Skip to main content
HHS Author Manuscripts logoLink to HHS Author Manuscripts
. Author manuscript; available in PMC: 2024 Sep 1.
Published in final edited form as: Food Chem Toxicol. 2023 Jul 17;179:113948. doi: 10.1016/j.fct.2023.113948

In silico modeling-based new alternative methods to predict drug and herb-induced liver injury: A review

Hyun Kil Shin 1, Ruili Huang 2,*, Minjun Chen 3,*
PMCID: PMC10640386  NIHMSID: NIHMS1921747  PMID: 37460037

Abstract

New approach methods (NAMs) have been developed to predict a wide range of toxicities through innovative technologies. Liver injury is one of the most extensively studied endpoints due to its severity and frequency, occurring among populations that consume drugs or dietary supplements. In this review, we focus on recent developments of in silico modeling for liver injury prediction using deep learning and in vitro data based on adverse outcome pathways (AOPs). Despite these models being mainly developed using datasets generated from drug-like molecules, they were also applied to the prediction of hepatotoxicity caused by herbal products. As deep learning has achieved great success in many different fields, advanced machine learning algorithms have been actively applied to improve the accuracy of in silico models. Additionally, the development of liver AOPs, combined with big data in toxicology, has been valuable in developing in silico models with enhanced predictive performance and interpretability. Specifically, one approach involves developing structure-based models for predicting molecular initiating events of liver AOPs, while others use in vitro data with structure information as model inputs for making predictions. Even though liver injury remains a difficult endpoint to predict, advancements in machine learning algorithms and the expansion of in vitro databases with relevant biological knowledge have made a huge impact on improving in silico modeling for drug-induced liver injury prediction.

1. Introduction

Due to the threats from severe disease outbreaks posed to human health (Tong et al., 2022), there is an urgent need for cost-effective and time-efficient toxicity testing methods to protect public health by accelerating the development of medicine (Baker et al., 2022). Traditional toxicity testing has relied on animal models; however, this approach has its inherent drawbacks, including substantial time and costs (Van Norman, 2019), as well as limited accuracy when extrapolating results to humans (Parish et al., 2020).

New approach methods (NAMs) are animal-free methods based on innovative technologies used to assess hazardous effects of chemicals. Cost-effectiveness and time efficiency are critical factors for NAMs, but their ability to accurately predict adverse outcomes in humans is also essential. NAMs have been widely used for cosmetics (Cronin et al., 2022), where animal testing for cosmetics and their active ingredients is banned in many countries (Sreedhar et al., 2020). The use of NAMs has also been considered for the safety evaluation of pesticides and industrial chemicals (Stucki et al., 2022; van der Zalm et al., 2022). For the evaluation of pharmaceutical toxicity, in vitro tests for predicting organ toxicity were developed (Brecklinghaus, 2020), and NAMs for biokinetics prediction can be applied (Punt et al., 2020).

Drug-induced liver injury (DILI) has caused significant loss of time and resources in drug development projects, since DILI liability is usually identified in the late stages of clinical trials (Kaplowitz, 2005). To predict DILI in the early phases of drug development, in vitro tests and in silico models have been developed. However, the lack of standardization in in vitro test often leads to contradictory results (Atienzar and Nicolas, 2018) and in silico models have yet to achieve sufficient sensitivity identifying DILI risk (Matthews et al., 2009). Despite the substantial progress made in this area, accurately predicting DILI remains a challenging endpoint.

Liver injury is also commonly associated with the use of herbal products (Lin et al., 2019). In line with the growing consumption of herbal products (Amadi and Orisakwe, 2018), reports of herb-induced liver injury (HILI) have significantly increased in past decades (Nunes et al., 2022). Herbal supplements are generally extracted from herbs, resulting in cocktails of multiple compounds with unknown toxic effects. The interactions between compounds in these mixtures could result in synergetic effects leading to HILI, highlighting the need of effective methods predicting this type of injury.

This review focuses on in silico models for the prediction of hepatotoxicity associated with the use of drugs and herbal dietary supplements. The majority of in silico models belong to (quantitative) structure-activity relationship, or (Q)SAR, models, which predict target endpoints based solely on molecular structures. To improve their prediction accuracy, in vitro data have been incorporated into these models as well. Recent developments in in vitro assay databases and the adverse outcome pathway (AOP) model have contributed significantly to the improvement of hepatotoxicity prediction by providing additional information on compounds’ mechanisms of action. Here, this review primarily concentrates on recent and notable achievements in DILI or HILI prediction to minimize redundancy with other reviews (Vall et al., 2021).

2. Hepatotoxicity phenotypes

A wide range of (Q)SAR models have been developed to predict hepatotoxicity phenotypes, which are typically categorized based on the endpoints being predicted. Most (Q)SAR studies begin with data curation; therefore, dataset curation and the model are introduced together in this section. A list of free or commercial software for predicting hepatotoxicity and relevant endpoints is shown in Table 1.

Table 1.

Selected liist of software for hepatotoxicity or relevant endpoint prediction

Name Endpoint License type Prediction models available at
ADMET predictor Elevation of DILI biomarkers
(ALP, AST, LDH)
Commercial software (GUI* program)
Derek Nexus Severity of DILI Commercial software (GUI* program)
VEGA Hepatotoxicity (Positive/Unknown/Negative) Freeware (GUI program) https://www.vegahub.eu/portfolio-item/vega-qsar/
Liver NOAEL and LOAEL
Hepatic steatosis MIEs (PXR/PPARα/PPARγ/NRF2)
ProTox–II Hepatotoxicity classification (Positive/Negative) Freeware (Web program) https://tox-new.charite.de/protox_II/
Nrf2/ARE classification
Mitochondrial membrane potential classification
ToxSTAR Binary classification of DILI indications (Cholestasis, Cirrhosis, Hepatitis, and Steatosis) Freeware (Web program) https://toxstar.kitox.re.kr/
admetSAR Hepatotoxicity classification Freeware (Web program) http://lmmd.ecust.edu.cn/admetsar2
BSEP inhibition
Mitochondrial toxicity
*

GUI: Graphical User Interface

2.1. DILI annotation

Three different data sources were used to annotate DILI risk in humans: U.S. Food and Drug Administration (FDA) labeling, clinical case reports, and literature. There is a wide range of datasets curated for DILI prediction; however, DILI annotations from different datasets were sometimes contradictory (Thakkar et al., 2018). Since there is no ground truth for DILI annotations on drugs, in silico model development studies sometimes go through data curation and define their own DILI annotations. Lack of a standardized DILI dataset is the major obstacle in validating a model since discrepancies among DILI annotations impose inherent prediction errors. To harmonize different DILI datasets, Thakkar et al. developed a DILI severity and toxicity (DILIst) dataset (Thakkar et al., 2020) by augmenting DILIrank (Chen et al., 2016b) with a large volume of human DILI datasets such as LiverTox (Hoofnagle, 2013), Suzuki et al. (Suzuki et al., 2010), Greene et al. (Greene et al., 2010), and Zhu et al. (Zhu and Kruhlak, 2014).

