2020 were approached for this substudy. SARS-CoV-2 antibodies were tested using a total antibody assay. RESULTS: Of the 1203 participants approached, 629 agreed to participate and were sent a fingerprick test; 565 participants collected a capillary blood sample, of which 562 were usable. Analysis showed that 57/202 (28.2%) of the tested IMIDT group with imed, 48/16 3(29.4%) of the IMIDT group without imed, and 69/197 (35.0%) of the control group tested positive for SARS-CoV-2 antibodies. Seroprevalences of SARS-CoV-2 antibodies between males and females, biological disease-modifying anti-rheumatic drug users and non-users, and those who had had a serious disease period (defined as an episode with dyspnoea and fever) and those who had not, were not statistically different between the three groups. CONCLUSIONS: Approximately 30% of patients who had reported COVID-19-like symptoms had SARS-CoV-2 antibodies. The seroprevalence of SARS-CoV-2 antibodies after reported COVID-19-like symptoms was similar in IMIDT patients with and without imed compared to controls.

3.
Journal of Neuromuscular Diseases ; 9:S109-S110, 2022.
Article in English | EMBASE | ID: covidwho-2043401

ABSTRACT

Background: Disease-specific studies have reported impaired humoral responses after SARS-CoV-2 vaccination in patients with immune-mediated inflammatory disorders (IMIDs) treated with specific immunosuppressants and immunomodulating agents. The objective of this study is to investigate the humoral immune response after SARS-CoV-2 vaccination in patients using immunosuppressive and immunomodulating mono- and combination therapies, focussing on frequently prescribed therapies for inflammatory neuromuscular diseases. Methods: National prospective observational cohort study in selected patients with prevalent IMIDs including neuromuscular disease, and immunosuppressive or immunomodulating monotherapy (n=1273), combination therapies (n=419), patients without immunosuppressants (n=473), and healthy controls (n=174). Anti-RBD IgG responses and neutralisation capacity were monitored following standard vaccination regimens and a three-vaccination regimen in subgroups. Hybrid immune responses, i.e. vaccination after previous SARS-CoV-2 infection, were studied as a proxy for recall responses. Findings: Sera from 1869 participants without and 470 participants with previous SARS-CoV-2 infection were analysed. We included 168 (7 2%) patients with inflammatory neuropathies and myopathies, and 127 (5 4%) patients with myasthenia gravis. Humoral responses did not differ between disorders. Anti-CD20 therapy and mycophenolate mofetil combined with corticosteroids were associated with lower relative risks (RR) for reaching seroconversion following standard vaccination (RR: 0 32 and 0 61 respectively). The monotherapies corticosteroids, purine antagonists, methotrexate, mycophenolate mofetil and IVIg were not associated with a lower RR for reaching seroconversion (RR: 0 97, 0 98, 1 01, 0 86, and 0 99, respectively). Similarly, corticosteroids combined with either methotrexate or purine antagonists was not associated with a lower RR for reaching seroconversion (RR 0 89). A third vaccination increased seroconversion for mycophenolate mofetil combination treatments but not for anti-CD20 therapies. Most immunosuppressant groups showed moderately reduced antibody titres after standard vaccination that, in subgroups, did not increase after a third vaccination, although seroconversion rates and neutralisation capacity were unaffected. In participants with previous SARS-CoV-2 infection, SARS-CoV-2 antibodies were boosted after vaccination, regardless of immunosuppressive treatment. Interpretation: Humoral responses following vaccination are impaired by specific immunosuppressants, most relevant for neuromuscular diseases being anti-CD20 and mycophenolate mofetil combination treatments. After standard vaccination regimens most immunosuppressants show equal seroconversion to controls although antibody titres may be moderately reduced. As neutralisation capacity and recall responses are also preserved in these patients, this is not likely to translate in loss of (short term) protection. Alternatively, in immunosuppressants showing poor humoral responses after standard vaccination regimens such as, a third vaccination resulted in additional seroconversion in mycophenolate mofetil combination treatments whereas the effect for anti-CD20 therapy was limited.

