1
|
He C, Yu L, Jiang Y, Xie L, Mai X, Ai P, Xue B. Deep-learning approach for developing bilayered electromagnetic interference shielding composite aerogels based on multimodal data fusion neural networks. J Colloid Interface Sci 2025; 688:79-92. [PMID: 39987843 DOI: 10.1016/j.jcis.2025.02.133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 02/10/2025] [Accepted: 02/19/2025] [Indexed: 02/25/2025]
Abstract
A non-experimental approach to developing high-performance EMI shielding materials is urgently needed to reduce costs and manpower. In this investigation, a multimodal data fusion neural network model is proposed to predict the EMI shielding performances of silver-modified four-pronged zinc oxide/waterborne polyurethane/barium ferrite (Ag@F-ZnO/WPU/BF) aerogels. First, 16 Ag@F-ZnO/WPU/BF samples with varying Ag@F-ZnO and BF contents were successfully prepared using the pre-casting and directional freezing techniques. The experimental results demonstrate that these aerogels perform well in terms of averaged EMI shielding effectiveness (SET) up to 78.6 dB and absorption coefficient as high as 0.96. On the basis of composite ingredients and microstructural images, the established multimodal neural network model can effectively predict the EMI shielding performances of Ag@F-ZnO/WPU/BF aerogels. Notably, the multimodal model of fully connected neural network (FCNN) and residual neural network (ResNet) utilizing GatedFusion method yields the best root mean squared error (RMSE) and mean absolute error (MAE) values of 0.7626 and 0.4918, respectively, and correlation coefficient (R) of 0.9885. In addition, this multimodal model successfully predicts the EMI performances of four new aerogels with an average error of less than 5 %, demonstrating its strong generalization capability. The accuracy and efficiency of material property prediction based on multimodal neural network model are largely improved by integrating multiple data sources, offering new possibility for reducing experimental burdens, accelerating the development of new materials, and gaining a deeper understanding of material mechanisms.
Collapse
Affiliation(s)
- Chenglei He
- College of Mechanical Engineering, Guizhou University, Guiyang 550025, China
| | - Liya Yu
- College of Mechanical Engineering, Guizhou University, Guiyang 550025, China.
| | - Yun Jiang
- College of Mechanical Engineering, Guizhou University, Guiyang 550025, China
| | - Lan Xie
- College of Mechanical Engineering, Guizhou University, Guiyang 550025, China; Department of Polymer Materials and Engineering, College of Materials and Metallurgy, Guizhou University, Guiyang 550025, China; State Key Laboratory of Public Big Data, Guizhou University, Guiyang 550025, China.
| | - Xiaoping Mai
- Department of Polymer Materials and Engineering, College of Materials and Metallurgy, Guizhou University, Guiyang 550025, China
| | - Peng Ai
- Department of Polymer Materials and Engineering, College of Materials and Metallurgy, Guizhou University, Guiyang 550025, China
| | - Bai Xue
- College of Mechanical Engineering, Guizhou University, Guiyang 550025, China; Department of Polymer Materials and Engineering, College of Materials and Metallurgy, Guizhou University, Guiyang 550025, China; State Key Laboratory of Public Big Data, Guizhou University, Guiyang 550025, China.
| |
Collapse
|
2
|
Wu L, Fang H, Qu Y, Xu J, Tong W. Leveraging FDA Labeling Documents and Large Language Model to Enhance Annotation, Profiling, and Classification of Drug Adverse Events with AskFDALabel. Drug Saf 2025; 48:655-665. [PMID: 39979771 PMCID: PMC12098182 DOI: 10.1007/s40264-025-01520-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/20/2025] [Indexed: 02/22/2025]
Abstract
BACKGROUND Drug adverse events (AEs) represent a significant public health concern. US Food and Drug Administration (FDA) drug labeling documents are an essential resource for studying drug safety such as assessing a drug's likelihood to cause certain organ toxicities; however, the manual extraction of AEs is labor-intensive, requires specialized expertise, and is challenging to maintain, due to frequent updates of the labeling documents. OBJECTIVE To automate the extraction of AE data from FDA drug labeling documents, we developed a workflow based on AskFDALabel, a large language model (LLM)-powered framework, and its demonstration in drug safety studies. METHODS This framework incorporates a retrieval-augmented generation (RAG) component based on FDALabel to enhance standard LLM inference. Key steps include (1) selection of a task-specific template, (2) FDALabel database querying, and (3) content preparation for LLM processing. We evaluated the performance of the framework in three benchmark experiments, including drug-induced liver injury (DILI) classification, drug-induced cardiotoxicity (DICT) classification, and AE term recognition. RESULTS AskFDALabel achieved F1-scores of 0.978 for DILI, 0.931 for DICT, and 0.911 for AE annotation, outperforming other traditional methods. It also provided cited labeling content and detailed explanations, facilitating manual verification. CONCLUSION AskFDALabel exhibited high consistency with human AE annotation, particularly in classifying and profiling DILI and DICT. Thus, it can significantly enhance the efficiency and accuracy of AE annotation, with promising potential for advanced AE surveillance and drug safety research.
Collapse
Affiliation(s)
- Leihong Wu
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, U.S. FDA, 3900 NCTR Rd, Jefferson, AR, 72079, USA.
| | - Hong Fang
- Office of Scientific Coordination, National Center for Toxicological Research, U.S. FDA, 3900 NCTR Rd, Jefferson, AR, 72079, USA
| | - Yanyan Qu
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, U.S. FDA, 3900 NCTR Rd, Jefferson, AR, 72079, USA
| | - Joshua Xu
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, U.S. FDA, 3900 NCTR Rd, Jefferson, AR, 72079, USA
| | - Weida Tong
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, U.S. FDA, 3900 NCTR Rd, Jefferson, AR, 72079, USA
| |
Collapse
|
3
|
Wu J, Zheng Z, Li J, Shen X, Huang B. Predicting treatment response to systemic therapy in advanced gallbladder cancer using multiphase enhanced CT images. Eur Radiol 2025:10.1007/s00330-025-11645-7. [PMID: 40341972 DOI: 10.1007/s00330-025-11645-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 03/21/2025] [Accepted: 04/08/2025] [Indexed: 05/11/2025]
Abstract
BACKGROUND Accurate estimation of treatment response can help clinicians identify patients who would potentially benefit from systemic therapy. This study aimed to develop and externally validate a model for predicting treatment response to systemic therapy in advanced gallbladder cancer (GBC). METHODS We recruited 399 eligible GBC patients across four institutions. Multivariable logistic regression analysis was performed to identify independent clinical factors related to therapeutic efficacy. This deep learning (DL) radiomics signature was developed for predicting treatment response using multiphase enhanced CT images. Then, the DL radiomic-clinical (DLRSC) model was built by combining the DL signature and significant clinical factors, and its predictive performance was evaluated using area under the curve (AUC). Gradient-weighted class activation mapping analysis was performed to help clinicians better understand the predictive results. Furthermore, patients were stratified into low- and high-score groups by the DLRSC model. The progression-free survival (PFS) and overall survival (OS) between the two different groups were compared. RESULTS Multivariable analysis revealed that tumor size was a significant predictor of efficacy. The DLRSC model showed great predictive performance, with AUCs of 0.86 (95% CI, 0.82-0.89) and 0.84 (95% CI, 0.80-0.87) in the internal and external test datasets, respectively. This model showed great discrimination, calibration, and clinical utility. Moreover, Kaplan-Meier survival analysis revealed that low-score group patients who were insensitive to systemic therapy predicted by the DLRSC model had worse PFS and OS. CONCLUSION The DLRSC model allows for predicting treatment response in advanced GBC patients receiving systemic therapy. The survival benefit provided by the DLRSC model was also assessed. KEY POINTS Question No effective tools exist for identifying patients who would potentially benefit from systemic therapy in clinical practice. Findings Our combined model allows for predicting treatment response to systemic therapy in advanced gallbladder cancer. Clinical relevance With the help of this model, clinicians could inform patients of the risk of potential ineffective treatment. Such a strategy can reduce unnecessary adverse events and effectively help reallocate societal healthcare resources.