2.2. Curation of post-marketing case reports

DILI was often reported during the post-market phase, and the frequency of post-marketing reports was considered as evidence for assigning DILI labels to drugs. Zhu et al. (Zhu and Kruhlak, 2014) curated the FDA’s Adverse Event Reporting System (FAERS) database to annotate drugs on four endpoints: liver damage, cholestasis, liver enzyme abnormalities, and bile duct disorders, based on the standardized terms from MeDRA (Medical Dictionary for Regulatory Activities) hierarchy. Shin et al. (Shin et al., 2020) used the frequencies of post-marketing reports on four indications (cholestasis, cirrhosis, hepatitis, and steatosis) collected from the PharmaPendium database to assign positive annotations to drugs, while the negative data was obtained from DILIrank (Chen et al., 2016b). However, no databases provided drug metabolite structures with their DILI annotations. Therefore, annotations from the parent drugs were assigned to the drug metabolites in this study. ToxSTAR is an available software product for classification of the liver injury indications for drugs and drug metabolites (Shin et al., 2022a).

2.3. DILI biomarker prediction

Since liver injury is diagnosed by serum biomarkers (Robles-Díaz et al., 2016), (Q)SAR models were developed to predict abnormal increases in these biomarkers. Rodgers et al. developed models for alkaline phosphatase (ALP), alanine aminotransferase (ALT), aspartate aminotransferase (AST), lactate dehydrogenase (LDH), and gamma-glutamyl transpeptidase (GGT) based on the Human Liver Adverse Effects Database (Rodgers et al., 2010).

An available software for the DILI biomarker prediction is ADMET Predictor. Liu used ADMET Predictor to predict HILI using molecular structures of natural products-derived compounds (NPCs) (Liu, 2018). In this study, only NPCs predicted to be absorbed in the gastrointestinal tract were selected, and possible metabolites from the selected NPCs were predicted. The structures of both parent compounds and their metabolites were used to predict the abnormal elevation of the biomarkers such as ALT, AST, and LDH.

3. Advances of in silico modeling for predicting liver toxicity

3.1. In silico modeling for predicting DILI in humans

Numerous (Q)SAR models for DILI prediction were developed for binary classification of DILI-positive (hepatotoxicant) or negative (no evidence of hepatotoxicity) using machine learning algorithms (Vall et al., 2021). Some DILI prediction software is publicly available, such as ProTox-II (Banerjee et al., 2018), VEGA, and Derek Nexus (Table 1). ProTox-II uses a random forest model, whereas VEGA and Derek Nexus use rule-based models. Recently, Li et al. developed DeepDILI for DILI prediction. Usually, DILI prediction models take molecular structure as an input; however, in their study the input of the deep neural network were the outputs from a set of (Q)SAR models (Li et al., 2021). Graph neural network (GNN) can be used for node classification, edge prediction, and graph classification. Molecular structure can be represented as a graph with atoms as nodes and covalent bonds as edges, and DILI prediction can be converted into a graph classification problem, where GNN predicts DILI annotation from a molecular graph.

Xu et al. used undirected graph recursive neural networks (UGRNN) for DILI prediction in which UGRNN gathers atomic information sequentially to each root atom for encoding, and then feeds encoded vectors to the output layer for prediction. In this study, multiple DILI annotation datasets were integrated to expand the sample size and improve model training (Xu et al., 2015). Similarly, Ma et al. used a multi-view graph recursive neural networks (MV-GNN) model to encode atom and bond-oriented input representation for predicting DILI risk. Since a large volume of data is required for MV-GNN to learn appropriate molecular representation, four toxicity datasets (hERG, phospholipidosis, Ames test, and mitochondrial membrane potential) were combined with DILI annotation data and multilabel training was applied. (Ma et al., 2021). These studies demonstrate the effectiveness of deep learning methods, such as UGRNN and MV-GNN, for improving DILI prediction.

Some studies applied (Q)SAR models to predict hepatotoxicity of NPCs (Kim and Nam, 2017; Li et al., 2018). In the study by Li et al., a support vector machine model with a MACCS (Molecular ACCess Systems keys) fingerprint achieved the highest accuracy, and the model was used to identify DILI-positive components in the herbal products. Kim and Nam proposed a novel fingerprint by assigning weights on each bit of PubChem fingerprint based on Bayesian probability calculated from DILI annotations. Because both studies applied (Q)SAR models derived from synthetic drug compounds for predicting HILI, there is a potential gap in model generalization considering differences in chemical space between NPCs and synthetic drug compounds (Stratton et al., 2015). Applicability domain analysis is suggested to reduce the model’s uncertainty and improve reliability of the prediction values (Weaver and Gleeson, 2008; Sutton et al., 2020).

3.2. in silico modeling for predicting liver toxicity in animals

Currently, in vivo animal studies are mandatory submissions to a regulatory agency in support of first-in-human studies. Considering animal studies’ high cost and time commitment, animal-free NAMs are proposed, including in silico models developed from accumulated animal studies to predict in vivo outcomes. Mulliner et al. curated data based on the hierarchical endpoint tree, in which hepatotoxicity was branched into clinical chemistry findings or histological reports for defining hepatobiliary or hepatocellular injury. Binary classification was defined for humans based on clinical and post-marketing data, and preclinical data with doses of less than 500 mg/kg, respectively. The investigators identified the best performance model for the internal use of early preclinical safety in-silico workflow within their institutes. (Mulliner et al., 2016).