4.
Journal of Neuromuscular Diseases ; 9:S110, 2022.
Article in English | EMBASE | ID: covidwho-2043397

ABSTRACT

Importance: Viral infection or vaccination has the potential to increase disease activity in immune-mediated neuromuscular diseases. Objective: We aimed to evaluate whether SARSCoV- 2 vaccination and infection leads to increase of disease activity in patients with immune-mediated neuromuscular diseases. Methods: This is an interim analysis of a subset of patients from an ongoing prospective multi-center cohort study on SARS-CoV-2 vaccination in patients with various immune mediated inflammatory diseases in the Netherlands, the Target to-B!-COVID study (T2B!). Patients received digital questionnaires every two months from study entry to assess disease activity compared to previous visit using a 5-point Likert scale. In addition, in case of SARS CoV-2 infection (prior to vaccination) patients received an extra questionnaire to assess disease activity in the four weeks after infection. In cases of self-reported increase of disease activity, medical files were used to assess whether disease activity was reported by the treating physician, and whether changes were made in type or dose of immunosuppressive or immunomodulating treatment. Results: In total, we included 303 patients with immune-mediated neuromuscular disease of which 127 patients with inflammatory neuropathies, 133 patients with myasthenia gravis, and 43 patients with myositis. In the four months after completed vaccination, 67 (22.1%) patients indicated an increase in disease activity, of which 62 (93%) was reported as worse and 5 (7%) as much worse. In 10 (3.3%) of the cases with self-reported increase, disease activity was also reported by the treating physician in the medical chart. In 4 (1.3%) of patients with self-reported increase disease activity treatment was adjusted because of the increase in disease activity. A SARS-CoV-2 infection prior to vaccination occurred in 24 (8%) patients, from which 3 (12.5%) indicated an increase in disease activity, not leading to change in treatment. Conclusion: Increase of disease activity after SARS-CoV-2 vaccination or infection was reported infrequently, and was self-limiting in most cases. Findings from our cohort may help physicians in neuromuscular disease to adequately inform patients on the risk of increased disease activity due to SARS-CoV-2 vaccination or infection. Full and verified results will be reported at the ICNMD 2022.

5.
Annals of the Rheumatic Diseases ; 81:118, 2022.
Article in English | EMBASE | ID: covidwho-2009141

ABSTRACT

Background: Concerns have been raised regarding risks of COVID-19 breakthrough infections in vaccinated patients with immune-mediated infammatory diseases (IMIDs) treated with immunosuppressants, but data on COVID-19 breakthrough infections in these patients are still scarce. Objectives: The primary objective was to compare the incidence and severity of COVID-19 breakthrough infections with the SARS-CoV-2 delta variant between fully vaccinated IMID patients with immunosuppressants, and controls (IMID patients without immunosuppressants and healthy controls). The secondary objective was to explore determinants of breakthrough infections. Methods: In this study we pooled data collected from two large ongoing prospective multi-center cohort studies (Target to-B! [T2B!] study and ARC study). Clinical data were collected between February and December 2021, using digital questionnaires, standardized electronic case record forms and medical files. Post-vaccination serum samples were analyzed for anti-RBD antibodies (T2B! study only) and anti-nucleocapsid antibodies to identify asymptomatic breakthrough infections (ARC study only). Logistic regression analyses were used to assess associations with the incidence of breakthrough infections. Multivariable models were adjusted for age, sex, cardiovascular disease, chronic pulmonary disease, obesity and vaccine type. Results: We included 3207 IMID patients with immunosuppressants and 1810 controls (985 IMID patients without immunosuppressants and 825 healthy controls). The incidence of COVID-19 breakthrough infections was comparable between patients with immunosuppressants (5%) and controls (5%). The absence of SARS-CoV-2 IgG antibodies after COVID-19 vaccination was independently associated with an increased incidence of breakthrough infections (P 0.044). The proportion of asymptomatic COVID-19 breakthrough cases that were additionally identifed serologically in the ARC cohort was comparable between IMID patients with immunosuppressants and controls;66 (10%) of 695 patients vs. 64 (10%) of 647 controls. Hospitalization was required in 8 (5%) of 149 IMID patients with immunosuppressants and 5 (6%) of 86 controls with a COVID-19 breakthrough infection. Hospitalized cases were generally older, and had more comorbidities compared with non-hospitalized cases (Table 1). Hospitalization rates were signifcantly higher among IMID patients treated with anti-CD20 therapy compared to IMID patients using any other immunosuppres-sant (3 [23%] of 13 patients vs. 5 [4%] of 128 patients, P 0.041;Table 1). Conclusion: The incidence of COVID-19 breakthrough infections in IMID patients with immunosuppressants was comparable to controls, and infections were mostly mild. Anti-CD20 therapy might increase patients' susceptibility to severe COVID-19 breakthrough infections, but traditional risk factors also continue to have a critical contribution to the disease course of COVID-19. Therefore, we argue that most patients with IMIDs should not necessarily be seen as a risk group for severe COVID-19, and that integrating other risk factors should become standard practice when discussing treatment options, COVID-19 vaccination, and adherence to infection prevention measures with patients.