Collapse
Affiliation(s)
- Ji Wu
- Department of General Surgery, Suzhou Ninth Hospital Affiliated to Soochow University, Suzhou, China
| | - Zhigang Zheng
- Department of Interventional Oncology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jian Li
- Department of Radiology, The Affiliated Changshu Hospital of Nantong University, Suzhou, China
| | - Xiping Shen
- Department of General Surgery, Suzhou Ninth Hospital Affiliated to Soochow University, Suzhou, China
| | - Bo Huang
- Department of Hepatobiliary Surgery, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China.
| |
Collapse
|
4
|
Zhou Y, Zhong Y, Lauschke VM. Evaluating the synergistic use of advanced liver models and AI for the prediction of drug-induced liver injury. Expert Opin Drug Metab Toxicol 2025; 21:563-577. [PMID: 39893552 DOI: 10.1080/17425255.2025.2461484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 01/29/2025] [Indexed: 02/04/2025]
Abstract
INTRODUCTION Drug-induced liver injury (DILI) is a leading cause of acute liver failure. Hepatotoxicity typically occurs only in a subset of individuals after prolonged exposure and constitutes a major risk factor for the termination of drug development projects. AREAS COVERED We provide an overview of available human liver models for DILI research and discuss how they have been used to aid in early risk assessments and to mitigate the risk of project closures due to DILI in clinical stages. We summarize the different data that can be provided by such models and illustrate how these diverse data types can be interfaced with machine learning strategies to improve predictions of liver safety liabilities. EXPERT OPINION Advanced human liver models closely mimic human liver phenotypes and functions for many weeks, allowing for the recapitulation of hepatotoxicity events in vitro. Integration of the biochemical, histological, and toxicogenomic output data from these models with physicochemical compound properties using different machine learning architectures holds promise to enhance preclinical DILI predictions. However, to realize this aim, it is important to benchmark the available liver models on test sets of DILI positive and negative compounds and to carefully annotate and share the resulting data.
Collapse
Affiliation(s)
- Yitian Zhou
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska Institutet and University Hospital, Stockholm, Sweden
| | - Yi Zhong
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska Institutet and University Hospital, Stockholm, Sweden
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Volker M Lauschke
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska Institutet and University Hospital, Stockholm, Sweden
- Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany
- University of Tübingen, Tübingen, Germany
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
5
|
Xia W, Shu J, Sang C, Wang K, Wang Y, Sun T, Xu X. The prediction of RNA-small-molecule ligand binding affinity based on geometric deep learning. Comput Biol Chem 2025; 115:108367. [PMID: 39904171 DOI: 10.1016/j.compbiolchem.2025.108367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 01/11/2025] [Accepted: 01/26/2025] [Indexed: 02/06/2025]
Abstract
Small molecule-targeted RNA is an emerging technology that plays a pivotal role in drug discovery and inhibitor design, with widespread applications in disease treatment. Consequently, predicting RNA-small-molecule ligand interactions is crucial. With advancements in computer science and the availability of extensive biological data, deep learning methods have shown great promise in this area, particularly in efficiently predicting RNA-small molecule binding sites. However, few computational methods have been developed to predict RNA-small molecule binding affinities. Meanwhile, most of these approaches rely primarily on sequence or structural representations. Molecular surface information, vital for RNA and small molecule interactions, has been largely overlooked. To address these gaps, we propose a geometric deep learning method for predicting RNA-small molecule binding affinity, named RNA-ligand Surface Interaction Fingerprinting (RLASIF). In this study, we create RNA-ligand interaction fingerprints from the geometrical and chemical features present on molecular surface to characterize binding affinity. RLASIF outperformed other computational methods across ten different test sets from PDBbind NL2020. Compared to the second-best method, our approach improves performance by 10.01 %, 6.67 %, 2.01 % and 1.70 % on four evaluation metrics, indicating its effectiveness in capturing key features influencing RNA-ligand binding strength. Additionally, RLASIF holds potential for virtual screening of potential ligands for RNA and predicting small molecule binding nucleotides within RNA structures.
Collapse
Affiliation(s)
- Wentao Xia
- Department of Physics, Zhejiang University of Science and Technology, Hangzhou 310008, China
| | - Jiasai Shu
- Department of Physics, Zhejiang University of Science and Technology, Hangzhou 310008, China
| | - Chunjiang Sang
- Department of Physics, Zhejiang University of Science and Technology, Hangzhou 310008, China
| | - Kang Wang
- Department of Physics, Zhejiang University of Science and Technology, Hangzhou 310008, China
| | - Yan Wang
- Department of Physics, Zhejiang University of Science and Technology, Hangzhou 310008, China
| | - Tingting Sun
- Department of Physics, Zhejiang University of Science and Technology, Hangzhou 310008, China.
| | - Xiaojun Xu
- Institute of Bioinformatics and Medical Engineering, Jiangsu University of Technology, Changzhou 213001, China.
| |
Collapse
|
6
|
Nan H, Liu X, Liu Z. Herbal-Induced Liver Injury Identification and Prevention. Liver Int 2025; 45:e16154. [PMID: 39605246 DOI: 10.1111/liv.16154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 10/23/2024] [Indexed: 11/29/2024]
Affiliation(s)
- Huang Nan
- Department of Clinical Pharmacy, Xiangtan Central Hospital (The Affiliated Hospital of Hunan University), Xiangtan, China
| | - Xiang Liu
- Department of Clinical Pharmacy, Xiangtan Central Hospital (The Affiliated Hospital of Hunan University), Xiangtan, China
| | - Zheng Liu
- Department of Clinical Pharmacy, Xiangtan Central Hospital (The Affiliated Hospital of Hunan University), Xiangtan, China
| |
Collapse
|
7
|
Bai C, Wu L, Li R, Cao Y, He S, Bo X. Machine Learning-Enabled Drug-Induced Toxicity Prediction. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2413405. [PMID: 39899688 PMCID: PMC12021114 DOI: 10.1002/advs.202413405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 12/25/2024] [Indexed: 02/05/2025]
Abstract
Unexpected toxicity has become a significant obstacle to drug candidate development, accounting for 30% of drug discovery failures. Traditional toxicity assessment through animal testing is costly and time-consuming. Big data and artificial intelligence (AI), especially machine learning (ML), are robustly contributing to innovation and progress in toxicology research. However, the optimal AI model for different types of toxicity usually varies, making it essential to conduct comparative analyses of AI methods across toxicity domains. The diverse data sources also pose challenges for researchers focusing on specific toxicity studies. In this review, 10 categories of drug-induced toxicity is examined, summarizing the characteristics and applicable ML models, including both predictive and interpretable algorithms, striking a balance between breadth and depth. Key databases and tools used in toxicity prediction are also highlighted, including toxicology, chemical, multi-omics, and benchmark databases, organized by their focus and function to clarify their roles in drug-induced toxicity prediction. Finally, strategies to turn challenges into opportunities are analyzed and discussed. This review may provide researchers with a valuable reference for understanding and utilizing the available resources to bridge prediction and mechanistic insights, and further advance the application of ML in drugs-induced toxicity prediction.
Collapse
Affiliation(s)
- Changsen Bai
- Academy of Medical Engineering and Translational MedicineTianjin UniversityTianjin300072China
- Department of Advanced & Interdisciplinary BiotechnologyAcademy of Military Medical SciencesBeijing100850China
- Tianjin Medical University Cancer Institute and HospitalTianjin300060China
| | - Lianlian Wu
- Academy of Medical Engineering and Translational MedicineTianjin UniversityTianjin300072China
- Department of Advanced & Interdisciplinary BiotechnologyAcademy of Military Medical SciencesBeijing100850China
| | - Ruijiang Li
- Department of Advanced & Interdisciplinary BiotechnologyAcademy of Military Medical SciencesBeijing100850China
| | - Yang Cao
- Department of Environmental MedicineAcademy of Military Medical SciencesTianjin300050China
| | - Song He
- Department of Advanced & Interdisciplinary BiotechnologyAcademy of Military Medical SciencesBeijing100850China
| | - Xiaochen Bo
- Academy of Medical Engineering and Translational MedicineTianjin UniversityTianjin300072China
- Department of Advanced & Interdisciplinary BiotechnologyAcademy of Military Medical SciencesBeijing100850China
| |
Collapse
|
8
|
Zhao L, Tan J, Su Q, Kuang Y. Integration of histopathological images and immunological analysis to predict M2 macrophage infiltration and prognosis in patients with serous ovarian cancer. Front Immunol 2025; 16:1505509. [PMID: 40165975 PMCID: PMC11955462 DOI: 10.3389/fimmu.2025.1505509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 02/28/2025] [Indexed: 04/02/2025] Open
Abstract
Objective Investigating the effect of M2 macrophage infiltration on overall survival and to use histopathological imaging features (HIF) to predict M2 macrophage infiltration in patients with serous ovarian cancer (SOC) is important for improving prognostic accuracy, identifying new therapeutic targets, and advancing personalized treatment approaches. Methods We downloaded data from 86 patients with SOC from The Cancer Genome Atlas (TCGA) and divided these patients into a training set and a validation set with a ratio of 8:2. In addition, tissue microarrays from 106 patients with SOC patients were included as an external validation set. HIF were recognized by deep multiple instance learning (MIL) to predict M2 macrophage infiltration via theResNet18 network in the training set. The final model was evaluated using the internal and external validation set. Results Using data acquired from the TCGA database, we applied univariate Cox analysis and determined that higher levels of M2 macrophage infiltration were associated with a poor prognosis (hazard ratio [HR]=6.8; 95% CI [confidence interval]: 1.6-28, P=0.0083). External validation revealed that M2 macrophage infiltration was an independent risk factor for the prognosis of patients with SOC (HR=3.986; 95% CI: 2.436-6.522; P<0.001). Next, we constructed four MIL strategies (Mean probability, Top-10 Mean, Top-100 Mean, and Maximum probability) to identify histopathological images that could predict M2 macrophage infiltration. The Mean Probability Method was the most suitable and was used to generate a HIF model with an AUC, recall rate, precision and F1 score of 0.7500, 0.6932, 0.600, 0.600, and 0.600, respectively. Conclusions Collectively, our findings indicated that M2 macrophage infiltration may increase prognostic prediction for SOC patients. Machine deep learning of pathological immunohistochemical images exhibited good potential for the direct prediction of M2 macrophage infiltration.
