BPG is committed to discovery and dissemination of knowledge
Review Open Access
Copyright ©The Author(s) 2025. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Dec 14, 2025; 31(46): 111176
Published online Dec 14, 2025. doi: 10.3748/wjg.v31.i46.111176
Artificial intelligence in hepatopathy diagnosis and treatment: Big data analytics, deep learning, and clinical prediction models
Jing-Ran Sun, Bao-Cheng Deng, The Second Department of Infectious Diseases, The First Affiliated Hospital, China Medical University, Shenyang 110001, Liaoning Province, China
Jing-Ran Sun, Bing-Jiu Lu, Department of Hepatology, Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang 110032, Liaoning Province, China
Xiao-Ning Sun, Department of Geriatrics, Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang 110032, Liaoning Province, China
ORCID number: Bao-Cheng Deng (0000-0003-4825-9794).
Co-first authors: Jing-Ran Sun and Xiao-Ning Sun.
Co-corresponding authors: Bing-Jiu Lu and Bao-Cheng Deng.
Author contributions: Sun JR and Sun XN contributed equally to this work, they participated in the literature review, data collection, and manuscript writing; Lu BJ and Deng BC contributed equally to this work, they designed the draft and critically reviewed the manuscript for academic rigor. All authors have read and approved the final manuscript.
Supported by the Science Planning Project of Liaoning Province, No. 2019JH2/10300031-05; and the National Natural Science Foundation of China, No. 12171074.
Conflict-of-interest statement: All the authors report no relevant conflicts of interest for this article.
Open Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Bao-Cheng Deng, PhD, The Second Department of Infectious Diseases, The First Affiliated Hospital, China Medical University, No. 155 Nanjing North Street, Shenyang 110001, Liaoning Province, China. sydengbc@163.com
Received: June 30, 2025
Revised: August 31, 2025
Accepted: October 21, 2025
Published online: December 14, 2025
Processing time: 165 Days and 13.5 Hours

Abstract

Artificial intelligence (AI) is rapidly transforming the landscape of hepatology by enabling automated data interpretation, early disease detection, and individualized treatment strategies. Chronic liver diseases, including non-alcoholic fatty liver disease, cirrhosis, and hepatocellular carcinoma, often progress silently and pose diagnostic challenges due to reliance on invasive biopsies and operator-dependent imaging. This review explores the integration of AI across key domains such as big data analytics, deep learning-based image analysis, histopathological interpretation, biomarker discovery, and clinical prediction modeling. AI algorithms have demonstrated high accuracy in liver fibrosis staging, hepatocellular carcinoma detection, and non-alcoholic fatty liver disease risk stratification, while also enhancing survival prediction and treatment response assessment. For instance, convolutional neural networks trained on portal venous-phase computed tomography have achieved area under the curves up to 0.92 for significant fibrosis (F2-F4) and 0.89 for advanced fibrosis, with magnetic resonance imaging-based models reporting comparable performance. Advanced methodologies such as federated learning preserve patient privacy during cross-center model training, and explainable AI techniques promote transparency and clinician trust. Despite these advancements, clinical adoption remains limited by challenges including data heterogeneity, algorithmic bias, regulatory uncertainty, and lack of real-time integration into electronic health records. Looking forward, the convergence of multi-omics, imaging, and clinical data through interpretable and validated AI frameworks holds great promise for precision liver care. Continued efforts in model standardization, ethical oversight, and clinician-centered deployment will be essential to realize the full potential of AI in hepatopathy diagnosis and treatment.

Key Words: Artificial intelligence; Hepatology; Liver disease diagnosis; Deep learning; Clinical prediction models

Core Tip: This review highlights how artificial intelligence is transforming hepatology by enabling early diagnosis, fibrosis staging, hepatocellular carcinoma detection, and personalized treatment. Key innovations include deep learning for imaging, multi-omics integration, and privacy-preserving federated learning. Explainable artificial intelligence builds clinician trust. Despite promising results, challenges like data heterogeneity, regulatory barriers, and limited real-time integration remain. Continued efforts in validation, ethical oversight, and user-centered design are essential for clinical adoption.



INTRODUCTION

Liver diseases are a major global health problem. They include viral hepatitis, nonalcoholic fatty liver disease (NAFLD), alcoholic liver disease, autoimmune hepatitis, cirrhosis, and hepatocellular carcinoma (HCC). In 2019, chronic liver diseases caused more than 2 million deaths worldwide. Cirrhosis is now one of the top ten causes of death, especially in low- and middle-income countries[1]. NAFLD affects over 25% of people worldwide. It is one of the most common chronic liver diseases[2]. These liver diseases often have no symptoms in the early stages. Many patients are not diagnosed until fibrosis or cancer has already developed. This makes early detection and treatment difficult.

Doctors often rely on blood tests like alanine aminotransferase and aspartate aminotransferase, imaging tools such as ultrasound, computed tomography (CT), or magnetic resonance imaging (MRI), and liver biopsy to make a diagnosis. Although biopsy is still the best way to assess fibrosis, it is invasive, expensive, and prone to sampling errors and disagreement between doctors[3]. Imaging also depends on the operator’s skill and often fails to detect early-stage disease. Most current clinical systems do not use detailed data like genetic profiles, long-term medical records, or imaging features, which limits precision diagnosis and personalized care.

Artificial intelligence (AI) is helping change this. AI uses large and complex datasets to detect patterns, make predictions, and support clinical decisions[4,5]. In liver disease, AI can analyze medical images, identify patients at risk, predict disease progression, and help guide treatment. For example, convolutional neural networks (CNNs) can classify liver tumors on scans with accuracy similar to expert radiologists[6]. Deep learning also improves the detection of HCC on CT images[7]. As an example, CT-based deep learning systems have reported area under the curve (AUC) of 0.89-0.92 for fibrosis staging[8,9], while multiphasic MRI CNNs reach approximately 0.91 for HCC differentiation, underscoring the clinical promise of AI[10]. This review discusses how AI is improving the diagnosis and treatment of liver disease. It covers AI applications in big data analytics, image analysis, pathology, biomarker discovery, and prediction models for diagnosis, staging, and outcome prediction. It also highlights current challenges such as model generalizability, interpretability, clinical use, and ethical concerns. Together, these topics show how AI is shaping the future of precision liver care.

Throughout this review, several technical terms are used. Explainable AI (XAI) refers to models that provide interpretable predictions, making them more acceptable in clinical practice. Federated learning (FL) enables decentralized training of models across hospitals without sharing raw patient data. Edge AI refers to running AI models locally on devices near data sources (e.g., imaging machines), which reduces latency and protects privacy. These concepts will be discussed in context within relevant sections.

In particular, given the rising clinical concern around drug-induced liver injury (DILI), and the growing application of AI-based models for hepatotoxicity prediction, this review includes a dedicated section on AI applications for DILI. We believe that separating DILI from general treatment decision modeling provides clearer thematic coherence and highlights its distinct importance in hepatology.

OVERVIEW OF AI TECHNOLOGIES IN MEDICINE

AI, including machine learning (ML), deep learning (DL), and big data analytics, is showing clear value in hepatology. These tools help extract useful clinical information from different types of data. Common data sources include medical images (such as CT, MRI, and ultrasound), multi-omics datasets (like genomics, proteomics, and metabolomics), and electronic health records (EHRs). Using these inputs, AI can support more accurate diagnosis, better disease staging, and personalized treatment planning.

AI-based clinical decision support systems (CDSS) usually follow several steps. First, they collect and clean the data. Then, they build models using algorithms that handle both structured and unstructured data. After model development, they test performance and integrate the system into daily clinical workflows[11-14]. For example, CNNs have reached high accuracy in detecting liver tumors and mapping liver structures on scans[15,16]. Other AI models built from EHR data can predict how liver disease will progress or how patients may respond to treatment, especially in cases of cirrhosis or HCC[12]. Studies also show that combining imaging with clinical or molecular data improves predictions more than using a single type of data alone[17].

Even with this progress, there are still important challenges. One issue is that many AI models do not work well across different hospitals or patient populations. Another is that doctors often cannot understand how the model makes decisions, which makes them less likely to use it. In addition, these tools must fit smoothly into existing hospital systems. Data privacy, fairness, and transparency are also major concerns[18]. To move forward, researchers and developers need to create clear standards for testing and reporting AI models. They also need to design systems that explain how results are generated. Finally, real-world studies are essential to show that AI can improve care in everyday clinical practice[19].

BIG DATA ANALYTICS IN HEPATOLOGY

The growing volume of clinical, molecular, and imaging data is pushing hepatology toward predictive, preventive, and personalized care. EHRs have been used with ML to find patients who are likely to have poor outcomes. These include disease progression from NAFLD to nonalcoholic steatohepatitis, liver failure due to cirrhosis, or the development of HCC[20,21]. These tools help guide targeted screening, which can lead to earlier treatment and reduce medical costs.

From a health-economics perspective, AI reduces costs through three complementary pathways. First, avoiding unnecessary invasive procedures: Noninvasive models for liver fibrosis - built on MRI/CT radiomics or routine laboratory data - support triage and can curb referrals for liver biopsy[22,23]. Second, enabling earlier diagnosis and less complex care: Opportunistic CT/MRI radiomics and primary-care AI screening surface fibrosis earlier, allowing timelier lifestyle or pharmacologic interventions; risk-stratified HCC surveillance further concentrates imaging where the expected benefit is highest[24,25]. Third, improving resource allocation and operational efficiency: In radiology services, implementation studies and early health-technology assessments show workflow gains (e.g., faster triage/reading) and favorable return on investment, with some tools projected to be cost-saving vs standard care when integrated into reporting workflows[26,27].