6.
Annals of the Rheumatic Diseases ; 81:963, 2022.
Article in English | EMBASE | ID: covidwho-2009067

ABSTRACT

Background: Many countries are promoting booster SARS-CoV-2 vaccination campaigns as the COVID-19 pandemic continues. Incremental short-term adverse events after two SARS-CoV-2 vaccinations have been reported in healthy individuals.1,2 However, data on incremental short-term adverse events in patients with various immune-mediated infammatory diseases (IMIDs) after repeated SARS-CoV-2 vaccination is scarce. Objectives: We report risk factors for short-term adverse events in IMID patients after SARS-CoV-2 vaccination. Methods: Self-reported daily questionnaires on adverse events in the frst seven days after SARS-CoV-2 vaccination were obtained from individuals participating in an ongoing prospective multi-arm multicenter cohort study on SARS-CoV-2 vaccination in patients with various IMIDs in the Netherlands (T2B! immunity after SARS-CoV-2). Clinically relevant adverse events were defned as systemic adverse advents lasting longer than two days or hindering daily activities. Adjusted relative risks for developing clinically relevant adverse events were calculated using a logistic mixed-effects model. Results: Data of 2081 patients and 178 healthy controls were obtained. Infammatory bowel disease (N:480), Multiple sclerosis (N:343) and Rheumatoid arthritis (N:266) were the largest disease groups. Adjusted relative risks for relevant adverse events are presented in Figure 1. Third vaccination was not associated with increased risk on adverse events when compared to a second vaccination (aRR: 0.93 95% CI: 0.84-1.02). Patients with IMIDs were at increased risk for developing adverse events after vaccination when compared to controls (aRR: 1.16 95% CI: 1.01-1.34). Female sex (aRR 1.43 95% CI: 1.32-1.56), age below 50 (aRR 1.14 95% CI: 1.06-1.23) and a preceding SARS-CoV-2 infection (aRR: 1.14 95% CI: 1.01-1.29) were also associated with increased risk of adverse events following vaccination. Allergic reactions and hospital admission were uncommon (0.67% and 0.19% respectively);7.4% and 6.8% of patients reported adverse events impacting daily life on day seven after second and third vaccination, respectively. Data on increase in disease activity of the IMID following vaccination are currently being investigated. Conclusion: A third SARS-CoV-2 vaccination was not associated with an increased risk on short-term clinically relevant adverse events when compared to a second vaccination. Although patients with IMIDs may be slightly more at risk to develop adverse events after SARS-CoV-2 vaccination, most adverse events were transient and disappeared within seven days. This message should reassure IMID patients who are hesitant on booster vaccination. Data on potential IMID fare-ups after vaccination will follow.

