Abstract
Reliable prediction and prevention of adverse drug reactions (ADRs) remains a key challenge in the development of new medicines. Advanced mathematical and computational modelling approaches, which incorporate cutting-edge mechanistic understanding of ADRs in concert with systematically collected data addressing knowledge gaps, are integral components of model-informed drug discovery and development (MID3). These approaches provide a precise, quantitative framework for predicting and mitigating safety risks in the earliest phases of drug development. Here, we highlight recent developments in the burgeoning field of quantitative systems toxicology (QST), including insights into the current state-of-the-art, as well as outcomes from the Innovative Medicines Initiative (IMI) 2 TransQST project. QST models that describe the disruption of cardiovascular, gastrointestinal, hepatic and renal physiological functions following drug exposure are presented, along with recommendations for their application in drug discovery and development.
This is a preview of subscription content, access via your institution
Access options
Access Nature and 54 other Nature Portfolio journals
Get Nature+, our best-value online-access subscription
$32.99 / 30 days
cancel any time
Subscribe to this journal
Receive 12 print issues and online access
$259.00 per year
only $21.58 per issue
Buy this article
- Purchase on SpringerLink
- Instant access to the full article PDF.
USD 39.95
Prices may be subject to local taxes which are calculated during checkout



Similar content being viewed by others
References
Guengerich, F. P. Mechanisms of drug toxicity and relevance to pharmaceutical development. Drug Metab. Pharmacokinet. 26, 3–14 (2011).
Cook, D. et al. Lessons learned from the fate of AstraZeneca’s drug pipeline: a five-dimensional framework. Nat. Rev. Drug Discov. 13, 419–431 (2014).
Mehta, K. et al. Modernizing preclinical drug development: the role of new approach methodologies. ACS Pharmacol. Transl. Sci. 8, 1513–1525 (2025).
Schmeisser, S. et al. New approach methodologies in human regulatory toxicology — not if, but how and when!. Environ. Int. 178, 108082 (2023).
Lauwereyns, J. et al. Toward a common interpretation of the 3Rs principles in animal research. Lab Anim. 53, 347–350 (2024).
Han, J. J. FDA Modernization Act 2.0 allows for alternatives to animal testing. Artif. Organs 47, 449–450 (2023).
Huang, S.-M., Abernethy, D. R., Wang, Y., Zhao, P. & Zineh, I. The utility of modeling and simulation in drug development and regulatory review. J. Pharm. Sci. 102, 2912–2923 (2013).
Alasmari, M. S. et al. Model-informed drug discovery and development approaches to inform clinical trial design and regulatory decisions: a primer for the MENA region. Saudi Pharm. J. 32, 102207 (2024).
Hütt, M.-T. in Emergence and Modularity in Life Sciences (eds Wegner, L. H. & Lüttge, U.) 37–49 (Springer, 2019).
Bloomingdale, P. et al. Quantitative systems toxicology. Curr. Opin. Toxicol. 4, 79–87 (2017).
Beattie, K. A. et al. Quantitative systems toxicology modeling in pharmaceutical research and development: an industry-wide survey and selected case study examples. CPT Pharmacomet. Syst. Pharmacol. 13, 2036–2051 (2024).
Ferreira, S. et al. Quantitative systems toxicology modeling to address key safety questions in drug development: a focus of the TransQST consortium. Chem. Res. Toxicol. 33, 7–9 (2020).
Bai, J. P. F. et al. Landscape of regulatory quantitative systems pharmacology submissions to the U.S. Food and Drug Administration: an update report. CPT Pharmacomet. Syst. Pharmacol. 13, 2102–2110 (2024).
Baker, R. E., Peña, J.-M., Jayamohan, J. & Jérusalem, A. Mechanistic models versus machine learning, a fight worth fighting for the biological community? Biol. Lett. 14, 20170660 (2018).
Upton, R. N. & Mould, D. R. Basic concepts in population modeling, simulation, and model-based drug development: part 3 — introduction to pharmacodynamic modeling methods. CPT Pharmacomet. Syst. Pharmacol. 3, e88 (2014).
