11
views
0
recommends
+1 Recommend
1 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Development and validation of multivariable prediction models of serological response to SARS-CoV-2 vaccination in kidney transplant recipients

      research-article

      Read this article at

      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

          Repeated vaccination against SARS-CoV-2 increases serological response in kidney transplant recipients (KTR) with high interindividual variability. No decision support tool exists to predict SARS-CoV-2 vaccination response to third or fourth vaccination in KTR. We developed, internally and externally validated five different multivariable prediction models of serological response after the third and fourth vaccine dose against SARS-CoV-2 in previously seronegative, COVID-19-naïve KTR. Using 20 candidate predictor variables, we applied statistical and machine learning approaches including logistic regression (LR), least absolute shrinkage and selection operator (LASSO)-regularized LR, random forest, and gradient boosted regression trees. For development and internal validation, data from 590 vaccinations were used. External validation was performed in four independent, international validation cohorts comprising 191, 184, 254, and 323 vaccinations, respectively. LASSO-regularized LR performed on the whole development dataset yielded a 20- and 10-variable model, respectively. External validation showed AUC-ROC of 0.840, 0.741, 0.816, and 0.783 for the sparser 10-variable model, yielding an overall performance 0.812. A 10-variable LASSO-regularized LR model predicts vaccination response in KTR with good overall accuracy. Implemented as an online tool, it can guide decisions whether to modulate immunosuppressive therapy before additional active vaccination, or to perform passive immunization to improve protection against COVID-19 in previously seronegative, COVID-19-naïve KTR.

          Related collections

          Most cited references33

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

          Neutralizing antibody levels are highly predictive of immune protection from symptomatic SARS-CoV-2 infection

          Predictive models of immune protection from COVID-19 are urgently needed to identify correlates of protection to assist in the future deployment of vaccines. To address this, we analyzed the relationship between in vitro neutralization levels and the observed protection from severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection using data from seven current vaccines and from convalescent cohorts. We estimated the neutralization level for 50% protection against detectable SARS-CoV-2 infection to be 20.2% of the mean convalescent level (95% confidence interval (CI) = 14.4-28.4%). The estimated neutralization level required for 50% protection from severe infection was significantly lower (3% of the mean convalescent level; 95% CI = 0.7-13%, P = 0.0004). Modeling of the decay of the neutralization titer over the first 250 d after immunization predicts that a significant loss in protection from SARS-CoV-2 infection will occur, although protection from severe disease should be largely retained. Neutralization titers against some SARS-CoV-2 variants of concern are reduced compared with the vaccine strain, and our model predicts the relationship between neutralization and efficacy against viral variants. Here, we show that neutralization level is highly predictive of immune protection, and provide an evidence-based model of SARS-CoV-2 immune protection that will assist in developing vaccine strategies to control the future trajectory of the pandemic.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            REGN-COV2, a Neutralizing Antibody Cocktail, in Outpatients with Covid-19

            Abstract Background Recent data suggest that complications and death from coronavirus disease 2019 (Covid-19) may be related to high viral loads. Methods In this ongoing, double-blind, phase 1–3 trial involving nonhospitalized patients with Covid-19, we investigated two fully human, neutralizing monoclonal antibodies against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike protein, used in a combined cocktail (REGN-COV2) to reduce the risk of the emergence of treatment-resistant mutant virus. Patients were randomly assigned (1:1:1) to receive placebo, 2.4 g of REGN-COV2, or 8.0 g of REGN-COV2 and were prospectively characterized at baseline for endogenous immune response against SARS-CoV-2 (serum antibody–positive or serum antibody–negative). Key end points included the time-weighted average change in viral load from baseline (day 1) through day 7 and the percentage of patients with at least one Covid-19–related medically attended visit through day 29. Safety was assessed in all patients. Results Data from 275 patients are reported. The least-squares mean difference (combined REGN-COV2 dose groups vs. placebo group) in the time-weighted average change in viral load from day 1 through day 7 was −0.56 log10 copies per milliliter (95% confidence interval [CI], −1.02 to −0.11) among patients who were serum antibody–negative at baseline and −0.41 log10 copies per milliliter (95% CI, −0.71 to −0.10) in the overall trial population. In the overall trial population, 6% of the patients in the placebo group and 3% of the patients in the combined REGN-COV2 dose groups reported at least one medically attended visit; among patients who were serum antibody–negative at baseline, the corresponding percentages were 15% and 6% (difference, −9 percentage points; 95% CI, −29 to 11). The percentages of patients with hypersensitivity reactions, infusion-related reactions, and other adverse events were similar in the combined REGN-COV2 dose groups and the placebo group. Conclusions In this interim analysis, the REGN-COV2 antibody cocktail reduced viral load, with a greater effect in patients whose immune response had not yet been initiated or who had a high viral load at baseline. Safety outcomes were similar in the combined REGN-COV2 dose groups and the placebo group. (Funded by Regeneron Pharmaceuticals and the Biomedical and Advanced Research and Development Authority of the Department of Health and Human Services; ClinicalTrials.gov number, NCT04425629.)
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Transparent Reporting of a multivariable prediction model for Individual Prognosis or Diagnosis (TRIPOD): the TRIPOD statement.