7.
Journal of Crohn's & colitis ; 16(Suppl 1):i079-i079, 2022.
Article in English | EuropePMC | ID: covidwho-1999590

ABSTRACT

Background The aim of this study was to investigate the effect of various immunosuppressants on the humoral immune responses after vaccination against SARS-CoV-2 in patients with immune-mediated inflammatory diseases (IMIDs). Methods The Target to B! SARS-CoV-2 study is a multicentre study, taking place in 7 Dutch academic hospitals. Patients with the following IMIDs were recruited: Crohn’s disease (CD), ulcerative colitis (UC), auto-immune hepatitis, rheumatic (e.g. rheumatoid arthritis), neurological (e.g. multiple sclerosis) and dermatological IMIDs (e.g. atopic dermatitis). Patients were recruited based on immunosuppressants (table 1) and previous SARS-CoV-2 infection. The control group consisted of healthy subjects and IMID patients without immunosuppressants. SARS-CoV-2 receptor binding domain (RBD) antibodies were measured 28 days after completed SARS-CoV-2 vaccination. Seroconversion was defined as anti-RBD IgG >4 AU/mL. In this , we focus on therapies relevant for inflammatory bowel diseases (IBD) and present results for these treatments from patients with IBD, but also other IMIDs. Results Numbers of recruited patients with each immunosuppressant are shown in table 1. Amongst these patients, 312 patients had CD and 176 UC, the rest was diagnosed with another IMID. Seroconversion was reduced in patients receiving sphingosine 1-phosphate (S1P) modulators (all multiple sclerosis patients) while seroconversion was similar to controls in the other treatment groups. However, use of Anti-tumour necrosis factor (TNF), methotrexate, janus kinase (JAK) inhibitor monotherapy and all combination therapies (except for corticosteroids combined with other immunosuppressants) were associated with reduced Sars-CoV-2 antibody titres. Patients with a previous SARS-CoV-2 infection had higher median antibody titres after second vaccination than those without a previous SARS-CoV-2 infection. The type of IMID did not affect seroconversion rates. Conclusion No immunosuppressant, registered for IBD, reduced the rates of seroconversion after vaccination against SARS-CoV-2. Some immunosuppressants were associated with lower antibody titres. However, the clinical relevance of lower antibody titres remains unknown. S1P modulators, had a clear negative impact on the humoral response against SARS-CoV-2 after vaccination. This might be relevant in the future as this therapy is currently being approved for UC. Disease aetiology did not impair immunity against SARS-CoV-2 immunity after vaccination. Disclaimer: Absolute numbers of antibody titres and rates of seroconversion will be reported at the conference and are not reported in this as this might negatively impact the current submission process.

8.
Vox Sanguinis ; 117(SUPPL 1):258-259, 2022.
Article in English | EMBASE | ID: covidwho-1916358

ABSTRACT

Background: SARS-CoV-2 antibody tests are variable using different antigens, reagent dilutions and units of reporting. Without calibration interlaboratory interpretation of assay results is not possible, SARS-CoV-2 antibody tests are variable using different antigens, reagent dilutions and units of reporting. Without calibration interlaboratory interpretation of assay results is not possible, slowing down progress in treatments with convalescent plasma. slowing down progress in treatments with convalescent plasma. Aims: The aim of this study, which is part of the SUPPORT-E consortium (Supporting high-quality evaluation of COVID-19 convalescent plasma throughout Europe), was to calibrate anti-SARS-CoV-2 antibody assays used by European laboratories to determine antibody titers in (convalescent) plasma. Methods: To achieve this we distributed a set of 23 reference samples to 26 participating blood establishments across Europe. This set served as a quality control round for SARS-CoV-2 antibody testing and consisted of SARS-CoV-2 pre-outbreak negative plasma samples and plasma samples from unvaccinated convalescent donors with low, medium, high and very high IgG titers against the receptor binding domain (RBD) of the SARS-CoV-2 Spike (S) protein. In addition, a serial dilution of one batch of pooled plasma (n = 3) was distributed that could serve as internal standard. This methodology allowed calibration to the WHO standard in IU/ml, so conversion factors could be calculated. Results: Twenty laboratories participated, including the qualitative Euroimmune (n = 7), Roche (n = 4) and Abbott (n = 4) SARS-CoV-2 IgG assay as most frequently used tests. Four laboratories, using commercial assays from WANTAI or DiaSorin, were not able to discriminate pre-outbreak samples from SARS-CoV-2 positive sera. In addition, the majority of the laboratories were not able to discriminate between plasma samples with high and very high titers, showing that the dynamic range of these commercial assays is limited. Using these data we calculated the conversion factor to IU/ml for the qualitative Euroimmune as 238, Roche as 0.98 and for the Abbott test as 0.9, which only applies when pre-vaccinated samples are used in the test. Summary/Conclusions: This initiative by the SUPPORT-E consortium aids in calibration of antibody testing across laboratories, allowing to compare SARS-CoV-2 antibody titers in (convalescent) plasma. For example, our conversion factor can now be used to calibrate Euroimmune units that were measured in the convalescent plasma samples within the Recovery (United Kingdom)1 Capsid (Germany)2 and CovEarly (Netherlands)3 clinical trial. .