Gabrielsson, J. Pharmacokinetic/Pharmacodynamic Data Analysis: Concepts and Applications (Apotekarsocieteten, 2000).
Jones, H. M., Gardner, I. B. & Watson, K. J. Modelling and PBPK simulation in drug discovery. AAPS J. 11, 155–166 (2009).
Jeon, J. Y., Ayyar, V. S. & Mitra, A. Pharmacokinetic and pharmacodynamic modeling of siRNA therapeutics — a minireview. Pharm. Res. 39, 1749–1759 (2022).
Choules, M. P. et al. Physiologically based pharmacokinetic model to predict drug-drug interactions with the antibody-drug conjugate enfortumab vedotin. J. Pharmacokinet. Pharmacodyn. 51, 417–428 (2024).
Bradshaw, E. L. et al. Applications of quantitative systems pharmacology in model-informed drug discovery: perspective on impact and opportunities. CPT Pharmacomet. Syst. Pharmacol. 8, 777–791 (2019).
Malik-Sheriff, R. S. et al. BioModels — 15 years of sharing computational models in life science. Nucleic Acids Res. 48, D407–D415 (2020).
Glont, M. et al. BioModels parameters: a treasure trove of parameter values from published systems biology models. Bioinformatics 36, 4649–4654 (2020).
Tiwari, K. et al. Reproducibility in systems biology modelling. Mol. Syst. Biol. 17, e9982 (2021).
Brennan, R. J. in Drug Safety Evaluation: Methods and Protocols (ed. Gautier, J.-C.) 213–228 (Springer, 2017).
Snelder, N. et al. Characterization and prediction of cardiovascular effects of fingolimod and siponimod using a systems pharmacology modeling approach. J. Pharmacol. Exp. Ther. 360, 356–367 (2017).
Su, H., Koomen, J. V., Eleveld, D. J., Struys, M. M. R. F. & Colin, P. J. Pharmacodynamic mechanism-based interaction model for the haemodynamic effects of remifentanil and propofol in healthy volunteers. Br. J. Anaesth. 131, 222–233 (2023).
Fu, Y., Snelder, N., Guo, T., van der Graaf, P. H. & van Hasselt, J. G. C. Evaluation of a cardiovascular systems model for design and analysis of hemodynamic safety studies. Pharmaceutics 15, 1175 (2023).
International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Human Use. General Principles for Model-Informed Drug Development (ICH Guideline M15: Step 2b Draft) (ICH, 2024).
Rudmann, D. G. On-target and off-target-based toxicologic effects. Toxicol. Pathol. 41, 310–314 (2013).
Gall, L. et al. A dynamic model of the intestinal epithelium integrates multiple sources of preclinical data and enables clinical translation of drug-induced toxicity. CPT Pharmacomet. Syst. Pharmacol. 12, 1511–1528 (2023).
Fang, H. et al. Exploring the FDA Adverse Event Reporting System (FAERS) to generate hypotheses for disease monitoring. Clin. Pharmacol. Ther. 95, 496–498 (2014).
Kuhn, M., Letunic, I., Jensen, L. J. & Bork, P. The SIDER database of drugs and side effects. Nucleic Acids Res. 44, D1075–D1079 (2016).
Sarkans, U. et al. The bioStudies database — one stop shop for all data supporting a life sciences study. Nucleic Acids Res. 46, D1266–D1270 (2018).
Ochoa, D. et al. The next-generation open targets platform: reimagined, redesigned, rebuilt. Nucleic Acids Res. 51, D1353–D1359 (2023).
Destere, A. et al. Drug-induced cardiac toxicity and adverse drug reactions, a narrative review. Therapies 79, 161–172 (2024).
Qu, Y., Li, T., Liu, Z., Li, D. & Tong, W. DICTrank: the largest reference list of 1318 human drugs ranked by risk of drug-induced cardiotoxicity using FDA labeling. Drug Discov. Today 28, 103770 (2023).
Mamoshina, P., Rodriguez, B. & Bueno-Orovio, A. Toward a broader view of mechanisms of drug cardiotoxicity. Cell Rep. Med. 2, 100216 (2021).
