16
views
0
recommends
+1 Recommend
1 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: not found

      COVID-19 vaccine-induced antibody and T-cell responses in immunosuppressed patients with inflammatory bowel disease after the third vaccine dose (VIP): a multicentre, prospective, case-control study

      research-article
      , PhD a , e , * , , PhD a , * , , PhD b , * , , PhD b , * , , MBBS a , e , , MBChB a , g , , BMBCh a , g , , PhD a , , BSc a , , MSc h , i , , MSc h , , BSc f , , MSc a , , PhD k , l , , MBChB a , e , , PhD c , m , , Prof, MD o , p , , MD n , q , , PhD a , e , , PhD a , e , , MBChB s , , BSc t , , Prof, DM t , u , , PhD v , , MBBS w , , Prof, MD a , e , , Prof, PhD d , , MBBS h , j , , Prof, PhD g , , Prof, PhD k , l , , Prof, PhD b , r , * , , PhD h , j , * , , PhD h , j , * , , PhD a , e , * , * , VIP study investigators
      The Lancet. Gastroenterology & Hepatology
      The Author(s). Published by Elsevier Ltd.

      Read this article at

      ScienceOpenPublisherPMC
      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Background

          COVID-19 vaccine-induced antibody responses are reduced in patients with inflammatory bowel disease (IBD) taking anti-TNF or tofacitinib after two vaccine doses. We sought to assess whether immunosuppressive treatments were associated with reduced antibody and T-cell responses in patients with IBD after a third vaccine dose.

          Methods

          VIP was a multicentre, prospective, case-control study done in nine centres in the UK. We recruited immunosuppressed patients with IBD and non-immunosuppressed healthy individuals. All participants were aged 18 years or older. The healthy control group had no diagnosis of IBD and no current treatment with systemic immunosuppressive therapy for any other indication. The immunosuppressed patients with IBD had an established diagnosis of Crohn's disease, ulcerative colitis, or unclassified IBD using standard definitions of IBD, and were receiving established treatment with one of six immunosuppressive regimens for at least 12 weeks at the time of first dose of SARS-CoV-2 vaccination. All participants had to have received three doses of an approved COVID-19 vaccine. SARS-CoV-2 spike antibody binding and T-cell responses were measured in all participant groups. The primary outcome was anti-SARS-CoV-2 spike (S1 receptor binding domain [RBD]) antibody concentration 28–49 days after the third vaccine dose, adjusted by age, homologous versus heterologous vaccine schedule, and previous SARS-CoV-2 infection. The primary outcome was assessed in all participants with available data.

          Findings

          Between Oct 18, 2021, and March 29, 2022, 352 participants were included in the study (thiopurine n=65, infliximab n=46, thiopurine plus infliximab combination therapy n=49, ustekinumab n=44, vedolizumab n=50, tofacitinib n=26, and healthy controls n=72). Geometric mean anti-SARS-CoV-2 S1 RBD antibody concentrations increased in all groups following a third vaccine dose, but were significantly lower in patients treated with infliximab (2736·8 U/mL [geometric SD 4·3]; p<0·0001), infliximab plus thiopurine (1818·3 U/mL [6·7]; p<0·0001), and tofacitinib (8071·5 U/mL [3·1]; p=0·0018) compared with the healthy control group (16 774·2 U/mL [2·6]). There were no significant differences in anti-SARS-CoV-2 S1 RBD antibody concentrations between the healthy control group and patients treated with thiopurine (12 019·7 U/mL [2·2]; p=0·099), ustekinumab (11 089·3 U/mL [2·8]; p=0·060), or vedolizumab (13 564·9 U/mL [2·4]; p=0·27). In multivariable modelling, lower anti-SARS-CoV-2 S1 RBD antibody concentrations were independently associated with infliximab (geometric mean ratio 0·15 [95% CI 0·11–0·21]; p<0·0001), tofacitinib (0·52 [CI 0·31–0·87]; p=0·012), and thiopurine (0·69 [0·51–0·95]; p=0·021), but not with ustekinumab (0·64 [0·39–1·06]; p=0·083), or vedolizumab (0·84 [0·54–1·30]; p=0·43). Previous SARS-CoV-2 infection (1·58 [1·22–2·05]; p=0·0006) was independently associated with higher anti-SARS-CoV-2 S1 RBD antibody concentrations and older age (0·88 [0·80–0·97]; p=0·0073) was independently associated with lower anti-SARS-CoV-2 S1 RBD antibody concentrations. Antigen-specific T-cell responses were similar in all groups, except for recipients of tofacitinib without evidence of previous infection, where T-cell responses were significantly reduced relative to healthy controls (p=0·021).