On the contrary, He et al. labeled drugs for hepatotoxicity by integrating human data (clinical data), animal experiment, and in vitro assay results. Thus, the model prediction is positive if evidence of hepatic injury is found in clinical studies, animal experiments, or cell-based assays (He et al., 2019). Cotterill et al. compiled hepatic steatosis datasets based on in vivo histology data and human clinical data retrieved from the literature, and a binary classification model was developed (Cotterill et al., 2020). Additionally, VEGA 1.2.0 provides a hepatotoxicity prediction model based on animal data (Table 1), which can predict NOAEL (No Observed Adverse Effect Level) and LOAEL (Lowest Observed Adverse Effect Level) in the liver.

4. In silico models for predicting hepatotoxic key events of AOPs

AOPs define series of biological events that lead to adverse outcomes, including a molecular initiating event (MIE), which is the first biological target that interacts with the molecule, and key events (KEs). AOPs could improve understanding of the systematic biological processes initiated by toxicants; thus, information defined and curated in AOPs provides a good starting point for developing in silico or in vitro NAMs (Ankley et al., 2010). The development of high-throughput screening technologies and the integration of AOPs and in vitro data has paved a way for developing mechanistically driven in silico models for DILI prediction.

Liver AOPs provide a wide range of MIEs and KEs relevant for DILI prediction. Multiple liver AOPs have been developed and are available in the AOP wiki (Table 2). The MIEs in the AOPs can be target proteins for DILI or HILI prediction. Gadaleta et al. developed an in silico model for hepatic steatosis prediction by compiling ToxCast in vitro assay data on MIEs of hepatic steatosis AOPs (Gadaleta et al., 2018). In this study, (Q)SAR models for predicting MIEs, such as PXR, LXR, AhR, NRF2, PPARα, and PPARγ, were developed and demonstrated the capability for virtually screening chemicals that can cause hepatic steatosis. Based on the key characteristics of hepatotoxicants (Rusyn et al., 2021), we selectively discuss certain target MIEs/KEs relevant to DILI, including bile salt export pump inhibition, oxidative stress, mitochondria dysfunction, and drug bioactivations, as well as the (Q)SAR models used for predicting these targets.

Table 2.

Selected list of liver AOPs from AOP wiki (accessed on 21 Mar 2023)

AOP ID Title Molecular initiating events (MIEs)
27 Cholestatic liver injury induced by inhibition of the bile salt export pump (ABCB11) Bile salt export pump inhibition (ABCB11)
34 LXR activation leading to hepatic steatosis LXR activation
PPAR promoter demethylation
36 Peroxisomal Fatty Acid Beta-Oxidation inhibition leading to steatosis PPAR-α, β, γ activation decrease
57 AhR activation leading to hepatic steatosis AhR activation
58 CAR suppression leading to hepatic steatosis CAR suppression
PPAR-α inhibition
LXR activation
PPAR promoter demethylation
59 HNF4 α suppression leading to hepatic steatosis HNF4 α suppression
60 PXR activation leading to hepatic steatosis PXR/SXR activation
61 NFE2L2/FXR activation leading to hepatic steatosis NRF2 activation
NR1H4 activation
62 AKT2 activation leading to hepatic steatosis Systemic inflammation leading to hepatic steatosis
220 CYP2E1 activation leading to liver cancer Activation of CYP2E1
232 NFE2/Nrf2 repression to steatosis NFE2/Nrf2 repression
273 Mitochondrial complex inhibition leading to liver injury Inhibition of any mitochondrial complexes (I, II, III,
IV, or V)
278 IKK complex inhibition leading to liver injury IKK complex inhibition
317 Glucocorticoid receptor activation leading to hepatic steatosis Glucocorticoid receptor activation

4.1. Bile salt export pump (BSEP) inhibition

Cholestasis is caused by disruption in bile flow, and the BSEP is one of the hepatic transporters responsible for excreting bile acids from hepatocytes. BSEP inhibition by drugs would lead to accumulation of drugs in hepatocytes and cause cholestasis as a consequence. (Chen et al., 2016c) Many drugs causing DILI in humans have shown the inhibitory activity to the BSEP. In AOP 27 (cholestatic liver injury induced by inhibition of the BSEP), BSEP inhibition is defined as a MIE since accumulated evidence suggests that BSEP inhibition is associated with the development of DILI. Currently, several (Q)SAR models were developed to predict BSEP inhibitors and non-inhibitors. Notably, different criteria were used to label compounds as BSEP inhibitors or non-inhibitors (Kenna et al., 2018); therefore, model predictions need to be interpreted carefully based on their specific definitions for BSEP inhibitors. A BSEP inhibition model is available in admetSAR (Table 1) (Yang et al., 2019).

Accurate prediction of BSEP inhibition using molecular structure alone is a challenging task due to the complicated mechanisms involved. BSEP inhibition can occur by interfering with ATP binding (competitive inhibitors) or BSEP kinetics (non-competitive inhibitors). To improve prediction performance, (Q)SAR models were used together with pharmacophore and molecular modeling techniques. Welch et al. used pharmacophore modeling to extract significant substructures present in the inhibitors. The (Q)SAR model was developed to predict inhibition of the BSEP and multidrug resistance protein 4 (MRP 4) as DILI was linked to the inhibition of both transporters (Welch et al., 2015). Jain et al. used molecular docking and molecular dynamics for analyzing BSEP protein structures generated through the homology modeling (Jain et al., 2017). Recently, crystal structures of BSEP complexed with inhibitors at the binding pocket have been reported (Wang et al., 2022), which provided a valuable resource for molecular modeling.

4.2. Oxidative stress and glutathione depletion

Oxidative stress can be induced by reactive oxygen species (ROS) generated in the hepatocytes. It is one of the critical events for the development of liver injury due to formation of reactive intermediates through hepatic metabolism (Villanueva-Paz et al., 2021). In AOP 220 (CYP2E1 activation leading to liver cancer), cytochrome P450 2E1 (CYP2E1) is identified as an MIE that subsequently induces oxidative stress. CYP2E1 bioactivates numerous molecules and generates reactive metabolites that can cause oxidative stress.