Strauss, D. G. et al. Comprehensive In Vitro Proarrhythmia Assay (CiPA) update from a Cardiac Safety Research Consortium/Health and Environmental Sciences Institute/FDA meeting. Ther. Innov. Regul. Sci. 53, 519–525 (2019).
Pang, P. D., Ahmed, S. M., Nishiga, M., Stockbridge, N. L. & Wu, J. C. Tackling the challenges of new approach methods for predicting drug effects from model systems. Nat. Rev. Drug Discov. 23, 565–566 (2024).
Passini, E. et al. The virtual assay software for human in silico drug trials to augment drug cardiac testing. J. Comput. Sci. 52, 101202 (2021).
Passini, E. et al. Drug-induced shortening of the electromechanical window is an effective biomarker for in silico prediction of clinical risk of arrhythmias. Br. J. Pharmacol. 176, 3819–3833 (2019).
Tylutki, Z., Mendyk, A. & Polak, S. Physiologically based pharmacokinetic-quantitative systems toxicology and safety (PBPK-QSTS) modeling approach applied to predict the variability of amitriptyline pharmacokinetics and cardiac safety in populations and in individuals. J. Pharmacokinet. Pharmacodyn. 45, 663–677 (2018).
Mohr, M., Chambard, J.-M., Ballet, V. & Schmidt, F. Accurate in silico simulation of the rabbit Purkinje fiber electrophysiological assay to facilitate early pharmaceutical cardiosafety assessment: dream or reality? J. Pharmacol. Toxicol. Methods 115, 107172 (2022).
Marian, A. J. Molecular genetic basis of hypertrophic cardiomyopathy. Circ. Res. 128, 1533–1553 (2021).
Margara, F. et al. In-silico human electro-mechanical ventricular modelling and simulation for drug-induced pro-arrhythmia and inotropic risk assessment. Prog. Biophys. Mol. Biol. 159, 58–74 (2021).
Trovato, C. et al. In silico predictions of drug-induced changes in human cardiac contractility align with experimental recordings. Front. Pharmacol. 16, 1500668 (2025).
Zhou, X. et al. Clinical phenotypes in acute and chronic infarction explained through human ventricular electromechanical modelling and simulations. eLife 13, RP93002 (2024).
Wang, Z. J. et al. Human biventricular electromechanical simulations on the progression of electrocardiographic and mechanical abnormalities in post-myocardial infarction. EP Eur. 23, i143–i152 (2021).
Margara, F. et al. Mechanism based therapies enable personalised treatment of hypertrophic cardiomyopathy. Sci. Rep. 12, 22501 (2022).
Trovato, C., Mohr, M., Schmidt, F., Passini, E. & Rodriguez, B. Cross clinical-experimental-computational qualification of in silico drug trials on human cardiac Purkinje cells for proarrhythmia risk prediction. Front. Toxicol. 4, 992650 (2022).
Beattie, K. A. et al. Evaluation of an in silico cardiac safety assay: using ion channel screening data to predict QT interval changes in the rabbit ventricular wedge. J. Pharmacol. Toxicol. Methods 68, 88–96 (2013).
Corral-Acero, J. et al. The ‘digital twin’ to enable the vision of precision cardiology. Eur. Heart J. 41, 4556–4564 (2020).
Viceconti, M. et al. In silico trials: verification, validation and uncertainty quantification of predictive models used in the regulatory evaluation of biomedical products. Methods San Diego Calif. 185, 120–127 (2021).
Musuamba, F. T. et al. Scientific and regulatory evaluation of mechanistic in silico drug and disease models in drug development: building model credibility. CPT Pharmacomet. Syst. Pharmacol. 10, 804–825 (2021).
US Food and Drug Administration. Assessment of Pressor Effects of Drugs Guidance for Industry (FDA, 2022).
Fu, Y. et al. A novel cardiovascular systems model to quantify drugs effects on the inter-relationship between contractility and other hemodynamic variables. CPT Pharmacomet. Syst. Pharmacol. 11, 640–652 (2022).