Using omics data - such as genomics, transcriptomics, and metabolomics - also helps define subtypes of liver disease. For example, genome-wide studies have linked specific genes (like MTARC1 and GPAM) to liver fat and fibrosis risk in NAFLD patients[28]. By combining multiple omics datasets, researchers have identified molecular subgroups of NAFLD and discovered key pathways involved in fibrosis[29].

Large imaging databases now support AI-based tools that detect liver lesions, measure liver size, and stage fibrosis. CT and MRI images have been used to train DL models that spot early signs of liver damage with high accuracy[3]. Radiomics, which analyzes image textures, can improve predictions when combined with clinical information. These models often perform better than standard imaging reports[30].

Still, several challenges limit clinical use. Medical data often vary between institutions. Some datasets lack labels or contain sensitive patient information. These issues make it hard to build models that work well across hospitals. Solutions such as FL, standardized data formats (like OMOP or FHIR), and secure computing systems can help researchers share data while protecting privacy[31,32]. Ethical issues also matter. AI tools must avoid bias and fairly represent all patient groups to support equal care.

Beyond individual care, big data also helps with public health planning. For example, EHR-based tools can estimate how common NAFLD or cirrhosis is in specific groups. This information supports health policy and resource planning. Tracking patient data over time also allows for flexible risk prediction, helping doctors adjust care based on how a disease changes[33]. In short, big data is changing hepatology by combining clinical records, omics, and imaging to better detect disease, divide patients into subgroups, and support population-level planning. To fully benefit from this approach, future work must overcome technical and ethical barriers and ensure broad, safe, and fair use of data.

DL APPLICATIONS IN LIVER DISEASE DIAGNOSIS

DL is changing how liver diseases are diagnosed. It is widely used in imaging, histology, and biomarker research.

Imaging: Fibrosis diagnosis and staging

In imaging, CNNs trained on contrast-enhanced CT or MRI scans have shown strong performance in staging liver fibrosis. For instance, one model using portal venous-phase CT achieved AUC values of 0.92 for significant fibrosis (F2-F4), 0.89 for advanced fibrosis (F3-F4), and 0.88 for cirrhosis (F4)[8]. A pilot study using contrast-enhanced CT found a moderate correlation with biopsy results (spearman ρ = 0.48) and reported AUCs between 0.73 and 0.76 for fibrosis staging[9]. Newer MRI-based methods have further improved detection. Some even apply XAI to show which image areas influence predictions[34].

Methodological details of key DL studies. Beyond general statements, pivotal imaging studies in hepatology report explicit architectures and external validation metrics. For fibrosis staging on contrast-enhanced CT, Choi et al[9] used a multi-phase 3D CNNs with residual blocks; on an external cohort (n = 100), AUCs reached 0.96 (≥ F2), 0.97 (≥ F3), and 0.95 (≥ F4) with balanced sensitivity/specificity profiles. On hepatobiliary-phase gadoxetic-acid MRI, Yasaka et al[35] trained a deep CNN that achieved high diagnostic performance for fibrosis staging in independent testing. More recently, deep residual networks on non-contrast or plain CT have also shown non-invasive staging capability with robust external testing[36]. In parallel, transformer-based models have emerged for liver tasks such as preoperative microvascular invasion prediction in HCC, showing competitive AUCs against strong CNN baselines and providing attention-based interpretability[37]. Together, these exemplars substantiate the claims of “high accuracy” with concrete architectures and validation-set performance.

Tumor detection (HCC and differentials)

DL also helps detect liver tumors. CNNs trained on multi-phase MRI have reached 91% accuracy (AUC = 0.912) in telling HCC apart from other liver lesions[10]. These tools offer fast and reliable options for tumor screening.

Pathology and biomarker modeling

In pathology, DL can analyze liver biopsy slides. One study used CNNs to grade how well HCC cells were differentiated, based on standard hematoxylin and eosin staining. The model helped doctors improve diagnostic accuracy[38]. For biomarker discovery, DL models have combined CT images and pathology data to assess the NAFLD activity score and fibrosis stage. These models provide non-invasive tools to help assess disease severity and guide treatment.

Translation toward clinical use and reporting standards

Some of these tools have already moved toward clinical use. A model trained on gadoxetic acid-enhanced MRI showed similar accuracy to MR elastography in staging fibrosis[39]. This suggests it could be used in regular clinical practice. But challenges remain. These include the need for large, high-quality datasets, making sure models work across different scanner types, and improving how well doctors can understand the model’s decisions. Guidelines like checklist for artificial intelligence in medical imaging and transparent reporting of a multivariable prediction model for individual prognosis or diagnosis-AI (TRIPOD-AI) now support better model reporting, testing, and sharing across hospitals[40].

Explainability in clinical review (XAI)

Overall, DL in liver disease is moving beyond early tests. New tools are becoming more useful in real-world settings. They can help with image reading, biopsy grading, and risk scoring - offering faster, more objective, and more consistent results in liver care. In practice, attention heatmaps on liver MRI can visually highlight sub-lesional rims, wash-in/wash-out zones, or peritumoral capsules that drove a malignant classification, enabling side-by-side review with radiologists[41,42]. LRP-style attributions on CT can mark parenchymal textures and periportal regions that contributed most to a fibrosis stage prediction, thereby increasing clinician confidence and facilitating error analysis[8,43,44].

CLINICAL PREDICTION MODELS FOR HEPATOPATHY
AI-based models for early diagnosis

AI is improving the early diagnosis of liver diseases. For NAFLD, Hsu et al[45] used a random forest (RF) model on large population datasets and reached an area under the receiver operating characteristic curve (AUROC) of 0.83 for identifying high-risk individuals. Zhang et al[46] built a CNN that analyzed ultrasound images and achieved an AUC of 0.89 for detecting ≥ F2 fibrosis. Yin et al[8] applied CNNs to portal-venous CT scans and obtained AUROC values of up to 0.92 for significant fibrosis. Yasaka et al[3] reported similar accuracy using CT-based DL tools with an AUC of about 0.89. Some multi-center studies that combine CT and MRI have shown improved generalization across patient populations[30].

AI also plays a growing role in early detection of HCC. For example, Xu et al’s ML tool improved the sensitivity of HCC screening in patients with hepatitis B-related cirrhosis[47]. Other studies using CNNs on multiphasic MRI achieved AUCs of 0.91 for distinguishing HCC from benign liver lesions[6]. These findings are supported by additional neural network approaches using medical images[48,49]. Together, these models offer non-invasive, fast, and consistent tools for population screening. They reduce the need for liver biopsy and lower the impact of operator-dependent variability.

Prognostic models for disease progression and survival

AI models are also used to predict how liver diseases will progress and how long patients may survive. Katzman et al[50] created an extreme gradient boosting model that outperformed traditional Cox regression in forecasting survival in HCC patients. Radiomics-based nomograms that combine clinical and imaging features can predict recurrence-free survival after liver surgery, with a C-index of about 0.76[51]. Other tools include improved versions of existing scores. For example, Zhu et al[52] modified the model for end-stage liver disease score to better predict outcomes in primary liver cancer. DeepSurv, a DL model, has been used to make personalized predictions in both cirrhosis and HCC[53]. Some research also focuses on dynamic models built from EHRs. These allow real-time updates of patient risk as new data becomes available[54,55]. Clinicians are beginning to use these models to adjust how often patients are monitored, how aggressive treatments should be, and whether liver transplant should be considered.

Treatment decision models and precision medicine

AI tools are helping doctors tailor treatment plans based on disease severity. For NAFLD, Zhang et al[56] developed a RF model using lab tests and achieved an AUROC of 0.91 for detecting moderate-to-severe disease, showing potential for guiding lifestyle or drug therapy. In HCC, Peng et al[57] used CT-based radiomics and DL to predict how patients would respond to transarterial chemoembolization. Their model showed high accuracy with AUC values up to 0.97 in training cohorts. External tests reported internal AUCs of 0.94 and external validation AUCs around 0.90[57].

ML is also being used to choose the right systemic treatment or immunotherapy. These models may include imaging features, genetic mutations, programmed death-ligand 1 status, and lab markers. Although many are still in testing, early results show they may predict treatment response better than traditional methods[58,59]. Reporting of model architecture and validation. For clinical prediction models that integrate laboratory tests and EHR features, we now explicitly report the learning algorithm and validation metrics whenever available (e.g., RF or gradient-boosted decision trees with AUROC, sensitivity, and specificity on external cohorts). Representative EHR-integrated or radiomics-augmented models in hepatology report external AUROCs (0.85-0.92) for advanced fibrosis/HCC-related endpoints, with calibration and decision-curve analyses complementing discrimination[60]. We have aligned our reporting with TRIPOD-AI/CONSORT-AI recommendations to enhance transparency and clinical interpretability.

AI for DILI prediction

DILI is a major cause of drug development failure and post-marketing drug withdrawal. Conventional toxicology approaches often lack predictive accuracy and mechanistic resolution, particularly in early-phase risk stratification. In this context, AI has emerged as a powerful tool to model, predict, and interpret DILI risk across diverse data modalities. Current AI applications in DILI prediction primarily include: Compound-based prediction using chemical descriptors, off-target profiling, and physicochemical properties, often implemented via RF, support vector machines, and deep neural networks; biological data modeling, such as transcriptomic or microarray analyses, to uncover gene-level predictors and regulatory cascade patterns preceding liver toxicity; integrative platforms that combine clinical laboratory data, pharmacogenomic inputs, and multi-source datasets for individualized DILI risk assessment; mechanistic and interpretable models (e.g., SHAP, virtual liver lobule simulations) that bridge black-box AI with biological interpretability and regulatory applicability (Table 1[61-69]).