A (Q)SAR model was developed to predict activation of the antioxidant responsive element (ARE) pathway, which help to alleviate oxidative stress (Zhang et al., 2020). In another study by Jia et al., structural alerts and ARE assay data were incorporated to predict hepatotoxicity (Jia et al., 2022). The nrf2 ARE assay was selected among 24 in vitro assays for its high correlation to hepatotoxicity. A (Q)SAR model was developed to predict ARE assay outcome to fill data gaps caused by the lack of testing results for certain compounds. Twenty-seven structural alerts for oxidative stress were identified and combined with ARE assays for classifying compounds into three categories: toxic, non-toxic, or inconclusive. The combination model predicted compounds as toxic or non-toxic when structural alerts and ARE assays were both positives or negatives, and inconclusive if they did not agree with each other. This mechanistically driven model demonstrated a high correlation to hepatotoxicity (positive predictive value=0.64) and confirmed the importance of oxidative stress as a critical toxicity event leading to hepatotoxicity. ProTox-II provides a nrf2/ARE classification model (Table 1).

4.3. Mitochondrial dysfunction

As adenosine triphosphates (ATPs) are produced in mitochondria, mitochondrial damage can lead to ATP depletion, and eventually to cell death. Mitochondrial impairment has been identified as a significant key event in the development of liver injury. In AOP 273 (mitochondrial complex inhibition leading to liver injury), an inhibitor binding to one of the mitochondrial complexes from I to V is defined as an MIE for liver injury. DILI drugs and certain herbal products were frequently reported to cause mitochondrial toxicity, including free radicals generation, membrane potential loss, and mitochondrial permeability transition (Ramachandran et al., 2018).

Several (Q)SAR models were developed to predict mitochondrial toxicity based on databases including Tox21, ChEMBL, PubChem, and DrugBank (Hemmerich et al., 2020; Bringezu et al., 2021; Zhao et al., 2021). Rana et al. used mitochondrial toxicity assays and physicochemical properties of drugs to predict hepatotoxicity, cardiotoxicity, and nephrotoxicity. Compared to other organ toxicities, more hepatotoxic compounds tested positive in the mitochondrial assay. Specifically, an isolated rat liver mitochondrial inhibition assay showed a higher correlation for hepatotoxicity prediction compared to that of other assays (Rana et al., 2019). DILIsym is a physiologically-based pharmacokinetic (PBPK) model to predict biomarkers of DILI. It runs simulations through the molecular properties, physiological parameters, and mechanistically driven in vitro assay data. DILIsym is particularly useful for examining the importance of mitochondrial toxicity involved in development of liver injury, since some of parameters were obtained from MITOsym, an in silico model for predicting mitochondrial dysfunction (Lin et al., 2022). A mitochondrial toxicity classification model is available in admetSAR, and a mitochondrial membrane potential classification model in ProTox-II. (Table 1)

4.4. Drug metabolism and bioactivation

Drug metabolism increases hydrophilicity of xenobiotics to create metabolites that are more easily excreted from the body. However, reactive metabolites (RMs), which are electrophilic species that can covalently bind to proteins and DNA and leads to toxicity, can also be formed via drug metabolism. RMs were well-known to be involved in development of liver injury (Weaver et al., 2020). Chen et al. developed a logistic regression model using daily dose/Cmax, logP, and RM formation for DILI prediction. RM formation was identified as the most significant factor in the model prediction because the highest coefficient was assigned to RM formation in the model (Chen et al., 2016a). A wide range of models are available for drug metabolism prediction, such as cytochrome P450 substrate classification, site of metabolism prediction, and drug metabolite structure prediction (Table 3). However, these models are still struggling with insufficient accuracy in drug metabolism prediction. For example, drug metabolites were usually generated through interactions with more than one drug metabolizing enzyme (DME), while the current developed models predict drug metabolism from a single DME; thus, significant gaps exist between the prediction and practice in drug metabolism.

Table 3.

Selected list of software for drug metabolism prediction

Name Endpoint License type Prediction models available at
ACD/Percepta CYP450 inhibitor/substrate Commercial (GUI program)
P-gP inhibitor/substrate
Phase I site of metabolism
ADMET predictor Phase I site of metabolism (Metabolite structure prediction) Commercial (GUI program)
Meteor Nexus Metabolic fate prediction Commercial (GUI program)
PreADMET CYP450 inhibitor/substrate Freeware (Web program) https://preadmet.webservice.bmdrc.org/
P-gP inhibitor
PreMetabo Phase I site of metabolism Freeware (Web program) https://premetabo.webservice.bmdrc.org/
Phase I & II drug metabolism inhibitor/substrate
P-gP substrate
admetSAR Phase I, II, and transporter inhibitor/substrate Freeware (Web program) http://lmmd.ecust.edu.cn/admetsar2
SMARTcyp CYP450 site of metabolism Freeware (Web program) https://smartcyp.sund.ku.dk/mol_to_som
Xenosite Site of metabolism
(Epoxidation, Quinonation, Reactivity, Phase I,
N-dealkylation, UGT conjugation)
Freeware (Web program) https://xenosite.org/

4.5. Identification of key events based on toxicogenomic database

Understanding the mechanism of DILI is as important as accurately predicting DILI. Toxicogenomic data provide a landscape of biological perturbations caused by toxicants. These data have been used to understand DILI mechanisms and identify possible key events (Shin et al., 2022b), as well as to design human liver cell models (Lauschke, 2021). Table 4 summarizes the toxicogenomics databases that have been used to enrich our understanding of the mechanisms underlying the adverse outcomes.

Table 4.

Selected list of toxicogenomics database (accessed on 6th July)

Abbreviations Descriptions Available at
BioPlanet Integration of human pathways with the healthy and disease state annotations and targets. https://tripod.nih.gov/bioplanet/
CHemDIS Chemical-disease inference system is deployed with interfaces for enrichment analysis for functions, pathways and diseases to identify chemicals with potential risks. http://cwtung.kmu.edu.tw/chemdis
CTD Comparative Toxicogenomics Database provides manually-curated chemical-gene/protein interactions, chemical-disease and gene-disease relationships curated manually. https://ctdbase.org/
DisGeNET DisGeNET is a platform to integrate information on human disease-associated genes and variants. https://www.disgenet.org/
DrugMatrix Toxicogenomic reference resources. Data is reported through ToxFX. It is no longer supported by National Toxicology Program; however, source code of DrugMatrix and ToxFX will be available in Github https://ntp.niehs.nih.gov/data/drugmatrix
LINCS The Library of Integrated Network-Based Cellular Signatures contains a variety of data based on the data level concept in the Cancer Genome Atlas. https://lincsproject.org/
MSigDB Molecular Signatures Database provides annotated gene sets for *GSEA software https://www.gsea-msigdb.org/gsea/msigdb
Reactome Reactome is a biological pathway database manually curated and peer-reviewed. https://reactome.org/
TG-GATEs Toxicogenomics Project-Genomics Assisted Toxicity Evaluation System is developed from the Japanese Toxicogenomics Project Consortium from 170 compounds. https://www.toxicodb.ca/datasets/1
*