Snelder, N. et al. Drug effects on the CVS in conscious rats: separating cardiac output into heart rate and stroke volume using PKPD modelling. Br. J. Pharmacol. 171, 5076–5092 (2014).
Venkatasubramanian, R., Collins, T. A., Lesko, L. J., Mettetal, J. T. & Trame, M. N. Semi-mechanistic modelling platform to assess cardiac contractility and haemodynamics in preclinical cardiovascular safety profiling of new molecular entities. Br. J. Pharmacol. 177, 3568–3590 (2020).
Stein, A., Voigt, W. & Jordan, K. Chemotherapy-induced diarrhea: pathophysiology, frequency and guideline-based management. Ther. Adv. Med. Oncol. 2, 51–63 (2010).
Saltz, L. B. et al. Irinotecan plus fluorouracil and leucovorin for metastatic colorectal cancer. N. Engl. J. Med. 343, 905–914 (2000).
Saltz, L. B. et al. Irinotecan plus fluorouracil/leucovorin for metastatic colorectal cancer: a new survival standard. Oncologist 6, 81–91 (2001).
McQuade, R. M. et al. Gastrointestinal dysfunction and enteric neurotoxicity following treatment with anticancer chemotherapeutic agent 5-fluorouracil. Neurogastroenterol. Motil. 28, 1861–1875 (2016).
Peters, M. F. et al. Developing in vitro assays to transform gastrointestinal safety assessment: potential for microphysiological systems. Lab Chip 20, 1177–1190 (2020).
Shankaran, H. et al. Systems pharmacology model of gastrointestinal damage predicts species differences and optimizes clinical dosing schedules. CPT Pharmacomet. Syst. Pharmacol. 7, 26–33 (2018).
Secombe, K. R. et al. Diarrhea induced by small molecule tyrosine kinase inhibitors compared with chemotherapy: potential role of the microbiome. Integr. Cancer Ther. 19, 1534735420928493 (2020).
Fragkos, K. C. & Forbes, A. Citrulline as a marker of intestinal function and absorption in clinical settings: a systematic review and meta-analysis. U Eur. Gastroenterol. J. 6, 181–191 (2018).
Peters, M. F. et al. Human 3D gastrointestinal microtissue barrier function as a predictor of drug-induced diarrhea. Toxicol. Sci. 168, 3–17 (2019).
Van Liedekerke, P., Palm, M. M., Jagiella, N. & Drasdo, D. Simulating tissue mechanics with agent-based models: concepts, perspectives and some novel results. Comput. Part. Mech. 2, 401–444 (2015).
Gehart, H. & Clevers, H. Tales from the crypt: new insights into intestinal stem cells. Nat. Rev. Gastroenterol. Hepatol. 16, 19–34 (2019).
Meineke, F. A., Potten, C. S. & Loeffler, M. Cell migration and organization in the intestinal crypt using a lattice-free model. Cell Prolif. 34, 253–266 (2001).
Pitt-Francis, J. et al. Chaste: a test-driven approach to software development for biological modelling. Comput. Phys. Commun. 180, 2452–2471 (2009).
Buske, P. et al. A comprehensive model of the spatio-temporal stem cell and tissue organisation in the intestinal crypt. PLoS Comput. Biol. 7, e1001045 (2011).
Gall, L. et al. Homeostasis, injury, and recovery dynamics at multiple scales in a self-organizing mouse intestinal crypt. eLife 12, e85478 (2023).
Zhang, M. et al. CDK inhibitors in cancer therapy, an overview of recent development. Am. J. Cancer Res. 11, 1913–1935 (2021).
Helleday, T., Petermann, E., Lundin, C., Hodgson, B. & Sharma, R. A. DNA repair pathways as targets for cancer therapy. Nat. Rev. Cancer 8, 193–204 (2008).
Tan, S. H. et al. A constant pool of Lgr5+ intestinal stem cells is required for intestinal homeostasis. Cell Rep. 34, 108633 (2021).