              Prediction models are developed to aid health care providers in estimating the probability or risk that a specific disease or condition is present (diagnostic models) or that a specific event will occur in the future (prognostic models), to inform their decision making. However, the overwhelming evidence shows that the quality of reporting of prediction model studies is poor. Only with full and clear reporting of information on all aspects of a prediction model can risk of bias and potential usefulness of prediction models be adequately assessed. The Transparent Reporting of a multivariable prediction model for Individual Prognosis Or Diagnosis (TRIPOD) Initiative developed a set of recommendations for the reporting of studies developing, validating, or updating a prediction model, whether for diagnostic or prognostic purposes. This article describes how the TRIPOD Statement was developed. An extensive list of items based on a review of the literature was created, which was reduced after a Web-based survey and revised during a 3-day meeting in June 2011 with methodologists, health care professionals, and journal editors. The list was refined during several meetings of the steering group and in e-mail discussions with the wider group of TRIPOD contributors. The resulting TRIPOD Statement is a checklist of 22 items, deemed essential for transparent reporting of a prediction model study. The TRIPOD Statement aims to improve the transparency of the reporting of a prediction model study regardless of the study methods used. The TRIPOD Statement is best used in conjunction with the TRIPOD explanation and elaboration document. To aid the editorial process and readers of prediction model studies, it is recommended that authors include a completed checklist in their submission (also available at www.tripod-statement.org).
                Bookmark

                Author and article information

                Contributors
                Journal
                Front Immunol
                Front Immunol
                Front. Immunol.
                Frontiers in Immunology
                Frontiers Media S.A.
                1664-3224
                04 October 2022
                2022
                04 October 2022
                : 13
                : 997343
                Affiliations
                [1] 1 Department of Nephrology and Medical Intensive Care, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health , Berlin, Germany
                [2] 2 Department of Nephrology, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University , Düsseldorf, Germany
                [3] 3 Division of Nephrology and Dialysis, Department of Internal Medicine III, Medical University Vienna , Vienna, Austria
                [4] 4 Department of Nephrology and Transplantation, University Hospitals of Strasbourg, INSERM Unit 1109 , Strasbourg, France
                [5] 5 Institut de Transplantation Urologie Néphrologie, Centre Hospitalier Universitaire de Nantes, Centre de Recherche en Transplantation et Immunologie, UMR 1064, INSERM, Nantes Université , Nantes, France
                [6] 6 Berlin Institute of Health , Berlin, Germany
                Author notes

                Edited by: Elke Bergmann-Leitner, Walter Reed Army Institute of Research, United States

                Reviewed by: Pinyi Lu, National Cancer Institute at Frederick (NIH), United States; Achmad Efendi, University of Brawijaya, Indonesia

                *Correspondence: Bilgin Osmanodja, bilgin.osmanodja@ 123456charite.de

                †ORCID: Bilgin Osmanodja, orcid.org/0000-0002-8660-0722; Johannes Stegbauer, orcid.org/0000-0001-8994-8102; Andreas Heinzel, orcid.org/0000-0002-8447-1795; Roman Reindl-Schwaighofer, orcid.org/0000-0002-4419-6282; Rainer Oberbauer, orcid.org/0000-0001-7544-6275; Ilies Benotmane, orcid.org/0000-0001-9113-2479; Sophie Caillard, orcid.org/0000-0002-0525-4291; Christophe Masset, orcid.org/0000-0002-7442-2164; Clarisse Kerleau, orcid.org/0000-0002-1487-5743; Gilles Blancho, orcid.org/0000-0003-0356-5069; Klemens Budde, orcid.org/0000-0002-7929-5942; Michael Mikhailov, orcid.org/0000-0002-7624-9668; Eva Schrezenmeier, orcid.org/0000-0002-0016-7885; Simon Ronicke, orcid.org/0000-0001-8822-4268

                This article was submitted to Vaccines and Molecular Therapeutics, a section of the journal Frontiers in Immunology

                Article
                10.3389/fimmu.2022.997343
                9576943
                36268021
                2ebc8984-261b-407d-954a-418edc94dbb1
                Copyright © 2022 Osmanodja, Stegbauer, Kantauskaite, Rump, Heinzel, Reindl-Schwaighofer, Oberbauer, Benotmane, Caillard, Masset, Kerleau, Blancho, Budde, Grunow, Mikhailov, Schrezenmeier and Ronicke

                This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

                History
                : 18 July 2022
                : 20 September 2022
                Page count
                Figures: 5, Tables: 8, Equations: 1, References: 33, Pages: 16, Words: 9330
                Categories
                Immunology
                Original Research

                Immunology
                kidney transplantation,covid-19,vaccination,clinical decision support,immunosuppression therapy

                Comments

                Comment on this article