          Interpretation

          A third dose of COVID-19 vaccine induced a boost in antibody binding in immunosuppressed patients with IBD, but these responses were reduced in patients taking infliximab, infliximab plus thiopurine, and tofacitinib. Tofacitinib was also associated with reduced T-cell responses. These findings support continued prioritisation of immunosuppressed groups for further vaccine booster dosing, particularly patients on anti-TNF and JAK inhibitors.

          Funding

          Pfizer.

          Related collections

          Most cited references32

          • Record: found
          • Abstract: found
          • Article: not found

          The REDCap consortium: Building an international community of software platform partners

          The Research Electronic Data Capture (REDCap) data management platform was developed in 2004 to address an institutional need at Vanderbilt University, then shared with a limited number of adopting sites beginning in 2006. Given bi-directional benefit in early sharing experiments, we created a broader consortium sharing and support model for any academic, non-profit, or government partner wishing to adopt the software. Our sharing framework and consortium-based support model have evolved over time along with the size of the consortium (currently more than 3200 REDCap partners across 128 countries). While the "REDCap Consortium" model represents only one example of how to build and disseminate a software platform, lessons learned from our approach may assist other research institutions seeking to build and disseminate innovative technologies.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found
            Is Open Access

            Fitting Linear Mixed-Effects Models Using lme4

            Maximum likelihood or restricted maximum likelihood (REML) estimates of the parameters in linear mixed-effects models can be determined using the lmer function in the lme4 package for R. As for most model-fitting functions in R, the model is described in an lmer call by a formula, in this case including both fixed- and random-effects terms. The formula and data together determine a numerical representation of the model from which the profiled deviance or the profiled REML criterion can be evaluated as a function of some of the model parameters. The appropriate criterion is optimized, using one of the constrained optimization functions in R, to provide the parameter estimates. We describe the structure of the model, the steps in evaluating the profiled deviance or REML criterion, and the structure of classes or types that represents such a model. Sufficient detail is included to allow specialization of these structures by users who wish to write functions to fit specialized linear mixed models, such as models incorporating pedigrees or smoothing splines, that are not easily expressible in the formula language used by lmer. Journal of Statistical Software, 67 (1) ISSN:1548-7660
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Impact and effectiveness of mRNA BNT162b2 vaccine against SARS-CoV-2 infections and COVID-19 cases, hospitalisations, and deaths following a nationwide vaccination campaign in Israel: an observational study using national surveillance data

              Background Following the emergency use authorisation of the Pfizer–BioNTech mRNA COVID-19 vaccine BNT162b2 (international non-proprietary name tozinameran) in Israel, the Ministry of Health (MoH) launched a campaign to immunise the 6·5 million residents of Israel aged 16 years and older. We estimated the real-world effectiveness of two doses of BNT162b2 against a range of SARS-CoV-2 outcomes and to evaluate the nationwide public-health impact following the widespread introduction of the vaccine. Methods We used national surveillance data from the first 4 months of the nationwide vaccination campaign to ascertain incident cases of laboratory-confirmed SARS-CoV-2 infections and outcomes, as well as vaccine uptake in residents of Israel aged 16 years and older. Vaccine effectiveness against SARS-CoV-2 outcomes (asymptomatic infection, symptomatic infection, and COVID-19-related hospitalisation, severe or critical hospitalisation, and death) was calculated on the basis of incidence rates in fully vaccinated individuals (defined as those for whom 7 days had passed since receiving the second dose of vaccine) compared with rates in unvaccinated individuals (who had not received any doses of the vaccine), with use of a negative binomial regression model adjusted for age group (16–24, 25–34, 35–44, 45–54, 55–64, 65–74, 75–84, and ≥85 years), sex, and calendar week. The proportion of spike gene target failures on PCR test among a nationwide convenience-sample of SARS-CoV-2-positive specimens was used to estimate the prevelance of the B.1.1.7 variant. Findings During the analysis period (Jan 24 to April 3, 2021), there were 232 268 SARS-CoV-2 infections, 7694 COVID-19 hospitalisations, 4481 severe or critical COVID-19 hospitalisations, and 1113 COVID-19 deaths in people aged 16 years or older. By April 3, 2021, 4 714 932 (72·1%) of 6 538 911 people aged 16 years and older were fully vaccinated with two doses of BNT162b2. Adjusted estimates of vaccine effectiveness at 7 days or longer after the second dose were 95·3% (95% CI 94·9–95·7; incidence rate 91·5 per 100 000 person-days in unvaccinated vs 3·1 per 100 000 person-days in fully vaccinated individuals) against SARS-CoV-2 infection, 91·5% (90·7–92·2; 40·9 vs 1·8 per 100 000 person-days) against asymptomatic SARS-CoV-2 infection, 97·0% (96·7–97·2; 32·5 vs 0·8 per 100 000 person-days) against symptomatic COVID-19, 97·2% (96·8–97·5; 4·6 vs 0·3 per 100 000 person-days) against COVID-19-related hospitalisation, 97·5% (97·1–97·8; 2·7 vs 0·2 per 100 000 person-days) against severe or critical COVID-19-related hospitalisation, and 96·7% (96·0–97·3; 0·6 vs 0·1 per 100 000 person-days) against COVID-19-related death. In all age groups, as vaccine coverage increased, the incidence of SARS-CoV-2 outcomes declined. 8006 of 8472 samples tested showed a spike gene target failure, giving an estimated prevalence of the B.1.1.7 variant of 94·5% among SARS-CoV-2 infections. Interpretation Two doses of BNT162b2 are highly effective across all age groups (≥16 years, including older adults aged ≥85 years) in preventing symptomatic and asymptomatic SARS-CoV-2 infections and COVID-19-related hospitalisations, severe disease, and death, including those caused by the B.1.1.7 SARS-CoV-2 variant. There were marked and sustained declines in SARS-CoV-2 incidence corresponding to increasing vaccine coverage. These findings suggest that COVID-19 vaccination can help to control the pandemic. Funding None.
                Bookmark