Collapse
Affiliation(s)
- Ling Zhao
- Department of Gynecology, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jiajia Tan
- Department of Gynecology, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Qiuyuan Su
- Department of Gynecology, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yan Kuang
- Department of Gynecology, First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Department of Gynecology, Guangzhou First People’s Hospital, Guangzhou, China
| |
Collapse
|
9
|
Wu J, Li J, Huang B, Dong S, Wu L, Shen X, Zheng Z. ConvXGB: A novel deep learning model to predict recurrence risk of early-stage cervical cancer following surgery using multiparametric MRI images. Transl Oncol 2025; 52:102281. [PMID: 39799749 PMCID: PMC11773201 DOI: 10.1016/j.tranon.2025.102281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 12/08/2024] [Accepted: 01/07/2025] [Indexed: 01/15/2025] Open
Abstract
BACKGROUND Accurate estimation of recurrence risk for cervical cancer plays a pivot role in making individualized treatment plans. We aimed to develop and externally validate an end-to-end deep learning model for predicting recurrence risk in cervical cancer patients following surgery by using multiparametric MRI images. METHODS The clinicopathologic data and multiparametric MRI images of 406 cervical cancer patients from three institutions were collected. We designed a novel deep learning model called "ConvXGB" for predicting recurrence risk by combining the convolutional neural network (CNN) and eXtreme Gradient Boost (XGBoost). The predictive performance of the ConvXGB model was evaluated using time-dependent area under curve (AUC), compared with the deep learning radio-clinical model, clinical model, conventional radiomics nomogram and an existing histology-specific tool. The potential of the ConvXGB model in predicting the recurrence-free survival (RFS) and overall survival (OS) was assessed. RESULTS The ConvXGB model outperformed other models in predicting recurrence risk, with AUCs for 1 and 3 year-RFS of 0.872(95% CI, 0.857-0.906) and 0.882(95% CI, 0.860-0.904) respectively in the test cohort. This model showed better discrimination, calibration and clinical utility. Grad-CAM analysis was adopted to help clinicians better understand the predictive results. Moreover, Kaplan-Meier survival analysis revealed that patients who were stratified into high-risk group by the ConvXGB model were significantly susceptible to higher cumulative recurrence risk rates and worse outcome. CONCLUSION The ConvXGB model allowed for predicting postoperative recurrence risk in cervical cancer patients and for stratifying the risk of RFS and OS.
Collapse
Affiliation(s)
- Ji Wu
- Department of General surgery, Suzhou Ninth Hospital Affiliated to Soochow University, Suzhou, Jiangsu Province, China
- Department of Radiology, Suzhou Ninth Hospital Affiliated to Soochow University, Suzhou, Jiangsu Province, China
| | - Jian Li
- Department of Radiology, Changshu No.2 People's Hospital, The Affiliated Changshu Hospital of Nantong University, Changshu, Jiangsu, China
| | - Bo Huang
- Department of Radiology, Municipal Hospital Affiliated to Nanjing Medical University, Suzhou, Jiangsu Province, China
| | - Sunbin Dong
- Department of Radiology, Municipal Hospital Affiliated to Nanjing Medical University, Suzhou, Jiangsu Province, China
| | - Luyang Wu
- Department of Radiology, Municipal Hospital Affiliated to Nanjing Medical University, Suzhou, Jiangsu Province, China
| | - Xiping Shen
- Department of General surgery, Suzhou Ninth Hospital Affiliated to Soochow University, Suzhou, Jiangsu Province, China
- Department of Radiology, Suzhou Ninth Hospital Affiliated to Soochow University, Suzhou, Jiangsu Province, China
| | - Zhigang Zheng
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Radiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
10
|
Yang X, Sun J, Jin B, Lu Y, Cheng J, Jiang J, Zhao Q, Shuai J. Multi-task aquatic toxicity prediction model based on multi-level features fusion. J Adv Res 2025; 68:477-489. [PMID: 38844122 PMCID: PMC11785906 DOI: 10.1016/j.jare.2024.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 05/21/2024] [Accepted: 06/02/2024] [Indexed: 06/09/2024] Open
Abstract
INTRODUCTION With the escalating menace of organic compounds in environmental pollution imperiling the survival of aquatic organisms, the investigation of organic compound toxicity across diverse aquatic species assumes paramount significance for environmental protection. Understanding how different species respond to these compounds helps assess the potential ecological impact of pollution on aquatic ecosystems as a whole. Compared with traditional experimental methods, deep learning methods have higher accuracy in predicting aquatic toxicity, faster data processing speed and better generalization ability. OBJECTIVES This article presents ATFPGT-multi, an advanced multi-task deep neural network prediction model for organic toxicity. METHODS The model integrates molecular fingerprints and molecule graphs to characterize molecules, enabling the simultaneous prediction of acute toxicity for the same organic compound across four distinct fish species. Furthermore, to validate the advantages of multi-task learning, we independently construct prediction models, named ATFPGT-single, for each fish species. We employ cross-validation in our experiments to assess the performance and generalization ability of ATFPGT-multi. RESULTS The experimental results indicate, first, that ATFPGT-multi outperforms ATFPGT-single on four fish datasets with AUC improvements of 9.8%, 4%, 4.8%, and 8.2%, respectively, demonstrating the superiority of multi-task learning over single-task learning. Furthermore, in comparison with previous algorithms, ATFPGT-multi outperforms comparative methods, emphasizing that our approach exhibits higher accuracy and reliability in predicting aquatic toxicity. Moreover, ATFPGT-multi utilizes attention scores to identify molecular fragments associated with fish toxicity in organic molecules, as demonstrated by two organic molecule examples in the main text, demonstrating the interpretability of ATFPGT-multi. CONCLUSION In summary, ATFPGT-multi provides important support and reference for the further development of aquatic toxicity assessment. All of codes and datasets are freely available online at https://github.com/zhaoqi106/ATFPGT-multi.
Collapse
Affiliation(s)
- Xin Yang
- School of Computer Science and Software Engineering, University of Science and Technology Liaoning, Anshan 114051, China; Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, China
| | - Jianqiang Sun
- School of Information Science and Engineering, Linyi University, Linyi 276000, China
| | - Bingyu Jin
- School of Computer Science and Software Engineering, University of Science and Technology Liaoning, Anshan 114051, China
| | - Yuer Lu
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, China
| | - Jinyan Cheng
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, China
| | - Jiaju Jiang
- College of Life Sciences, Sichuan University, Chengdu 610064, China
| | - Qi Zhao
- School of Computer Science and Software Engineering, University of Science and Technology Liaoning, Anshan 114051, China.
| | - Jianwei Shuai
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou 325001, China.
| |
Collapse
|
11
|
He W, Han Y, Zuo Y, Bai Y, Guo F. NBCR-ac4C: A Deep Learning Framework Based on Multivariate BERT for Human mRNA N4-Acetylcytidine Sites Prediction. J Chem Inf Model 2024; 64:8074-8081. [PMID: 39367830 DOI: 10.1021/acs.jcim.4c01415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/07/2024]
Abstract
N4-acetylcytidine (ac4C) plays a crucial role in regulating cellular biological processes, particularly in gene expression regulation and disease development. However, experiments to identify ac4C in a wet lab are time-consuming and costly, and the learning-based methods struggle to capture the underlying semantic knowledge and relations within sequences. To address this, we propose a deep learning approach called NBCR-ac4C based on pretrained models. Specifically, we employ Nucleotide Transformer and DNABERT2 to construct contextual embedding of nucleotide sequences, which effectively mine and express context relations between different features in the sequence. Convolutional neural network (CNN) and ResNet18 are then applied to further extract shallow and deep knowledge from context embedding. Depending on extensive experiments for the prediction of ac4C sites in nucleotide sequences, we observe that NBCR-ac4C outperforms general learning-based models. It achieves the highest accuracy (ACC) of 83.51% and an Area Under the Receiver Operating Characteristic Curve (AUROC) of 89.58% on an independent test set. Moreover, the proposed model, compared to the current state-of-the-art (SOTA) model LSA-ac4C, demonstrates higher ACC and AUROC by 0.81-3.7% and 0.05-1.58%, respectively. The data set and code are available on https://github.com/2103374200/NBCR to facilitate further discussion on NBCR-ac4C.