Table 1 Artificial intelligence-based studies on drug-induced liver injury prediction, highlighting their methodological frameworks and key performance outcomes.
Ref.
Methodology
Results
Mostafa et al[61], 2024RF & MLP on large human DILI datasets, externally validated on failed drug candidatesRF accuracy 63%, MLP MCC 0.245; models flagged failed drugs in external test set
Lesiński et al[62], 2021RF combining gene expression & molecular descriptorsAUC approximately 0.73 (high vs low-risk classification)
Liu et al[63], 2022Gene-expression cascade modeling preceding DILI histopathologyMechanistic insights into pathways & TFs
Wang et al[64], 2022ML on microarray dataAUC > 0.80 for genes DDIT3, GADD45A, SLC3A2, RBM24
Rao et al[65], 2023SVM, RF, ANN on physicochemical & offtarget features for small moleculesAUC 0.88; sensitivity 0.73; specificity 0.90
Li et al[66], 2021DeepDILI: Deep learning combining coupled ML + Mold2 descriptorsMCC 0.331; outperformed conventional ML (RF, SVM)
Li et al[67], 20208-layer deep neural network on human cell-line transcriptomics (L1000)Training/IV AUC 0.802/0.798; balanced accuracies approximately 0.74
Xiao et al[68], 2024XGBoost, RF, LASSO for TB treatment DILI prediction with SHAP interpretabilityAUROC 0.89 in validation; strong model interpretability
Lee and Yoo[69], 2024InterDILI interpretable RF model on multi-dataset integration (substructures, descriptors)AUROC 0.88-0.97; AUPRC 0.81-0.95; feature insights
INTEGRATION OF AI IN CLINICAL PRACTICE
CDSS in hepatology

AI-powered CDSS are being developed to assist with liver disease management. These systems help predict HCC risk, monitor complications in cirrhosis, and guide treatment choices by analyzing imaging and clinical data. For example, Malik et al[10] outlined the expanding role of AI across early diagnosis, prognosis, and therapy selection in liver diseases. One CT-based tool, PLAN-B-DF, achieved strong predictive power, with a C-index of 0.91 in internal validation and 0.89 in external datasets. This outperformed traditional scoring models in predicting HCC risk among patients with chronic hepatitis B[70]. However, most of these systems remain in early development stages. External validation and widespread clinical use are still limited[71].

Regulatory, ethical, and interpretability considerations

Bringing AI-CDSS into clinical settings requires meeting regulatory standards and addressing ethical concerns. In the United States, the Food and Drug Administration (FDA) has introduced its software as a medical device action plan. This includes guidance on algorithm transparency, lifecycle monitoring, and good ML practices. In the European Union, the General Data Protection Regulation enforces strict rules for data privacy in healthcare AI[72]. Ethical issues remain a major concern. These include risks of algorithmic bias, lack of fairness, and automation bias, where clinicians may over-rely on AI decisions. Studies show that unless bias is actively addressed, AI systems may reinforce health disparities[73].

To improve transparency and model quality, guidelines like TRIPOD-AI and CONSORT-AI have been developed. These frameworks promote better reporting, validation, and reproducibility of AI-based clinical prediction models[74]. Operationally, saliency- or attention-based visualizations reviewed with radiologists can document whether the model relies on clinically credible cues (e.g., arterial rim, peritumoral capsule), supporting model verification in the imaging report workflow[41]. Pixel-wise relevance maps (e.g., LRP) further enable case-level audits on CT by localizing fibrosis-related textures (e.g., periportal change), aligning with governance requirements for post-hoc explainability and error analysis[8,43,75,76]. Despite these advances, common barriers still exist. These include alert fatigue, poor integration with EHRs, and limited real-time interpretability. Such issues continue to hinder routine clinical adoption.

Clinical translation and deployment challenges

Despite promising results in experimental and retrospective settings, the readiness of AI systems for clinical hepatology remains limited. Several barriers must be addressed before widespread deployment.

Regulatory hurdles: Both the United States FDA and the European Medicines Agency now provide frameworks for AI/ML-enabled medical devices, requiring continuous performance monitoring and transparent reporting of algorithm updates. FDA’s 2021 action plan for AI/ML-based software as a medical device emphasizes real-world performance monitoring and change control protocols[77].

Real-world barriers: Clinical translation is constrained by privacy regulations (Health Insurance Portability and Accountability Act/General Data Protection Regulation), lack of interoperability between hospital EHRs, and low clinician trust in ‘black-box’ models. For example, across-site validation studies show significant performance drops due to domain shifts, while surveys reveal that hepatologists express concerns over liability and interpretability[78,79].

Cost-effectiveness: AI can potentially reduce costs by avoiding unnecessary liver biopsies, enabling earlier disease detection and treatment, and optimizing imaging resource allocation. For instance, cost-effectiveness analyses of AI-enabled imaging in oncology indicate that early detection strategies reduce downstream treatment expenditures by up to 30%[80]. Similar modeling studies are beginning to emerge for chronic liver disease, though prospective economic evaluations remain limited. Taken together, these considerations underscore the importance of regulatory compliance, interoperability, and health economic validation to ensure safe, equitable, and sustainable adoption of AI in hepatology practice (Table 2).

Table 2 Artificial intelligence-augmented vs human-only diagnostic accuracy: Current evidence.
Task
AI model/dataset
AI performance
Comparator
Outcome
CT-based HCC detection[6]CNN on CT (deep segmentation, auto segment)Sensitivity approximately 92%, specificity approximately 97%RadiologistsOutperformed (AI Sn/Sp 92/98 vs 82.5/96.5); supports workflow
PLAN-B-DF (internal/external validation)[70]Auto segmentation + clinical dataC-index 0.91; 0.89Traditional risk scoresOutperformed
Ultrasound focal lesion detection[81]DL on B-mode USAUC approximately 0.93SonographersComparable performance
Radiomics MVI in HCC[82]Deep learning (large meta analysis)AUC approximately 0.97Non-DL ML (AUC 0.82)DL superior
Histopathology slide review[38]DL assistanceAccuracy approximately 0.885PathologistsAssisted improvements but risks of misguidance noted
User acceptance and workflow adaptation

Successful AI implementation depends on clinician trust and workflow fit. Doctors-especially transplant hepatologists-often stress that AI should support, not replace, human judgment[81,82]. Transparency and ease of use are essential. Practical design solutions can mitigate these barriers. For instance, interactive dashboards such as the GutGPT system were shown to reduce alert fatigue by prioritizing clinically relevant notifications[76]. Similarly, case studies of application program interfaces-based integration with EHRs have demonstrated smoother adoption by embedding AI predictions directly into radiology reports or hepatology consult notes, minimizing workflow disruption[27]. Some systems use dashboard interfaces, similar to “GutGPT”, to guide decisions and improve compliance with clinical guidelines. While these tools can enhance care quality, they may also disrupt workflow or lead to alert fatigue. Many reviews have found that lack of EHR integration and limited real-time feedback are key reasons why clinicians hesitate to adopt AI system.

CHALLENGES AND FUTURE DIRECTIONS
Data quality, bias, and generalizability

Reliable AI tools in hepatology depend on access to diverse and high-quality data. But most current datasets are retrospective and collected from single centers. These datasets often lack variation in patient ethnicity, liver disease causes, and imaging methods. As a result, AI models may show poor performance when tested in new settings. Ghosh et al[83] reported that some models lost more than 20% accuracy when applied to data from different hospitals.

Beyond performance drops across centers, several studies have highlighted real-world failures of AI in hepatology. For example, Yin et al[8] reported that a DL model for liver fibrosis staging, developed on predominantly Western image datasets, when applied in an Asian cohort exhibited notable performance drop-off (e.g., lower AUCs), thereby highlighting the risk of ethnic and etiological bias in model generalisation. Similarly, Abràmoff et al[73] emphasized that insufficient subgroup validation can reinforce health inequities if AI is deployed without fairness audits. In addition to ethnic variability, gender bias has also been documented - for instance, routine clinical and demographic feature-based ML tools like FibrAIm under-detect early steatosis and fibrosis in certain subpopulations, raising concerns about subgroup performance disparities in early screening tools for metabolic dysfunction-associated steatotic liver disease/steatohepatitis patients[84]. These examples caution against premature clinical deployment and highlight the need for prospective, multi-ethnic validation before routine use.

Cross-site domain shifts (scanner vendors, acquisition protocols, disease etiologies, and ethnicity) remain major sources of performance drop; model bias has been well documented in medical AI and requires pre-specifying sub-group analyses and fairness audits[72,73]. Adopting TRIPOD-AI for transparent reporting and conducting prospective, multi-center external validation are therefore essential to avoid spectrum bias and improve real-world reliability[74].

Moreover, evidence suggests that AI performance in hepatology is uneven across underrepresented populations. For instance, Obermeyer et al[85] showed that an algorithm widely used in United States healthcare underestimated risk in Black patients due to reliance on healthcare costs as a proxy for illness, demonstrating how systemic bias in training data can exacerbate disparities. Similarly, Nam et al[86] emphasized that most hepatology AI studies are derived from Western cohorts, with markedly lower accuracy when applied to Asian populations, underscoring the need for multi-ethnic validation before clinical deployment. Gender bias has also been reported - for example, blood-test-based AI models missed 44% of female liver disease cases compared to 23% in males, highlighting subgroup-specific risks that could worsen inequities if unaddressed[87]. Standard data pipelines and formats, such as OMOP and FHIR, are rarely used in liver studies. To improve generalizability, future research should focus on building multicenter, prospective datasets and on harmonizing metadata to reduce bias across institutions.