Azkanaz, M. et al. Retrograde movements determine effective stem cell numbers in the intestine. Nature 607, 548–554 (2022).
Jadalannagari, S. & Ewart, L. Beyond the hype and toward application: liver complex in vitro models in preclinical drug safety. Expert Opin. Drug Metab. Toxicol. 20, 607–619 (2024).
Siramshetty, V. B. et al. WITHDRAWN — a resource for withdrawn and discontinued drugs. Nucleic Acids Res. 44, D1080–D1086 (2016).
Weaver, R. J. et al. Managing the challenge of drug-induced liver injury: a roadmap for the development and deployment of preclinical predictive models. Nat. Rev. Drug Discov. 19, 131–148 (2020).
Watkins, P. B. The DILI-sim initiative: insights into hepatotoxicity mechanisms and biomarker interpretation. Clin. Transl. Sci. 12, 122–129 (2019).
Watkins, P. B. Quantitative systems toxicology approaches to understand and predict drug-induced liver injury. Clin. Liver Dis. 24, 49–60 (2020).
Battista, C., Howell, B. A., Siler, S. Q. & Watkins, P. B. An introduction to DILIsym® software, a mechanistic mathematical representation of drug-induced liver injury. Methods Pharmacol. Toxicol. https://doi.org/10.1007/978-1-4939-7677-5_6 (2018).
Woodhead, J. L. et al. Application of a mechanistic model to evaluate putative mechanisms of tolvaptan drug-induced liver injury and identify patient susceptibility factors. Toxicol. Sci. 155, 61–74 (2017).
Woodhead, J. L., Siler, S. Q., Howell, B. A., Watkins, P. B. & Conway, C. Comparing the liver safety profiles of 4 next-generation CGRP receptor antagonists to the hepatotoxic CGRP inhibitor telcagepant using quantitative systems toxicology modeling. Toxicol. Sci. 188, 108–116 (2022).
Beaudoin, J. J., Brock, W. J., Watkins, P. B. & Brouwer, K. L. R. Quantitative systems toxicology modeling predicts that reduced biliary efflux contributes to tolvaptan hepatotoxicity. Clin. Pharmacol. Ther. 109, 433–442 (2021).
Generaux, G. et al. Quantitative systems toxicology (QST) reproduces species differences in PF-04895162 liver safety due to combined mitochondrial and bile acid toxicity. Pharmacol. Res. Perspect. 7, e00523 (2019).
Meier, M. J. et al. Progress in toxicogenomics to protect human health. Nat. Rev. Genet. 26, 105–122 (2025).
Russomanno, G. et al. A systems approach reveals species differences in hepatic stress response capacity. Toxicol. Sci. 196, 112–125 (2023).
Thiel, C. et al. A comparative analysis of drug-induced hepatotoxicity in clinically relevant situations. PLoS Comput. Biol. 13, e1005280 (2017).
Thiel, C. et al. Model-based contextualization of in vitro toxicity data quantitatively predicts in vivo drug response in patients. Arch. Toxicol. 91, 865–883 (2017).
Maldonado, E. M. et al. Integration of genome scale metabolic networks and gene regulation of metabolic enzymes with physiologically based pharmacokinetics. CPT Pharmacomet. Syst. Pharmacol. 6, 732–746 (2017).
Berndt, N. et al. HEPATOKIN1 is a biochemistry-based model of liver metabolism for applications in medicine and pharmacology. Nat. Commun. 9, 2386 (2018).
DeGracia, D. J., Huang, Z.-F. & Huang, S. A nonlinear dynamical theory of cell injury. J. Cereb. Blood Flow. Metab. 32, 1000–1013 (2012).
Zhang, Q., Pi, J., Woods, C. G. & Andersen, M. E. Phase I to II cross-induction of xenobiotic metabolizing enzymes: a feedforward control mechanism for potential hormetic responses. Toxicol. Appl. Pharmacol. 237, 345–356 (2009).
Wijaya, L. S. et al. Integration of temporal single cell cellular stress response activity with logic-ODE modeling reveals activation of ATF4-CHOP axis as a critical predictor of drug-induced liver injury. Biochem. Pharmacol. 190, 114591 (2021).