GSEA: Gene Set Enrichment Analysis

Since the cost of toxicogenomic database generation is high, Chen et al. proposed ToxGAN, a generative adversarial network (GAN) using artificial intelligence, as an alternative method to generate toxicogenomic data for chemicals (Chen et al., 2022). The ToxGAN model uses molecular structure, dose, and exposure time as inputs and generates predicted toxicogenomic profiles through a deep neural network. While the data gap remains, deep learning and biological big data present new opportunities for improving the accuracy of in silico models for DILI prediction.

5. Integration of in silico and in vitro data

Since both in silico and in vitro models alone are insufficiently accurate for predicting DILI in humans, there has been increasing interest in the integration of these two data types. Chen et al. suggested a two-tiered approach for DILI prediction based on rule of two (RO2) and high content screening (HCS) assays. The RO2 classifies a drug as positive if a daily dose is equal to or more than 100 mg/day and its logP is greater or equal to 3. HCS would be only applied to those classified as negative by RO2, which significantly reduced the number of molecules to be screened (Chen et al., 2014). This study showed that the integration of in vitro data can improve hepatotoxicity prediction compared to use of the RO2 alone. Another benefit of incorporating in vitro assay data is its interpretability, enabling the mechanism of toxicity to be understood, while (Q)SAR models were usually “black-box” due to use of chemical descriptors together with complicated machine learning/deep learning algorithms. As in vitro assay data in the public domain has been increasingly accumulated through projects such as Tox21 and ToxCast, in silico models can be developed together with in vitro data to improve toxicity prediction.

Xu et al. developed multiple models for predicting organ toxicities by using chemical structure and in vitro assay data (Xu et al., 2020). The liver toxicity prediction model achieved the highest performance using structural information and in vitro assay data together. Interestingly, more in vitro data were used than structural data to achieve good accuracy, whereas other organ toxicity models used less in vitro data. Khadka et al. developed a DILI prediction model using drug properties and in vitro data, and relevant KEs from liver AOPs were used to guide the selection of in vitro assays from Tox21 and L1000 datasets (Khadka et al., 2020). Williams et al. developed a Bayesian model using a mechanistically relevant hepatic safety in vitro assay together with logP and Cmax values (Williams et al., 2020). In this study, DILI occurrence at different doses was tested by modifying the exposure of the chemical through Cmax, which is one of the model variables.

The integration of in silico and in vitro data has not always been reported to improve prediction accuracy in DILI models. Ye et al. showed that DILI prediction models based on in vitro and structural information achieved similar prediction accuracies with the model based on structural information alone. One possible reason for this could be insufficient coverage of the biological response space by available in vitro data (Ye et al., 2022). In some senses, biological information is critical for achieving good prediction of DILI. Kohonen et al. developed a DILI prediction model from toxicogenomic and cytotoxicity data, assuming that transcriptional response patterns could be shared among similar hepatotoxic drugs. (Kohonen et al., 2017). The developed genomic model was reported to consistently outperform predictions generated from (Q)SAR analysis.

6. Challenges

In silico modeling for liver injury prediction has made significant progress in the past decade; however, further improvements are still needed to accurately predict liver liabilities caused by drugs and dietary supplements. Existing DILI prediction models often suffered from low sensitivity, typically 55–65%. Additionally, the general applicability of the models to other datasets is limited since the models were trained and tested on specific datasets. Recently, advancements in machine learning algorithms and expansion of in vitro databases have made significant impacts on in silico modeling for DILI prediction. However, due to the complicated mechanisms underlying the development of DILI, even the models of integrating in silico and in vitro data are still limited by insufficient prediction performance.

DILI is often reported with the administration of multiple drugs and dietary supplements. Currently, the methods covered here are not sufficient for predicting liver injury caused by mixtures of chemicals. Moreover, many DILI events are immune-mediated and cannot be easily modeled through in silico or in vitro systems. Growing evidence has suggested that DILI results from interactions between drug properties, as well as environmental and host factors (Chen et al., 2015). Variations in the human leukocyte antigen genes are also reported to be associated with DILI susceptibility (Daly, 2023). These factors, in addition to individual differences in DMEs and hepatic transporter expression, should be considered in order to develop an improved model for DILI prediction in humans.

Data availability

None

Acknowledgements

This work was supported in part by the Korea Institute of Toxicology, Republic of Korea (KK-2303), and by the Intramural Research Programs of the National Center for Advancing Translational Sciences, National Institutes of Health. The authors thank Joanne Berger, FDA Library, for manuscript editing assistance.

Footnotes

CReDiT authorship contribution statement

MC and RH conceived the concept. HS wrote the draft with input from all authors, and MC and RH revised and approved the manuscript.

Declaration of competing interest

None conflicted interest needs to be declared.

Disclaimer: This article reflects the views of the authors and does not necessarily reflect those of the U.S. Food and Drug Administration. Any mention of commercial products is for clarification only and is not intended as approval, endorsement, or recommendation.