Multi-omics integration and real-time analytics

AI has enabled the integration of multi-omics data - such as genomics, transcriptomics, and metabolomics - into liver disease models. These tools can improve diagnosis, risk assessment, and treatment planning. A recent review in gut highlighted how AI turns omics data into meaningful clinical insights[83]. By combining single-cell and bulk omics data using DL or graph-based models, researchers can better classify disease subtypes and understand how they progress[88,89]. For example, by integrating genomic variants (e.g., PNPLA3[90], MTARC1[28]), transcriptomic signatures of fibrogenic activation[91], metabolomic lipid pathway shifts[92], and routine labs, an AI meta-model can identify NAFLD subtypes that respond preferentially to glucagon-like peptide 1 analogs[93] vs pioglitazone[94], enabling tailored therapy selection in clinics. However, technical challenges remain. These include differences in data types, timing, and limited interpretability. Some early-stage solutions use bio-inspired AI frameworks to link genotype with phenotype, but they are not yet widely adopted[95]. In addition, real-time AI tools at the bedside (edge AI) are limited due to hardware constraints and data processing speed. More research is needed to bring these systems into routine use.

FL and privacy-preserving models

Protecting patient privacy is a key challenge in AI model development. Centralized data sharing is often not allowed, especially in liver disease research where multi-institutional data is critical. FL offers a solution. It allows models to be trained across sites without moving patient data. Instead, only the model updates are shared. Several studies show that FL works well for liver imaging tasks. Bernecker et al[96] developed a method called Federated Normalization, which adapts FL to both CT and MRI data. The model achieved near-centralized performance, with Dice coefficients close to 0.96 across six liver imaging datasets[97]. In pathology, Lusnig et al[98] built a hybrid quantum FL model to grade hepatic steatosis. Their approach used quantum neural networks and achieved over 90% accuracy without any data sharing. These methods are especially useful in settings where biopsy data is sensitive.

Even with these successes, challenges remain. Medical data from different centers are not always distributed evenly. This can make training unstable and hurt performance[99]. Other barriers include high communication costs, synchronization issues, and the need for strong computing power. To address this, researchers are testing strategies like Federated Averaging, FedSGD, and split learning to improve speed and stability[100].

Towards explainable and trustworthy AI in hepatology

Building trust in AI requires transparency and interpretability. Tools like SHAP values and attention maps are now being used to explain AI decisions in omics and imaging[101]. For multi-omics models, surveys have shown how XAI techniques can point to key features and explain which data types are most important[102,103]. Combining explainable models with FL can lead to systems that are both secure and interpretable[74,97]. This makes it easier for clinicians to trust and adopt these tools. Regulatory groups are also supporting this shift. Guidelines like TRIPOD-AI and DECIDE-AI push for better reporting and validation of AI models[104]. Future deployment should also include clinician-centered design, regular feedback loops, and real-world testing of how AI affects workload and decision-making[105].

FUTURE DIRECTIONS

AI is transforming the landscape of hepatopathy diagnosis and treatment by enabling automated data analysis, improved disease stratification, and personalized therapeutic decision-making. This review comprehensively highlights AI applications across multiple domains: Big data analytics, DL-based imaging interpretation, histopathological analysis, biomarker discovery, and clinical prediction modeling. AI has demonstrated high diagnostic accuracy in liver fibrosis staging, HCC detection, and NAFLD stratification using CT, MRI, and ultrasound. Prognostic models integrating radiomics and EHR data offer improved survival predictions and facilitate treatment selection. AI-driven decision support systems have shown promise in enhancing the efficiency and precision of clinical workflows.

Furthermore, AI has enabled scalable early screening tools and non-invasive biomarkers, thus minimizing reliance on liver biopsy. FL has addressed data privacy issues while maintaining model performance across decentralized datasets. Advances in XAI have contributed to clinician trust by enhancing transparency in complex models. However, widespread clinical integration remains limited by issues such as data heterogeneity, regulatory ambiguity, and lack of real-time interpretability.

Looking forward, the promise of AI in hepatology lies in its potential to integrate multi-omics, imaging, and clinical data into unified, interpretable, and actionable models. This requires the development of robust, externally validated algorithms trained on large, ethnically diverse, and longitudinal datasets. Interoperability standards, such as OMOP and FHIR, should be adopted to harmonize data input across institutions. FL and edge AI represent promising frameworks for ensuring privacy-preserving, real-time analytics at the point of care. Moreover, regulatory frameworks such as TRIPOD-AI and DECIDE-AI should be universally implemented to standardize AI model reporting and validation. Clinician-in-the-loop design, user-centered interface development, and alert burden mitigation are critical to promote AI adoption in hepatology. Equally important is the incorporation of bioethical safeguards to ensure algorithmic fairness, accountability, and transparency.

CONCLUSION

This review summarizes the current progress and challenges of AI in the diagnosis and management of liver diseases. AI technologies - including big data analytics, DL, and clinical prediction modeling - have demonstrated promising potential across multiple domains, such as fibrosis staging, HCC detection, and non-invasive risk stratification. These tools support earlier diagnosis, individualized therapy, and more efficient clinical workflows. In addition, FL offers privacy-preserving solutions for multicenter model training, while XAI improves transparency and builds clinician trust. Despite these advances, barriers such as data heterogeneity, lack of real-time interpretability, and regulatory uncertainty remain. Therefore, we believe AI should be actively integrated but critically evaluated in hepatology. Future efforts should focus on large-scale validation, harmonized data standards, and user-centered design. With sustained investment in clinical translation, interpretability, and infrastructure, AI is poised to become a central component in the precision management of liver diseases. Looking ahead, we envision a learning hepatology ecosystem in which harmonized EHR, imaging, and multi-omics streams continuously update validated, explainable models at the bedside. In this future state, AI serves as an accountable clinical co-pilot - auditable, bias-aware, interoperable, and aligned with practice guidelines - supporting prevention, earlier diagnosis, and individualized therapy while reducing unwarranted variation. Progress should be judged not only by benchmark AUCs but by patient-centered outcomes, equity, safety, and efficiency. Realizing this vision will require shared standards, prospective trials, and governance that earns durable trust among patients, clinicians, and regulators.

Footnotes

Provenance and peer review: Invited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Gastroenterology and hepatology

Country of origin: China

Peer-review report’s classification

Scientific Quality: Grade B, Grade B, Grade B, Grade C, Grade C

Novelty: Grade B, Grade B, Grade B, Grade D, Grade D

Creativity or Innovation: Grade B, Grade B, Grade B, Grade D, Grade D

Scientific Significance: Grade B, Grade B, Grade B, Grade C, Grade D

P-Reviewer: Huang X, PhD, Associate Professor, Senior Researcher, China; Naeem MR, PhD, Lecturer, Researcher, Visiting Professor, Pakistan; Peng WL, MD, Lecturer, Researcher, China S-Editor: Wang JJ L-Editor: A P-Editor: Lei YY