Perazella, M. A. Drug-induced acute kidney injury: diverse mechanisms of tubular injury. Curr. Opin. Crit. Care 25, 550 (2019).
Mody, H. et al. A review on drug-induced nephrotoxicity: pathophysiological mechanisms, drug classes, clinical management, and recent advances in mathematical modeling and simulation approaches. Clin. Pharmacol. Drug Dev. 9, 896–909 (2020).
Rowland Yeo, K. & Gil Berglund, E. An integrated approach for assessing the impact of renal impairment on pharmacokinetics of drugs in development: pivotal role of PBPK modelling. Clin. Pharmacol. Ther. 110, 1168–1171 (2021).
Li, Z. et al. Modeling exposure to understand and predict kidney injury. Semin. Nephrol. 39, 176–189 (2019).
Burt, H. J. et al. Metformin and cimetidine: physiologically based pharmacokinetic modelling to investigate transporter mediated drug–drug interactions. Eur. J. Pharm. Sci. 88, 70–82 (2016).
Scotcher, D. et al. Mechanistic models as framework for understanding biomarker disposition: prediction of creatinine-drug interactions. CPT Pharmacomet. Syst. Pharmacol. 9, 282–293 (2020).
Li, Z. et al. A physiologically based in silico tool to assess the risk of drug-related crystalluria. J. Med. Chem. 63, 6489–6498 (2020).
McSweeney, K. R. et al. Mechanisms of cisplatin-induced acute kidney injury: pathological mechanisms, pharmacological interventions, and genetic mitigations. Cancers 13, 1572 (2021).
Fukushima, K. et al. Population pharmacokinetic–toxicodynamic modeling and simulation of cisplatin-induced acute renal injury in rats: effect of dosing rate on nephrotoxicity. J. Pharm. Sci. 105, 324–332 (2016).
Zhou, J. et al. The drug-resistance mechanisms of five platinum-based antitumor agents. Front. Pharmacol. 11, 343 (2020).
Gebremichael, Y. et al. Multiscale mathematical model of drug-induced proximal tubule injury: linking urinary biomarkers to epithelial cell injury and renal dysfunction. Toxicol. Sci. 162, 200–211 (2018).
Okada, A. et al. Alterations in cisplatin pharmacokinetics and its acute/sub-chronic kidney injury over multiple cycles of cisplatin treatment in rats. Biol. Pharm. Bull. 40, 1948–1955 (2017).
Gebremichael, Y. et al. Quantitative prediction of cisplatin-induced AKI using RENAsym, a mechanistic quantitative systems toxicology model, and renal proximal tubule epithelial cell in vitro assays: PO0234. J. Am. Soc. Nephrol. 31, 124 (2020).
Wijaya, L. S. et al. Spatio-temporal transcriptomic analysis reveals distinct nephrotoxicity, DNA damage, and regeneration response after cisplatin. Cell Biol. Toxicol. 41, 49 (2025).
Wilkinson, M. D. et al. The FAIR guiding principles for scientific data management and stewardship. Sci. Data 3, 160018 (2016).
Venkatakrishnan, K. & van der Graaf, P. H. Model-informed drug development: connecting the dots with a totality of evidence mindset to advance therapeutics. Clin. Pharmacol. Ther. 110, 1147–1154 (2021).
Adashi, E. Y., O’Mahony, D. P. & Cohen, I. G. The FDA Modernization Act 2.0: drug testing in animals is rendered optional. Am. J. Med. 136, 853–854 (2023).
Passini, E. et al. Human in silico drug trials demonstrate higher accuracy than animal models in predicting clinical pro-arrhythmic cardiotoxicity. Front. Physiol. 8, 668 (2017).
Smith, B. et al. Mechanistic investigations support liver safety of ubrogepant. Toxicol. Sci. 177, 84–93 (2020).
Pichardo-Almarza, C. & Kimko, H. Modeling adeno-associated virus (AAV) gene therapy: when systems biology met systems pharmacology. IFAC PapersOnline 55, 127–128 (2022).