                Author and article information

                Journal
                Lancet Gastroenterol Hepatol
                Lancet Gastroenterol Hepatol
                The Lancet. Gastroenterology & Hepatology
                The Author(s). Published by Elsevier Ltd.
                2468-1253
                9 September 2022
                9 September 2022
                Affiliations
                [a ]Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
                [b ]Department of Infectious Disease, Imperial College London, London, UK
                [c ]Department of Surgery and Cancer, Imperial College London, London, UK
                [d ]Department of Immunology and Inflammation, Imperial College London, London, UK
                [e ]Department of Gastroenterology, Imperial College Healthcare NHS Trust, London, UK
                [f ]Division of Medicine and Integrated Care, Imperial College Healthcare NHS Trust, London, UK
                [g ]Department of Gastroenterology, St Mark's Hospital and Academic Institute, London, UK
                [h ]Exeter Inflammatory Bowel Disease and Pharmacogenetics Research Group, University of Exeter, Exeter, UK
                [i ]Department of Clinical Chemistry, Biochemistry, Exeter Clinical Laboratory International, Royal Devon and Exeter NHS Foundation Trust, Exeter, UK
                [j ]Department of Gastroenterology, Royal Devon and Exeter NHS Foundation Trust, Exeter, UK
                [k ]Department of Gastroenterology, Western General Hospital, NHS Lothian, Edinburgh, UK
                [l ]Centre for Inflammation Research, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, UK
                [m ]Nightingale-Saunders Clinical Trials and Epidemiology Unit, King's Clinical Trials Unit, King's College London, London, UK
                [n ]School of Immunology and Microbial Sciences, King's College London, London, UK
                [o ]Department of Gastroenterology, Hull University Teaching Hospitals NHS Trust, Hull, UK
                [p ]Hull York Medical School, University of Hull, Hull, UK
                [q ]Department of Gastroenterology, Guy's and St Thomas' NHS Foundation Trust, London, UK
                [r ]Lung Division, Royal Brompton and Harefield Hospitals, Guy's and St Thomas' NHS Foundation Trust, London, UK
                [s ]Department of Gastroenterology, King's College Hospital, London, UK
                [t ]The NIHR Bioresource, University of Cambridge, Cambridge, UK
                [u ]Department of Gastroenterology, Cambridge University Hospitals NHS Trust, Cambridge, UK
                [v ]Department of Gastroenterology, Bart's Health NHS Trust, London, UK
                [w ]Department of Gastroenterology, St George's Hospital NHS Trust, London, UK
                Author notes
                [* ]Correspondence to: Dr Nick Powell, Department of Metabolism, Digestion and Reproduction, Imperial College, London W12 0NN, UK
                [*]

                Equal contribution

                [†]

                A full list of VIP study investigators is in the appendix (p 9)

                Article
                S2468-1253(22)00274-6
                10.1016/S2468-1253(22)00274-6
                9458592
                36088954
                abd733e7-65c2-4ac0-9e1c-66c046b20d19
                © 2022 The Author(s). Published by Elsevier Ltd. This is an Open Access article under the CC BY 4.0 license

                Since January 2020 Elsevier has created a COVID-19 resource centre with free information in English and Mandarin on the novel coronavirus COVID-19. The COVID-19 resource centre is hosted on Elsevier Connect, the company's public news and information website. Elsevier hereby grants permission to make all its COVID-19-related research that is available on the COVID-19 resource centre - including this research content - immediately available in PubMed Central and other publicly funded repositories, such as the WHO COVID database with rights for unrestricted research re-use and analyses in any form or by any means with acknowledgement of the original source. These permissions are granted for free by Elsevier for as long as the COVID-19 resource centre remains active.

                History
                Categories
                Articles

                Comments

                Comment on this article

                scite_

                Similar content172

                Cited by24

                Most referenced authors1,665