References
1.  Asrani SK, Devarbhavi H, Eaton J, Kamath PS. Burden of liver diseases in the world. J Hepatol. 2019;70:151-171.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1382]  [Cited by in RCA: 2381]  [Article Influence: 396.8]  [Reference Citation Analysis (0)]
2.  Younossi ZM, Koenig AB, Abdelatif D, Fazel Y, Henry L, Wymer M. Global epidemiology of nonalcoholic fatty liver disease-Meta-analytic assessment of prevalence, incidence, and outcomes. Hepatology. 2016;64:73-84.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 5322]  [Cited by in RCA: 7755]  [Article Influence: 861.7]  [Reference Citation Analysis (0)]
3.  Yasaka K, Akai H, Abe O, Kiryu S. Deep Learning with Convolutional Neural Network for Differentiation of Liver Masses at Dynamic Contrast-enhanced CT: A Preliminary Study. Radiology. 2018;286:887-896.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 293]  [Cited by in RCA: 411]  [Article Influence: 51.4]  [Reference Citation Analysis (0)]
4.  Topol EJ. High-performance medicine: the convergence of human and artificial intelligence. Nat Med. 2019;25:44-56.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 2376]  [Cited by in RCA: 3185]  [Article Influence: 530.8]  [Reference Citation Analysis (4)]
5.  LeCun Y, Bengio Y, Hinton G. Deep learning. Nature. 2015;521:436-444.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 36149]  [Cited by in RCA: 20659]  [Article Influence: 2065.9]  [Reference Citation Analysis (0)]
6.  Hamm CA, Wang CJ, Savic LJ, Ferrante M, Schobert I, Schlachter T, Lin M, Duncan JS, Weinreb JC, Chapiro J, Letzen B. Deep learning for liver tumor diagnosis part I: development of a convolutional neural network classifier for multi-phasic MRI. Eur Radiol. 2019;29:3338-3347.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 117]  [Cited by in RCA: 221]  [Article Influence: 36.8]  [Reference Citation Analysis (0)]
7.  Okimoto N, Yasaka K, Kaiume M, Kanemaru N, Suzuki Y, Abe O. Improving detection performance of hepatocellular carcinoma and interobserver agreement for liver imaging reporting and data system on CT using deep learning reconstruction. Abdom Radiol (NY). 2023;48:1280-1289.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 16]  [Cited by in RCA: 15]  [Article Influence: 7.5]  [Reference Citation Analysis (0)]
8.  Yin Y, Yakar D, Dierckx RAJO, Mouridsen KB, Kwee TC, de Haas RJ. Liver fibrosis staging by deep learning: a visual-based explanation of diagnostic decisions of the model. Eur Radiol. 2021;31:9620-9627.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 4]  [Cited by in RCA: 29]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
9.  Choi KJ, Jang JK, Lee SS, Sung YS, Shim WH, Kim HS, Yun J, Choi JY, Lee Y, Kang BK, Kim JH, Kim SY, Yu ES. Development and Validation of a Deep Learning System for Staging Liver Fibrosis by Using Contrast Agent-enhanced CT Images in the Liver. Radiology. 2018;289:688-697.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 108]  [Cited by in RCA: 156]  [Article Influence: 22.3]  [Reference Citation Analysis (0)]
10.  Oestmann PM, Wang CJ, Savic LJ, Hamm CA, Stark S, Schobert I, Gebauer B, Schlachter T, Lin M, Weinreb JC, Batra R, Mulligan D, Zhang X, Duncan JS, Chapiro J. Deep learning-assisted differentiation of pathologically proven atypical and typical hepatocellular carcinoma (HCC) versus non-HCC on contrast-enhanced MRI of the liver. Eur Radiol. 2021;31:4981-4990.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 49]  [Cited by in RCA: 55]  [Article Influence: 13.8]  [Reference Citation Analysis (0)]
11.  Kalapala R, Rughwani H, Reddy DN. Artificial Intelligence in Hepatology- Ready for the Primetime. J Clin Exp Hepatol. 2023;13:149-161.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 2]  [Cited by in RCA: 8]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
12.  Su TH, Wu CH, Kao JH. Artificial intelligence in precision medicine in hepatology. J Gastroenterol Hepatol. 2021;36:569-580.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 19]  [Cited by in RCA: 40]  [Article Influence: 10.0]  [Reference Citation Analysis (0)]
13.  Malik S, Das R, Thongtan T, Thompson K, Dbouk N. AI in Hepatology: Revolutionizing the Diagnosis and Management of Liver Disease. J Clin Med. 2024;13:7833.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 5]  [Reference Citation Analysis (0)]
14.  Le Berre C, Sandborn WJ, Aridhi S, Devignes MD, Fournier L, Smaïl-Tabbone M, Danese S, Peyrin-Biroulet L. Application of Artificial Intelligence to Gastroenterology and Hepatology. Gastroenterology. 2020;158:76-94.e2.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 230]  [Cited by in RCA: 342]  [Article Influence: 68.4]  [Reference Citation Analysis (1)]
15.  Yin C, Zhang H, Du J, Zhu Y, Zhu H, Yue H. Artificial intelligence in imaging for liver disease diagnosis. Front Med (Lausanne). 2025;12:1591523.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 6]  [Reference Citation Analysis (0)]
16.  Schattenberg JM, Chalasani N, Alkhouri N. Artificial Intelligence Applications in Hepatology. Clin Gastroenterol Hepatol. 2023;21:2015-2025.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1]  [Cited by in RCA: 29]  [Article Influence: 14.5]  [Reference Citation Analysis (0)]
17.  Mohsen F, Ali H, El Hajj N, Shah Z. Artificial intelligence-based methods for fusion of electronic health records and imaging data. Sci Rep. 2022;12:17981.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 1]  [Cited by in RCA: 72]  [Article Influence: 24.0]  [Reference Citation Analysis (0)]
18.  Ahn JC, Connell A, Simonetto DA, Hughes C, Shah VH. Application of Artificial Intelligence for the Diagnosis and Treatment of Liver Diseases. Hepatology. 2021;73:2546-2563.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 49]  [Cited by in RCA: 107]  [Article Influence: 26.8]  [Reference Citation Analysis (0)]
19.  Khalifa A, Obeid JS, Erno J, Rockey DC. The role of artificial intelligence in hepatology research and practice. Curr Opin Gastroenterol. 2023;39:175-180.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 4]  [Cited by in RCA: 7]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
20.  Chen JH, Asch SM. Machine Learning and Prediction in Medicine - Beyond the Peak of Inflated Expectations. N Engl J Med. 2017;376:2507-2509.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 547]  [Cited by in RCA: 630]  [Article Influence: 78.8]  [Reference Citation Analysis (4)]
21.  Thrift AP, Nguyen Wenker TH, Godwin K, Balakrishnan M, Duong HT, Loomba R, Kanwal F, El-Serag HB. An Electronic Health Record Model for Predicting Risk of Hepatic Fibrosis in Primary Care Patients. Dig Dis Sci. 2024;69:2430-2436.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Reference Citation Analysis (0)]
22.  Gotta J, Gruenewald LD, Reschke P, Booz C, Mahmoudi S, Stieltjes B, Choi MH, D'Angelo T, Bernatz S, Vogl TJ, Sinkus R, Grimm R, Strecker R, Haberkorn S, Koch V. Noninvasive Grading of Liver Fibrosis Based on Texture Analysis From MRI-Derived Radiomics. NMR Biomed. 2025;38:e5301.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 2]  [Reference Citation Analysis (0)]
23.  Yoo JJ, Namdar K, Carey S, Fischer SE, McIntosh C, Khalvati F, Rogalla P. Non-invasive liver fibrosis screening on CT images using radiomics. BMC Med Imaging. 2025;25:285.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 3]  [Reference Citation Analysis (0)]
24.  Blanes-Vidal V, Lindvig KP, Thiele M, Nadimi ES, Krag A. Artificial intelligence outperforms standard blood-based scores in identifying liver fibrosis patients in primary care. Sci Rep. 2022;12:2914.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 21]  [Reference Citation Analysis (0)]
25.  Sarkar S, Alurwar A, Ly C, Piao C, Donde R, Wang CJ, Meyers FJ. A Machine Learning Model to Predict Risk for Hepatocellular Carcinoma in Patients With Metabolic Dysfunction-Associated Steatotic Liver Disease. Gastro Hep Adv. 2024;3:498-505.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 6]  [Reference Citation Analysis (0)]
26.  Wenderott K, Krups J, Zaruchas F, Weigl M. Effects of artificial intelligence implementation on efficiency in medical imaging-a systematic literature review and meta-analysis. NPJ Digit Med. 2024;7:265.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 23]  [Reference Citation Analysis (0)]
27.  van Leeuwen KG, Meijer FJA, Schalekamp S, Rutten MJCM, van Dijk EJ, van Ginneken B, Govers TM, de Rooij M. Cost-effectiveness of artificial intelligence aided vessel occlusion detection in acute stroke: an early health technology assessment. Insights Imaging. 2021;12:133.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 4]  [Cited by in RCA: 30]  [Article Influence: 7.5]  [Reference Citation Analysis (0)]
28.  Sveinbjornsson G, Ulfarsson MO, Thorolfsdottir RB, Jonsson BA, Einarsson E, Gunnlaugsson G, Rognvaldsson S, Arnar DO, Baldvinsson M, Bjarnason RG; DBDS Genomic consortium, Eiriksdottir T, Erikstrup C, Ferkingstad E, Halldorsson GH, Helgason H, Helgadottir A, Hindhede L, Hjorleifsson G, Jones D, Knowlton KU, Lund SH, Melsted P, Norland K, Olafsson I, Olafsson S, Oskarsson GR, Ostrowski SR, Pedersen OB, Snaebjarnarson AS, Sigurdsson E, Steinthorsdottir V, Schwinn M, Thorgeirsson G, Thorleifsson G, Jonsdottir I, Bundgaard H, Nadauld L, Bjornsson ES, Rulifson IC, Rafnar T, Norddahl GL, Thorsteinsdottir U, Sulem P, Gudbjartsson DF, Holm H, Stefansson K. Multiomics study of nonalcoholic fatty liver disease. Nat Genet. 2022;54:1652-1663.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 6]  [Cited by in RCA: 153]  [Article Influence: 51.0]  [Reference Citation Analysis (0)]
