TY - JOUR T1 - A mathematical model to identify optimal combinations of drug targets for dupilumab poor responders in atopic dermatitis JF - medRxiv DO - 10.1101/2021.02.08.21251317 SP - 2021.02.08.21251317 AU - Takuya Miyano AU - Alan D Irvine AU - Reiko J Tanaka Y1 - 2021/01/01 UR - http://medrxiv.org/content/early/2021/02/10/2021.02.08.21251317.abstract N2 - Background Several biologic drugs for atopic dermatitis (AD) have demonstrated good efficacy in clinical trials, but with a substantial proportion of patients being identified as poor responders. This study aims to understand the pathophysiological backgrounds of patient variability in drug response, especially for dupilumab, and to identify promising drug targets in dupilumab poor responders.Methods We conducted model-based meta-analysis of recent clinical trials of AD biologics and developed a mathematical model that reproduces reported clinical efficacies for nine biological drugs (dupilumab, lebrikizumab, tralokinumab, secukinumab, fezakinumab, nemolizumab, tezepelumab, GBR 830, and recombinant interferon-gamma) by describing systems-level AD pathogenesis. Using this model, we simulated the clinical efficacy of hypothetical therapies on virtual patients.Results IL-13 in the skin was affirmed, by the global sensitivity analysis of our model, as a potential predictive biomarker to stratify dupilumab good responders. The model simulation identified simultaneous inhibition of IL-13 and IL-22 as a promising drug target for dupilumab poor responders, whereas inhibition of either IL-13 or IL-22 alone in these non-responders was ineffective.Conclusion We present a mathematical model of AD pathogenesis developed by integration of clinical efficacy data of multiple drugs. This model will serve as a computational platform for model-informed drug development for precision medicine, as it allows evaluation of the effects of new potential drug targets, including combination therapeutics, at an individual patient level and the mechanisms behind patient variability in drug response. Similar mathematical models can be developed for other diseases and drugs, for patient stratification and identification of predictive biomarkers.Competing Interest StatementADI has received honorarium for consultancy from AbbVie, Arena Pharmaceuticals, Benevolent AI, Chugai, Dermavant, Genentech, LEO Pharma, Lilly, Menlo Therapeutics, Novartis, Pfizer, Regeneron, Sanofi, and UCB. The other authors declare no competing interests for this work.Funding StatementThis work was funded by the British Skin Foundation (005/R/18).Author DeclarationsI confirm all relevant ethical guidelines have been followed, and any necessary IRB and/or ethics committee approvals have been obtained.YesThe details of the IRB/oversight body that provided approval or exemption for the research described are given below:N/AAll necessary patient/participant consent has been obtained and the appropriate institutional forms have been archived.YesI understand that all clinical trials and any other prospective interventional studies must be registered with an ICMJE-approved registry, such as ClinicalTrials.gov. I confirm that any such study reported in the manuscript has been registered and the trial registration ID is provided (note: if posting a prospective study registered retrospectively, please provide a statement in the trial ID field explaining why the study was not registered in advance).YesI have followed all appropriate research reporting guidelines and uploaded the relevant EQUATOR Network research reporting checklist(s) and other pertinent material as supplementary files, if applicable.YesThe code of the QSP model is available. https://github.com/Tanaka-Group/AD_QSP_model ER -