Shah, D. K. & Betts, A. M. Antibody biodistribution coefficients: inferring tissue concentrations of monoclonal antibodies based on the plasma concentrations in several preclinical species and human. mAbs 5, 297–305 (2013).
Haddish-Berhane, N. et al. On translation of antibody drug conjugates efficacy from mouse experimental tumors to the clinic: a PK/PD approach. J. Pharmacokinet. Pharmacodyn. 40, 557–571 (2013).
Hardiansyah, D. & Ng, C. M. Quantitative systems pharmacology model of chimeric antigen receptor T-cell therapy. Clin. Transl. Sci. 12, 343–349 (2019).
Jiang, X. et al. Development of a target cell-biologics-effector cell (TBE) complex-based cell killing model to characterize target cell depletion by T cell redirecting bispecific agents. mAbs 10, 876–889 (2018).
Betts, A. & van der Graaf, P. H. Mechanistic quantitative pharmacology strategies for the early clinical development of bispecific antibodies in oncology. Clin. Pharmacol. Ther. 108, 528–541 (2020).
Verbeke, R., Hogan, M. J., Loré, K. & Pardi, N. Innate immune mechanisms of mRNA vaccines. Immunity 55, 1993–2005 (2022).
Lynch, J. J., Van Vleet, T. R., Mittelstadt, S. W. & Blomme, E. A. G. Potential functional and pathological side effects related to off-target pharmacological activity. J. Pharmacol. Toxicol. Methods 87, 108–126 (2017).
Brennan, R. J. et al. The state of the art in secondary pharmacology and its impact on the safety of new medicines. Nat. Rev. Drug Discov. 23, 525–545 (2024).
Monticello, T. M. et al. Current nonclinical testing paradigm enables safe entry to first-in-human clinical trials: the IQ consortium nonclinical to clinical translational database. Toxicol. Appl. Pharmacol. 334, 100–109 (2017).
Mendez, D. et al. ChEMBL: towards direct deposition of bioassay data. Nucleic Acids Res. 47, D930–D940 (2019).
Papatheodorou, I. et al. Expression Atlas update: from tissues to single cells. Nucleic Acids Res. 48, D77–D83 (2020).
Füzi, B., Malik-Sheriff, R. S., Manners, E. J., Hermjakob, H. & Ecker, G. F. KNIME workflow for retrieving causal drug and protein interactions, building networks, and performing topological enrichment analysis demonstrated by a DILI case study. J. Cheminform. 14, 37 (2022).
Wishart, D. S. et al. DrugBank 5.0: a major update to the DrugBank database for 2018. Nucleic Acids Res. 46, D1074–D1082 (2018).
Alexander, S. P. H. et al. The concise guide to pharmacology 2019/20: introduction and other protein targets. Br. J. Pharmacol. 176, S1–S20 (2019).
Whirl-Carrillo, M. et al. An evidence-based framework for evaluating pharmacogenomics knowledge for personalized medicine. Clin. Pharmacol. Ther. 110, 563–572 (2021).
Zhou, Y. et al. Therapeutic Target Database update 2022: facilitating drug discovery with enriched comparative data of targeted agents. Nucleic Acids Res. 50, D1398–D1407 (2022).
Valeur, E. et al. New modalities for challenging targets in drug discovery. Angew. Chem. Int. Ed. 56, 10294–10323 (2017).
Yuan, S. et al. New drug approvals for 2021: synthesis and clinical applications. Eur. J. Med. Chem. 245, 114898 (2023).
Békés, M., Langley, D. R. & Crews, C. M. PROTAC targeted protein degraders: the past is prologue. Nat. Rev. Drug Discov. 21, 181–200 (2022).
Kulkarni, J. A. et al. The current landscape of nucleic acid therapeutics. Nat. Nanotechnol. 16, 630–643 (2021).
Al Shaer, D., Al Musaimi, O., Albericio, F. & de la Torre, B. G. 2021 FDA TIDES (peptides and oligonucleotides) harvest. Pharmaceuticals 15, 222 (2022).