29.  Ding J, Liu H, Zhang X, Zhao N, Peng Y, Shi J, Chen J, Chi X, Li L, Zhang M, Liu WY, Zhang L, Ouyang J, Yuan Q, Liao M, Tan Y, Li M, Xu Z, Tang W, Xie C, Li Y, Pan Q, Xu Y, Cai SY, Byrne CD, Targher G, Ouyang X, Zhang L, Jiang Z, Zheng MH, Sun F, Chai J. Integrative multiomic analysis identifies distinct molecular subtypes of NAFLD in a Chinese population. Sci Transl Med. 2024;16:eadh9940.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 8]  [Cited by in RCA: 12]  [Article Influence: 12.0]  [Reference Citation Analysis (0)]
30.  Tang M, Wu Y, Hu N, Lin C, He J, Xia X, Yang M, Lei P, Luo P. A combination model of CT-based radiomics and clinical biomarkers for staging liver fibrosis in the patients with chronic liver disease. Sci Rep. 2024;14:20230.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 6]  [Reference Citation Analysis (0)]
31.  Dayan I, Roth HR, Zhong A, Harouni A, Gentili A, Abidin AZ, Liu A, Costa AB, Wood BJ, Tsai CS, Wang CH, Hsu CN, Lee CK, Ruan P, Xu D, Wu D, Huang E, Kitamura FC, Lacey G, de Antônio Corradi GC, Nino G, Shin HH, Obinata H, Ren H, Crane JC, Tetreault J, Guan J, Garrett JW, Kaggie JD, Park JG, Dreyer K, Juluru K, Kersten K, Rockenbach MABC, Linguraru MG, Haider MA, AbdelMaseeh M, Rieke N, Damasceno PF, E Silva PMC, Wang P, Xu S, Kawano S, Sriswasdi S, Park SY, Grist TM, Buch V, Jantarabenjakul W, Wang W, Tak WY, Li X, Lin X, Kwon YJ, Quraini A, Feng A, Priest AN, Turkbey B, Glicksberg B, Bizzo B, Kim BS, Tor-Díez C, Lee CC, Hsu CJ, Lin C, Lai CL, Hess CP, Compas C, Bhatia D, Oermann EK, Leibovitz E, Sasaki H, Mori H, Yang I, Sohn JH, Murthy KNK, Fu LC, de Mendonça MRF, Fralick M, Kang MK, Adil M, Gangai N, Vateekul P, Elnajjar P, Hickman S, Majumdar S, McLeod SL, Reed S, Gräf S, Harmon S, Kodama T, Puthanakit T, Mazzulli T, de Lavor VL, Rakvongthai Y, Lee YR, Wen Y, Gilbert FJ, Flores MG, Li Q. Federated learning for predicting clinical outcomes in patients with COVID-19. Nat Med. 2021;27:1735-1743.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 278]  [Cited by in RCA: 250]  [Article Influence: 62.5]  [Reference Citation Analysis (0)]
32.  Hripcsak G, Duke JD, Shah NH, Reich CG, Huser V, Schuemie MJ, Suchard MA, Park RW, Wong IC, Rijnbeek PR, van der Lei J, Pratt N, Norén GN, Li YC, Stang PE, Madigan D, Ryan PB. Observational Health Data Sciences and Informatics (OHDSI): Opportunities for Observational Researchers. Stud Health Technol Inform. 2015;216:574-578.  [PubMed]  [DOI]
33.  Abbas SR, Abbas Z, Zahir A, Lee SW. Federated Learning in Smart Healthcare: A Comprehensive Review on Privacy, Security, and Predictive Analytics with IoT Integration. Healthcare (Basel). 2024;12:2587.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 26]  [Reference Citation Analysis (0)]
34.  Yu Y, Wang J, Ng CW, Ma Y, Mo S, Fong ELS, Xing J, Song Z, Xie Y, Si K, Wee A, Welsch RE, So PTC, Yu H. Deep learning enables automated scoring of liver fibrosis stages. Sci Rep. 2018;8:16016.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 48]  [Cited by in RCA: 65]  [Article Influence: 9.3]  [Reference Citation Analysis (0)]
35.  Yasaka K, Akai H, Kunimatsu A, Abe O, Kiryu S. Liver Fibrosis: Deep Convolutional Neural Network for Staging by Using Gadoxetic Acid-enhanced Hepatobiliary Phase MR Images. Radiology. 2018;287:146-155.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 112]  [Cited by in RCA: 155]  [Article Influence: 19.4]  [Reference Citation Analysis (1)]
36.  Li Q, Kang H, Zhang R, Guo Q. Non-invasive precise staging of liver fibrosis using deep residual network model based on plain CT images. Int J Comput Assist Radiol Surg. 2022;17:627-637.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 2]  [Cited by in RCA: 2]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
37.  Cao L, Wang Q, Hong J, Han Y, Zhang W, Zhong X, Che Y, Ma Y, Du K, Wu D, Pang T, Wu J, Liang K. MVI-TR: A Transformer-Based Deep Learning Model with Contrast-Enhanced CT for Preoperative Prediction of Microvascular Invasion in Hepatocellular Carcinoma. Cancers (Basel). 2023;15:1538.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 14]  [Cited by in RCA: 17]  [Article Influence: 8.5]  [Reference Citation Analysis (0)]
38.  Kiani A, Uyumazturk B, Rajpurkar P, Wang A, Gao R, Jones E, Yu Y, Langlotz CP, Ball RL, Montine TJ, Martin BA, Berry GJ, Ozawa MG, Hazard FK, Brown RA, Chen SB, Wood M, Allard LS, Ylagan L, Ng AY, Shen J. Impact of a deep learning assistant on the histopathologic classification of liver cancer. NPJ Digit Med. 2020;3:23.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 181]  [Cited by in RCA: 167]  [Article Influence: 33.4]  [Reference Citation Analysis (0)]
39.  Hectors SJ, Kennedy P, Huang KH, Stocker D, Carbonell G, Greenspan H, Friedman S, Taouli B. Fully automated prediction of liver fibrosis using deep learning analysis of gadoxetic acid-enhanced MRI. Eur Radiol. 2021;31:3805-3814.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 13]  [Cited by in RCA: 49]  [Article Influence: 9.8]  [Reference Citation Analysis (0)]
40.  Lambin P, Leijenaar RTH, Deist TM, Peerlings J, de Jong EEC, van Timmeren J, Sanduleanu S, Larue RTHM, Even AJG, Jochems A, van Wijk Y, Woodruff H, van Soest J, Lustberg T, Roelofs E, van Elmpt W, Dekker A, Mottaghy FM, Wildberger JE, Walsh S. Radiomics: the bridge between medical imaging and personalized medicine. Nat Rev Clin Oncol. 2017;14:749-762.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1825]  [Cited by in RCA: 3774]  [Article Influence: 471.8]  [Reference Citation Analysis (0)]
41.  Li M, Zhang Z, Chen Z, Chen X, Liu H, Xiao Y, Chen H, Zong X, Chen J, Chen J, Wang X, Xiao X, Yang Z, Han L, Wang J, Wu B. Interactive Explainable Deep Learning Model for Hepatocellular Carcinoma Diagnosis at Gadoxetic Acid-enhanced MRI: A Retrospective, Multicenter, Diagnostic Study. Radiol Imaging Cancer. 2025;7:e240332.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 1]  [Cited by in RCA: 1]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
42.  Dong X, Jia X, Zhang W, Zhang J, Xu H, Xu L, Ma C, Hu H, Luo J, Zhang J, Wang Z, Ji W, Yang D, Yang Z. Interpretable and generalizable deep learning model for preoperative assessment of microvascular invasion and outcome in hepatocellular carcinoma based on MRI: a multicenter study. Insights Imaging. 2025;16:151.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Reference Citation Analysis (0)]
43.  Böhle M, Eitel F, Weygandt M, Ritter K. Layer-Wise Relevance Propagation for Explaining Deep Neural Network Decisions in MRI-Based Alzheimer's Disease Classification. Front Aging Neurosci. 2019;11:194.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 159]  [Cited by in RCA: 111]  [Article Influence: 18.5]  [Reference Citation Analysis (0)]
44.  Budai BK, Tóth A, Borsos P, Frank VG, Shariati S, Fejér B, Folhoffer A, Szalay F, Bérczi V, Kaposi PN. Three-dimensional CT texture analysis of anatomic liver segments can differentiate between low-grade and high-grade fibrosis. BMC Med Imaging. 2020;20:108.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 29]  [Cited by in RCA: 25]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
45.  Hsu C, Caussy C, Imajo K, Chen J, Singh S, Kaulback K, Le MD, Hooker J, Tu X, Bettencourt R, Yin M, Sirlin CB, Ehman RL, Nakajima A, Loomba R. Magnetic Resonance vs Transient Elastography Analysis of Patients With Nonalcoholic Fatty Liver Disease: A Systematic Review and Pooled Analysis of Individual Participants. Clin Gastroenterol Hepatol. 2019;17:630-637.e8.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 258]  [Cited by in RCA: 311]  [Article Influence: 51.8]  [Reference Citation Analysis (0)]
46.  Zhang L, Tan Z, Li C, Mou L, Shi YL, Zhu XX, Luo Y. A Deep Learning Model Based on High-Frequency Ultrasound Images for Classification of Different Stages of Liver Fibrosis. Liver Int. 2025;45:e70148.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 2]  [Reference Citation Analysis (0)]
47.  Xu Y, Zhang B, Zhou F, Yi YP, Yang XL, Ouyang X, Hu H. Development of machine learning-based personalized predictive models for risk evaluation of hepatocellular carcinoma in hepatitis B virus-related cirrhosis patients with low levels of serum alpha-fetoprotein. Ann Hepatol. 2024;29:101540.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 5]  [Reference Citation Analysis (1)]
48.  Cao SE, Zhang LQ, Kuang SC, Shi WQ, Hu B, Xie SD, Chen YN, Liu H, Chen SM, Jiang T, Ye M, Zhang HX, Wang J. Multiphase convolutional dense network for the classification of focal liver lesions on dynamic contrast-enhanced computed tomography. World J Gastroenterol. 2020;26:3660-3672.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in CrossRef: 23]  [Cited by in RCA: 32]  [Article Influence: 6.4]  [Reference Citation Analysis (3)]
49.  Yang CJ, Wang CK, Fang YD, Wang JY, Su FC, Tsai HM, Lin YJ, Tsai HW, Yeh LR. Clinical application of mask region-based convolutional neural network for the automatic detection and segmentation of abnormal liver density based on hepatocellular carcinoma computed tomography datasets. PLoS One. 2021;16:e0255605.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 1]  [Cited by in RCA: 9]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
50.  Katzman JL, Shaham U, Cloninger A, Bates J, Jiang T, Kluger Y. DeepSurv: personalized treatment recommender system using a Cox proportional hazards deep neural network. BMC Med Res Methodol. 2018;18:24.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 398]  [Cited by in RCA: 809]  [Article Influence: 115.6]  [Reference Citation Analysis (0)]