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  1. Amadi CN, Orisakwe OE, 2018. Herb-induced liver injuries in developing nations: an update. Toxics 6, 24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Ankley GT, Bennett RS, Erickson RJ, Hoff DJ, Hornung MW, Johnson RD, Mount DR, Nichols JW, Russom CL, Schmieder PK, Serrrano JA, Tietge JE, Villeneuve DL, 2010. Adverse outcome pathways: A conceptual framework to support ecotoxicology research and risk assessment. Environmental Toxicology and Chemistry 29, 730–741. [DOI] [PubMed] [Google Scholar]
  3. Atienzar FA, Nicolas J-M, 2018. Prediction of human liver toxicity using in vitro assays: limitations and opportunities. in: Chen M, Will Y. (Eds.). Drug-induced liver toxicity. Springer; New York, New York, NY, pp. 125–150. [Google Scholar]
  4. Baker RE, Mahmud AS, Miller IF, Rajeev M, Rasambainarivo F, Rice BL, Takahashi S, Tatem AJ, Wagner CE, Wang L-F, Wesolowski A, Metcalf CJE, 2022. Infectious disease in an era of global change. Nature Reviews Microbiology 20, 193–205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Banerjee P, Eckert AO, Schrey AK, Preissner R, 2018. ProTox-II: a webserver for the prediction of toxicity of chemicals. Nucleic Acids Research 46, W257–W263. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Brecklinghaus T, 2020. Roadmap for the development of alternative test methods. Archives of Toxicology 94, 3597–3598. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Bringezu F, Carlos Gómez-Tamayo J, Pastor M, 2021. Ensemble prediction of mitochondrial toxicity using machine learning technology. Computational Toxicology 20, 100189. [Google Scholar]
  8. Chen M, Borlak J, Tong W, 2016a. A model to predict severity of drug-induced liver injury in humans. Hepatology 64, 931–940. [DOI] [PubMed] [Google Scholar]
  9. Chen M, Suzuki A, Borlak J, Andrade RJ, Lucena MI, 2015. Drug-induced liver injury: Interactions between drug properties and host factors. Journal of Hepatology 63, 503–514. [DOI] [PubMed] [Google Scholar]
  10. Chen M, Suzuki A, Thakkar S, Yu K, Hu C, Tong W, 2016b. DILIrank: the largest reference drug list ranked by the risk for developing drug-induced liver injury in humans. Drug Discovery Today 21, 648–653. [DOI] [PubMed] [Google Scholar]
  11. Chen M, Tung C-W, Shi Q, Guo L, Shi L, Fang H, Borlak J, Tong W, 2014. A testing strategy to predict risk for drug-induced liver injury in humans using high-content screen assays and the ‘rule-of-two’ model. Archives of Toxicology 88, 1439–1449. [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Chen Q, Zheng J, Shao Y, Ai N, Wu L, Liu Y, Lu X, Fan X, 2016c. Network-based Assessment on Chemical-induced Cholestatic Liver Injury. Current Topics in Medicinal Chemistry 16, 3668–3677. [DOI] [PubMed] [Google Scholar]
  13. Chen X, Roberts R, Tong W, Liu Z, 2022. Tox-GAN: An artificial intelligence approach alternative to animal studies—a case study with toxicogenomics. Toxicological Sciences 186, 242–259. [DOI] [PubMed] [Google Scholar]
  14. Cotterill J, Price N, Rorije E, Peijnenburg A, 2020. Development of a QSAR model to predict hepatic steatosis using freely available machine learning tools. Food and Chemical Toxicology 142, 111494. [DOI] [PubMed] [Google Scholar]
  15. Cronin MTD, Enoch SJ, Madden JC, Rathman JF, Richarz A-N, Yang C, 2022. A review of in silico toxicology approaches to support the safety assessment of cosmetics-related materials. Computational Toxicology 21, 100213. [Google Scholar]
  16. Daly AK, 2023. Genetics of drug-induced liver injury: Current knowledge and future prospects. Clinical and Translational Science 16, 37–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Gadaleta D, Manganelli S, Roncaglioni A, Toma C, Benfenati E, Mombelli E, 2018. QSAR modeling of ToxCast assays relevant to the molecular initiating events of AOPs leading to hepatic steatosis. Journal of Chemical Information and Modeling 58, 1501–1517. [DOI] [PubMed] [Google Scholar]
  18. Greene N, Fisk L, Naven RT, Note RR, Patel ML, Pelletier DJ, 2010. Developing structure−activity relationships for the prediction of hepatotoxicity. Chemical Research in Toxicology 23, 1215–1222. [DOI] [PubMed] [Google Scholar]
  19. He S, Ye T, Wang R, Zhang C, Zhang X, Sun G, Sun X, 2019. An in silico model for predicting drug-induced hepatotoxicity. International Journal of Molecular Sciences 20, 1897. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Hemmerich J, Troger F, Füzi B, F.Ecker G, 2020. Using machine learning methods and structural alerts for prediction of mitochondrial toxicity. Molecular Informatics 39, 2000005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Hoofnagle JH, 2013. Chapter 40 - LiverTox: A website on drug-induced liver injury. in: Kaplowitz N, DeLeve LD (Eds.). Drug-induced liver disease (Third Edition). Academic Press, Boston, pp. 725–732. [Google Scholar]
  22. Jain S, Grandits M, Richter L, Ecker GF, 2017. Structure based classification for bile salt export pump (BSEP) inhibitors using comparative structural modeling of human BSEP. Journal of Computer-Aided Molecular Design 31, 507–521. [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Jia X, Wen X, Russo DP, Aleksunes LM, Zhu H, 2022. Mechanism-driven modeling of chemical hepatotoxicity using structural alerts and an in vitro screening assay. Journal of Hazardous Materials 436, 129193. [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Kaplowitz N, 2005. Idiosyncratic drug hepatotoxicity. Nature Reviews Drug Discovery 4, 489–499. [DOI] [PubMed] [Google Scholar]
  25. Kenna JG, Taskar KS, Battista C, Bourdet DL, Brouwer KLR, Brouwer KR, Dai D, Funk C, Hafey MJ, Lai Y, Maher J, Pak YA, Pedersen JM, Polli JW, Rodrigues AD, Watkins PB, Yang K, Yucha RW, On behalf of the International Transporter, C., 2018. Can bile salt export pump inhibition testing in drug discovery and development reduce liver injury risk? An international transporter consortium perspective. Clinical Pharmacology & Therapeutics 104, 916–932. [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Khadka KK, Chen M, Liu Z, Tong W, Wang D, 2020. Integrating adverse outcome pathways (AOPs) and high throughput in vitro assays for better risk evaluations, a study with drug-induced liver injury (DILI). ALTEX - Alternatives to animal experimentation 37, 187–196. [DOI] [PubMed] [Google Scholar]