Collapse
Affiliation(s)
- Wenying He
- School of Artificial Intelligence, Hebei University of Technology, Tianjin 300400, China
| | - Yu Han
- School of Artificial Intelligence, Hebei University of Technology, Tianjin 300400, China
| | - Yun Zuo
- School of Artificial Intelligence and Computer Science, Jiangnan University, Wuxi 214000, China
| | - Yude Bai
- School of Software, Tiangong University, Tianjin 300387, China
| | - Fei Guo
- School of Computer Science and Engineering, Central South University, Changsha 410083, China
| |
Collapse
|
12
|
Wei W, Xu J, Xia F, Liu J, Zhang Z, Wu J, Wei T, Feng H, Ma Q, Jiang F, Zhu X, Zhang X. Deep learning-assisted diagnosis of benign and malignant parotid gland tumors based on automatic segmentation of ultrasound images: a multicenter retrospective study. Front Oncol 2024; 14:1417330. [PMID: 39184051 PMCID: PMC11341398 DOI: 10.3389/fonc.2024.1417330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Accepted: 07/18/2024] [Indexed: 08/27/2024] Open
Abstract
Objectives To construct deep learning-assisted diagnosis models based on automatic segmentation of ultrasound images to facilitate radiologists in differentiating benign and malignant parotid tumors. Methods A total of 582 patients histopathologically diagnosed with PGTs were retrospectively recruited from 4 centers, and their data were collected for analysis. The radiomics features of six deep learning models (ResNet18, Inception_v3 etc) were analyzed based on the ultrasound images that were obtained under the best automatic segmentation model (Deeplabv3, UNet++, and UNet). The performance of three physicians was compared when the optimal model was used and not. The Net Reclassification Index (NRI) and Integrated Discrimination Improvement (IDI) were utilized to evaluate the clinical benefit of the optimal model. Results The Deeplabv3 model performed optimally in terms of automatic segmentation. The ResNet18 deep learning model had the best prediction performance, with an area under the receiver-operating characteristic curve of 0.808 (0.694-0.923), 0.809 (0.712-0.906), and 0.812 (0.680-0.944) in the internal test set and external test sets 1 and 2, respectively. Meanwhile, the optimal model-assisted clinical and overall benefits were markedly enhanced for two out of three radiologists (in internal validation set, NRI: 0.259 and 0.213 [p = 0.002 and 0.017], IDI: 0.284 and 0.201 [p = 0.005 and 0.043], respectively; in external test set 1, NRI: 0.183 and 0.161 [p = 0.019 and 0.008], IDI: 0.205 and 0.184 [p = 0.031 and 0.045], respectively; in external test set 2, NRI: 0.297 and 0.297 [p = 0.038 and 0.047], IDI: 0.332 and 0.294 [p = 0.031 and 0.041], respectively). Conclusions The deep learning model constructed for automatic segmentation of ultrasound images can improve the diagnostic performance of radiologists for PGTs.
Collapse
Affiliation(s)
- Wei Wei
- Department of Ultrasound, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital), Wuhu, China
| | - Jingya Xu
- Department of Radiology, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital), Wuhu, China
| | - Fei Xia
- Department of Ultrasound, WuHu Hospital, East China Normal University (The Second People’s Hospital, WuHu), Wuhu, Anhui, China
| | - Jun Liu
- Department of Ultrasound, Linyi Central Hospital, Linyi, Shandong, China
| | - Zekai Zhang
- Department of Ultrasound, Zibo Central Hospital, Zibo, Shandong, China
| | - Jing Wu
- Department of Ultrasound, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital), Wuhu, China
| | - Tianjun Wei
- Department of Ultrasound, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital), Wuhu, China
| | - Huijun Feng
- Department of Ultrasound, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital), Wuhu, China
| | - Qiang Ma
- Department of Ultrasound, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital), Wuhu, China
| | - Feng Jiang
- Department of Ultrasound, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital), Wuhu, China
| | - Xiangming Zhu
- Department of Ultrasound, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital), Wuhu, China
| | - Xia Zhang
- Department of Ultrasound, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital), Wuhu, China
| |
Collapse
|
13
|
Zhao Y, Zhang Z, Kong X, Wang K, Wang Y, Jia J, Li H, Tian S. Prediction of Drug-Induced Liver Injury: From Molecular Physicochemical Properties and Scaffold Architectures to Machine Learning Approaches. Chem Biol Drug Des 2024; 104:e14607. [PMID: 39179521 DOI: 10.1111/cbdd.14607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 07/24/2024] [Accepted: 08/01/2024] [Indexed: 08/26/2024]
Abstract
The process of developing new drugs is widely acknowledged as being time-intensive and requiring substantial financial investment. Despite ongoing efforts to reduce time and expenses in drug development, ensuring medication safety remains an urgent problem. One of the major problems involved in drug development is hepatotoxicity, specifically known as drug-induced liver injury (DILI). The popularity of new drugs often poses a significant barrier during development and frequently leads to their recall after launch. In silico methods have many advantages compared with traditional in vivo and in vitro assays. To establish a more precise and reliable prediction model, it is necessary to utilize an extensive and high-quality database consisting of information on drug molecule properties and structural patterns. In addition, we should also carefully select appropriate molecular descriptors that can be used to accurately depict compound characteristics. The aim of this study was to conduct a comprehensive investigation into the prediction of DILI. First, we conducted a comparative analysis of the physicochemical properties of extensively well-prepared DILI-positive and DILI-negative compounds. Then, we used classic substructure dissection methods to identify structural pattern differences between these two different types of chemical molecules. These findings indicate that it is not feasible to establish property or substructure-based rules for distinguishing between DILI-positive and DILI-negative compounds. Finally, we developed quantitative classification models for predicting DILI using the naïve Bayes classifier (NBC) and recursive partitioning (RP) machine learning techniques. The optimal DILI prediction model was obtained using NBC, which combines 21 physicochemical properties, the VolSurf descriptors and the LCFP_10 fingerprint set. This model achieved a global accuracy (GA) of 0.855 and an area under the curve (AUC) of 0.704 for the training set, while the corresponding values were 0.619 and 0.674 for the test set, respectively. Moreover, indicative substructural fragments favorable or unfavorable for DILI were identified from the best naïve Bayesian classification model. These findings may help prioritize lead compounds in the early stage of drug development pipelines.