51.  Li N, Wan X, Zhang H, Zhang Z, Guo Y, Hong D. Tumor and peritumor radiomics analysis based on contrast-enhanced CT for predicting early and late recurrence of hepatocellular carcinoma after liver resection. BMC Cancer. 2022;22:664.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 21]  [Reference Citation Analysis (0)]
52.  Zhu HB, Zheng ZY, Zhao H, Zhang J, Zhu H, Li YH, Dong ZY, Xiao LS, Kuang JJ, Zhang XL, Liu L. Radiomics-based nomogram using CT imaging for noninvasive preoperative prediction of early recurrence in patients with hepatocellular carcinoma. Diagn Interv Radiol. 2020;26:411-419.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 13]  [Cited by in RCA: 27]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
53.  Kartoun U, Corey KE, Simon TG, Zheng H, Aggarwal R, Ng K, Shaw SY. The MELD-Plus: A generalizable prediction risk score in cirrhosis. PLoS One. 2017;12:e0186301.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 32]  [Cited by in RCA: 47]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
54.  Martinez Chanza N, Werner L, Plimack E, Yu EY, Alva AS, Crabb SJ, Powles T, Rosenberg JE, Baniel J, Vaishampayan UN, Berthold DR, Ladoire S, Hussain SA, Milowsky MI, Agarwal N, Necchi A, Pal SK, Sternberg CN, Bellmunt J, Galsky MD, Harshman LC; RISC Investigators. Incidence, Patterns, and Outcomes with Adjuvant Chemotherapy for Residual Disease After Neoadjuvant Chemotherapy in Muscle-invasive Urinary Tract Cancers. Eur Urol Oncol. 2020;3:671-679.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 7]  [Cited by in RCA: 8]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
55.  Zhang P, Shi Y, Zhou M, Mao Q, Tao Y, Yang L, Zhang X. A CECT-Based Radiomics Nomogram Predicts the Overall Survival of Patients with Hepatocellular Carcinoma After Surgical Resection. Biomedicines. 2025;13:1237.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Reference Citation Analysis (0)]
56.  Zhang L, Huang Y, Huang M, Zhao CH, Zhang YJ, Wang Y. Development of Cost-Effective Fatty Liver Disease Prediction Models in a Chinese Population: Statistical and Machine Learning Approaches. JMIR Form Res. 2024;8:e53654.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Reference Citation Analysis (0)]
57.  Peng J, Huang J, Huang G, Zhang J. Predicting the Initial Treatment Response to Transarterial Chemoembolization in Intermediate-Stage Hepatocellular Carcinoma by the Integration of Radiomics and Deep Learning. Front Oncol. 2021;11:730282.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 34]  [Reference Citation Analysis (0)]
58.  Miranda J, Horvat N, Fonseca GM, Araujo-Filho JAB, Fernandes MC, Charbel C, Chakraborty J, Coelho FF, Nomura CH, Herman P. Current status and future perspectives of radiomics in hepatocellular carcinoma. World J Gastroenterol. 2023;29:43-60.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in CrossRef: 1]  [Cited by in RCA: 22]  [Article Influence: 11.0]  [Reference Citation Analysis (4)]
59.  Cui H, Zeng L, Li R, Li Q, Hong C, Zhu H, Chen L, Liu L, Zou X, Xiao L. Radiomics signature based on CECT for non-invasive prediction of response to anti-PD-1 therapy in patients with hepatocellular carcinoma. Clin Radiol. 2023;78:e37-e44.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 8]  [Reference Citation Analysis (0)]
60.  Sarvestany SS, Kwong JC, Azhie A, Dong V, Cerocchi O, Ali AF, Karnam RS, Kuriry H, Shengir M, Candido E, Duchen R, Sebastiani G, Patel K, Goldenberg A, Bhat M. Development and validation of an ensemble machine learning framework for detection of all-cause advanced hepatic fibrosis: a retrospective cohort study. Lancet Digit Health. 2022;4:e188-e199.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 3]  [Cited by in RCA: 18]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
61.  Mostafa F, Howle V, Chen M. Machine Learning to Predict Drug-Induced Liver Injury and Its Validation on Failed Drug Candidates in Development. Toxics. 2024;12:385.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 3]  [Reference Citation Analysis (0)]
62.  Lesiński W, Mnich K, Golińska AK, Rudnicki WR. Integration of human cell lines gene expression and chemical properties of drugs for Drug Induced Liver Injury prediction. Biol Direct. 2021;16:2.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 4]  [Cited by in RCA: 5]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
63.  Liu A, Han N, Munoz-Muriedas J, Bender A. Deriving time-concordant event cascades from gene expression data: A case study for Drug-Induced Liver Injury (DILI). PLoS Comput Biol. 2022;18:e1010148.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 4]  [Reference Citation Analysis (0)]
64.  Wang K, Zhang L, Li L, Wang Y, Zhong X, Hou C, Zhang Y, Sun C, Zhou Q, Wang X. Identification of Drug-Induced Liver Injury Biomarkers from Multiple Microarrays Based on Machine Learning and Bioinformatics Analysis. Int J Mol Sci. 2022;23:11945.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 10]  [Reference Citation Analysis (0)]
65.  Rao M, Nassiri V, Alhambra C, Snoeys J, Van Goethem F, Irrechukwu O, Aleo MD, Geys H, Mitra K, Will Y. AI/ML Models to Predict the Severity of Drug-Induced Liver Injury for Small Molecules. Chem Res Toxicol. 2023;36:1129-1139.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 19]  [Reference Citation Analysis (0)]
66.  Li T, Tong W, Roberts R, Liu Z, Thakkar S. DeepDILI: Deep Learning-Powered Drug-Induced Liver Injury Prediction Using Model-Level Representation. Chem Res Toxicol. 2021;34:550-565.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 13]  [Cited by in RCA: 52]  [Article Influence: 10.4]  [Reference Citation Analysis (0)]
67.  Li T, Tong W, Roberts R, Liu Z, Thakkar S. Deep Learning on High-Throughput Transcriptomics to Predict Drug-Induced Liver Injury. Front Bioeng Biotechnol. 2020;8:562677.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 7]  [Cited by in RCA: 29]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
68.  Xiao Y, Chen Y, Huang R, Jiang F, Zhou J, Yang T. Interpretable machine learning in predicting drug-induced liver injury among tuberculosis patients: model development and validation study. BMC Med Res Methodol. 2024;24:92.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 8]  [Reference Citation Analysis (0)]
69.  Lee S, Yoo S. InterDILI: interpretable prediction of drug-induced liver injury through permutation feature importance and attention mechanism. J Cheminform. 2024;16:1.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 16]  [Reference Citation Analysis (0)]
70.  Shin H, Hur MH, Song BG, Park SY, Kim GA, Choi G, Nam JY, Kim MA, Park Y, Ko Y, Park J, Lee HA, Chung SW, Choi NR, Park MK, Lee YB, Sinn DH, Kim SU, Kim HY, Kim JM, Park SJ, Lee HC, Lee DH, Chung JW, Kim YJ, Yoon JH, Lee JH. AI model using CT-based imaging biomarkers to predict hepatocellular carcinoma in patients with chronic hepatitis B. J Hepatol. 2025;82:1080-1088.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 2]  [Cited by in RCA: 9]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
71.  Kawka M, Dawidziuk A, Jiao LR, Gall TMH. Artificial intelligence in the detection, characterisation and prediction of hepatocellular carcinoma: a narrative review. Transl Gastroenterol Hepatol. 2022;7:41.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 1]  [Cited by in RCA: 18]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
72.  Mittermaier M, Raza MM, Kvedar JC. Bias in AI-based models for medical applications: challenges and mitigation strategies. NPJ Digit Med. 2023;6:113.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 144]  [Reference Citation Analysis (0)]
73.  Abràmoff MD, Tarver ME, Loyo-Berrios N, Trujillo S, Char D, Obermeyer Z, Eydelman MB; Foundational Principles of Ophthalmic Imaging and Algorithmic Interpretation Working Group of the Collaborative Community for Ophthalmic Imaging Foundation, Washington, D. C, Maisel WH. Considerations for addressing bias in artificial intelligence for health equity. NPJ Digit Med. 2023;6:170.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 1]  [Cited by in RCA: 133]  [Article Influence: 66.5]  [Reference Citation Analysis (0)]
74.  Collins GS, Moons KGM, Dhiman P, Riley RD, Beam AL, Van Calster B, Ghassemi M, Liu X, Reitsma JB, van Smeden M, Boulesteix AL, Camaradou JC, Celi LA, Denaxas S, Denniston AK, Glocker B, Golub RM, Harvey H, Heinze G, Hoffman MM, Kengne AP, Lam E, Lee N, Loder EW, Maier-Hein L, Mateen BA, McCradden MD, Oakden-Rayner L, Ordish J, Parnell R, Rose S, Singh K, Wynants L, Logullo P. TRIPOD+AI statement: updated guidance for reporting clinical prediction models that use regression or machine learning methods. BMJ. 2024;385:e078378.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 114]  [Cited by in RCA: 686]  [Article Influence: 686.0]  [Reference Citation Analysis (0)]
75.  Ludwig DR, Fraum TJ, Ballard DH, Narra VR, Shetty AS. Imaging Biomarkers of Hepatic Fibrosis: Reliability and Accuracy of Hepatic Periportal Space Widening and Other Morphologic Features on MRI. AJR Am J Roentgenol. 2021;216:1229-1239.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 4]  [Cited by in RCA: 10]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
76.  Vasey B, Nagendran M, Campbell B, Clifton DA, Collins GS, Denaxas S, Denniston AK, Faes L, Geerts B, Ibrahim M, Liu X, Mateen BA, Mathur P, McCradden MD, Morgan L, Ordish J, Rogers C, Saria S, Ting DSW, Watkinson P, Weber W, Wheatstone P, McCulloch P; DECIDE-AI expert group. Reporting guideline for the early-stage clinical evaluation of decision support systems driven by artificial intelligence: DECIDE-AI. Nat Med. 2022;28:924-933.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 243]  [Cited by in RCA: 238]  [Article Influence: 79.3]  [Reference Citation Analysis (0)]