  27. Kim E, Nam H, 2017. Prediction models for drug-induced hepatotoxicity by using weighted molecular fingerprints. BMC Bioinformatics 18, 227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Kohonen P, Parkkinen JA, Willighagen EL, Ceder R, Wennerberg K, Kaski S, Grafström RC, 2017. A transcriptomics data-driven gene space accurately predicts liver cytopathology and drug-induced liver injury. Nature Communications 8, 15932. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Lauschke VM, 2021. Toxicogenomics of drug induced liver injury – from mechanistic understanding to early prediction. Drug Metabolism Reviews 53, 245–252. [DOI] [PubMed] [Google Scholar]
  30. Li T, Tong W, Roberts R, Liu Z, Thakkar S, 2021. DeepDILI: Deep learning-powered drug-induced liver injury prediction using model-level representation. Chemical Research in Toxicology 34, 550–565. [DOI] [PubMed] [Google Scholar]
  31. Li X, Chen Y, Song X, Zhang Y, Li H, Zhao Y, 2018. The development and application of in silico models for drug induced liver injury. RSC Advances 8, 8101–8111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Lin J, Li M, Mak W, Shi Y, Zhu X, Tang Z, He Q, Xiang X, 2022. Applications of in silico models to predict drug-induced liver injury. Toxics 10, 788. [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Lin NH, Yang HW, Su YJ, Chang CW, 2019. Herb induced liver injury after using herbal medicine: A systemic review and case-control study. Medicine 98, e14992. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Liu Y, 2018. Incorporation of absorption and metabolism into liver toxicity prediction for phytochemicals: A tiered in silico QSAR approach. Food and Chemical Toxicology 118, 409–415. [DOI] [PubMed] [Google Scholar]
  35. Ma H, An W, Wang Y, Sun H, Huang R, Huang J, 2021. Deep graph learning with property augmentation for predicting drug-induced liver injury. Chemical Research in Toxicology 34, 495–506. [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Matthews EJ, Ursem CJ, Kruhlak NL, Benz RD, Sabaté DA, Yang C, Klopman G, Contrera JF, 2009. Identification of structure-activity relationships for adverse effects of pharmaceuticals in humans: Part B. Use of (Q)SAR systems for early detection of drug-induced hepatobiliary and urinary tract toxicities. Regulatory Toxicology and Pharmacology 54, 23–42. [DOI] [PubMed] [Google Scholar]
  37. Mulliner D, Schmidt F, Stolte M, Spirkl H-P, Czich A, Amberg A, 2016. Computational models for human and animal hepatotoxicity with a global application scope. Chemical Research in Toxicology 29, 757–767. [DOI] [PubMed] [Google Scholar]
  38. Nunes DR, Monteiro CS, dos Santos JL, 2022. Herb-induced liver injury—a challenging diagnosis. Healthcare 10, 278. [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Parish ST, Aschner M, Casey W, Corvaro M, Embry MR, Fitzpatrick S, Kidd D, Kleinstreuer NC, Lima BS, Settivari RS, Wolf DC, Yamazaki D, Boobis A, 2020. An evaluation framework for new approach methodologies (NAMs) for human health safety assessment. Regulatory Toxicology and Pharmacology 112, 104592. [DOI] [PubMed] [Google Scholar]
  40. Punt A, Bouwmeester H, Blaauboer BJ, Coecke S, Hakkert B, Hendriks DFG, Jennings P, Kramer NI, Neuhoff S, Masereeuw R, Paini A, Peijnenburg AACM, Rooseboom M, Shuler ML, Sorrell I, Spee B, Strikwold M, van der Meer AD, van der Zande M, Vinken M, Yang H, Bos PMJ, Heringa MB, 2020. New approach methodologies (NAMs) for human-relevant biokinetics predictions: Meeting the paradigm shift in toxicology towards an animal-free chemical risk assessment. ALTEX - Alternatives to animal experimentation 37, 607–622. [DOI] [PubMed] [Google Scholar]
  41. Ramachandran A, Visschers RGJ, Duan L, Akakpo JY, Jaeschke H, 2018. Mitochondrial dysfunction as a mechanism of drug-induced hepatotoxicity: current understanding and future perspectives. Journal of clinical and translational research 4, 75–100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Rana P, Aleo MD, Gosink M, Will Y, 2019. Evaluation of in vitro mitochondrial toxicity assays and physicochemical properties for prediction of organ toxicity using 228 pharmaceutical drugs. Chemical Research in Toxicology 32, 156–167. [DOI] [PubMed] [Google Scholar]
  43. Robles-Díaz M, Medina-Caliz I, Stephens C, Andrade RJ, Lucena MI, 2016. Biomarkers in DILI: One more step forward. Frontiers in Pharmacology 7, 267. [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Rodgers AD, Zhu H, Fourches D, Rusyn I, Tropsha A, 2010. Modeling liver-related adverse effects of drugs using knearest neighbor quantitative structure−activity relationship method. Chemical Research in Toxicology 23, 724–732. [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Rusyn I, Arzuaga X, Cattley RC, Corton JC, Ferguson SS, Godoy P, Guyton KZ, Kaplowitz N, Khetani SR, Roberts RA, Roth RA, Smith MT, 2021. Key characteristics of human hepatotoxicants as a basis for identification and characterization of the causes of liver toxicity. Hepatology 74, 3486–3496. [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Shin HK, Chun H-S, Lee S, Park S-M, Park D, Kang M-G, Hwang S, Oh J-H, Han H-Y, Kim W-K, Yoon S, 2022a. ToxSTAR: drug-induced liver injury prediction tool for the web environment. Bioinformatics 38, 4426–4427. [DOI] [PubMed] [Google Scholar]
  47. Shin HK, Florean O, Hardy B, Doktorova T, Kang M-G, 2022b. Semi-automated approach for generation of biological networks on drug-induced cholestasis, steatosis, hepatitis, and cirrhosis. Toxicological Research 38, 393–407. [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Shin HK, Kang M-G, Park D, Park T, Yoon S, 2020. Development of prediction models for drug-induced cholestasis, cirrhosis, hepatitis, and steatosis based on drug and drug metabolite structures. Frontiers in Pharmacology 11, 67. [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Sreedhar D, Manjula N, Pise A, Pise S, Ligade VS, 2020. Ban of cosmetic testing on animals: A brief overview. International Journal of Current Research and Review 12, 113–116. [Google Scholar]