Collapse
Affiliation(s)
- Yulong Zhao
- College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Zhoudong Zhang
- College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Xiaotian Kong
- Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, Soochow University, Suzhou, China
| | - Kai Wang
- College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Yaxuan Wang
- College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Jie Jia
- College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Huanqiu Li
- College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Sheng Tian
- College of Pharmaceutical Sciences, Soochow University, Suzhou, China
- College of Chemistry and Life Science, Beijing University of Technology, Beijing, China
| |
Collapse
|
14
|
Amorim AM, Piochi LF, Gaspar AT, Preto A, Rosário-Ferreira N, Moreira IS. Advancing Drug Safety in Drug Development: Bridging Computational Predictions for Enhanced Toxicity Prediction. Chem Res Toxicol 2024; 37:827-849. [PMID: 38758610 PMCID: PMC11187637 DOI: 10.1021/acs.chemrestox.3c00352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 04/29/2024] [Accepted: 05/07/2024] [Indexed: 05/19/2024]
Abstract
The attrition rate of drugs in clinical trials is generally quite high, with estimates suggesting that approximately 90% of drugs fail to make it through the process. The identification of unexpected toxicity issues during preclinical stages is a significant factor contributing to this high rate of failure. These issues can have a major impact on the success of a drug and must be carefully considered throughout the development process. These late-stage rejections or withdrawals of drug candidates significantly increase the costs associated with drug development, particularly when toxicity is detected during clinical trials or after market release. Understanding drug-biological target interactions is essential for evaluating compound toxicity and safety, as well as predicting therapeutic effects and potential off-target effects that could lead to toxicity. This will enable scientists to predict and assess the safety profiles of drug candidates more accurately. Evaluation of toxicity and safety is a critical aspect of drug development, and biomolecules, particularly proteins, play vital roles in complex biological networks and often serve as targets for various chemicals. Therefore, a better understanding of these interactions is crucial for the advancement of drug development. The development of computational methods for evaluating protein-ligand interactions and predicting toxicity is emerging as a promising approach that adheres to the 3Rs principles (replace, reduce, and refine) and has garnered significant attention in recent years. In this review, we present a thorough examination of the latest breakthroughs in drug toxicity prediction, highlighting the significance of drug-target binding affinity in anticipating and mitigating possible adverse effects. In doing so, we aim to contribute to the development of more effective and secure drugs.
Collapse
Affiliation(s)
- Ana M.
B. Amorim
- Department
of Life Sciences, University of Coimbra, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal
- CNC-UC—Center
for Neuroscience and Cell Biology, University
of Coimbra, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal
- CIBB—Centre
for Innovative Biomedicine and Biotechnology, University of Coimbra, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal
- PhD
Programme in Biosciences, Department of Life Sciences, University of Coimbra, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal
- PURR.AI,
Rua Pedro Nunes, IPN Incubadora, Ed C, 3030-199 Coimbra, Portugal
| | - Luiz F. Piochi
- Department
of Life Sciences, University of Coimbra, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal
- CNC-UC—Center
for Neuroscience and Cell Biology, University
of Coimbra, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal
- CIBB—Centre
for Innovative Biomedicine and Biotechnology, University of Coimbra, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal
| | - Ana T. Gaspar
- Department
of Life Sciences, University of Coimbra, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal
- CNC-UC—Center
for Neuroscience and Cell Biology, University
of Coimbra, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal
- CIBB—Centre
for Innovative Biomedicine and Biotechnology, University of Coimbra, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal
| | - António
J. Preto
- CNC-UC—Center
for Neuroscience and Cell Biology, University
of Coimbra, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal
- CIBB—Centre
for Innovative Biomedicine and Biotechnology, University of Coimbra, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal
- PhD Programme
in Experimental Biology and Biomedicine, Institute for Interdisciplinary
Research (IIIUC), University of Coimbra, Casa Costa Alemão, 3030-789 Coimbra, Portugal
| | - Nícia Rosário-Ferreira
- CNC-UC—Center
for Neuroscience and Cell Biology, University
of Coimbra, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal
- CIBB—Centre
for Innovative Biomedicine and Biotechnology, University of Coimbra, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal
| | - Irina S. Moreira
- Department
of Life Sciences, University of Coimbra, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal
- CNC-UC—Center
for Neuroscience and Cell Biology, University
of Coimbra, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal
- CIBB—Centre
for Innovative Biomedicine and Biotechnology, University of Coimbra, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal
| |
Collapse
|
15
|
Li Y, Liu B, Deng J, Guo Y, Du H. Image-based molecular representation learning for drug development: a survey. Brief Bioinform 2024; 25:bbae294. [PMID: 38920347 PMCID: PMC11200195 DOI: 10.1093/bib/bbae294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 05/19/2024] [Accepted: 06/08/2024] [Indexed: 06/27/2024] Open
Abstract
Artificial intelligence (AI) powered drug development has received remarkable attention in recent years. It addresses the limitations of traditional experimental methods that are costly and time-consuming. While there have been many surveys attempting to summarize related research, they only focus on general AI or specific aspects such as natural language processing and graph neural network. Considering the rapid advance on computer vision, using the molecular image to enable AI appears to be a more intuitive and effective approach since each chemical substance has a unique visual representation. In this paper, we provide the first survey on image-based molecular representation for drug development. The survey proposes a taxonomy based on the learning paradigms in computer vision and reviews a large number of corresponding papers, highlighting the contributions of molecular visual representation in drug development. Besides, we discuss the applications, limitations and future directions in the field. We hope this survey could offer valuable insight into the use of image-based molecular representation learning in the context of drug development.
Collapse
Affiliation(s)
- Yue Li
- Division of Gastroenterology, Dongzhimen Hospital, Beijing University of Chinese Medicine, No. 5 Haiyun Warehouse, 100700, Beijing, China
| | - Bingyan Liu
- School of Computer Science, Beijing University of Posts and Telecommunications, No.10 Xituchen Street, 100876, Beijing, China
| | - Jinyan Deng
- Division of Gastroenterology, Dongzhimen Hospital, Beijing University of Chinese Medicine, No. 5 Haiyun Warehouse, 100700, Beijing, China
| | - Yi Guo
- Division of Gastroenterology, Dongzhimen Hospital, Beijing University of Chinese Medicine, No. 5 Haiyun Warehouse, 100700, Beijing, China
| | - Hongbo Du
- Division of Gastroenterology, Dongzhimen Hospital, Beijing University of Chinese Medicine, No. 5 Haiyun Warehouse, 100700, Beijing, China
- Institute of Liver Disease, Beijing University of Chinese Medicine, No. 5 Haiyun Warehouse, 100700, Beijing, China
| |
Collapse
|
16
|
Mostafa F, Chen M. Computational models for predicting liver toxicity in the deep learning era. FRONTIERS IN TOXICOLOGY 2024; 5:1340860. [PMID: 38312894 PMCID: PMC10834666 DOI: 10.3389/ftox.2023.1340860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Accepted: 12/22/2023] [Indexed: 02/06/2024] Open
Abstract
Drug-induced liver injury (DILI) is a severe adverse reaction caused by drugs and may result in acute liver failure and even death. Many efforts have centered on mitigating risks associated with potential DILI in humans. Among these, quantitative structure-activity relationship (QSAR) was proven to be a valuable tool for early-stage hepatotoxicity screening. Its advantages include no requirement for physical substances and rapid delivery of results. Deep learning (DL) made rapid advancements recently and has been used for developing QSAR models. This review discusses the use of DL in predicting DILI, focusing on the development of QSAR models employing extensive chemical structure datasets alongside their corresponding DILI outcomes. We undertake a comprehensive evaluation of various DL methods, comparing with those of traditional machine learning (ML) approaches, and explore the strengths and limitations of DL techniques regarding their interpretability, scalability, and generalization. Overall, our review underscores the potential of DL methodologies to enhance DILI prediction and provides insights into future avenues for developing predictive models to mitigate DILI risk in humans.
Collapse
Affiliation(s)
- Fahad Mostafa
- Department of Mathematics and Statistics, Texas Tech University, Lubbock, TX, United States
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, United States
| | - Minjun Chen
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, United States
| |
Collapse
|
17
|
Wang J, Zhang L, Sun J, Yang X, Wu W, Chen W, Zhao Q. Predicting drug-induced liver injury using graph attention mechanism and molecular fingerprints. Methods 2024; 221:18-26. [PMID: 38040204 DOI: 10.1016/j.ymeth.2023.11.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Revised: 11/14/2023] [Accepted: 11/25/2023] [Indexed: 12/03/2023] Open
Abstract
Drug-induced liver injury (DILI) is a significant issue in drug development and clinical treatment due to its potential to cause liver dysfunction or damage, which, in severe cases, can lead to liver failure or even fatality. DILI has numerous pathogenic factors, many of which remain incompletely understood. Consequently, it is imperative to devise methodologies and tools for anticipatory assessment of DILI risk in the initial phases of drug development. In this study, we present DMFPGA, a novel deep learning predictive model designed to predict DILI. To provide a comprehensive description of molecular properties, we employ a multi-head graph attention mechanism to extract features from the molecular graphs, representing characteristics at the level of compound nodes. Additionally, we combine multiple fingerprints of molecules to capture features at the molecular level of compounds. The fusion of molecular fingerprints and graph features can more fully express the properties of compounds. Subsequently, we employ a fully connected neural network to classify compounds as either DILI-positive or DILI-negative. To rigorously evaluate DMFPGA's performance, we conduct a 5-fold cross-validation experiment. The obtained results demonstrate the superiority of our method over four existing state-of-the-art computational approaches, exhibiting an average AUC of 0.935 and an average ACC of 0.934. We believe that DMFPGA is helpful for early-stage DILI prediction and assessment in drug development.