9.
Nephrology Dialysis Transplantation ; 37(SUPPL 3):i246, 2022.
Article in English | EMBASE | ID: covidwho-1915713

ABSTRACT

BACKGROUND AND AIMS: Lower antibody responses after SARS-Cov-2 vaccination have been reported in patients with severely impaired kidney function or patients with kidney replacement treatment. We compared humoral responses and reported adverse events of three vaccines (mRNA-1273, BNT162b2 and AZD1222) in kidney transplant recipients (KTRs), dialysis patients, patients with CKD stages G4-G5 and control subjects without kidney disease. METHOD: KTRs, dialysis patients and patients with CKD stages G4-G5 were vaccinated with either mRNA-1273, BNT162b2 or AZD1222 during the Dutch SARSCoV- 2 vaccination program. Control subjects were all vaccinated with mRNA-1273. Blood samples were obtained at 1 month after two vaccinations by home-based finger prick tests and were analysed for the presence of IgG antibodies against the receptorbinding domain of the spike protein of SARS-CoV-2 using the Sanquin anti-SARSCoV- 2 RBD IgG ELISA assay. Primary endpoints were the antibody titer and reported systemic adverse events (AEs) at 1 month after the second vaccination. Multivariate regression analysis was performed on the difference between vaccines with respect to antibody titer and AEs after correction for sex, ethnicity, BMI, eGFR, dialysis vintage, transplantation characteristics and use of immunosuppressive drugs. RESULTS: A total of 2468 KTRs, 480 dialysis patients, 400 patients with CKD stages G4-G5 and 186 control subjects were enrolled. KTRs had lower antibody titers (66 [8-573] BAU/mL) in comparison to dialysis patients [1375 (431-2896) BAU/mL], patients with CKD stages G4-G5 [2097 (828-4077) BAU/mL] and control subjects [3713 (2291-6451) BAU/mL]. mRNA-1273 demonstrated a higher antibody titer compared with BNT162b2 in KTR [72 (9-638) versus 21 (6-128) BAU/mL;P < .001), dialysis patients [1675 (573-3031) versus 636 (216-1416) BAU/mL;P < .001] and patients with CKD stages G4-G5 [2879 (1425-5311) versus 1063 (389-1939) BAU/mL;P < .001). In a similar pattern, mRNA-1273 demonstrated a higher antibody titer compared with AZD1222 (P < .001 in all groups). Multivariate analysis revealed that BNT162b2 and AZD1222 were significantly associated with lower antibody levels compared with mRNA-1273 in all 3 patient groups. BNT162b2 demonstrated less frequently systemic AEs compared with mRNA-1273 in KTRs (12% versus 27%;P < .001), dialysis patients (12% versus 29%;P = .007) and in patients with CKD G4- G5 (18% versus 67%, P < .001). AZD1222 demonstrated less systemic AEs compared with mRNA-1273 only in patients with CKD stages G4-G5 (39% versus 67%;P = .03). Multivariate analysis revealed that BNT162b2 was associated with fewer systemic AEs in only dialysis patients (P = .04) and patients with CKD stages G4-G5 (P = .02). CONCLUSION: mRNA-1273 demonstrated significantly higher antibody levels at 1 month after 2 vaccinations as compared with BNT162b2 and AZD1222 in high-risk patients with kidney disease. BNT162b2 was associated with a fewer systemic AEs in dialysis patients and patients with CKD stages G4-G5, although these AEs were mild and self-limiting. mRNA-1273 may therefore be considered as the preferred SARS-CoV-2 vaccine in high-risk patients with kidney disease. Whether the higher antibody response following vaccination with mRNA-1273 sustains and results in a better protection against COVID-19 is yet to be analysed.