  50. Stratton CF, Newman DJ, Tan DS, 2015. Cheminformatic comparison of approved drugs from natural product versus synthetic origins. Bioorganic & Medicinal Chemistry Letters 25, 4802–4807. [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Stucki AO, Barton-Maclaren TS, Bhuller Y, Henriquez JE, Henry TR, Hirn C, Miller-Holt J, Nagy EG, Perron MM, Ratzlaff DE, Stedeford TJ, Clippinger AJ, 2022. Use of new approach methodologies (NAMs) to meet regulatory requirements for the assessment of industrial chemicals and pesticides for effects on human health. Frontiers in Toxicology 4, 964553. [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Sutton C, Boley M, Ghiringhelli LM, Rupp M, Vreeken J, Scheffler M, 2020. Identifying domains of applicability of machine learning models for materials science. Nature Communications 11, 4428. [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Suzuki A, Andrade RJ, Bjornsson E, Lucena MI, Lee WM, Yuen NA, Hunt CM, Freston JW, 2010. Drugs associated with hepatotoxicity and their reporting frequency of liver adverse events in VigiBase. Drug Safety 33, 503–522. [DOI] [PubMed] [Google Scholar]
  54. Thakkar S, Chen M, Hong H, Liu Z, Fang H, Tong W, 2018. Drug-induced liver injury (DILI) classification and its application on human DILI risk prediction. in: Chen M, Will Y. (Eds.). Drug-induced liver toxicity. Springer; New York, New York, NY, pp. 45–59. [Google Scholar]
  55. Thakkar S, Li T, Liu Z, Wu L, Roberts R, Tong W, 2020. Drug-induced liver injury severity and toxicity (DILIst): binary classification of 1279 drugs by human hepatotoxicity. Drug Discovery Today 25, 201–208. [DOI] [PubMed] [Google Scholar]
  56. Tong S, Bambrick H, Beggs PJ, Chen L, Hu Y, Ma W, Steffen W, Tan J, 2022. Current and future threats to human health in the Anthropocene. Environment International 158, 106892. [DOI] [PubMed] [Google Scholar]
  57. Vall A, Sabnis Y, Shi J, Class R, Hochreiter S, Klambauer G, 2021. The promise of AI for DILI prediction. Frontiers in Artificial Intelligence 4, 638410. [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. van der Zalm AJ, Barroso J, Browne P, Casey W, Gordon J, Henry TR, Kleinstreuer NC, Lowit AB, Perron M, Clippinger AJ, 2022. A framework for establishing scientific confidence in new approach methodologies. Archives of Toxicology 96, 2865–2879. [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. Van Norman GA, 2019. Limitations of animal studies for predicting toxicity in clinical trials: Is it time to rethink our current approach? JACC: Basic to Translational Science 4, 845–854. [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Villanueva-Paz M, Morán L, López-Alcántara N, Freixo C, Andrade RJ, Lucena MI, Cubero FJ, 2021. Oxidative stress in drug-induced liver injury (DILI): From mechanisms to biomarkers for use in clinical practice. Antioxidants 10, 390. [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Wang L, Hou W-T, Wang J, Xu D, Guo C, Sun L, Ruan K, Zhou C-Z, Chen Y, 2022. Structures of human bile acid exporter ABCB11 reveal a transport mechanism facilitated by two tandem substrate-binding pockets. Cell Research 32, 501–504. [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Weaver RJ, Blomme EA, Chadwick AE, Copple IM, Gerets HHJ, Goldring CE, Guillouzo A, Hewitt PG, Ingelman-Sundberg M, Jensen KG, Juhila S, Klingmüller U, Labbe G, Liguori MJ, Lovatt CA, Morgan P, Naisbitt DJ, Pieters RHH, Snoeys J, van de Water B, Williams DP, Park BK, 2020. Managing the challenge of drug-induced liver injury: a roadmap for the development and deployment of preclinical predictive models. Nature Reviews Drug Discovery 19, 131–148. [DOI] [PubMed] [Google Scholar]
  63. Weaver S, Gleeson MP, 2008. The importance of the domain of applicability in QSAR modeling. Journal of Molecular Graphics and Modelling 26, 1315–1326. [DOI] [PubMed] [Google Scholar]
  64. Welch MA, Köck K, Urban TJ, Brouwer KLR, Swaan PW, 2015. Toward predicting drug-induced liver injury: Parallel computational approaches to identify multidrug resistance protein 4 and bile salt export pump inhibitors. Drug Metabolism and Disposition 43, 725. [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Williams DP, Lazic SE, Foster AJ, Semenova E, Morgan P, 2020. Predicting drug-induced liver injury with bayesian machine learning. Chemical Research in Toxicology 33, 239–248. [DOI] [PubMed] [Google Scholar]
  66. Xu T, Ngan DK, Ye L, Xia M, Xie HQ, Zhao B, Simeonov A, Huang R, 2020. Predictive models for human organ toxicity based on in vitro bioactivity data and chemical structure. Chemical Research in Toxicology 33, 731–741. [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. Xu Y, Dai Z, Chen F, Gao S, Pei J, Lai L, 2015. Deep learning for drug-induced liver injury. Journal of Chemical Information and Modeling 55, 2085–2093. [DOI] [PubMed] [Google Scholar]
  68. Yang H, Lou C, Sun L, Li J, Cai Y, Wang Z, Li W, Liu G, Tang Y, 2019. admetSAR 2.0: Web-service for prediction and optimization of chemical ADMET properties. Bioinformatics 35, 1067–1069. [DOI] [PubMed] [Google Scholar]
  69. Ye L, Ngan DK, Xu T, Liu Z, Zhao J, Sakamuru S, Zhang L, Zhao T, Xia M, Simeonov A, Huang R, 2022. Prediction of drug-induced liver injury and cardiotoxicity using chemical structure and in vitro assay data. Toxicology and Applied Pharmacology 454, 116250. [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Zhang S, Khan WA, Su L, Zhang X, Li C, Qin W, Zhao Y, 2020. Predicting oxidative stress induced by organic chemicals by using quantitative Structure–Activity relationship methods. Ecotoxicology and Environmental Safety 201, 110817. [DOI] [PubMed] [Google Scholar]
  71. Zhao P, Peng Y, Xu X, Wang Z, Wu Z, Li W, Tang Y, Liu G, 2021. In silico prediction of mitochondrial toxicity of chemicals using machine learning methods. Journal of Applied Toxicology 41, 1518–1526. [DOI] [PubMed] [Google Scholar]
  72. Zhu X, Kruhlak NL, 2014. Construction and analysis of a human hepatotoxicity database suitable for QSAR modeling using post-market safety data. Toxicology 321, 62–72. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

None

RESOURCES