Collapse
Affiliation(s)
- Jifeng Wang
- School of Computer Science and Software Engineering, University of Science and Technology Liaoning, Anshan 114051, China
| | - Li Zhang
- School of Life Science, Liaoning University, Shenyang 110036, China
| | - Jianqiang Sun
- School of Information Science and Engineering, Linyi University, Linyi 276000, China
| | - Xin Yang
- School of Computer Science and Software Engineering, University of Science and Technology Liaoning, Anshan 114051, China
| | - Wei Wu
- School of Computer Science and Software Engineering, University of Science and Technology Liaoning, Anshan 114051, China
| | - Wei Chen
- Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Qi Zhao
- School of Computer Science and Software Engineering, University of Science and Technology Liaoning, Anshan 114051, China.
| |
Collapse
|
18
|
Zahra U, Khan MA, Alhaisoni M, Alasiry A, Marzougui M, Masood A. An Integrated Framework of Two-Stream Deep Learning Models Optimal Information Fusion for Fruits Disease Recognition. IEEE JOURNAL OF SELECTED TOPICS IN APPLIED EARTH OBSERVATIONS AND REMOTE SENSING 2024; 17:3038-3052. [DOI: 10.1109/jstars.2023.3339297] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/25/2024]
Affiliation(s)
- Unber Zahra
- Department of Computer Science, HITEC University, Taxila, Pakistan
| | | | - Majed Alhaisoni
- Department of Computer Sciences, College of Computer and Information Sciences, Princess Nourah Bint Abdul Rahman University, Riyadh, Saudi Arabia
| | - Areej Alasiry
- College of Computer Science, King Khalid University, Abha, Saudi Arabia
| | - Mehrez Marzougui
- College of Computer Science, King Khalid University, Abha, Saudi Arabia
| | - Anum Masood
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway
| |
Collapse
|
19
|
Björnsson HK, Björnsson ES. Review of human risk factors for idiosyncratic drug-induced liver injury: latest advances and future goals. Expert Opin Drug Metab Toxicol 2023; 19:969-977. [PMID: 37997265 DOI: 10.1080/17425255.2023.2288260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 11/22/2023] [Indexed: 11/25/2023]
Abstract
INTRODUCTION Idiosyncratic drug-induced liver injury (DILI) is a common cause of acute liver injury and can lead to death from acute liver failure or require liver transplantation. Although the total burden of liver injury is high, the frequency of DILI caused by specific agents is often low. As the liver injury is by per definition idiosyncratic, the prediction of which patients will develop liver injury from specific drugs is currently a very difficult challenge. AREAS COVERED The current paper highlights the most important studies on prediction of DILI published in 2019-2023, including studies on genetic, metabolomic, and demographic risk factors, concomitant medication, and the role of comorbid liver diseases. Risk stratification using demographic, metabolomic, and multigenetic risk factors is discussed. EXPERT OPINION Great advances have been made in identifying genetic risk factors for DILI. Combining these risk factors with demographic information and other biomarkers into multigenetic risk models might become highly useful in risk stratifying patients exposed to DILI. However, a more detailed mapping of genetic risk factors is needed. Results of these studies need to be validated in the selected ethnic groups before applicability and cost-effectiveness can be determined.
Collapse
Affiliation(s)
- Helgi Kristinn Björnsson
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Einar Stefan Björnsson
- Division of Gastroenterology and Hepatology, Landspitali - The National University Hospital of Iceland, Reykjavik, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| |
Collapse
|
20
|
Wu W, Qian J, Liang C, Yang J, Ge G, Zhou Q, Guan X. GeoDILI: A Robust and Interpretable Model for Drug-Induced Liver Injury Prediction Using Graph Neural Network-Based Molecular Geometric Representation. Chem Res Toxicol 2023; 36:1717-1730. [PMID: 37839069 DOI: 10.1021/acs.chemrestox.3c00199] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2023]
Abstract
Drug-induced liver injury (DILI) is a significant cause of drug failure and withdrawal due to liver damage. Accurate prediction of hepatotoxic compounds is crucial for safe drug development. Several DILI prediction models have been published, but they are built on different data sets, making it difficult to compare model performance. Moreover, most existing models are based on molecular fingerprints or descriptors, neglecting molecular geometric properties and lacking interpretability. To address these limitations, we developed GeoDILI, an interpretable graph neural network that uses a molecular geometric representation. First, we utilized a geometry-based pretrained molecular representation and optimized it on the DILI data set to improve predictive performance. Second, we leveraged gradient information to obtain high-precision atomic-level weights and deduce the dominant substructure. We benchmarked GeoDILI against recently published DILI prediction models, as well as popular GNN models and fingerprint-based machine learning models using the same data set, showing superior predictive performance of our proposed model. We applied the interpretable method in the DILI data set and derived seven precise and mechanistically elucidated structural alerts. Overall, GeoDILI provides a promising approach for accurate and interpretable DILI prediction with potential applications in drug discovery and safety assessment. The data and source code are available at GitHub repository (https://github.com/CSU-QJY/GeoDILI).
Collapse
Affiliation(s)
- Wenxuan Wu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jiayu Qian
- School of Mathematics and Statistics, Central South University, Changsha, Hunan 410083, China
| | - Changjie Liang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jingya Yang
- School of Mathematics and Statistics, Central South University, Changsha, Hunan 410083, China
| | - Guangbo Ge
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Qingping Zhou
- School of Mathematics and Statistics, Central South University, Changsha, Hunan 410083, China
| | - Xiaoqing Guan
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| |
Collapse
|
21
|
Rodrigues K, Hussain R, Cooke S, Zhang G, Zhang D, Yin L, Tong X. Fructose as a novel nutraceutical for acetaminophen (APAP)-induced hepatotoxicity. METABOLISM AND TARGET ORGAN DAMAGE 2023; 3:20. [PMID: 39193224 PMCID: PMC11349303 DOI: 10.20517/mtod.2023.28] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/29/2024]
Abstract
Acetaminophen (APAP) is the most widely used analgesic in the world. APAP overdose can cause severe hepatotoxicity and therefore is the most common cause of drug-induced liver injury. The only approved treatment for APAP overdose is N-acetyl-cysteine (NAC) supplementation. However, the narrow efficacy window of the drug severely limits its clinical use, prompting the search for other therapeutic options to counteract APAP toxicity. Recent research has pointed to fructose as a novel nutraceutical for APAP-induced liver injury. This review summarizes the current understanding of the molecular mechanisms underlying APAP-induced liver injury, introduces how fructose supplementation could prevent and treat APAP liver toxicity with a focus on the ChREBPα-FGF21 pathway, and proposes possible future directions of study.
Collapse
Affiliation(s)
- Kyle Rodrigues
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48105, USA
- Caswell Diabetes Institute, University of Michigan Medical School, Ann Arbor, MI 48105, USA
| | - Rawdat Hussain
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48105, USA
- Caswell Diabetes Institute, University of Michigan Medical School, Ann Arbor, MI 48105, USA
| | - Sarah Cooke
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48105, USA
- Caswell Diabetes Institute, University of Michigan Medical School, Ann Arbor, MI 48105, USA
| | - Gary Zhang
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48105, USA
- Caswell Diabetes Institute, University of Michigan Medical School, Ann Arbor, MI 48105, USA
| | - Deqiang Zhang
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48105, USA
- Caswell Diabetes Institute, University of Michigan Medical School, Ann Arbor, MI 48105, USA
| | - Lei Yin
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48105, USA
- Caswell Diabetes Institute, University of Michigan Medical School, Ann Arbor, MI 48105, USA
| | - Xin Tong
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48105, USA
- Caswell Diabetes Institute, University of Michigan Medical School, Ann Arbor, MI 48105, USA
| |
Collapse
|
22
|
Li X, Ni J, Chen L. Advances in the study of acetaminophen-induced liver injury. Front Pharmacol 2023; 14:1239395. [PMID: 37601069 PMCID: PMC10436315 DOI: 10.3389/fphar.2023.1239395] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 07/28/2023] [Indexed: 08/22/2023] Open
Abstract
Acetaminophen (APAP) overdose is a significant cause of drug-induced liver injury and acute liver failure. The diagnosis, screening, and management of APAP-induced liver injury (AILI) is challenging because of the complex mechanisms involved. Starting from the current studies on the mechanisms of AILI, this review focuses on novel findings in the field of diagnosis, screening, and management of AILI. It highlights the current issues that need to be addressed. This review is supposed to summarize the recent research progress and make recommendations for future research.