14.
BMC Nephrol ; 23(1): 55, 2022 02 05.
Article in English | MEDLINE | ID: covidwho-1690947

ABSTRACT

BACKGROUND: COVID-19 is associated with increased morbidity and mortality in patients with chronic kidney disease (CKD) stages G4-G5, on dialysis or after kidney transplantation (kidney replacement therapy, KRT). SARS-CoV-2 vaccine trials do not elucidate if SARS-CoV-2 vaccination is effective in these patients. Vaccination against other viruses is known to be less effective in kidney patients. Our objective is to assess the efficacy and safety of various types of SARS-CoV-2 vaccinations in patients with CKD stages G4-G5 or on KRT. METHODS: In this national prospective observational cohort study we will follow patients with CKD stages G4-G5 or on KRT (n = 12,000) after SARS-CoV-2 vaccination according to the Dutch vaccination program. Blood will be drawn for antibody response measurements at day 28 and month 6 after completion of vaccination. Patient characteristics and outcomes will be extracted from registration data and questionnaires during 2 years of follow-up. Results will be compared with a control group of non-vaccinated patients. The level of antibody response to vaccination will be assessed in subgroups to predict protection against COVID-19 breakthrough infection. RESULTS: The primary endpoint is efficacy of SARS-CoV-2 vaccination determined as the incidence of COVID-19 after vaccination. Secondary endpoints are the antibody based immune response at 28 days after vaccination, the durability of this response at 6 months after vaccination, mortality and (serious) adverse events. CONCLUSION: This study will fulfil the lack of knowledge on efficacy and safety of SARS-CoV-2 vaccination in patients with CKD stages G4-G5 or on KRT. TRIAL REGISTRATION: The study protocol has been registered in clinicaltrials.gov ( NCT04841785 ). Current knowledge about this subject COVID-19 has devastating impact on patients with CKD stages G4-G5, on dialysis or after kidney transplantation. Effective SARS-CoV-2 vaccination is very important in these vulnerable patient groups. Recent studies on vaccination in these patient groups are small short-term studies with surrogate endpoints. Contribution of this study Assessment of incidence and course of COVID-19 after various types of SARS-CoV-2 vaccination during a two-year follow-up period in not only patients on dialysis or kidney transplant recipients, but also in patients with CKD stages G4-G5. Quantitative analysis of antibody response after SARS-CoV-2 vaccination and its relationship with incidence and course of COVID-19 in patients with CKD stages G4-G5, on dialysis or after kidney transplantation compared with a control group. Monitoring of (serious) adverse events and development of anti-HLA antibodies. Impact on practice or policy Publication of the study design contributes to harmonization of SARS-CoV-2 vaccine study methodology in kidney patients at high-risk for severe COVID-19. Data on efficacy of SARS-CoV-2 vaccination in patients with CKD will provide guidance for future vaccination policy.


Subject(s)
COVID-19 Vaccines , Kidney Transplantation , Renal Dialysis , Renal Insufficiency, Chronic/therapy , COVID-19 Vaccines/administration & dosage , Cohort Studies , Humans , Netherlands , Observational Studies as Topic , Prospective Studies , Time Factors
15.
Mult Scler Relat Disord ; 57: 103416, 2022 Jan.
Article in English | MEDLINE | ID: covidwho-1611928

ABSTRACT

OBJECTIVE: The objective of this study was to measure humoral responses after SARS-CoV-2 vaccination in MS patients treated with ocrelizumab (OCR) compared to MS patients without disease modifying therapies (DMTs) in relation to timing of vaccination and B-cell count. METHODS: OCR treated patients were divided into an early and a late group (cut-off time 12 weeks between infusion and first vaccination). Patients were vaccinated with mRNA-1273 (Moderna). B-cells were measured at baseline (time of first vaccination) and SARS-CoV-2 antibodies were measured at baseline, day 28, 42, 52 and 70. RESULTS: 87 patients were included (62 OCR patients, 29 patients without DMTs). At day 70, seroconversion occurred in 39.3% of OCR patients compared to 100% of MS patients without DMTs. In OCR patients, seroconversion varied between 26% (early group) to 50% (late group) and between 27% (low B-cells) to 56% (at least 1 detectable B-cell/µL). CONCLUSIONS: Low B-cell counts prior to vaccination and shorter time between OCR infusion and vaccination may negatively influence humoral response but does not preclude seroconversion. We advise OCR treated patients to get their first vaccination as soon as possible. In case of an additional booster vaccination, timing of vaccination based on B-cell count and time after last infusion may be considered.


Subject(s)
COVID-19 , Multiple Sclerosis , Antibodies, Monoclonal, Humanized , COVID-19 Vaccines , Humans , SARS-CoV-2 , Vaccination
SELECTION OF CITATIONS
SEARCH DETAIL