Beaudoin, J. J. et al. The combination of a human biomimetic liver microphysiology system with BIOLOGXsym, a quantitative systems toxicology (QST) modeling platform for macromolecules, provides mechanistic understanding of tocilizumab- and GGF2-induced liver injury. Int. J. Mol. Sci. 24, 9692 (2023).
Li, X., George, S. M., Vernetti, L., Gough, A. H. & Taylor, D. L. A glass-based, continuously zonated and vascularized human liver acinus microphysiological system (vLAMPS) designed for experimental modeling of diseases and ADME/TOX. Lab Chip 18, 2614–2631 (2018).
Longo, D. et al. Refining liver safety risk assessment: application of mechanistic modeling and serum biomarkers to cimaglermin alfa (GGF2) clinical trials. Clin. Pharmacol. Ther. 102, 961–969 (2017).
Mosedale, M. et al. Transient changes in hepatic physiology that alter bilirubin and bile acid transport may explain elevations in liver chemistries observed in clinical trials of GGF2 (cimaglermin alfa). Toxicol. Sci. 161, 401–411 (2018).
Schiff, M. H. et al. Integrated safety in tocilizumab clinical trials. Arthritis Res. Ther. 13, R141 (2011).
Acknowledgements
This project has received funding from the Innovative Medicines Initiative 2 Joint Undertaking under grant agreement number 116030. This Joint Undertaking receives support from the European Union’s Horizon 2020 research and innovation programme and EFPIA. The authors declare that this work reflects only the views of the authors and that IMI-JU is not responsible for any use that may be made of the information it contains. This paper is dedicated to K. Park, a leader in the field of drug safety, who led the work of the TransQST Consortium from 2018 to 2020. B.R. is funded by a Wellcome Trust Fellowship in Basic Biomedical Sciences (214290/Z/18/Z).
Author information
Authors and Affiliations
Corresponding authors
Ethics declarations
Competing interests
C.P. and L.G. are employees and shareholders of AstraZeneca. K.A.B., C.P.F. and E.I.R. are employees of GSK. F.S., M.M. and P.T. are employees and shareholders, and R.J.B. is a retired employee and shareholder of Sanofi. D.C., S.T. and L.L. are employees and shareholders of AbbVie. M.K. is an employee of Orion Corporation. L.L. is a shareholder of Johnson & Johnson. J.S.-R. reports in the past 3 years funding from GSK and Pfizer and fees or honoraria from Travere Therapeutics, Stadapharm, Astex, Owkin, Pfizer, Grunenthal, Tempus and Moderna. F.V. is an employee of Servier. J.A.W. is an employee of Vertex Pharmaceuticals. D.J.L. is an employee of Eli Lilly and Company. All other authors declared no competing interests for this work.
Peer review
Peer review information
Nature Reviews Drug Discovery thanks Richard Allen, Paul Watkins and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.
Additional information
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Related links
ChEMBL: https://www.ebi.ac.uk/chembl/
DrugBank data sets: https://www.drugbank.com/datasets
ExpressionAtlas: https://www.ebi.ac.uk/gxa/home
GastroPlus PBPK and PBBM software: https://www.simulations-plus.com/software/gastroplus/
GitHub PKSim: https://github.com/Open-Systems-Pharmacology/PK-Sim
IUPHAR/BPS Guide to PHARMACOLOGY: https://www.guidetopharmacology.org/
PharmGKB (ClinPGx): https://www.clinpgx.org/
Simcyp PBPK: https://www.certara.com/software/simcyp-pbpk/
Simulations Plus: https://www.simulations-plus.com/
Therapeutic Target Database: https://db.idrblab.net/ttd/
Supplementary information
Rights and permissions
Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.
About this article
Cite this article
Goldring, C.E., Russomanno, G., Pin, C. et al. Quantitative systems toxicology: modelling to mechanistically understand and predict drug safety. Nat Rev Drug Discov (2025). https://doi.org/10.1038/s41573-025-01308-z
Accepted:
Published:
Version of record:
DOI: https://doi.org/10.1038/s41573-025-01308-z