Collapse
Affiliation(s)
- Xinghui Li
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Jiaqi Ni
- West China School of Pharmacy, Sichuan University, Chengdu, China
- Department of Pharmacy, Evidence-Based Pharmacy Center, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| | - Li Chen
- Department of Pharmacy, Evidence-Based Pharmacy Center, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| |
Collapse
|
23
|
Adesokan M, Alamu EO, Fawole S, Maziya-Dixon B. Prediction of functional characteristics of gari (cassava flakes) using near-infrared reflectance spectrometry. Front Chem 2023; 11:1156718. [PMID: 37234202 PMCID: PMC10206270 DOI: 10.3389/fchem.2023.1156718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 05/02/2023] [Indexed: 05/27/2023] Open
Abstract
Gari is a creamy, granular flour obtained from roasting fermented cassava mash. Its preparation involves several unit operations, including fermentation, which is essential in gari production. Fermentation brings about specific biochemical changes in cassava starch due to the actions of lactic acid bacteria. Consequently, it gives rise to organic acids and a significant reduction in the pH. Consumer preferences for gari are influenced by these changes and impact specific functional characteristics, which are often linked to cassava genotypes. Measurement of these functional characteristics is time-consuming and expensive. Therefore, this study aimed to develop high-throughput and less expensive prediction models for water absorption capacity, swelling power, bulk density, and dispersibility using Near-Infrared Reflectance Spectroscopy (NIRS). Gari was produced from 63 cassava genotypes using the standard method developed in the RTB foods project. The prediction model was developed by dividing the gari samples into two sets of 48 samples for calibration and 15 samples as the validation set. The gari samples were transferred into a ring cell cup and scanned on the NIRS machine within the Vis-NIR range of 400-2,498 nm wavelength, though only the NIR range of 800-2,400 nm was used to build the model. Calibration models were developed using partial least regression algorithms after spectra pre-processing. Also, the gari samples were analysed in the laboratory for their functional properties to generate reference data. Results showed an excellent coefficient of determination in calibrations (R2 Cal) of 0.99, 0.97, 0.97, and 0.89 for bulk density, swelling power, dispersibility, and water absorption capacity, respectively. Also, the performances of the prediction models were tested using an independent set of 15 gari samples. A good prediction coefficient (R2 pred) and low standard error of prediction (SEP) was obtained as follows: Bulk density (0.98), Swelling power (0.93), WAC (0.68), Dispersibility (0.65), and solubility index (0.62), respectively. Therefore, NIRS prediction models in this study could provide a rapid screening tool for cassava breeding programs and food scientists to determine the food quality of cassava granular products (Gari).
Collapse
Affiliation(s)
- Michael Adesokan
- Food and Nutrition Sciences Laboratory, International Institute of Tropical Agriculture (IITA), Ibadan, Nigeria
| | - Emmanuel Oladeji Alamu
- Food and Nutrition Sciences Laboratory, International Institute of Tropical Agriculture (IITA), Ibadan, Nigeria
- International Institute of Tropical Agriculture, Southern Africa Research and Administration Hub (SARAH) Campus, Lusaka, Zambia
| | - Segun Fawole
- Food and Nutrition Sciences Laboratory, International Institute of Tropical Agriculture (IITA), Ibadan, Nigeria
| | - Busie Maziya-Dixon
- Food and Nutrition Sciences Laboratory, International Institute of Tropical Agriculture (IITA), Ibadan, Nigeria
| |
Collapse
|
24
|
Tran TTV, Surya Wibowo A, Tayara H, Chong KT. Artificial Intelligence in Drug Toxicity Prediction: Recent Advances, Challenges, and Future Perspectives. J Chem Inf Model 2023; 63:2628-2643. [PMID: 37125780 DOI: 10.1021/acs.jcim.3c00200] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
Toxicity prediction is a critical step in the drug discovery process that helps identify and prioritize compounds with the greatest potential for safe and effective use in humans, while also reducing the risk of costly late-stage failures. It is estimated that over 30% of drug candidates are discarded owing to toxicity. Recently, artificial intelligence (AI) has been used to improve drug toxicity prediction as it provides more accurate and efficient methods for identifying the potentially toxic effects of new compounds before they are tested in human clinical trials, thus saving time and money. In this review, we present an overview of recent advances in AI-based drug toxicity prediction, including the use of various machine learning algorithms and deep learning architectures, of six major toxicity properties and Tox21 assay end points. Additionally, we provide a list of public data sources and useful toxicity prediction tools for the research community and highlight the challenges that must be addressed to enhance model performance. Finally, we discuss future perspectives for AI-based drug toxicity prediction. This review can aid researchers in understanding toxicity prediction and pave the way for new methods of drug discovery.
Collapse
Affiliation(s)
- Thi Tuyet Van Tran
- Department of Electronics and Information Engineering, Jeonbuk National University, Jeonju 54896, Republic of Korea
- Faculty of Information Technology, An Giang University, Long Xuyen 880000, Vietnam
- Vietnam National University - Ho Chi Minh City, Ho Chi Minh 700000, Vietnam
| | - Agung Surya Wibowo
- Department of Electronics and Information Engineering, Jeonbuk National University, Jeonju 54896, Republic of Korea
- Department of Electrical Engineering, Telkom University, Bandung 40257, Indonesia
| | - Hilal Tayara
- School of International Engineering and Science, Jeonbuk National University, Jeonju 54896, Republic of Korea
| | - Kil To Chong
- Advances Electronics and Information Research Center, Jeonbuk National University, Jeonju 54896, Republic of Korea
| |
Collapse
|
25
|
Sun J, Xu X, Feng S, Zhang H, Xu L, Jiang H, Sun B, Meng Y, Chen W. Rapid identification of salmonella serovars by using Raman spectroscopy and machine learning algorithm. Talanta 2023; 253:123807. [PMID: 36115103 DOI: 10.1016/j.talanta.2022.123807] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/26/2022] [Accepted: 07/29/2022] [Indexed: 12/13/2022]
Abstract
A widespread and escalating public health problem worldwide is foodborne illness, and foodborne Salmonella infection is one of the most common causes of human illness.For the three most pathogenic Salmonella serotypes, Raman spectroscopy was employed to acquire spectral data.As machine learning offers high efficiency and accuracy, we have chosen the convolutional neural network(CNN), which is suitable for solving multi-classification problems, to do in-depth mining and analysis of Raman spectral data.To optimize the instrument parameters, we compared three laser wavelengths: 532, 638, and 785 nm.Ultimately, the 532 nm wavelength was chosen as the most effective for detecting Salmonella.A pre-processing step is necessary to remove interference from the background noise of the Raman spectrum.Our study compared the effects of five spectral preprocessing methods, Savitzky-Golay smoothing (SG), Multivariate Scatter Correction (MSC), Standard Normal Variate (SNV), and Hilbert Transform (HT), on the predictive power of CNN models.Accuracy(ACC), Precision, Recall, and F1-score 4 machine learning evaluation indicators are used to evaluate the model performance under different preprocessing methods.In the results, SG combined with SNV was found to be the most accurate spectral pre-processing method for predicting Salmonella serotypes using Raman spectroscopy, achieving an accuracy of 98.7% for the training set and over 98.5% for the test set in CNN model.Pre-processing spectral data using this method yields higher accuracy than other methods.As a conclusion, the results of this study demonstrate that Raman spectroscopy when used in conjunction with a convolutional neural network model enables the rapid identification of three Salmonella serotypes at the single-cell level, and that the model has a great deal of potential for distinguishing between different serotypes of pathogenic bacteria and closely related bacterial species.This is vital to preventing outbreaks of foodborne illness and the spread of foodborne pathogens.