77.  United States Food and Drug Administration  Artificial Intelligence/Machine Learning (AI/ML)-Based Software as a Medical Device (SaMD) Action Plan. 2021. [cited 24 April 2025]. Available from: https://www.fda.gov/media/145022/download.  [PubMed]  [DOI]
78.  Amann J, Blasimme A, Vayena E, Frey D, Madai VI; Precise4Q consortium. Explainability for artificial intelligence in healthcare: a multidisciplinary perspective. BMC Med Inform Decis Mak. 2020;20:310.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 496]  [Cited by in RCA: 602]  [Article Influence: 120.4]  [Reference Citation Analysis (0)]
79.  Wiens J, Saria S, Sendak M, Ghassemi M, Liu VX, Doshi-Velez F, Jung K, Heller K, Kale D, Saeed M, Ossorio PN, Thadaney-Israni S, Goldenberg A. Do no harm: a roadmap for responsible machine learning for health care. Nat Med. 2019;25:1337-1340.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 245]  [Cited by in RCA: 416]  [Article Influence: 69.3]  [Reference Citation Analysis (0)]
80.  Parikh RB, Teeple S, Navathe AS. Addressing Bias in Artificial Intelligence in Health Care. JAMA. 2019;322:2377-2378.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 197]  [Cited by in RCA: 339]  [Article Influence: 56.5]  [Reference Citation Analysis (0)]
81.  Parra NS, Ross HM, Khan A, Wu M, Goldberg R, Shah L, Mukhtar S, Beiriger J, Gerber A, Halegoua-DeMarzio D. Advancements in the Diagnosis of Hepatocellular Carcinoma. Int J Transl Med. 2023;3:51-65.  [PubMed]  [DOI]  [Full Text]
82.  Calderaro J, Seraphin TP, Luedde T, Simon TG. Artificial intelligence for the prevention and clinical management of hepatocellular carcinoma. J Hepatol. 2022;76:1348-1361.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 19]  [Cited by in RCA: 168]  [Article Influence: 56.0]  [Reference Citation Analysis (0)]
83.  Ghosh S, Zhao X, Alim M, Brudno M, Bhat M. Artificial intelligence applied to 'omics data in liver disease: towards a personalised approach for diagnosis, prognosis and treatment. Gut. 2025;74:295-311.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 10]  [Cited by in RCA: 30]  [Article Influence: 30.0]  [Reference Citation Analysis (0)]
84.  Ginter-Matuszewska B, Adamek A, Majchrzak M, Rozplochowski B, Zientarska A, Kowala-Piaskowska A, Lukasiak P. FibrAIm - The machine learning approach to identify the early stage of liver fibrosis and steatosis. Int J Med Inform. 2025;197:105837.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1]  [Reference Citation Analysis (0)]
85.  Obermeyer Z, Powers B, Vogeli C, Mullainathan S. Dissecting racial bias in an algorithm used to manage the health of populations. Science. 2019;366:447-453.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1119]  [Cited by in RCA: 2110]  [Article Influence: 422.0]  [Reference Citation Analysis (0)]
86.  Nam D, Chapiro J, Paradis V, Seraphin TP, Kather JN. Artificial intelligence in liver diseases: Improving diagnostics, prognostics and response prediction. JHEP Rep. 2022;4:100443.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 10]  [Cited by in RCA: 99]  [Article Influence: 33.0]  [Reference Citation Analysis (0)]
87.  Straw I, Wu H. Investigating for bias in healthcare algorithms: a sex-stratified analysis of supervised machine learning models in liver disease prediction. BMJ Health Care Inform. 2022;29:e100457.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 18]  [Cited by in RCA: 29]  [Article Influence: 9.7]  [Reference Citation Analysis (0)]
88.  Lin B, Ma Y, Wu S. Multi-Omics and Artificial Intelligence-Guided Data Integration in Chronic Liver Disease: Prospects and Challenges for Precision Medicine. OMICS. 2022;26:415-421.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 5]  [Cited by in RCA: 13]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
89.  Sedano R, Solitano V, Vuyyuru SK, Yuan Y, Hanžel J, Ma C, Nardone OM, Jairath V. Artificial intelligence to revolutionize IBD clinical trials: a comprehensive review. Therap Adv Gastroenterol. 2025;18:17562848251321915.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 17]  [Reference Citation Analysis (0)]
90.  Romeo S, Kozlitina J, Xing C, Pertsemlidis A, Cox D, Pennacchio LA, Boerwinkle E, Cohen JC, Hobbs HH. Genetic variation in PNPLA3 confers susceptibility to nonalcoholic fatty liver disease. Nat Genet. 2008;40:1461-1465.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 2701]  [Cited by in RCA: 2654]  [Article Influence: 156.1]  [Reference Citation Analysis (0)]
91.  Govaere O, Cockell S, Tiniakos D, Queen R, Younes R, Vacca M, Alexander L, Ravaioli F, Palmer J, Petta S, Boursier J, Rosso C, Johnson K, Wonders K, Day CP, Ekstedt M, Orešič M, Darlay R, Cordell HJ, Marra F, Vidal-Puig A, Bedossa P, Schattenberg JM, Clément K, Allison M, Bugianesi E, Ratziu V, Daly AK, Anstee QM. Transcriptomic profiling across the nonalcoholic fatty liver disease spectrum reveals gene signatures for steatohepatitis and fibrosis. Sci Transl Med. 2020;12:eaba4448.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 192]  [Cited by in RCA: 292]  [Article Influence: 58.4]  [Reference Citation Analysis (0)]
92.  McGlinchey AJ, Govaere O, Geng D, Ratziu V, Allison M, Bousier J, Petta S, de Oliviera C, Bugianesi E, Schattenberg JM, Daly AK, Hyötyläinen T, Anstee QM, Orešič M. Metabolic signatures across the full spectrum of non-alcoholic fatty liver disease. JHEP Rep. 2022;4:100477.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 1]  [Cited by in RCA: 59]  [Article Influence: 19.7]  [Reference Citation Analysis (0)]
93.  Sanyal AJ, Newsome PN, Kliers I, Østergaard LH, Long MT, Kjær MS, Cali AMG, Bugianesi E, Rinella ME, Roden M, Ratziu V; ESSENCE Study Group. Phase 3 Trial of Semaglutide in Metabolic Dysfunction-Associated Steatohepatitis. N Engl J Med. 2025;392:2089-2099.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 81]  [Cited by in RCA: 174]  [Article Influence: 174.0]  [Reference Citation Analysis (0)]
94.  Kawaguchi-Suzuki M, Cusi K, Bril F, Gong Y, Langaee T, Frye RF. A Genetic Score Associates With Pioglitazone Response in Patients With Non-alcoholic Steatohepatitis. Front Pharmacol. 2018;9:752.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 15]  [Cited by in RCA: 27]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
95.  Wu Y, Xie L. AI-driven multi-omics integration for multi-scale predictive modeling of genotype-environment-phenotype relationships. Comput Struct Biotechnol J. 2025;27:265-277.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 17]  [Cited by in RCA: 35]  [Article Influence: 35.0]  [Reference Citation Analysis (0)]
96.  Bernecker T, Peters A, Schlett CL, Bamberg F, Fabian Theis, Rueckert D, Weiß J, Albarqouni S.   FedNorm: Modality-Based Normalization in Federated Learning for Multi-Modal Liver Segmentation. 2022 Preprint. Available from: arXiv:2205.11096.  [PubMed]  [DOI]  [Full Text]
97.  Rieke N, Hancox J, Li W, Milletarì F, Roth HR, Albarqouni S, Bakas S, Galtier MN, Landman BA, Maier-Hein K, Ourselin S, Sheller M, Summers RM, Trask A, Xu D, Baust M, Cardoso MJ. The future of digital health with federated learning. NPJ Digit Med. 2020;3:119.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 608]  [Cited by in RCA: 766]  [Article Influence: 153.2]  [Reference Citation Analysis (0)]
98.  Lusnig L, Sagingalieva A, Surmach M, Protasevich T, Michiu O, McLoughlin J, Mansell C, De' Petris G, Bonazza D, Zanconati F, Melnikov A, Cavalli F. Hybrid Quantum Image Classification and Federated Learning for Hepatic Steatosis Diagnosis. Diagnostics (Basel). 2024;14 558 [PMID:38473030 DOI: 10.3390/diagnostics14050558.  [PubMed]  [DOI]
99.  Xu J, Glicksberg BS, Su C, Walker P, Bian J, Wang F. Federated Learning for Healthcare Informatics. J Healthc Inform Res. 2021;5:1-19.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 151]  [Cited by in RCA: 275]  [Article Influence: 55.0]  [Reference Citation Analysis (0)]
100.  Lv Y, Ding H, Wu H, Zhao Y, Zhang L. FedRDS: Federated Learning on Non-IID Data via Regularization and Data Sharing. Appl Sci. 2023;13:12962.  [PubMed]  [DOI]  [Full Text]
101.  Toussaint PA, Leiser F, Thiebes S, Schlesner M, Brors B, Sunyaev A. Explainable artificial intelligence for omics data: a systematic mapping study. Brief Bioinform. 2023;25:bbad453.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 4]  [Cited by in RCA: 14]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
102.  Giuste F, Shi W, Zhu Y, Naren T, Isgut M, Sha Y, Tong L, Gupte M, Wang MD. Explainable Artificial Intelligence Methods in Combating Pandemics: A Systematic Review. IEEE Rev Biomed Eng. 2023;16:5-21.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1]  [Cited by in RCA: 37]  [Article Influence: 12.3]  [Reference Citation Analysis (0)]
103.  Wan L, Liu R, Sun L, Nie H, Wang X. UAV swarm based radar signal sorting via multi-source data fusion: A deep transfer learning framework. Inf Fusion. 2022;78:90-101.  [PubMed]  [DOI]  [Full Text]
104.  Linardatos P, Papastefanopoulos V, Kotsiantis S. Explainable AI: A Review of Machine Learning Interpretability Methods. Entropy (Basel). 2020;23:18.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 253]  [Cited by in RCA: 623]  [Article Influence: 124.6]  [Reference Citation Analysis (0)]
105.  Bai K, Yang L, Xue J, Zhao L, Hao F. Pathogenicity classification of missense mutations based on deep generative model. Comput Biol Med. 2024;170:107980.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Reference Citation Analysis (0)]