Collapse
Affiliation(s)
- Jiazheng Sun
- College of Criminal Investigation, People's Public Security University of China, Beijing, 100038, PR China
| | - Xuefang Xu
- State Key Laboratory of Communicable Disease Prevention and Control, Institute for Communicable Disease Prevention and Control, Chinese Center for Disease Control and Prevention, Beijing, 102206, PR China
| | - Songsong Feng
- College of Information and Cyber Security,People's Public Security University of China, Beijing, 100038, PR China
| | - Hanyu Zhang
- School of Criminology,People's Public Security University of China, Beijing, 100038, PR China
| | - Lingfeng Xu
- College of Criminal Investigation, People's Public Security University of China, Beijing, 100038, PR China
| | - Hong Jiang
- College of Criminal Investigation, People's Public Security University of China, Beijing, 100038, PR China.
| | - Baibing Sun
- College of Information and Cyber Security,People's Public Security University of China, Beijing, 100038, PR China
| | - Yuyan Meng
- College of Information and Cyber Security,People's Public Security University of China, Beijing, 100038, PR China
| | - Weizhou Chen
- School of Law,People's Public Security University of China, Beijing, 100038, PR China
| |
Collapse
|
26
|
Yasar A. Benchmarking analysis of CNN models for bread wheat varieties. Eur Food Res Technol 2022. [DOI: 10.1007/s00217-022-04172-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
27
|
Liu Z, Li T, Connor S, Thakkar S, Roberts R, Tong W. Best practice and reproducible science are required to advance artificial intelligence in real-world applications. Brief Bioinform 2022; 23:6618241. [PMID: 35848999 DOI: 10.1093/bib/bbac237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 05/19/2022] [Accepted: 05/19/2022] [Indexed: 11/13/2022] Open
Abstract
Drug-induced liver injury (DILI) is one of the most significant concerns in medical practice but yet it still cannot be fully recapitulated with existing in vivo, in vitro and in silico approaches. To address this challenge, Chen et al. [ 1] developed a deep learning-based DILI prediction model based on chemical structure information alone. The reported model yielded an outstanding prediction performance (i.e. 0.958, 0.976, 0.935, 0.947, 0.926 and 0.913 for AUC, accuracy, recall, precision, F1-score and specificity, respectively, on a test set), far outperforming all publicly available and similar in silico DILI models. This extraordinary model performance is counter-intuitive to what we know about the underlying biology of DILI and the principles and hypothesis behind this type of in silico approach. In this Letter to the Editor, we raise awareness of several issues concerning data curation, model validation and comparison practices, and data and model reproducibility.
Collapse
Affiliation(s)
- Zhichao Liu
- National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, USA
| | - Ting Li
- National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, USA
| | - Skylar Connor
- National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, USA
| | - Shraddha Thakkar
- Center for Drug Evaluation and Research, US FDA, Silver Spring, MD 20993, USA
| | - Ruth Roberts
- ApconiX Ltd, Alderley Park, Alderley Edge, SK10 4TG, UK.,University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Weida Tong
- National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, USA
| |
Collapse
|
28
|
Chen Z, Jiang Y, Zhang X, Zheng R, Qiu R, Sun Y, Zhao C, Shang H. The prediction approach of drug-induced liver injury: response to the issues of reproducible science of artificial intelligence in real-world applications. Brief Bioinform 2022; 23:6598880. [PMID: 35656709 DOI: 10.1093/bib/bbac196] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 04/12/2022] [Accepted: 04/27/2022] [Indexed: 11/12/2022] Open
Abstract
Abstract
In the previous study, we developed the generalized drug-induced liver injury (DILI) prediction model—ResNet18DNN to predict DILI based on multi-source combined DILI dataset and achieved better performance than that of previously published described DILI prediction models. Recently, we were honored to receive the invitation from the editor to response the Letter to Editor by Liu Zhichao, et al. We were glad that our research has attracted the attention of Liu’s team and they has put forward their opinions on our research. In this response to Letter to the Editor, we will respond to these comments.
Collapse
Affiliation(s)
- Zhao Chen
- Key Laboratory of Chinese Internal Medicine of Ministry of Education , Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yin Jiang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education , Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaoyu Zhang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education , Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Rui Zheng
- Key Laboratory of Chinese Internal Medicine of Ministry of Education , Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Ruijin Qiu
- Key Laboratory of Chinese Internal Medicine of Ministry of Education , Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yang Sun
- Key Laboratory of Chinese Internal Medicine of Ministry of Education , Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Chen Zhao
- Institute of Basic Research in Clinical Medicine , China Academy of Chinese Medical Sciences, Beijing, China
| | - Hongcai Shang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education , Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- College of Integrated Traditional Chinese and Western Medicine , Hunan University of Chinese Medicine, Changsha, Hunan 410208 , China
| |
Collapse
|
29
|
Chen CI, Lu NH, Huang YH, Liu KY, Hsu SY, Matsushima A, Wang YM, Chen TB. Segmentation of liver tumors with abdominal computed tomography using fully convolutional networks. JOURNAL OF X-RAY SCIENCE AND TECHNOLOGY 2022; 30:953-966. [PMID: 35754254 DOI: 10.3233/xst-221194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
BACKGROUND Dividing liver organs or lesions depicting on computed tomography (CT) images could be applied to help tumor staging and treatment. However, most existing image segmentation technologies use manual or semi-automatic analysis, making the analysis process costly and time-consuming. OBJECTIVE This research aims to develop and apply a deep learning network architecture to segment liver tumors automatically after fine tuning parameters. METHODS AND MATERIALS The medical imaging is obtained from the International Symposium on Biomedical Imaging (ISBI), which includes 3D abdominal CT scans of 131 patients diagnosed with liver tumors. From these CT scans, there are 7,190 2D CT images along with the labeled binary images. The labeled binary images are regarded as gold standard for evaluation of the segmented results by FCN (Fully Convolutional Network). The backbones of FCN are extracted from Xception, InceptionresNetv2, MobileNetv2, ResNet18, ResNet50 in this study. Meanwhile, the parameters including optimizers (SGDM and ADAM), size of epoch, and size of batch are investigated. CT images are randomly divided into training and testing sets using a ratio of 9:1. Several evaluation indices including Global Accuracy, Mean Accuracy, Mean IoU (Intersection over Union), Weighted IoU and Mean BF Score are applied to evaluate tumor segmentation results in the testing images. RESULTS The Global Accuracy, Mean Accuracy, Mean IoU, Weighted IoU, and Mean BF Scores are 0.999, 0.969, 0.954, 0.998, 0.962 using ResNet50 in FCN with optimizer SGDM, batch size 12, and epoch 9. It is important to fine tuning the parameters in FCN model. Top 20 FNC models enable to achieve higher tumor segmentation accuracy with Mean IoU over 0.900. The occurred frequency of InceptionresNetv2, MobileNetv2, ResNet18, ResNet50, and Xception are 9, 6, 3, 5, and 2 times. Therefore, the InceptionresNetv2 has higher performance than others. CONCLUSIONS This study develop and test an automated liver tumor segmentation model based on FCN. Study results demonstrate that many deep learning models including InceptionresNetv2, MobileNetv2, ResNet18, ResNet50, and Xception have high potential to segment liver tumors from CT images with accuracy exceeding 90%. However, it is still difficult to accurately segment tiny and small size tumors by FCN models.
Collapse
Affiliation(s)
- Chih-I Chen
- Division of Colon and Rectal Surgery, Department of Surgery, E-DA Hospital, Kaohsiung City, Taiwan
- Division of General Medicine Surgery, Department of Surgery, E-DA Hospital, Kaohsiung City, Taiwan
- School of Medicine, College of Medicine, I-Shou University, Kaohsiung City, Taiwan
- Department of Information Engineering, I-Shou University, Kaohsiung City, Taiwan
- The School of Chinese Medicine for Post Baccalaureate, I-Shou University, Kaohsiung City, Taiwan
| | - Nan-Han Lu
- Department of Pharmacy, Tajen University, Pingtung City, Taiwan
- Department of Radiology, E-DA Hospital, I-Shou University, Kaohsiung City, Taiwan
- Department of Medical Imaging and Radiological Science, I-Shou University, Kaohsiung City, Taiwan
| | - Yung-Hui Huang
- Department of Medical Imaging and Radiological Science, I-Shou University, Kaohsiung City, Taiwan
| | - Kuo-Ying Liu
- Department of Radiology, E-DA Hospital, I-Shou University, Kaohsiung City, Taiwan
| | - Shih-Yen Hsu
- Department of Information Engineering, I-Shou University, Kaohsiung City, Taiwan
| | - Akari Matsushima
- Department of Radiological Technology Faculty of Medical Technology, Teikyo University, Tokyo, Japan
| | - Yi-Ming Wang
- Department of Information Engineering, I-Shou University, Kaohsiung City, Taiwan
- Department of Critical Care Medicine, E-DA hospital, I-Shou University, Kaohsiung City, Taiwan
| | - Tai-Been Chen
- Department of Medical Imaging and Radiological Science, I-Shou University, Kaohsiung City, Taiwan
- Institute of Statistics, National Yang Ming Chiao Tung University, Hsinchu, Taiwan
| |
Collapse
|